18272
18272
18272
DETAILS
CONTRIBUTORS
GET THIS BOOK Gillian J. Buckley and Lawrence O. Gostin, Editors; Committee on Understanding
the Global Public Health Implications of Substandard, Falsified, and
Counterfeit Medical Products; Board on Global Health; Institute of Medicine
FIND RELATED TITLES
SUGGESTED CITATION
Visit the National Academies Press at NAP.edu and login or register to get:
Distribution, posting, or copying of this PDF is strictly prohibited without written permission of the National Academies Press.
(Request Permission) Unless otherwise indicated, all materials in this PDF are copyrighted by the National Academy of Sciences.
THE NATIONAL ACADEMIES PRESS 500 Fifth Street, NW Washington, DC 20001
NOTICE: The project that is the subject of this report was approved by the Govern-
ing Board of the National Research Council, whose members are drawn from the
councils of the National Academy of Sciences, the National Academy of Engineer-
ing, and the Institute of Medicine. The members of the committee responsible for
the report were chosen for their special competences and with regard for appropri-
ate balance.
This study was supported by Contract No. HHSF22301024T, TO #25 between the
National Academy of Sciences and the U.S. Food and Drug Administration. Any
opinions, findings, conclusions, or recommendations expressed in this publication
are those of the author(s) and do not necessarily reflect the view of the organizations
or agencies that provided support for this project.
Additional copies of this report are available from the National Academies Press,
500 Fifth Street, NW, Keck 360, Washington, DC 20001; (800) 624-6242 or (202)
334-3313; http://www.nap.edu.
For more information about the Institute of Medicine, visit the IOM home page
at: www.iom.edu.
The serpent has been a symbol of long life, healing, and knowledge among almost
all cultures and religions since the beginning of recorded history. The serpent ad-
opted as a logotype by the Institute of Medicine is a relief carving from ancient
Greece, now held by the Staatliche Museen in Berlin.
Cover image from 1900 calendar produced by the Antikamnia (“Opposed to Pain”)
Chemical Company of St. Louis, Missouri (http://unitproj.library.ucla.edu/biomed/
his/antikamnia/antikamnia.html).
The National Academy of Engineering was established in 1964, under the charter
of the National Academy of Sciences, as a parallel organization of outstanding engi
neers. It is autonomous in its administration and in the selection of its members,
sharing with the National Academy of Sciences the responsibility for advising the
federal government. The National Academy of Engineering also sponsors engineer-
ing programs aimed at meeting national needs, encourages education and research,
and recognizes the superior achievements of engineers. Dr. Charles M. Vest is presi-
dent of the National Academy of Engineering.
The National Research Council was organized by the National Academy of Sci-
ences in 1916 to associate the broad community of science and technology with the
Academy’s purposes of furthering knowledge and advising the federal government.
Functioning in accordance with general policies determined by the Academy, the
Council has become the principal operating agency of both the National Academy
of Sciences and the National Academy of Engineering in providing services to
the government, the public, and the scientific and engineering communities. The
Council is administered jointly by both Academies and the Institute of Medicine.
Dr. Ralph J. Cicerone and Dr. Charles M. Vest are chair and vice chair, respectively,
of the National Research Council.
www.national-academies.org
IOM Staff
GILLIAN BUCKLEY, Program Officer, Study Director
KENISHA PETERS, Research Associate
MEGAN GINIVAN, Research Assistant
KATHLEEN BURNS, Intern
JULIE WILTSHIRE, Financial Associate
PATRICK W. KELLEY, Director, Boards on Global Health and African
Science Academy Development
Reviewers
vii
viii REVIEWERS
Acknowledgments
ix
x ACKNOWLEDGMENTS
ACKNOWLEDGMENTS xi
xiii
Contents
ACKNOWLEDGMENTS ix
SUMMARY 1
1 INTRODUCTION 15
Background and Terms, 17
Intellectual Property and Public Health, 18
TRIPS and the Doha Declaration, 18
Patent and Trademark Infringement, 20
Competing Meanings of the Term Counterfeit, 22
Substandard and Falsified Drugs, 24
The Problem of Unregistered Medicines, 26
A Proposed Vocabulary, 28
Drug Quality Standards, 30
Pharmacopeia, 30
Registration Agencies and National Pharmaceutical
Authorities, 31
References, 48
xvii
xviii CONTENTS
CONTENTS xix
xx CONTENTS
Spectroscopy, 265
Mass Spectrometry, 271
Emerging Technologies, 273
Using Technology, 274
Combining Techniques, 277
Using Technology in Developing Countries, 279
Field Technologies, 279
Detection in Every Setting, 281
The Technological Landscape, 284
References, 290
APPENDIXES
A GLOSSARY 309
B COMMITTEE BIOGRAPHIES 331
C MEETING AGENDAS 339
BOXES
1-1 Statement of Task, 18
1-2 Definitions of Terms, 29
xxi
FIGURES
1-1 A two-dimensional description of medicine quality and
registration, 27
TABLES
1-1 Definitions of Counterfeit Pharmaceuticals, 34
1-2 National Definitions of Counterfeit Pharmaceuticals, 37
1-3 Definitions of Substandard Pharmaceuticals, 42
1-4 National Definitions of Substandard Pharmaceuticals, 44
1-5 Other Terms of Interest, 45
2-1 Medicines Use to Treat the Most Common Causes of Child Death
Are Compromised in Developing Countries, 75
Summary
1 The terms medicine, drug, and pharmaceutical are used interchangeably in this report in
accordance with the definitions listed in the American Heritage Stedman’s Medical Dictionary.
2 Illegitimate, as explained later in the report, is a parent category for falsified and substan-
dard medicines.
dealt with by courts, case by case, the report does not discuss the problem
of counterfeit medicines.
The trade in illegitimate drugs is, however, a problem of public health
consequence and the topic of this report. In order to discuss this problem
more precisely, the report distinguishes two main categories of poor-quality
drugs. First, there are substandard drugs, those that do not meet the specifi-
cations given in the accepted pharmacopeia or in the manufacturer’s dossier.
The other main category of illegitimate products is falsified drugs, those
that carry a false representation of identity or source or both. Many coun-
tries also have problems with unregistered medicines, those not granted
market authorization in a country. Unregistered drugs may be of good
quality, though some research indicates they often are not. Unregistered
medicines usually circulate outside the controlled distribution chain and
are therefore suspect.
The drug failures of public health concern can be divided into two main
categories: falsified and substandard. Admittedly, the distinction between
the two categories is not always clear. Falsified drugs are usually also sub-
standard; national specifications referenced in the definition of a substan-
dard drug can vary.3 However, these terms cover the two main divisions of
interest with sufficient precision. International endorsement of these two
categories could advance public discourse on the topic.
The spirit of these definitions and the exclusion of the term counterfeit
are central to this recommendation. The exact wording suggested is not.
and was removed from the recommendation after the report release. The supporting text
describes the committee’s understanding of a substandard drug.
SUMMARY 3
5 As the report explains later, fake is a commonly used synonym for falsified.
SUMMARY 5
turing practices to sell in these markets. United Nations agencies and larger
international aid organizations will also refuse to do business with com-
panies that cannot meet stringent regulatory authority quality standards.
Manufacturers are aware, however, that low- and middle-income countries
are less likely to enforce these standards. When a manufacturer produces
medicines of inferior quality for less exacting markets it is known as tiered
or parallel production.
When regulatory checks on production are inconsistent, good procure-
ment practices can ensure that quality medicines get the largest market
share. The firms that offer the cheapest prices do so by buying impure
ingredients and cutting corners in formulation. Good procurement dictates
that the cheapest tenders are not accepted if they are of dubious quality,
but it is difficult not to be swayed by price. Proper precaution in medicines
procurement can prevent poor-quality products from infiltrating the mar-
ket. Good procurement puts a strong emphasis on controlling corruption
and promoting transparency. The WHO’s Model Quality Assurance System
for procurement agencies lays out the steps necessary for efficient and open
procurement of the best-quality medicines possible.
SUMMARY 7
SUMMARY 9
secondary wholesale market to vetted firms would improve the U.S. drug
supply. The National Association of Boards of Pharmacy (NABP) whole-
saler accreditation process requires criminal background checks on senior
staff and proof of professional standards in record keeping and drug stor-
age and handling. Some states require NABP accreditation of wholesalers,
but unscrupulous businesses can seek out states with lower standards for
their headquarters. And, because the wholesale trade is national, weak-
nesses in one state’s system can become vulnerabilities in another.
SUMMARY 11
The private sector will invest in medicines retail if there is a good busi-
ness reason to do so. Governments can take steps that would encourage
private sector investment and create an environment where responsible
private drug sellers will thrive. Governments can provide low-interest loans
for improving drug shops and encourage private-sector accreditation or
franchising programs. They can also work with their national pharmacy
councils to set out tiers of training, including vocational training, for phar-
maceutical personnel. Governments can also give incentives to keep trained
staff in underserved areas.
Disorganized medicines retail is not confined to developing countries.
Through the internet, unlicensed drug vendors sell around the world, mostly
in middle- and high-income countries. Unlicensed internet pharmacies are
similar to street drug bazaars, both in the quality of the products they stock,
which is poor, and in the lack of official oversight of their operations.
In the United States the NABP runs the Verified Internet Pharmacy
Practice Sites (VIPPS) accreditation program to recognize safe online drug
stores. Accredited online pharmacies comply with state licensing require-
ments for both the state that the pharmacy is in and all the states in which
it sells. Chief among these requirements are the authentication of prescrip-
tions, observance of quality-assurance standards, and submission to regular
state inspection. Accredited pharmacies display the VIPPS seal, and, because
this seal could be copied, the project website lists both certified pharmacies
and known fraudulent ones.
DETECTION TECHNOLOGY
The main categories of techniques for pharmaceutical analysis can
be broken down as visual inspection of product and packaging; tests for
physical properties such as reflectance and refractive index; chemical tests
including colorimetry, disintegration, and dissolution; chromatography;
spectroscopic techniques; and mass spectrometry. Within each of these
categories, some technologies are appropriate for field use, while others
require sophisticated lab equipment and a high level of technical expertise.
Understanding when, where, and why to use the various techniques
can be difficult. The information a technique provides, as well as its reli-
ability, cost, speed, and portability, make it more or less appropriate in any
given situation. While any one test may suffice to label a drug substandard
or falsified, no single analytical technique provides enough information
SUMMARY 13
to confirm that a drug is genuine. One challenge in both field and labora-
tory testing is determining how to combine tests for maximum efficiency.
It is usually best to work through tests beginning with the easiest or least
expensive ones. Only if samples pass these tests should the inspector move
on to more difficult or expensive ones.
Making detection technology more accessible in low- and middle-
income countries would be invaluable to controlling the trade in falsified
and substandard drugs. Technologies can protect consumers and are useful
to surveillance staff working to generate accurate estimates of the magni-
tude of the problem of poor-quality drugs. An understanding of the tech-
nological landscape, the range and gaps in available technologies, and the
likely improvements in the near future is essential for using technologies in
developing countries.
CODE OF PRACTICE
Individual countries have the responsibility for protecting the national
drug supply. This includes regulating good-quality manufacturers, prevent-
ing poor-quality drugs from entering the market, detecting them when they
do, and punishing those who manufacture and trade them. Drug regulation,
surveillance, and law enforcement are the necessary components of any
national response to the problem.
A voluntary soft law such as an international code of practice could
encourage international action against falsified and substandard drugs. The
code of practice would contain guidelines on surveillance and international
reporting of drug quality problems. The code would facilitate passage of
national laws on how to punish and, when necessary, extradite those re-
sponsible for falsified drugs and criminally negligent manufacture. It would
also promote harmonized regulatory standards for drug manufacture and
licensing.
Introduction
In the 1949 film The Third Man and the novel of the same name, Holly
Martin learns that his childhood friend Harry Lime has made a fortune
diluting stolen penicillin and selling it on the black market. In a dramatic
confrontation on the Vienna Ferris wheel, Martin refers to Lime’s earlier
racketeering, asking, “Couldn’t you have stuck to tires?” No, explains
Lime, one of the American Film Institute’s 100 greatest villains, “I’ve al-
ways been ambitious” (AFI, 2003).
The theft, adulteration, careless manufacture, and fraudulent label-
ing of medicines1 continue to attract villains who, like Harry Lime, grow
wealthy off their business. Although the problem is most widespread in
poor countries with weak regulatory oversight, it is no longer confined to
underground economies as in postwar Vienna. As of January 2013, gross
manufacturing negligence at a compounding pharmacy in Massachusetts
had sickened 693 Americans and killed 45 (CDC, 2013). Less than a year
earlier, 76 doctors in the United States unknowingly treated cancer patients
with a fake version of the drug Avastin (Weaver and Whalen, 2012).
International trade and manufacturing systems obscure connections
between the crime and the criminal; in modern supply chains, medicines
may change hands many times in many countries before reaching a patient.
To complicate the problem, medicines are mostly for sick people. The ef-
fects of inactive, even toxic, drugs can go unnoticed or be mistaken for the
1 The terms medicine, drug, and pharmaceutical are used interchangeably in this report in
accordance with the definitions listed in the American Heritage Stedman’s Medical Dictionary
(2012a,b,c).
15
natural course of the underlying disease. This is most true in parts of the
world with weak pharmacovigilance systems, poor clinical record keeping,
and high all-cause mortality, where “friends or relatives of those who die
are obviously saddened, but not necessarily shocked” (Bate, 2010).
Deaths from fake drugs go largely uncounted, to say nothing of the
excess morbidity and the time and money wasted by using them. The manu-
facture and trade in fake pharmaceuticals is illegal and hence almost impos-
sible to measure precisely. Even crude copies can blend in with legitimate
products in the market. The camouflage succeeds because drug quality is
not something consumers can accurately judge. This imbalance, also called
information asymmetry, makes the medicines trade vulnerable to market
failure (Mackintosh et al., 2011). In short, “At every step of the supply
chain there is this unequal knowledge, and people are exploited because of
[it]” (Mackintosh et al., 2011, p. 2).
Market controls and oversight aim to correct the information imbal-
ance in the medicines market, but supervising sprawling multinational dis-
tribution chains is a “regulatory nightmare” (Economist, 2012). National
drugs regulatory agencies (hereafter, regulatory agencies) are responsible
for assuring drug quality in their countries, a job that increasingly requires
cooperation with their counterpart agencies around the world (IOM, 2012).
The World Health Organization (WHO) has worked to facilitate this co-
operation since 1985, but advancing the public discourse on this topic has
proven more difficult than anyone would have predicted then (Clift, 2010).
To start, different countries and international stakeholders cannot agree
on how to define the problem. When it is framed as one of counterfeit
and legitimate drugs, many civil society groups and emerging manufactur-
ing nations see a thinly veiled excuse to persecute generic drug industries
(Clift, 2010; Economist, 2012). Large innovator pharmaceutical companies
have the most experience in finding and prosecuting pharmaceutical crime.
This expertise brought them a place in the WHO’s International Medical
Products Anti-Counterfeiting Taskforce (IMPACT), the largest interna-
tional working group on drug safety to date. Involving these companies
with a WHO program, however, raised suspicions of civil society groups
(TWN, 2010). Objections to the taskforce’s inception and confusion about
its mandate from WHO governing bodies further eroded support (TWN,
2010). The WHO distanced itself from IMPACT after 2010; the taskforce’s
secretariat moved to the Italian drugs regulatory authority (Seear, 2012;
Taylor, 2012).
IMPACT may no longer be active, but criminals and unscrupulous drug
manufacturers are. The Economist recently described the 21st century as “a
golden age for bad drugs” (Economist, 2012). There is an urgent need for
international public discourse on the problem. In an effort to advance this
discourse, the U.S. Food and Drug Administration (FDA) commissioned
INTRODUCTION 17
BOX 1-1
Statement of Task
INTRODUCTION 19
INTRODUCTION 21
2 Anti-Counterfeiting
Trade Agreement (ACTA). October 1, 2011.
3 TRIPS:
Agreement on Trade-Related Aspects of Intellectual Property Rights, Apr. 15,
1994, Marrakesh Agreement Establishing the World Trade Organization, Annex 1C, THE
LEGAL TEXTS: THE RESULTS OF THE URUGUAY ROUND OF MULTILATERAL TRADE
NEGOTIATIONS 320 (1999), 1869 U.N.T.S. 299, 33 I.L.M. 1197 (1994).
INTRODUCTION 23
4 For amodiaquine hydrochloride tablets, the acceptable drug concentration range under
U.S. Pharmacopeia is 93 percent to 107 percent of labeled amount (USP, 2011a); under Inter-
national Pharmacopoeia it is 90 percent to 110 percent (WHO, 2011c). For quinine sulfate
tablets, the acceptable drug concentration range under U.S. Pharmacopeia is 90 percent to 110
percent of the labeled amount (USP, 2011b); under British Pharmacopoeia, it is 95 percent to
105 percent (British Pharmacopoeia, 2012c).
INTRODUCTION 25
quality drug is not in fact its own. In such a case, the manufacturer is the
victim of fraud. The drug in question was falsified and therefore in the
domain of law enforcement.
The committee considers a drug falsified when there is false represen-
tation of the product’s identity or source or both. Falsified medicines may
contain the wrong ingredients in the wrong doses. A fake product in legiti-
mate packaging is falsified, as is a good-quality product in fake packaging
(EMA, 2012). The producer’s intention is theoretically important to the
understanding of a falsified drug, though in practice it is often impossible to
known what these intentions were. That is, when a licensed manufacturer
makes bad drugs, the deliberateness of the mistake is at least debatable.
When an underground producer makes a bad-quality product there is not
even a pretense of adhering to drug quality standards. This understanding
of a falsified medicine is consistent with the broad definition of counterfeit
used by WHO and other organizations. A falsified drug may also be called
fake, a synonym used in this report and by some scholars, governments,
and international NGOs (Bate, 2011; Björkman-Nyqvist et al., 2012; MSF,
2012; Newton et al., 2011). (See Table 1-5.)
Often, the difference between a substandard and a falsified medicine is
the difference between a known and unknown manufacturer. Manufactur-
ers may produce substandard drugs because they failed to adhere to good
manufacturing practices or because their internal quality systems failed.
Degraded or expired products are also substandard; in some ways, failure
to pull these drugs from the market is a quality system failure. Inspection
of the manufacturer’s records can usually distinguish between a degraded or
expired drug and one that left the factory already outside of specifications.
Falsified drugs are usually also substandard. Drug regulators have no
authority over underground manufacturers; nothing can be said about
The Indian generics house V.S. International’s authentic ciprofloxacin (left) and a
falsified version (right).
SOURCE: Bate, 2012b.
INTRODUCTION 27
Legal
Illegal – Negligent
Illegal – Willful
Fails standard
(bad treatment)
Illegitimate drugs
Manufacturing error
(intentional wrong)
Degradation
Expiration
Legitimate drugs
Meets standard
(good treatment)
A field survey of uterotonic drug quality in Ghana found that all unreg-
istered drug samples tested were substandard; one of the unregistered
products contained no active ingredient at all (Stanton et al., 2012). Many
of the samples might have degraded during disorganized transport, but the
explanation is never clear with unregistered drugs.
Unregistered medicines are vulnerable to quality failures. They do not
enter the market through reputable channels and are often transported un-
der poor conditions. These problems can easily go undetected. Postmarket
surveillance is, by definition, a way to monitor the safety of those drugs
authorized for a particular market. Therefore, the quality failures of un-
registered medicines resist detection in postmarket surveillance (Amin and
Snow, 2005). The proliferation of unregistered medicines suggests problems
with the market authorization process in a country and, more generally,
with regulatory oversight. Although unregistered drugs are not by defini-
tion falsified or substandard, they are conceptually related and part of the
problem.
A Proposed Vocabulary
The lack of a consistent vocabulary has held back public discourse on
the problem of poor quality medicines in the market. As Tables 1-1 through
1-5 indicate, different countries often have widely different interpretations
of the same terms, creating a confusion that holds back international co-
operation (Clift, 2010). Defining a common vocabulary is important, not
just for this report but for all discourse on the topic.
Box 1-2 presents the definitions of the terms falsified, substandard,
counterfeit, and unregistered used in this report. As this chapter explains,
distinguishing between substandard and falsified medicines in the field can
be difficult. In practice, there is often considerable ambiguity in real-life
examples of unlabeled, poor-quality drugs. Nevertheless, falsified and sub-
standard are good categories to describe problems with poor-quality drugs.
Consistent use of these terms would ease the measuring of trends, analysis
of causes, and discussion of proposed solutions to the problem.
5 An emphasis on quality system failures is not essential to the idea of a substandard drug
and was removed from the recommendation after the report release. The supporting text
describes the committee’s understanding of a substandard drug.
INTRODUCTION 29
BOX 1-2
Definitions of Terms
The committee agrees with the emerging consensus that falsified and
substandard are the two main categories of poor-quality drugs (Bate,
2012a; Clift, 2010; MSF, 2012; Newton, 2012; Oxfam International,
2011). The World Health Assembly (WHA) is the decision-making body
of the WHO (WHO, 2013) and the international authority on questions
of health policy. WHA endorsement of these two main categories would
advance public discourse on the topic. The spirit of the definitions, not
the exact wording suggested in Box 1-2, are key to this recommendation,
as is the exclusion of the term counterfeit. Counterfeit is an overly broad
term and should be used only to describe trademark infringement, which
is not a problem of primary concern to public health organizations. As
WHO Director-General Margaret Chan explained in the opening remarks
of the November 2012 member state meeting on illegitimate drugs, “trade
and intellectual property considerations are explicitly excluded” from the
WHO’s discussions (Chan, 2012).
Falsified and substandard products are two useful categories in think-
ing about drug quality problems. There is overlap between these catego-
ries, but they are sufficiently precise for public discussion. Similarly, the
problem of unregistered medicines is intimately linked to problems of drug
quality.
Pharmacopeia
National governments have long created officially recognized lists of le-
gal drugs. Starting in the 16th and 17th centuries, city-based pharmacopeia
attempted to standardize the apothecaries’ products (Brockbank, 1964).
Modern pharmacopeias have been published since the 19th century: the
U.S. Pharmacopeia in 1820 and the British Pharmacopoeia in 1864 (British
Pharmacopoeia, 2012a; USP, 2012).
The strength of regulation by pharmacopeial standards depends on
the regulatory agency to enforce the standards. In the United States, the
Drug Import Act of 1848 made the U.S. Pharmacopeia the national drug
compendium (USP, 2012). This recognition made the drug quality standards
legally binding. In the latter half of the 19th century, state governments
created licensing boards for pharmacists and pharmacies, and these boards
emphasized the importance of the pharmacopeial standards (USP, 2012).
The Pure Food and Drugs Act of 1906 recognized the U.S. Pharmacopeia
standards as official and to be enforced by the Bureau of Chemistry in the
U.S. Department of Agriculture, the forerunner of today’s FDA (Swann,
2009; USP, 2012).
Terra silligata, medicinal clay from the Greek island of Lemnos (left), was
stamped with a seal of authenticity (right), an early example of a drug
trademark.
SOURCE: Wellcome Library, London.
INTRODUCTION 31
The passage of the Food, Drug, and Cosmetic Act in 1938 gave the
FDA new authorities and recognized the quality, packaging, and labeling
standards published in the pharmacopeia and the national formulary (USP,
2012). The act also gave the FDA inspectorate the authority to enforce
these standards (USP, 2012). The 1960s saw several changes to accepted
drug regulation, including the creation of an Adopted Names Council, an
organization that establishes the United States Adopted Names, unique
nonproprietary names for drugs (AMA, 2012; USP, 2012). Eventually the
U.S. Pharmacopeia purchased the National Formulary and Drug Standards
Laboratory from the American Pharmacists’ Association (USP, 2012). They
merged the formulary and pharmacopeia in 1975, creating a collection of
more than 4,000 monographs (USP, 2008).
Around the same time in Europe, the unified economic community was
encouraging the use of regional pharmacopeial standards (EDQM, 2012).
The first official European pharmacopeia was published in 1964 and is
currently in its seventh edition (EDQM, 2012; European Pharmacopoeia,
2012). The European Directorate for the Quality of Medicines maintains
and revises the European Pharmacopoeia and runs chemical and biologi-
cal laboratories devoted to testing pharmaceutical products intended for
the EU market (EDQM, 2012). While the FDA enforces pharmacopeial
standards in the United States, both the national regulatory authorities and
the European Directorate enforce the pharmacopeial standards in Europe
(AVMA, 2012; EDQM, 2012). The European Medicines Agency (EMA),
often described as the counterpart of the FDA, is primarily a medicines
registration (review and approval) agency. The European Directorate has
more responsibility for enforcing quality standards (EDQM, 2012).
In China and India, national pharmacopeial standards and organiza-
tions have developed rapidly in the last two decades. The government of
India began enforcing pharmaceutical standards more systematically after
the Drugs and Cosmetics Act of 1940 (Gothoskar, 1983). Only in 2009,
however, did the Indian Pharmacopoeia Commission became an indepen-
dent agency under the Ministry of Health, separate from the drug regula-
tory authority (Indian Pharmacopoeia Commission, 2011). The Indian
government also maintains a pharmacopeia on ayurvedic medicines, first
published in a single volume in 1978 (Pharmacopoeial Laboratory for In-
dian Medicine, 2011).
cacy reviews. They also conduct inspections and enforce quality control
regulations.
The USDA Bureau of Chemistry was the forerunner of the FDA and
one of the first agencies dedicated to quality enforcement for food and
drugs (FDA, 2010). This agency enforced drug quality and antiadulteration
standards in accordance with the Pure Food and Drugs Act of 1906. In
1927 it became a separate agency in the Department of Agriculture (Swann,
2009). Premarket review for drugs was not part of the drug registration
process in the United States until the Federal Food, Drug, and Cosmetic
Act of 1938, though premarket authorization of vaccines was mandatory
after 1902 (FDA, 2012c).
The development of national registration entered a new phase in the
two decades following World War II. In the 1940s and 1950s, before the
thalidomide crisis of the early 1960s, federal drug regulators in the United
States began to regulate efficacy (Carpenter, 2010; FDA, 2012c). In 1958,
the Netherlands Medicines Act created an advanced administrative drug
registration system and established, but did not yet use, the Medicines
Evaluation Board to regulate market approval of new drugs (Carpenter,
2010; MSH, 2012).
The global thalidomide tragedy in the early 1960s changed all these
institutions. Thalidomide was a sedative and antiemetic developed in Ger-
many, used widely throughout Australia, Europe, and Japan in the late
1950s (Kim and Scialli, 2011). It was effective against morning sickness
and commonly prescribed to pregnant women (Bren, 2001; Kim and Scialli,
2011). By 1961, however, thalidomide was identified as the cause of severe
birth defects in more than 10,000 children. Birth defects included abnor-
mally short limbs, toes sprouting directly from the hips, flipper-like arms,
or no limbs at all; eye and ear defects; and congenital heart disease (Bren,
2001; Kim and Scialli, 2011). The drug was pulled from the market in 1961
and 1962 (Fintel et al., 2009; Kim and Scialli, 2011). Thalidomide had been
licensed in 46 countries, but in the United States, the FDA had refused to
approve its application and the drug never entered the market (Bren, 2001).
After the tragedy, governments worldwide revamped their drug regula-
tion systems. The Drug Amendments of 1962 officially added efficacy to
safety as a basis for FDA regulation and as a necessity for marketing au-
thorization in the United States and imposed clinical trial requirements on
drug development (FDA, 2012b). Australia, Britain, and Germany changed
their systems of drug regulation in 1963 and 1964 (Daermmrich, 2003;
Rägo and Santoso, 2008; TGA, 2003). Following the European Economic
Community resolutions in 1965 and the 1970s, Britain, France, Germany,
and other European nations took further steps to build stronger, more sci-
entific regulatory agencies based in part upon the FDA model (Carpenter,
2010; ECHAMP, 2012). Hence, although thalidomide was not a problem of
INTRODUCTION 33
Organization Definition
WHO 1992 “A counterfeit medicine is one which is deliberately and fraudulently mislabeled with respect to identity and/or
source. Counterfeiting can apply to both branded and generic products and counterfeit products may include
products with correct ingredients, wrong ingredients, without active ingredients, with insufficient quantity of active
ingredient or with fake packaging” (WHO, 1992, p. 1).
2003 “Counterfeit medicines are part of the broader phenomenon of substandard pharmaceuticals. The difference is
that they are deliberately and fraudulently mislabeled with respect to identity and/or source. Counterfeiting can
apply to both branded and generic products and counterfeit medicines may include products with the correct
ingredients but fake packaging, with the wrong ingredients, without active ingredients or with insufficient active
ingredients” (WHO, 2003a).
Countering the Problem of Falsified and Substandard Drugs
2006 “Counterfeit medicines are part of the broader phenomenon of substandard pharmaceuticals. . . . They are
deliberately and fraudulently mislabeled with respect to identity and/or source. Counterfeiting can apply to both
branded and generic products and counterfeit medicines may include products with the correct ingredients but
fake packaging, with the wrong ingredients, without active ingredients or with insufficient active ingredients”
(WHO, 2006).
2009 “A counterfeit medicine is one which is deliberately and fraudulently mislabeled with respect to identity and/or
source. Counterfeiting can apply to both branded and generic products and counterfeit products may include
products with the correct ingredients or with the wrong ingredients, without active ingredients, with insufficient
active ingredients or with fake packaging” (WHO, 2009).
2011 Never explicitly defined, except as part of the so-called spurious, substandard, falsified, falsely labeled, counterfeit
(SFFC). “There are no good quality SSFC medicines. By definition SSFC medicines are products whose true identify
and/or source are unknown or hidden. They are mislabeled . . . and produced by criminals” (WHO, 2011a).
World Trade “Unauthorized representation of a registered trademark carried on goods identical or similar to goods for which the
Organization trademark is registered, with a view to deceiving the purchaser into believing that he/she is buying the original goods”
(WTO, 2012).
World Bank “Counterfeits are usually defined as drugs that are deliberately made as fake copies of the original branded or generic
drugs, imitating design, colors and other visible features. In many cases they contain only filling materials without any active
ingredients. Or they may contain insufficient or an excess of active ingredients, or active drug substances other than the ones
specified in the label” (Siter, 2005).
World Medical “Counterfeit medicines are drugs manufactured below established standards of safety, quality and efficacy and therefore
Association create serious health risks, including death” (WMA, 2012).
continued
TABLE 1–1 Continued
36
Organization Definition
TRIPS “[Counterfeit trademark goods] shall mean any goods, including packaging, bearing without authorization a trademark which
Agreement is identical to the trademark validly registered in respect of such goods, or which cannot be distinguished in its essential
aspects from such a trademark, and which thereby infringes the rights of the owner of the trademark in question under the
law of the country of importation” (WTO, 1994).
Doctors Without “Counterfeit medicines are products that are presented in such a way as to look like a legitimate product although they are
Borders not that product. In legal terms this is called trademark infringement. They are the result of deliberate criminal activity that
has nothing to do with legitimate pharmaceutical producers—be it generic or brand producers” (MSF, 2009).
Pharmaceutical Counterfeit medicines are products deliberately and fraudulently produced and/or mislabeled with respect to identify and/
Security or source to make it appear to be a genuine product. This applies to both branded and generic products. They can have
Institute more or less than the required amount of active pharmaceutical ingredients (APIs) or have the correct amount of API, but
manufactured in unsanitary, unsafe conditions. This definition can also be extended to genuine medicines. Genuine medicines
can be placed in counterfeit packaging to extend its expiry date (PSI-Inc., 2012).
The Partnership “Counterfeit drugs are fake medicines intentionally made by unknown manufacturers who hide their identity. These drugs do
Countering the Problem of Falsified and Substandard Drugs
for Safe not meet established standards of quality. Counterfeit drugs deceive consumers by closely resembling the looks of a genuine
Medicines drug. They are made without approval of the regulator, such as the U.S. Food and Drug Administration (FDA). Counterfeiters
create fake versions of branded, generic and over-the-counter drugs. Counterfeit medicines have been found to be made
missing key ingredients; too strong or too weak; with the wrong active ingredient; with dangerous contaminants; in unsanitary
or unsterile conditions; using unsafe methods; and with improper labels” (PSM, 2012).
International “Counterfeiting in relation to medicinal products means the deliberate and fraudulent mislabeling with respect to the
Pharmaceutical identity, composition and/or source of a finished medicinal product, or ingredient for the preparation of a medicinal product.
Federation Counterfeiting can apply to both branded and generic products and to traditional remedies. Counterfeit products may
include products with the correct ingredients, wrong ingredients, without active ingredients, with insufficient quantity of
active ingredient or with false or misleading packing; they may also contain different, or different quantities of, impurities
both harmless and toxic” (FIP, 2003).
International “Counterfeit medicines threaten the full spectrum of legitimate medicines. They can be falsified versions of patented
Federation of medicines, generic medicines or over-the-counter medicines and exist in all therapeutic areas (even traditional medicine).
Country Definition
United States Food, Drug, and “A drug which, or the container or labeling of which, without authorization, bears the trademark,
Cosmetic Act trade name, or other identifying mark, imprint, or device, or any likeness thereof, of a drug
manufacturer, processor, packer, or distributor other than the person or persons who in fact
manufactured, processed, packed, or distributed such drug and which thereby falsely purports
or is represented to be the product of, or to have been packed or distributed by, such other drug
manufacturer, processor, packer, or distributor.”a
Countering the Problem of Falsified and Substandard Drugs
U.S. Food and Drug “Counterfeit medicine is fake medicine. It may be contaminated or contain the wrong or no active
Administration ingredient. They could have the right active ingredient but at the wrong dose. Counterfeit drugs
Website are illegal and may be harmful to your health” (FDA, 2012a).
United States Code Having a spurious trademark, not genuine or authentic, identical with, or substantially
indistinguishable from the genuine trademark, registered on the principal register in the U.S.
Patent and Trademark Office and in use. The counterfeit mark is likely to cause confusion, to cause
mistakes, or to deceive. [general trademark counterfeit]b
Country Definition
China Drug Administration “A drug is a counterfeit drug in any of the following cases:
Law of the People’s 1. the ingredients in the drug are different from those specified by the national drug standards; or
Republic of China
2. a non-drug substance is simulated as a drug or one drug is simulated as another.
Philippines “Medicinal products with correct ingredients but not in the amounts as provided there under, wrong ingredients, without active
ingredients, with insufficient quantity of active ingredients, which results in the reduction of the drug’s safety, efficacy, quality,
strength, or purity. It is a drug which is deliberately and fraudulently mislabeled with respect to identity and/or source or with
fake packaging and can apply to both branded and generic products. It shall also refer to: 1) the drug itself, or the container or
labeling thereof or any part of such drug, container, or labeling bearing without authorization the trademark, trade name, or
other identification mark or imprint or any likeness to that which is owned or registered in the Bureau of Patent, Trademark, and
Technology transfer in the name of another natural or juridical person; 2) a drug product refilled in containers by unauthorized
persons if the legitimate labels or marks are used; 3) an unregistered imported drug product, except drugs brought in the
country for personal us as confirmed and justified by accompanying medical records; and 4) a drug which contains no amount
of or a different active ingredient, or less than 80% of the active ingredient it purports to possess, as distinguished from an
adulterated drug including reduction or loss of efficacy due to expiration” (Clift, 2010, p. 15).
Pakistan “A drug, the label or outer packaging of which is an imitation of, resembles as to be calculated to deceive, the label or outer
packing of a drug manufacturer” (Clift, 2010, p. 15).
India Mashelkar Report “The term, ‘counterfeit’ that is commonly used worldwide for spurious drugs does not appear in
Drugs and Cosmetic Act but the definition of spurious drug comprehensively covers counterfeit
drugs. According to the Drugs and Cosmetic Act (by the Amendment Act of 1982, section 17-B)
spurious drugs are:
a) manufactured under a name which belongs to another drug; or
b) an intimation of, or a substitute for, another drug or resembles another drug in a manner likely
Countering the Problem of Falsified and Substandard Drugs
to deceive or bear upon it or upon its label or container the name of another drug unless it is
plainly and conspicuously marked so as to reveal its true character and its lack of identity with
such other drug; or
c) labeled or in a container bearing the name of an individual or company purporting to be the
manufacturer of the drug, which individual or company is fictitious or does not exist; or
d) substituted wholly or in part by another drug or substance; or
e) purporting to be the product of a manufacturer of whom it is not truly a product”
(Government of India, 2003).
Country Definition
Indonesia 2000 Per the Republic of Indonesia’s Ministry of Health (MOH) Regulation No. 242/2000, counterfeit
medicine(s) is/are the medicine(s) that are produced by the party/parties who has/have no
authority to produce it based on the government’s act.
There are five kinds of counterfeit medicines:
1. Product containing API with required concentrations; produced, packaged, and labeled as the
original product, but this product is produced by the party without license.
2. The medicine contains API, but the concentration is outside of requirements.
3. Product is made as the original form and package, but no content of API.
4. The product is similar to the original, but content is of different substances/materials.
5. Products that are produced without a permit from the MOH. Per Republic Indonesian MOH
Regulation No. 949/MenKes/SK/VI/2000 imported product/s that are illegal can be grouped
as counterfeit without a permit for circulation issued by the National Agency of Food and Drug
Control.
Countering the Problem of Falsified and Substandard Drugs
2008 Per the republic of Indonesia’s Regulation No. 1010/2008, counterfeit medicines are produced by
the party/ies who has/have no authority to produce the medicines by the government’s act, or
medicines whose identities are imitated by other medicines that already have a circulating permit.
There are three categories of counterfeit medicine:
1. The volume of substance (API) and the trade name is the same as the original medicine, but
produced by the party who has no license to produce it.
2. The trade name is the same as the original medicine, but the volume of substance (API) is
different and produced by the other producer.
3. The trade name is the same as the original, but the content of substance (API) is not medicine
and not clear how the processing produced the drug.
Kenya 2008 Anti- “‘Counterfeiting’ means taking the following actions without the authority of the owner of any
Counterfeit Bill intellectual property right subsisting in Kenya or elsewhere in respect of protected goods—
a) the manufacture, production, packaging, re-packaging, labeling or making, whether in Kenya
or elsewhere, of any goods whereby those protected goods are imitated in such manner and
to such a degree that those other goods are identical or substantially similar copies of the
protected goods;
b) the manufacture, production or making, whether in Kenya or elsewhere, the subject matter of
that intellectual property, or a colorable imitation thereof so that the other goods are calculated
to be confused with or to be taken as being the protected goods of the said owner or any
goods manufactured, produced or made under his license;
c) the manufacturing, producing or making of copies, in Kenya or elsewhere, in violation of an
author’s rights or related rights.”d
Mexico It is considered a falsificado product when it is manufactured, packaged, or sold with reference to an authorization that does
not exist, or uses a permit granted by law to another or imitation of legally manufactured and registered products.e
Europe Council of Europe A false representation as regards identity and/or source (Council of Europe, 2011).
Countering the Problem of Falsified and Substandard Drugs
European Medicines “Counterfeit medicines are medicines that do not comply with intellectual-property rights or that
Agency infringe trademark law” (EMA, 2012).
Vietnam “Counterfeit drugs mean products manufactured in any form of drug with a deceitful intention, and falling into one of the
following cases:
a) they have no pharmaceutical ingredients;
b) they have pharmaceutical ingredients, which are, however, not at registered contents;
c) they have pharmaceutical ingredients different from those listed in their labels;
d) they imitate names and industrial designs of drugs which have been registered for industrial property protection of other
manufacturing establishments.”f
a Federal Food, Drug, and Cosmetic Act. As amended December 19, 2002. Chap. II, Sec. 201, (g)(2).
b 18 U.S.C. § 2320. (f)(1). (2012).
c Drug Administration Law of the People’s Republic of China. (China). 2001. Chap. V, Art. 48.
Organization Definition
World Health 2003 “Substandard medicines are products whose composition and ingredients do not meet the correct scientific
Organization specifications and which are consequently ineffective and often dangerous to the patient. Substandard products
may occur as a result of negligence, human error, insufficient human and financial resources or counterfeiting”
(WHO, 2003a).
2006 “Substandard pharmaceuticals [are] medicines manufactured below established standards of safety, quality and
efficacy” (WHO, 2006).
2009 “Substandard medicines (also called out of specification [OOS] products) are genuine medicines produced
by manufacturers authorized by the [national medicines regulatory authority] which do not meet quality
specifications set for them by national standards” (WHO, 2009).
2010 “Each pharmaceutical product that a manufacturer produces has to comply with quality standards and
specifications at release and throughout the product shelf-life required by the territory of use. Normally, these
standards and specifications are reviewed, assessed and approved by the applicable national medicines regulatory
authority before the product is authorized for marketing. Substandard medicines are pharmaceutical products
Countering the Problem of Falsified and Substandard Drugs
that do not meet their quality standards and specifications” (WHO, 2010).
2011 Substandard medicines are pharmaceutical products that do not meet their quality standards and specifications.
“They arise mostly due to the application of poor manufacturing practices by the producer or when a good
quality medicine is stored and distributed under improper conditions leading to deterioration of the quality of the
product” (WHO, 2011a).
Oxfam “Substandard medicines do not meet the scientific specifications for the product as laid down in the WHO standards. They
may contain the wrong type or concentration of active ingredient, or they may have deteriorated during distribution in the
supply chain and thus become ineffective or dangerous” (Brant and Malpani, 2011).
The Partnership “Substandard drugs are produced by a known manufacturer, but they do not meet the quality standards of the drug
for Safe regulator. In the United States, these high standards are set by the United States Pharmacopeia and the National Formulary.
Medicines There is no intent to fool or defraud the consumer. Substandard medications are a result of manufacturer that do not
follow approved Good Manufacturing Practices, which is regulated by the FDA. Simply stated, these drugs fall below the
established standard—hence the term ‘substandard drugs’” (PSM, 2012).
World Bank “Substandard drugs are manufactured with the intent of making a genuine pharmaceutical product, but the manufacturer
saves costs by not following GMP (Good Manufacturing Practice) or using poor quality raw materials. Another potential
problem relates to inadequate storage or transport conditions, leading to deterioration of the product. The performance of
such medicines is questionable” (Siter, 2005).
International “All substandards are not counterfeits. A medicine which is approved and legally manufactured but does not meet all
Federation of quality criteria is substandard, and may pose a significant health risk, but should not be regarded as counterfeit. However, all
Pharmaceutical counterfeits are, by their nature, at high risk of being substandard” (IFPMA, 2010).
Manufacturers
Countering the Problem of Falsified and Substandard Drugs
and Associations
Country Definition
Cambodia A substandard drug is a registered product, whose specifications are outside of accepted standards as defined by reference
pharmacopoeias (Phana, 2007).
China “A drug with content not up to the national drug standards is a substandard drug. A drug shall be treated as a substandard
drug in any of the following cases:
1. the date of expiry is not indicated or is altered;
2. the batch number is not indicated or is altered;
3. it is beyond the date of expiry;
4. no approval is obtained for the immediate packaging material or container;
5. colorants, preservatives, spices, flavorings or other excipients are added without authorization; or
6. other cases where the drug standard are not conformed.”a
Philippines “Substandard product means the product fails to comply, with an applicable risk of injury to the public.”b
“1. Drugs produced with active substances which quantity or strength are lower than the minimum or higher than the
maximum standards prescribed in the registered formula to a degree less than the stated in Section 73 (5) [of the Thai
Drug Act of 1967].
2. Drugs produced so that their purity or other characteristics which are important to their quality differ from the standards
prescribed in the registered formula under Section 79 or drug formulas which the Minister has ordered the drug formula
registry under Section 86 bis [of the Thai Drug Act of 1967].”c
a Drug Administration Law of the People’s Republic of China. (China). 2001. Chap. V, Art. 49.
b Republic Act No. 7394, The Consumer Act of the Philippines. (Philippines). Tit. I, Art. 4 (bt).
c Thailand Drug Act, B.E. 2510 (1967). (Thailand). Chap. VIII, Sec. 74.
Oxfam Falsified Medicines for which the identity, source, or history was misrepresented (parent category for fake and
falsely labeled) (Brant and Malpani, 2011).
Falsely labeled The true properties of the product do not correspond to the information provided (Brant and Malpani,
2011).
Fake Does not contain the correct type of concentration of active and/or other ingredients (Brant and Malpani,
2011).
European Falsified “Falsified medicines are fake medicines that pass themselves off as real, authorized medicines. Falsified
Medicines medicines may:
Agency • contain ingredients of low quality or in the wrong doses;
• be deliberately and fraudulently mislabeled with respect to their identity or source;
• have fake packaging, the wrong ingredients, or low levels of the active ingredients.
Falsified medicines do not pass through the usual evaluation of quality, safety and efficacy, which is
Countering the Problem of Falsified and Substandard Drugs
required for the European Union (EU) authorization procedure. Because of this, they can be a health
threat” (EMA, 2012).
Brazil Falsification “Illicit reproduction of registered medicine, made by [a] third [party], with the fraudulent intention of
of medicines giving a legitimate appearance of what is not legitimate” (Anvisa, 2006).
Adulteration “Intervention of [a] third [party], with the purpose of altering legitimate medicine in [a] way to commit
therapeutic effectiveness and/or to turn it noxious to the health; or intervention that modifies the
specifications of the registration fraudulently, changing its registered formulation” (Anvisa, 2006).
Alteration “Modification for addition or subtraction of components of the medicine and/or if the pharmaceutical
formula, without previous and expressed approval of the National Agency of Health Surveillance” (Anvisa,
2006).
Council of the Falsified “Falsified medicinal product [is] any medicinal product with a false representation of:
European Union a) its identity, including its packaging and labeling, its name or its composition as regards any of the
ingredients including excipients and the strength of those ingredients;
b) its source, including its manufacturer, its country of manufacturing, its country of origin or its marketing
authorization holder; or
c) its history, including the records and documents relating to the distribution channels used.
This definition does not include unintentional quality defects and is without prejudice to infringements of
intellectual property rights.”a
2. A drug which shows the name of another drug, or an expiry date which is false;
3. A drug which shows a name or mark of a producer, or the location of the producer [of] the drug, which
is false;
4. Drugs which falsely show that they are in accordance with a formula which has been registered; and
5. Drugs produced with active substances which quantity or strength [is] lower than the minimum
or higher than the maximum standards prescribed in the registered formula by more than twenty
percent.”c
a Directive 2011/62/EU on the community code relating to medicinal products for human use, as regards the prevention of the entry into the legal supply chain of falsified
medicinal products [2011] OJ L174/77-78.
b Thailand Drug Act, B.E. 2510 (1967). (Thailand). Chap. VII, Sec. 75.
c Thailand Drug Act, B.E. 2510 (1967). (Thailand). Chap. VII, Sec. 73.
d The Drugs and Cosmetics Act and Rules. (India). As corrected up to 30th April, 2003. Chap. III, 9C.
e The Drugs and Cosmetics Act and Rules. (India). As corrected up to 30th April, 2003. Chap. III, 9A.
Countering the Problem of Falsified and Substandard Drugs
REFERENCES
AFI (American Film Institute). 2003. AFI’s 100 years . . . 100 heroes & villains. http://www.
afi.com/100years/handv.aspx (accessed November 7, 2012).
AMA (American Medical Association). 2012. About us. http://www.ama-assn.org/ama/pub/
physician-resources/medical-science/united-states-adopted-names-council/about-us.page?
(accessed November 16, 2012 ).
American Heritage Stedman’s Medical Dictionary. 2012a. “Drug.” Boston, MA: Houghton
Mifflin.
———. 2012b. “Medicine.” Boston, MA: Houghton Mifflin.
———. 2012c. “Pharmaceutical.” Boston, MA: Houghton Mifflin.
Amin, A., and R. Snow. 2005. Brands, costs and registration status of antimalarial drugs in the
Kenyan retail sector. Malaria Journal 4(36). DOI: 10.1186/1475-2875-4-36.
Anvisa. 2006. The Anvisa effort on combating counterfeit medicines in Brazil. Paper presented
at World Congress of Pharmacy and Pharmaceutical Sciences 2006: 66th International
Congress of FIP, Salvador Bahia, Brazil.
Attaran, A., D. Barry, S. Basheer, R. Bate, D. Benton, J. Chauvin, L. Garrett, I. Kickbusch,
J. C. Kohler, K. Midha, P. N. Newton, S. Nishtar, P. Orhii, and M. McKee. 2012. How
to achieve international action on falsified and substandard medicines. British Medical
Journal 345. DOI: 10.1136/bmj.e7381.
Attaran, A., and R. Bate. 2010. A counterfeit drug treaty: Great idea, wrong implementation.
Lancet 376(July):1446-1448.
AVMA (American Veterinary Medical Association). 2012. Representation to the United States Phar-
macopeial Convention (USP). https://www.avma.org/KB/Policies/Pages/Representation-
to-the-United-States-Pharmacopeial-Convention-USP.aspx (accessed Novemeber 19,
2012).
Barton, J. H. 2004. TRIPS and the global pharmaceutical market. Health Affairs 23(3):146-154.
Bate, R. 2010. Lessons from a Syrian drug bust. Wall Street Journal, April 28.
———. 2011. The market for inferior medicines: Comparing the price of falsified and sub-
standard poducts with the legitimate medicines in emerging markets. Washington, DC:
American Enterprise Institute.
———. 2012a. The deadly world of falsified and substandard medicine. http://www.aei.org/
article/health/global-health/the-deadly-world-of-falsified-and-sub-standard-medicine (ac-
cessed November 9, 2012).
———. 2012b (March 13). Defining dangerous drugs. Presentation at Committee on Under-
standing the Global Public Health Implications of Substandard, Falsified, and Counterfeit
Medical Products: Meeting One, Institute of Medicine, Washington, DC.
BBC (British Broadcasting Corporation). 2001a. Court battle over AIDS drugs. BBC News,
March 5.
———. 2001b. SA victory in AIDS drugs case. BBC News, April 19.
Beebe, B. 2006. An empirical study of the multifactor tests for trademark infringement. Cali-
fornia Law Review 94:1581-1654.
Björkman-Nyqvist, M., J. Svensson, and D. Yanagizawa-Drott. 2012. Can good products
drive out bad? Evidence from local markets for (fake?) antimalarial medicine in Uganda.
Cambridge, MA: Harvard University Center for International Development.
Brant, J., and R. Malpani. 2011. Eye on the ball medicine regulation—not IP enforcement—
can best deliver quality medicines. Oxford, UK: Oxfam International.
Bren, L. 2001. Frances Oldham Kelsey: FDA medical reviewer leaves her mark on history.
FDA Consumer Magazine 35(2):24-29.
INTRODUCTION 49
INTRODUCTION 51
MSF (Médecins Sans Frontières). 2009. Counterfeit, substandard and generic drugs. http://
www.msfaccess.org/content/counterfeit-substandard-and-generic-drugs (accessed May 1,
2012).
———. 2012. Spotlight on . . . substandard & counterfeit medicines. http://www.msfaccess.
org/spotlight-on/substandard-counterfeit-medicines (accessed November 9, 2012).
MSH (Management Sciences for Health). 2012. Pharmaceutical legislation and regulation. In
MDS-3: Managing access to medicines and health technologies. Arlington, VA: Manage-
ment Sciences for Health.
New, W. 2012. “Counterfeit” dropped from new WHO Protocol on Illicit Tobacco Trade.
Intellectual Property Watch, April 4.
Newton, P. 2012. Interview. Pathogens and Global Health 106(2):69-71.
Newton, P. N., A. A. Amin, C. Bird, P. Passmore, G. Dukes, G. Tomson, B. Simons, R. Bate,
P. J. Guerin, and N. J. White. 2011. The primacy of public health considerations in de-
fining poor quality medicines. PLoS Medicine 8(12). :e1001139. DOI: 10.1371/journal.
pmed.1001139.
Newton, P. N., M. D. Green, and F. M. Fernández. 2010. Impact of poor-quality medicines in
the “developing” world. Trends in Pharmacological Sciences 31(3):99-101.
Osmond, D. 2003. Epidemiology of HIV/AIDS in the United States. HIV InSite—University
of California San Francisco Medical Center.
Oxfam International. 2011. Crisis of poor quality medicines being used as an excuse to
push up prices for poor. http://www.oxfam.org/en/pressroom/pressrelease/2011-02-02/
crisis-poor-quality-medicines-being-used-excuse-push-prices-poor (accessed November
9, 2012).
Oxford Dictionaries. 2012. “Counterfeit.” http://oxforddictionaries.com/definition/counterfeit
(accessed April 27, 2012).
Paleshnuik, L. 2009. Dissolution. Paper presented at Training Workshop on Regulatory Re-
quirements for Registration of Artemisinin Based Combined Medicines and Assessment
of Data Submitted to Regulatory Authorities, Kampala, Uganda, February 23-27.
Phana, C. 2007. Country presentation: Cambodia. Paper presented at First ASEAN–China
Conference on Combating Counterfeit Medicinal Products, Jakarta, Indonesia.
Pharmacopoeial Laboratory for Indian Medicine. 2011. Publications. http://www.plimism.nic.
in/publication0.html (accessed November 19, 2012).
Pollack, A. 2001. Defensive drug industry: Fueling clash over patents. New York Times, April
20.
PSI-Inc. (Pharmaceutical Security Institute). 2012. Counterfeit situation. http://www.psi-inc.
org/counterfeitSituation.cfm (accessed April 25, 2012).
PSM (Partnership for Safe Medicines). 2012. Counterfeit vs. substandard drugs. http://
www.safemedicines.org/resources/PSM%20-%20Counterfeit%20vs.%20%20
Substandard%20Drugs.pdf (accessed April 26, 2012).
Rägo, L., and B. Santoso. 2008. Drug regulation: History, present and future. In Drug benefits
and risks: International textbook of clinical pharmacolog. 2nd ed., edited by C. van
Boxtel, B. Santoso, and I. Edwards. IOS Press and Uppsala Monitoring Centre.
Rai, A. 2001. The information revolution reaches pharmaceuticals: Balancing innovation
incentives, cost, and access. University of Illinois Law Review 1:173-210.
Ratanawijitrasin, S., and E. Wondemagegnehu. 2002. Effective drug regulation: A multi-
country study. Geneva: WHO.
Ravinetto, R. M., M. Boelaert, J. Jacobs, C. Pouget, and C. Luyckx. 2012. Poor-quality medi-
cal products: Time to address substandards, not only counterfeits. Tropical Medicine &
International Health 17(11):1412-1416.
Ruse-Khan, H. G. 2011. The (non) use of treaty object and purpose in intellectual property
disputes in the WTO. In Sustainable development principles in the decisions of interna-
tional courts and tribunals 1992-2012. Max Planck Institute for Intellectual Property and
Competition Law, Research Paper No. 11-15. Oxford: Oxford University Press.
SADC (South African Development Community). 2007. Guidelines for submitting application
for registration of medicine. Gaborone, Botswana: SADC.
Seear, M. 2012. Pharmaceutical quality: An urgent and unresolved issue. Lancet Infectious
Diseases 12(6):428-429.
Simmons, A. 2001. Suit against cheap AIDS drugs ends in S. Africa. Los Angeles Times,
April 20.
Singh, K. 2009. Row over generic drugs intensifies after seizure in Germany. Economic Times,
June 8.
Siter, A. 2005. Pharmaceuticals: Counterfeits, substandard drugs and drug diversion. Wash-
ington, DC: World Bank.
Smine, A. 2002. Malaria sentinel surveillance site assessment. Rockville, MD: U.S. Pharmacopeia.
Stanton, C., A. Koski, P. Cofie, E. Mirzabagi, B. L. Grady, and S. Brooke. 2012. Uterotonic
drug quality: An assessment of the potency of injectable uterotonic drugs purchased by
simulated clients in three districts in Ghana. BMJ Open 2(3):1-7.
Swann, J. 2009. FDA’s origin. http://www.fda.gov/AboutFDA/WhatWeDo/History/Origin/
ucm124403.htm (accessed November 19, 2012).
Swarns, R. 2001. Drug makers drop South Africa suit over AIDS medicine. New York Times,
April 20.
Taylor, L. 2009. India to reimburse costs of drug shipments which avoid EU. PharmaTimes,
August 7.
Taylor, P. 2012. WHO adopts new resolution on counterfeit medicines. http://www.securing
industry.com/pharmaceuticals/who-adopts-new-resolution-on-counterfeit-medicines/s40/
a1181 (accessed November 7, 2012).
TGA (Therapeutic Goods Administration). 2003. History of the Australian Drug Evalua-
tion Committee. http://www.tga.gov.au/archive/committees-adec-history.htm (accessed
November 28, 2012).
t’Hoen, E., J. Berger, A. Calmy, and S. Moon. 2011. Driving a decade of change: HIV/AIDS,
patents and access to medicines for all. Journal of the International AIDS Society
14(1):15.
TWN (Third World Network). 2010. The IMPACT Counterfeit Taskforce, intellectual prop-
erty rights enforcement and seizure of medicines. Penang, Malaysia: TWN.
USP (U.S. Pharmacopeia) 2008. USP standards development.
———. 2011a. Amodiaquine hydrochloride tablets. USP 35-NF 30.
———. 2011b. Quinine sulfate tablets. USP 35-NF 30.
———. 2012. Mission & history. http://www.usp.org/about-usp/our-impact/mission-history
(accessed November 16, 2012).
———. 2013. International recognition. http://www.usp.org/about-usp/legal-recognition/
international-recognition (accessed January 8, 2013).
USTR (Office of the United States Trade Representative). 2011. Anti-Counterfeiting Trade
Agreement (ACTA). http://www.ustr.gov/acta (accessed November 12, 2012).
Weaver, C., and J. Whalen. 2012. How fake cancer drugs entered U.S. Wall Street Journal,
July 20.
WHO (World Health Organization). 1992. Counterfeit drugs: Report of a WHO/IFPMA
workshop 1-3 April 1992. Geneva: WHO.
———. 2002. Coverage of selected health services for HIV/AIDS prevention and care in less
developed countries in 2001. Geneva: WHO.
INTRODUCTION 53
———. 2003a. Fact sheet n°275: Substandard and counterfeit medicines. http://www.who.int/
mediacentre/factsheets/2003/fs275/en (accessed April 12, 2012).
———. 2003b. WHO launches campaign against counterfeit medicines. Bulletin of the World
Health Organization 81(12):921-922.
———. 2006. Fact sheet n°275: Counterfeit medicines. http://web.archive.org/web/2006
1012143704/http:/www.who.int/mediacentre/factsheets/fs275/en/index.html (accessed
April 19, 2012).
———. 2009. Medicines: Frequently asked questions. Geneva: WHO.
———. 2010. New definition for “substandard medicines.” Working Document QAS/10.344/
Rev.1. http://www.who.int/medicines/services/expertcommittees/pharmprep/14052010
NewDefinitionSubstandardMeds-QAS10-344Rev1.pdf (accessed January 7, 2012).
———. 2011a. Frequently asked questions. http://www.who.int/medicines/services/counterfeit/
faqs/QandAsUpdateJuly11.pdf (accessed April 20, 2012).
———. 2011b. Marketing authorization of pharmaceutical products with special reference to
multisource (generic) products: A manual for national medicines regulatory authorities
(NMRAs)—2nd ed. Geneva: WHO.
———. 2011c. Monographs: Dosage forms: Specific monographs: Amodiaquini compressi-
amodiaquine tablets. In The international pharmacopoeia: Fourth edition. Geneva:
WHO.
———. 2013. Media centre: The World Health Assembly. http://www.who.int/mediacentre/
events/governance/wha/en/index.html (accessed January 8, 2013).
WMA (World Medical Association). 2012. Counterfeit medical products. http://www.wma.
net/en/20activities/30publichealth/50counterfeits (accessed May 1, 2012).
WTO (World Trade Organization). 1994. Agreement on trade-related aspects of intellectual
property rights. Geneva: WTO.
———. 2012. Glossary. http://www.wto.org/english/thewto_e/glossary_e/glossary_e.htm (ac-
cessed April 26, 2012).
Yadav, P., and L. Smith. 2012. Pharmaceutical company strategies and distribution systems in
emerging markets. Elsevier Encyclopedia on Health Economics.
Poisoning
Some of the most compelling stories of pharmaceutical fraud are
those of frank poisoning. Between November 2008 and February 2009,
84 Nigerian children died from acute kidney failure brought on by the
industrial solvent diethylene glycol in teething syrup (Akuse et al., 2012;
55
problem is medicine that simply does not work. Ineffective medicines of-
ten contain benign ingredients, such as chalk, pollen, or flour, instead of
medicinal chemicals. More dangerously, some contain substances intended
to mask the illness and feign treatment, such as paracetemol added to fake
antimalarials to lower fever. Patients taking ineffective drugs die of appar-
ently natural causes, making these products more difficult to identify.
1 The Partnership for Safe Medicines uses the term counterfeit broadly, the way this report
uses falsified. See page 23.
They were able to draw conclusions about uterotonic drug quality because
their data represented a random sample of drugs from an almost exhaustive
sampling frame of known pharmacies, chemical shops, and other dispensa-
ries in their study area (Stanton et al., 2012). The identification of falsified
and substandard medicines is more often incidental, found in newspaper
accounts or uncovered in research that had a different primary aim.
BOX 2-1
Defective Glucose Test Strips
In October 2006, the FDA recalled two batches of blood glucose test
strips used in LifeScan, Inc. OneTouch Ultra brand blood glucose moni-
tors after LifeScan notified the agency that it had received a number of
customer complaints. The strips produced inaccurate blood glucose level
readings, the results of which are used by diabetics to monitor their condi-
tion and determine medication dosing (Bloomberg News, 2007). Diabetics
rely on their blood glucose monitors to manage their self-treatment, and
incorrect readings can lead patients to administer the wrong dosage of in-
sulin or induce unnecessary panic. Improper insulin dosing is a potentially
fatal error. In the LifeScan recall, the FDA identified the problem strips and
instructed consumers to inspect the serial numbers on their boxes and
replace any fake or unidentifiable strips (FDA, 2006; WHO, 2006).
Investigation traced the strips back to Halson Pharmaceuticals in
Shanghai. The manufacturer sold approximately one million substandard
test strips to importers, and from there the strips went through the sup-
ply chain to reach U.S. and Canadian pharmacies. Over the course of the
next year, the test strips made their way to 8 countries and 35 U.S. states.
The Chinese authorities eventually arrested and imprisoned Henry Fu,
owner of Halson Pharmaceuticals (Bloomberg News, 2007).
The LifeScan recall is a reminder that substandard medical products
can find their way into countries with strong regulatory systems. The
United States and Canada have systems in place for prompt recalls, al-
lowing them to mitigate the threat the product poses to public health.
Within 2 years the fake test strips were fully recalled in the United States,
but between 2009 and 2011 customers and investigators still found them
in other countries, including Egypt, India, and Pakistan (Loftus, 2011).
As the prevalence of diabetes increases rapidly in the developing
world, new, loosely regulated markets attract potential counterfeiters.
India is home to more than 50 million diabetics, more than any other
country, and the number is expected to increase dramatically over the
coming years (World Diabetes Foundation, 2013). In 2007, not long after
the first bad test strips appeared in the United States, there were ap-
proximately 40.9 million diabetics in India; by the time they reached the
country’s growing diabetic population the number had risen by more
than 10 million (Mohan et al., 2007). As the chronic disease burden in-
creases in developing countries, falsified and substandard versions of the
expensive products used to treat them pose new risks.
general, the vaccine supply chain is simpler, if only because Unicef manages
the parts of the chain between the manufacturer and the national port of
entry (Kauffmann et al., 2011). Cases of falsified and substandard vaccines
are rare, but Box 2-2 describes some.
If antibiotics are some of the oldest and most widely used medicines in
the world, antiretrovirals are their opposites: new medicines, prescribed in
complicated regimes, to a relatively small segment of the population. An
exhaustive WHO survey of antiretroviral drug quality in seven sub-Saharan
African countries and a variety of treatment centers found reliable good
quality in HIV medications (WHO, 2007). Only 1.8 percent of the drugs
tested failed to meet quality specifications, and even those were “[not] seri-
ous failures, i.e., no critical deficiencies which would pose a serious risk to
patients” (WHO, 2007, p. 19).
BOX 2-2
Deaths from Substandard and Falsified Vaccines
Some more recent reports suggest that falsified antiretroviral drugs may
circulate in African countries. In September 2011, falsified and substandard
versions of the triple combination therapy Zidolam-N surfaced in Kenya,
many samples molding and crumbling in the packages (Taylor, 2011). A
year later, in Tanzania, the regulatory authority uncovered falsified anti-
retrovirals at a district hospital (Athumani, 2012). These failures put HIV
patients at risk for disease progression and favor the selection of resistant
virus strains (WHO, 2003). As their viral loads increase, these patients are
also more likely to transmit the infection, impeding efforts to control the
virus.
Although data suggesting compromised antiretroviral drug quality are
mixed, there is substantial evidence, presented in Chapter 3, that antima-
larials are often of poor quality. Substandard and falsified malaria drugs
are especially common in malaria-endemic parts of Africa and Asia. In
2003, substandard sulfadoxine-pyrimethamine was used to treat a malaria
epidemic in northwest Pakistan refugee camps (Leslie et al., 2009). Re-
searchers concluded that, as the strain of P. falciparum they identified was
90 percent curable when using standard sulfadoxine-pyrimethamine drugs,
the substandard medicines, procured from local manufacturers because of
drug shortages, were a causal factor in what initially presented as drug-
resistant malaria (Leslie et al., 2009). In this example, the effects of the
substandard medicine were promptly mitigated. Health workers diagnosed
the parasite with microscopy, monitored drug resistance, and checked drug
quality using procedures described in the U.S. Pharmacopeia monograph.
Good care during initial infection and treatment with an effective second-
line drug prevented any deaths, and the onset of cooler weather stopped
transmission (Leslie et al., 2009). The prognosis for most people treated
with poor-quality antimalarial drugs is worse. Not only will their malaria
be untreated, but inadequate treatment favors the selection of resistant
parasites, which threaten their entire communities.
Treatment Failure
Individual patients have much to lose from substandard and falsified
medicines. These products also encourage drug resistance and thereby
threaten population health today and for future generations. This is a par-
ticular concern with substandard products where the dose of active ingredi-
ent is low and variable and with falsified products diluted by criminals in
an effort to pass screening assays. Drug resistance is common in pathogens
with short life cycles: viruses, bacteria, and protozoa. Poor-quality anti-
microbial medications, taken frequently and, in poor countries, generally
taken without professional supervision, contribute to drug resistance.
Antimicrobial Resistance
Antibiotics should be used only when indicated, in the appropriate
dose, and for the correct length of time. Ensuring the proper treatment
with the right combination of drugs is the underlying principle of Directly
Observed Treatment–Short Course (DOTS), the internationally accepted
method of tuberculosis surveillance and treatment (WHO SEARO, 2006).
DOTS also depends on a safe and reliable drug supply. Poor-quality drugs
have been cited as a causal factor for the rise of multidrug-resistant tubercu-
losis (Kelland, 2012). Over time, the bacteria causing tuberculosis have be-
come increasingly drug resistant. Multidrug-resistant tuberculosis precedes
extensively drug-resistant tuberculosis, and finally, sometimes, totally drug-
resistant tuberculosis (Udwadia, 2012). Extensively drug-resistant strains of
tuberculosis account for about 6 percent of incident infections worldwide,
but much more in China, India, and the former Soviet Union (Jain and
Mondal, 2008). Figure 2-1 shows the increasing incidence of multidrug-
resistant tuberculosis around the world.
Drug-resistant bacteria often surface in hospitals, causing infections
that are difficult to treat and are an important killer of adults in low-
and middle-income countries (Okeke et al., 2005b; WHO, 2012a). It is
difficult to estimate the burden of antimicrobial resistance in low- and
middle-income countries, in part because of the dearth of data, especially
from francophone Africa, the Asian Pacific, and the former Soviet Union
(Okeke et al., 2005a). The data that do exist are grim. Multidrug-resistant
staphylococcus, an emerging problem in India and sub-Saharan Africa
(Parasa et al., 2010; Vincent et al., 2009), accounts for more than half of
all nosocomial infections in parts of Latin America (Guzmán-Blanco et al.,
2009). (See Figure 2-2.)
In a qualitative study in Orissa, India, doctors, veterinarians, and phar-
macists cited poor-quality antibiotics as a cause of drug resistance, but men-
tioned it only in passing, focusing more on patient and provider behaviors
(Sahoo et al., 2010). This is consistent with most public health literature,
which gives great deal of attention to the overuse of antibiotics as contrib-
uting to the rise of antimicrobial resistance in general (Byarugaba, 2010;
Okeke et al., 2005b) and drug-resistant pneumonia in particular (Unicef
and WHO, 2006). Comparatively little work, however, discusses the role
of drug quality in encouraging bacterial resistance. Antibiotics that contain
low doses of active ingredient cause low circulating levels of the drug in
the patient. This contributes to treatment failure and selectively favors the
growth of drug-resistant organisms (Okeke et al., 2005b). Resistance is
most common among the oldest and least expensive families of antibiotics
(Okeke et al., 2005b).
MDR-TB among
new TB cases (%)
0-2.9
3-5.9
6-11.9
12-17.9
> 18
No data
Subnational data only
Not applicable
Antimalarial Resistance
Through a conceptually similar mechanism, selectively allowing the
growth of drug-resistant parasites by exposing them to subtherapeutic doses
of medicines, falsified and substandard drugs favor survival and spread of
resistance to antimalarial medicines. Drug-resistant parasites of particular
concern are the malaria parasites Plasmodium falciparum and Plasmodium
vivax.
Artemisinin combination treatments are effective in treating falciparum
malaria (WHO, 2011d). They have been the recommended first-line treat-
ment for falciparum malaria everywhere in the world since 2001 (WWARN,
2012d). In areas where these drugs are available and appropriately used,
malaria deaths have dropped dramatically (WHO, 2011d).
Drug resistance could undo the success that artemisinin therapies have
won, however (see Box 2-3). A recent review estimates that about 35 per-
cent of the antimalarial medicines in Southeast Asia are substandard, and
36 percent can be classified as falsified (Nayyar et al., 2012). The same
researchers found similar patterns in sub-Saharan Africa, where about
35 percent of antimalarials are substandard and 20 percent are falsified
(Nayyar et al., 2012). In both regions, underdosing the active ingredients
is far more common than overdosing (Nayyar et al., 2012). Already, 8
of the 12 major antimalarial drugs used in the world have been falsified,
including products labeled as of mefloquine, but containing sulphadoxine-
pyrimethamine and no mefloquine, and a product labeled as artesunate, but
containing 6 percent chloroquine and no artesunate (Newton et al., 2006).
Poor-quality medicines supply a subtherapeutic dose that selectively encour-
ages the emergence of partially resistant pathogens (Talisuna et al., 2012).
Underdosing with antimalarials causes low concentrations of ac-
tive drugs in patients and selective pressure to breed resistant parasites
(Dondorp et al., 2011; Sengaloundeth et al., 2009; White et al., 2009). In
Thailand investigators have observed a progressive lengthening of the time
it takes for patients to clear malaria parasites from their bloodstream dur-
ing treatment, suggesting that the parasites are becoming more resistant to
artemisinin (Phyo et al., 2012). Resistance is heritable from one generation
Mexico
32–52% Cuba
6%
Hondoras 12%
Guatemala Nicaragua 20%
64%
Panama Venezuela
Costa Rica 28% 25–36%
58%
Colombia
34–47%
Ecuador
25%
Brazil
Peru 54–93%
80–85%
Bolivia
36–55%
Paraguay
30–44%
Chile Argentina
29–80% 42–51%
Uruguay
24–59%
of parasite to the next; the relatively resistant parasites persist and are
transmitted (Anderson et al., 2010). So far, artemisinin resistance has been
documented only in Southeast Asia, but its persistence and spread could
threaten global malaria control programs. No other antimalarial drugs are
available as alternatives. If the current first-line therapy is lost because of
resistance, malaria deaths will again increase.
Key Findings
BOX 2-3
The WorldWide Antimalarial Resistance Network
Pharmacy in Cambodia.
SOURCE: Hen Sophal in Pharmacide Arts, an exhibit of Southeast Asian artists.
sold in rich countries. It is not yet clear how much patients and insurance
companies paid for falsified Avastin during the 2012 crisis, but the Wall
Street Journal found that the fake product sold for almost $2,000 per vial
(Weaver and Whalen, 2012).
Drug resistance will increase costs to the health system, and not only
because of increased clinical attention. Drug resistance reduces the effective
life of a drug. Already the cheapest, oldest classes of anti-infective drugs
are becoming useless. Society must bear the expense of new drug develop-
ment, an ever-increasing cost (see Figure 2-3), because resistant pathogens
require treatment with more complex drugs. A 2010 estimate put the cost
of developing a single drug at $1.3 billion (Kaitin, 2010), and a 2003 study
showed that the cost of drug development grew 7.4 percent faster than
inflation (DiMasi et al., 2003).
Aside from the direct financial costs of treatment, there are opportunity
costs incurred to patients who miss work for additional doctors’ visits or
become too sick to work. Chapter 3 will explain that the burden of falsi-
fied and substandard medicines is borne mostly by the poor in South and
Southeast Asia and sub-Saharan Africa. Transport costs and opportunity
costs are a known obstacle to health care for these patients (Whitty et al.,
2008). Customers at gray pharmaceutical markets, including flea markets,
$1.4
$1.3B
$1.2
Billions (Constant dollars, 2000)
$1.0
$0.8
$800M
$0.6
$0.4
$300M
$0.2
$100M
$0.0
1975 1987 2000 2005
Fiscal Year
unlicensed medicine shops, and bazaars, are often there because they can-
not afford to miss work for a formal consultation (Whitty et al., 2008).
For example, participants at the São Paulo site visit for this study explained
that although medicines are free through the public health system in Brazil,
miners and other daily-wage workers circumvent this system. They continue
working and self-treat with medicines of dubious quality from the gray
market. In Brazil, as in many parts of the world, falsified and substandard
medicines extract the highest costs from those who can afford the least.
Scientists and policy makers in developing countries are aware of the
toll falsified and substandard drugs take on their health systems. A 2009
WHO expert working group rated fake medicines as a top priority for
research in developing countries (Bates et al., 2009).
Patients may begin to distrust modern pharmacies after experiences
with falsified and substandard drugs. In Ugandan villages, the proportion
of positive responses to the question “Do you expect that the antimalarial
medicines sold by the nearest drug shop are fake?” correlates roughly
1,200
1,000
1,000 1,010
800
774
US$ millions
600
400 438
285
200
167
95
75
0 Cocaine
Workers
Toxic waste
Arms
Trafficking victims
Antimalarials
Cigarettes
Oil
Development and Crime (UNODC) reckons that in West Africa the sale
in falsified medicines may be worth as much as the billion-dollar oil and
cocaine trafficking industries; their estimate of the worth of trafficked anti-
malarials alone is more than $400 million (see Figure 2-4).
Chapter 4 describes why medicines fraud is sometimes called the perfect
crime. Fake medicines generate income for criminals, and only the weakest
evidence, if any, ties them to their crime. Acute cases of medicine poisoning
can elicit public outcry, but more often bad drugs go unnoticed, blending
in with lawful business. Victims of falsified and substandard drugs usually
do not even know they are victims and are therefore deprived of their right
to redress. The UNODC described the traffic in fake drugs as both as cause
and an effect of political instability, explaining, “Living in a society where
such widespread and serious fraud can occur undermines confidence in gov-
ernment, but the effects are so diffuse and uncertain that they are unlikely
to generate an organized political response” (UNODC, 2009, p. 6). In many
parts of the world, falsified and substandard medicines further erode the
already weak political infrastructure that allows them to circulate, part of
a vicious cycle of poverty and crime.
Known Falsified or
Substandard or
Common Pathogen Essential Medicine Both?
Ampicillin Yes a,b,e
Trimethoprim-sulfamethoxazole
Yes a
Pneumocystis carinii Trimethoprim-sulfamethoxazole
Erythromycin Yes a,f,g
Erythromycin Yes a,f,g
Doxycycline Yes a,h
Ampicillin Yes a,b,e
Trimethoprim-sulfamethoxazole Yes a
Known Falsified or
Substandard or
Common Pathogen Essential Medicine Both?
Artesunate Yes k,l,m
Sulfadoxine/pyrimethamine Yes l,j,o
Countering the Problem of Falsified and Substandard Drugs
Quinine Yes j,k,o
Primaquine
Yes q,r
Plasmodium ovale Primaquine
i Bate, R., P. Coticelli, R. Tren, and A. Attaran. 2008. Antimalarial drug quality in the nel. Lancet 348(9039):1453-1454.
r Petralanda, I. 1995. Quality of antimalarial drugs and resistance to Plasmodium
most severely malarious parts of Africa—a six country study. PLoS ONE 3(5):e2132.
j Kaur, H., C. Goodman, E. Thompson et al. 2008. A nationwide survey of the quality vivax in Amazonian region. Lancet 345(8962):1433.
of antimalarials in retail outlets in Tanzania. PLoS ONE 3(10).
REFERENCES
Akuse, R. M., F. U. Eke, A. D. Ademola, I. B. Fajolu, A. O. Asinobi, H. U. Okafor, S. I.
Adeleke, L. I. Audu, A. Otuneye, E. Disu, H. Idis, H. Aikhonbare, A. Yakubu, W. Ogala,
O. Ogunrinde, R. Wammanda, A. Orogade, J. Anyiam, E. Eseigbe, L. Umar, H. Musa,
R. Onalo, B. West, N. Paul, F. Lesi, T. Ladapo, O. Boyede, R. Okeowo, A. Mustapha,
I. Akinola, O. Chima-Oduko, and O. Awobusuyi. 2012. Diagnosing renal failure due
to diethylene glycol in children in a resource-constrained setting. Pediatric Nephrology
27(6):1021-1028.
Anderson, T. J. C., S. Nair, S. Nkhoma, J. T. Williams, M. Imwong, P. Yi, D. Socheat, D. Das,
K. Chotivanich, N. P. J. Day, N. J. White, and A. M. Dondorp. 2010. High heritability
of malaria parasite clearance rate indicates a genetic basis for artemisinin resistance in
Western Cambodia. Journal of Infectious Diseases 201(9):1326-1330.
Asamoah, B. O., K. M. Moussa, M. Stafstrom, and G. Musinguzi. 2011. Distribution of causes
of maternal mortality among different socio-demographic groups in Ghana: A descriptive
study. BMC Public Health 11(159). DOI: 10.1186/1471-2458-11-159.
Associated Press. 1998. Phony birth control pills sold in Brazil. Augusta Chronicle, July 4.
———. 2010a. China probes vaccines after child deaths reported. http://www.boston.com/
news/world/asia/articles/2010/03/18/china_probes_vaccines_after_child_deaths_reported
(accessed March 5, 2013).
———. 2010b. China rabies vaccine recall prompts changes. http://www.cbc.ca/news/health/
story/2010/04/02/china-vaccine.html (accessed March 5, 2013).
Athumani, R. 2012. Tanzania: Factory closed over fake ARVs. Tanzania Daily News, October
11.
BASCAP (Business Action to Stop Counterfeiting and Pharmacy). 1996. Fake vaccine leads
to 3,000 deaths in Niger. http://www.icc-ccs.co.uk/bascap/article.php?articleid=363 (ac-
cessed February 21, 2013).
Bates, D. W., I. Larizgoitia, N. Prasopa-Plaizier, and A. K. Jha. 2009. Global priorities for
patient safety research. British Medical Journal 338. DOI: 10.1136/bmj.b1775.
BBC (British Broadcasting Corporation). 2010. Global Fund freezes Zambia aid over corrup-
tion concern. BBC Africa, June 16.
———. 2012. Tanzania investigates fake HIV drugs. BBC News Africa, October 15.
Berendes, S., P. Heywood, S. Oliver, and P. Garner. 2011. Quality of private and public ambu-
latory health care in low and middle income countries: Systematic review of comparative
studies. PLoS Medicine 8(4):e1000433.
Björkman-Nyqvist, M., J. Svensson, and D. Yanagizawa-Drott. 2012. Can good products
drive out bad? Evidence from local markets for (fake?) antimalarial medicine in Uganda.
Cambridge, MA: Harvard University Center for International Development.
Bloomberg News. 2007. Bogus diabetes test strips traced to Chinese distributor. New York
Times, August 17.
Bogdanich, W. 2007. F.D.A. tracked poisoned drugs, but trail went cold in China. New York
Times, June 17.
Bogdanich, W., and J. Hooker. 2007. From China to Panama, a trail of poisoned medicine.
New York Times, May 6.
Byarugaba, D. K. 2004. Antimicrobial resistance in developing countries and responsible risk
factors. International Journal of Antimicrobial Agents 24(2):105-110.
———. 2010. Mechanisms of antimicrobial resistance. In Antimicrobial resistance in devel-
oping countries, edited by A. de J. Sosa, D. K. Byarugaba, C. F. Amábile-Cuevas, P.-R.
Hsueh, S. Kariuki and I. N. Okeke. New York: Springer. Pp. 15-26.
Cabaret, J. 2010. False resistance to antiparasitic drugs: Causes from shelf availability to
patient compliance. Anti-Infective Agents in Medicinal Chemistry 9(3):161-167.
Cameron, A., M. Ewen, D. Ross-Degnan, D. Ball, and R. Laing. 2008. Medicines prices,
availability, and affordability in 36 developing and middle-income countries: A secondary
analysis. Lancet 373(9659):240-249.
Campbell, O. M. R., and W. J. Graham. 2006. Strategies for reducing maternal mortality:
Getting on with what works. Lancet 368(9543):1284-1299.
CDC (Centers for Disease Control and Prevention). 2009. Fatal poisoning among young
children from diethylene glycol-contaminated acetaminophen. Morbidity and Mortality
Weekly Report 58(48):1345-1347.
Cheng, M. M. 2009. Is the drugstore safe? Counterfeit diabetes products on the shelves. Jour-
nal of Diabetes Science and Technology Online 3(6):1516-1520.
Cockburn, R. 2005. Death by dilution. American Prospect, http://prospect.org/article/death-
dilution (accessed January 11, 2013).
Csillag, C. 1998. São Paulo: Epidemic of counterfeit drugs causes concern in Brazil. Lancet
352(9127):553.
DiMasi, J., R. Hansen, and H. Grabowski. 2003. The price innovation: New estimates of drug
devopment. Journal of Health Economics 22:151-185.
Dondorp, A. M., R. M. Fairhurst, L. Slutsker, J. R. MacArthur, J. G. Breman, P. J. Guerin, T. E.
Wellems, P. Ringwald, R. D. Newman, and C. V. Plowe. 2011. The threat of artemisinin-
resistant malaria. New England Journal of Medicine 365(12):1073-1075.
Dorlo, T. P. C., T. A. Eggelte, P. J. De Vries, and J. H. Beijnen. 2012. Characterization and
identification of suspected counterfeit miltefosine capsules. Analyst 137(5):1265-1274.
Eichie, F. E., M. I. Arhewoh, J. E. Isesele, and K. T. Olatunji. 2011. In vitro assessment of qual-
ity control parameters of some commercially available generics of amlodipine besylate in
Nigerian drug market. International Journal of Health Research 4(1):57-61.
Fackler, M. 2002. China’s fake drugs kill thousands. San Francisco Examiner, July 31.
FDA (U.S. Food and Drug Administration). 2006. FDA updates its nationwide alert on coun-
terfeit One Touch blood glucose test strips. http://www.fda.gov/NewsEvents/Newsroom/
PressAnnouncements/2006/ucm108809.htm (accessed June 21, 2012).
Findlay, B. 2011. Counterfeit drugs and national security. Washington, DC: The Stimson
Center.
Ganyaglo, G. Y. K., and W. C. Hill. 2012. A 6-year (2004-2009) review of maternal mortality
at the eastern regional hospital, Koforidua, Ghana. Seminars in Perinatology 36(1):79-83.
Gaziano, T. A. 2007. Reducing the growing burden of cardiovascular disease in the developing
world. Health Affairs 26(1):13-24.
Geerts, S., and B. Gryseels. 2001. Anthelmintic resistance in human helminths: A review.
Tropical Medicine and International Health 6(11):915-921.
Goering, L. 1998. Baby boom forces Brazil to confront fake medicines. Chicago Tribune
News, July 14.
Guzmán-Blanco, M., C. Mejía, R. Isturiz, C. Alvarez, L. Bavestrello, E. Gotuzzo, J. Labarca,
C. M. Luna, E. Rodríguez-Noriega, M. J. C. Salles, J. Zurita, and C. Seas. 2009. Epide-
miology of meticillin-resistant Staphylococcus aureus (MRSA) in Latin America. Inter-
national Journal of Antimicrobial Agents 34(4):304-308.
Jain, A., and R. Mondal. 2008. Extensively drug-resistant tuberculosis: Current challenges and
threats. FEMS Immunology & Medical Microbiology 53(2):145-150.
Jia, H., and K. Carey. 2011. Chinese vaccine developers gain WHO imprimatur. Nature Bio-
technology 29(6):471-472.
Kaitin, K. 2010. Deconstructing the drug development process: The new face of innovation.
Clinical Pharmacology and Therapeutics 87(3):356-361.
Kauffmann, J. R., R. Miller, and J. Cheyne. 2011. Vaccine supply chains need to be better
funded and strengthened, or lives will be at risk. Health Affairs 30(6):1113-1121.
Kelland, K. 2012. Drug-resistant “white plague” lurks among rich and poor. Reuters,
March 19.
Kyriacos, S., M. Mroueh, R. P. Chahine, and O. Khouzam. 2008. Quality of amoxicillin
formulations in some Arab countries. Journal of Clinical Pharmacy and Therapeutics
33(4):375-379.
Lee, Q. Y., A. T. Odoi, H. Opare-Addo, and E. T. Dassah. 2012. Maternal mortality in Ghana:
A hospital-based review. Acta Obstetricia et Gynecologica Scandinavica 91(1):87-92.
Lemonick, M. D. 2005. Drug warrior. Time, October 31.
Leslie, T., H. Kaur, N. Mohammed, K. Kolaczinski, R. L. Ord, and M. Rowland. 2009.
Epidemic of plasmodium falciparum malaria involving substandard antimalarial drugs,
Pakistan, 2003. Emerging Infectious Diseases 15(11):1753-1759.
Lim, M.-K., H. Yang, T. Zhang, W. Feng, and Z. Zhou. 2004. Public perceptions of private
health care in socialist China. Health Affairs 23(6):222-234.
Loftus, P. 2011. J&J finds more fake diabetes test strips. Wall Street Journal, April 26.
Mohan, V., R. Sandeep, R. Deepa, B. Shah, and C. Varghese. 2007. Epidemiology of type 2
diabetes. Indian Journal of Medical Research 125:217-230.
Murray, C. J. L., L. C. Rosenfeld, S. S. Lim, K. G. Andrews, K. J. Foreman, D. Haring, N.
Fullman, M. Naghavi, R. Lozano, and A. D. Lopez. 2012. Global malaria mortality
between 1980 and 2010: A systematic analysis. Lancet 379(9814):413-431.
Nair, A., S. Strauch, J. Lauwo, R. W. O. Jähnke, and J. Dressman. 2011. Are counterfeit or
substandard anti-infective products the cause of treatment failure in Papua New Guinea?
Journal of Pharmaceutical Sciences 100(11):5059-5068.
Nayyar, G. M. L., J. G. Breman, P. N. Newton, and J. Herrington. 2012. Poor-quality an-
timalarial drugs in southeast Asia and sub-Saharan Africa. Lancet Infectious Diseases
12(6):488-496.
Newton, P. N., M. D. Green, F. M. Fernández, N. P. J. Day, and N. J. White. 2006. Counterfeit
anti-infective drugs. Lancet Infectious Diseases 6(9):602-613.
Núñez, E. 2011. Institute of legal medicine advances in review of pending cases: 219 are killed
by syrup. Prensa, June 28.
O’Brien, K. L., L. J. Wolfson, J. P. Watt, E. Henkle, M. Deloria-Knoll, N. McCall, E. Lee,
K. Mulholland, O. S. Levine, and T. Cherian. 2009. Burden of disease caused by
streptococcus pneumoniae in children younger than 5 years: Global estimates. Lancet
374(9693):893-902.
Okeke, I. N., K. P. Klugman, Z. A. Bhutta, A. G. Duse, P. Jenkins, T. F. O’Brien, A. Pablos-
Mendez, and R. Laxminarayan. 2005a. Antimicrobial resistance in developing countries.
Part II: Strategies for containment. Lancet Infectious Diseases 5(9):568-580.
Okeke, I. N., R. Laxminarayan, Z. A. Bhutta, A. G. Duse, P. Jenkins, T. F. O’Brien, A. Pablos-
Mendez, and K. P. Klugman. 2005b. Antimicrobial resistance in developing countries.
Part I: Recent trends and current status. Lancet Infectious Diseases 5(8):481-493.
Olajide, A. S., O. C. Chidinnma, and U. E. Dennis. 2010. Comparative assessment of the qual-
ity control measurements of multisource amlodipine tablets marketed in Nigeria. Inter-
national Journal of Biomedical and Advance Research 1(4). DOI: 10.7439/ijbar.v1i4.10.
Olusola, A. M., A. I. Adekoya, and O. J. Olanrewaju. 2012. Comparative evaluation of physi-
cochemical properties of some commercially available brands of metformin HCL tablets
in lagos, Nigeria. Journal of Applied Pharmaceutical Science 2(2):41-44.
Parasa, L. S., L. C. A. Kumar, S. Para, V. S. R. Atluri, K. Santhisree, P. R. Kumar, and C. R.
Setty. 2010. Epidemiological survey of mithicillin resistant Staphylococcus aureus in the
community and hospital, Gannavaram, Andhra Pradesh, South India. Reviews in Infec-
tion 1(2):117-123.
PhRMA (Pharmaceutical Research and Manufacturers of America). 2012. Chart pack: Bio-
pharmaceuticals in perspective. http://www.phrma.org/default/files/159/phrma_chart_
pack.pdf (accessed April 10, 2013).
Phyo, A. P., S. Nkhoma, K. Stepniewska, E. A. Ashley, S. Nair, R. McGready, C. ler Moo,
S. Al-Saai, A. M. Dondorp, K. M. Lwin, P. Singhasivanon, N. P. J. Day, N. J. White,
T. J. C. Anderson, and F. Nosten. 2012. Emergence of artemisinin-resistant malaria on
the western border of Thailand: A longitudinal study. Lancet 379(9830):1960-1966.
Poisoned teething drug arrests. 2009. Sky News, February 11.
Polgreen, L. 2009. 84 children are killed by medicine in Nigeria. New York Times, February 6.
PSM (Partnership for Safe Medicines). n.d. L.E.A.D.E.R.’S guide for physicians. Vienna, VA:
PSM.
Ratanawijitrasin, S., and E. Wondemagegnehu. 2002. Effective drug regulation: A multicoun-
try study. Geneva: WHO.
Rentz, E. D., L. Lewis, O. J. Mujica, D. B. Barr, J. G. Schier, G. Weeraskera, P. Kuklenyik,
M. McGeehin, J. Osterloh, J. Wamsley, W. Lum, C. Alleyne, N. Sosa, J. Motta, and C.
Rubin. 2008. Outbreak of acute renal failure in Panama in 2006: A case-control study.
Bulletin of the World Health Organization 86(10):737-816.
Rickman, J. 2012. China shutters shoddy drugmakes in wake of chromium capsule scare.
International Pharmaceutical Regulatory Monitor 40(6).
Sahoo, K. C., A. J. Tamhankar, E. Johansson, and C. S. Lundborg. 2010. Antibiotic use, re-
sistance development and environmental factors: A qualitative study among healthcare
professionals in Orissa, India. BMC Public Health 10. DOI: 10.1186/1471-2458-10-629.
Sengaloundeth, S., M. Green, F. Fernandez, O. Manolin, K. Phommavong, V. Insixiengmay, C.
Hampton, L. Nyadong, D. Mildenhall, D. Hostetler, L. Khounsaknalath, L. Vongsack, S.
Phompida, V. Vanisaveth, L. Syhakhang, and P. Newton. 2009. A stratified random sur-
vey of the proportion of poor quality oral artesunate sold at medicine outlets in the Lao
PDR—implications for therapeutic failure and drug resistance. Malaria Journal 8(1):172.
Stanton, C., A. Koski, P. Cofie, E. Mirzabagi, B. L. Grady, and S. Brooke. 2012. Uterotonic
drug quality: An assessment of the potency of injectable uterotonic drugs purchased by
simulated clients in three districts in Ghana. BMJ Open 2(3):1-7.
Sundar, S. 2001. Drug resistance in Indian visceral leishmaniasis. Tropical Medicine and In-
ternational Health 6(11):849-854.
Sundar, S., P. R. Sinha, N. K. Agrawal, R. Srivastava, P. M. Rainey, J. D. Berman, H. W.
Murray, and V. P. Singh. 1998. A cluster of cases of severe cardiotoxicity among kala-
azar patients treated with a high-osmolarity lot of sodium antimony gluconate. American
Journal of Tropical Medicine and Hygiene 59(1):139-143.
Tabernero, P., and P. N. Newton. 2012. The WWARN antimalarial quality surveyor. Patho-
gens and Global Health 106(2):77.
Talisuna, A. O., C. Karema, B. Ogutu, E. Juma, J. Logedi, A. Nyandigisi, M. Mulenga, W. F.
Mbacham, C. Roper, P. J. Guerin, U. D’Alessandro, and R. W. Snow. 2012. Mitigating
the threat of artemisinin resistance in Africa: Improvement of drug-resistance surveillance
and response systems. Lancet Infectious Diseases 12(11):888-896.
Taylor, P. 2011. Fake HIV medications uncovered in Kenya. http://www.securingindustry.com/
pharmaceuticals/fake-hiv-medications-uncovered-in-kenya/s40/a1095 (accessed January
2, 2013).
Twagirumukiza, M., A. Cosijns, E. Pringels, J. P. Remon, C. Vervaet, and L. Van Bortel.
2009. Influence of tropical climate conditions on the quality of antihypertensive drugs
from Rwandan pharmacies. American Journal of Tropical Medicine and Hygiene 81(5):
776-781.
Udwadia, Z. F. 2012. MDR, XDR, TDR tuberculosis: Ominous progression. Thorax 67(4):
286-288.
Unachukwu, C. N., D. I. Uchenna, and E. Young. 2008. Mortality among diabetes in-patients
in Port-Harcourt, Nigeria. African Journal of Endocrinology and Metabolism 7(1). http://
www.ajol.info/index.php/ajem/article/view/57567/45947 (accessed May 1, 2013).
Unicef (United Nations Children’s Fund). 2003. At a glance: Ghana. http://www.unicef.org/
infobycountry/ghana_statistics.html (accessed June 21, 2012).
Unicef and WHO (World Health Organization). 2006. Pneumonia: The forgotten killer of
children. Geneva: Unicef and WHO.
UNODC (United Nations Office on Drugs and Crime). 2009. Transnational trafficking and the
rule of law in West Africa: A threat assessment. Vienna, Austria: UNODC.
Vincent, J.-L., J. Rello, J. Marshall, E. Silva, A. Anzueto, C. D. Martin, R. Moreno, J. Lipman,
C. Gomersall, Y. Sakr, and K. Reinhart. 2009. International study of the prevalence
and outcomes of infection in intensive care units. Caring for the Critically Ill Patient
302(21):2323-2329.
Weaver, C., and J. Whalen. 2012. How fake cancer drugs entered U.S. Wall Street Journal,
July 20.
White, N., W. Pongtavornpinyo, R. Maude, S. Saralamba, R. Aguas, K. Stepniewska, S. Lee,
A. Dondorp, L. White, and N. Day. 2009. Hyperparasitaemia and low dosing are an
important source of anti-malarial drug resistance. Malaria Journal 8(1):253.
Whitty, C., C. Chandler, E. Ansah, T. Leslie, and S. Staedke. 2008. Deployment of ACT
antimalarials for treatment of malaria: Challenges and opportunities. Malaria Journal
7(Suppl 1):S7.
WHO (World Health Organization). 2003. Alert no. 110: Counterfeit triple antiretroviral
combination product (Ginovir 3D) detected in Côte d’Ivoire. Geneva: WHO.
———. 2006. Fact sheet n°275: Counterfeit medicines. http://web.archive.org/web/2006
1012143704/http:/www.who.int/mediacentre/factsheets/fs275/en/index.html (accessed
April 19, 2012).
———. 2007. Survey of the quality of antiretroviral medicines circulating in selected African
countries. Geneva: WHO.
———. 2009. Fact sheet n°335: Medicines—corruption and pharmaceuticals. http://www.
who.int/mediacentre/factsheets/fs335/en/index.html (accessed September 10, 2012).
———. 2011a. Diabetes: Fact sheet n°312. http://www.who.int/mediacentre/factsheets/fs312/
en (accessed June 21, 2012).
———. 2011b. Pneumonia: Fact sheet n°331. http://www.who.int/mediacentre/factsheets/
fs331/en/index.html (accessed June 21, 2012).
———. 2011c. WHO model list of essential medicines: 17th list. Geneva: WHO.
———. 2011d. World malaria report: 2011. Geneva: WHO.
———. 2012a. Antimicrobial resistance: Fact sheet n°194. http://www.who.int/mediacentre/
factsheets/fs194/en (accessed March 5, 2013).
———. 2012b. Global tuberculosis report. Geneva: WHO.
———. 2012c. Malaria: Fact sheet n°94. Geneva: WHO.
WHO SEARO (Regional Office for South-East Asia). 2006. Tuberculosis factsheets: What is
DOTS? http://209.61.208/233/en/Section10/Section2097/Section2106_10678.htm (ac-
cessed June 21, 2012).
Wondemagegnehu, E. 1999. Counterfeit and substandard drugs in Myanmar and Viet Nam:
Report of a study carried out in cooperation with the governments of Myanmar and Viet
Nam. Geneva: WHO.
World Diabetes Foundation. 2013. Gestational diabetes among rural and tribal people
WDF08-381: Objectives and approaches. http://www.worlddiabetesfoundation.org/
projects/karnataka-India-wdf08-381 (accessed April 1, 2013).
85
1
Detection means confirming though chemical or package analysis that the product is not
what it purports to be.
Country Incidents
1 China 279
3 Japan 79
4 Germany 62
5 Pakistan 61
5 Peru 61
7 Colombia 59
8 United Kingdom 56
9 South Korea 47
10 Brazil 45
10 Russia 45
12 Taiwan 44
of medicines move into the top three most commonly targeted (PSI data
shared with the committee, Thomas Kubic, PSI-Inc., July 11, 2012). This
is consistent with other industry reports that drugs sold and restocked fre-
quently are most often targeted (Mukherjee, 2012).
PSI member companies identified 1,623 counterfeiting incidents in
2011. In about half of these incidents (n = 810) companies were able to
do product and packaging analysis. Investigators found that most samples
were fraudulent in both product and packaging (see Figure 3-1). A false
product in legitimate packaging was the second most common result; Chap-
ter 5 discusses this problem in more detail.
Analysis of PSI data supports two main conclusions. First, falsified and
substandard drugs are a global problem that affected at least 124 countries
in 2011 (PSI data shared with the committee, Thomas Kubic, PSI-Inc., July
11, 2012). Twelve more countries were affected in 2011 than in 2010;
African countries accounted for most of this increase (PSI data shared
with the committee, Thomas Kubic, PSI-Inc., July 11, 2012). The data do
not suggest anything about the relative burden of the problem in different
countries, however. Indeed, countries with lax enforcement attract illegal
manufacturers, and countries with vigorous law enforcement repel them.
TABLE 3–2 Ten Countries Most Named in PSI Incident Reports, 2011
Total
Country Counterfeit a Diversion Theft b Incidents
3 India 95 23 0 118
4 Brazil 47 47 3 97
5 Colombia 62 32 0 94
6 Japan 81 0 0 81
7 United Kingdom 61 17 2 80
8 Germany 64 10 0 74
9 Uzbekistan 35 37 0 72
10 Pakistan 64 7 0 71
SOURCE: PSI data shared with the committee, Thomas Kubic, PSI-Inc., July 11, 2012.
34 12
70
Fake product,
fake packaging
Fake product,
legitimate packaging
Legitimate product,
145 549 fake packaging
Undeclared active
ingredient
North America
$358
Europe
$329
Latin America
16%
Middle East 5%
Africa 3%
Asia
51%
North America
10%
Europe
15%
19972 (Devine and Jung, 2012). Solid oral dosage forms (pills and tablets)
are the most commonly investigated product types (FDA, 2011). Zyprexa,
Viagra, Lipitor, Zoloft, and Risperdal are the top 5 brand-name products
implicated in the 10 highest-volume cases (FDA, 2011). Problems with
individual criminals are more common than problems with negligent busi-
nesses; the FDA’s 2003-2008 review found that 86 percent of criminal in-
vestigations were of individual suspects, and 14 percent were of companies
(FDA, 2011).
2 The FDA refers to these as counterfeit drug cases. The agency uses a definition of coun-
terfeit that includes both falsified and substandard as this report defines them. See Table 1-2.
Number of
Operation Countries
Name Participating Duration Year Results
Number of
Operation Countries
Name Participating Duration Year Results
Case Reports
A great deal of literature on falsified and substandard drugs describes
problems that emerge only after patients have been harmed (Ravinetto et
al., 2012). These reports do not set out to comment on the regional burden
of poor drug quality or trends in compromised products, but they are useful
for other reasons. Many of these stories receive significant media attention,
encouraging public interest in the problem. Case studies also give under-
standing of the social and environmental conditions that foster problems
with falsified and substandard drugs (Pew, 2012).
Patient case studies are a common type of incidental investigation.
For example, the rapid deterioration and death of a Burmese patient with
uncomplicated malaria triggered a drug analysis that found the medicines
used to treat him were both falsified and substandard (Newton et al., 2006).
A postmortem investigation in a previously healthy, 58-year-old Canadian
woman found that her death was from acute metal poisoning from a variety
of falsified and substandard drugs, many of them antianxiety and antide-
pressive medications she bought from the internet (Solomon, 2007).
Individual deaths can trigger drug quality investigations; mass causali-
ties are clearly more likely to rouse suspicion. Chapter 2 describes one such
incident, when a Panamanian physician reported on a spike in cases of
acute renal failure, accompanied by neurological dysfunction, abdominal
symptoms, urinary irregularities, anorexia, and fatigue (Rentz et al., 2008).
A case-control investigation found diethyelene glycol poisoning to be the
cause of the outbreak (Rentz et al., 2008). Later investigations, including
a Pulitzer Prize–winning New York Times series, implicated falsified ingre-
dients from China in an international poisoning crisis (Bogdanich, 2007;
Rentz et al., 2008).
Newspaper reports and other gray literature sources also contain a
wealth of information about drug quality problems. Monitoring this litera-
ture is a valuable way to follow what drugs are compromised and where.
The U.S. Pharmacopeia’s Media Reports on Medicine Quality Focusing on
Convenience Samples
A convenience sample is a no-probability sample chosen for its acces-
sibility to researchers, not from an a priori sampling frame. Research on
drug quality often uses convenience samples of pharmacies or dispensaries.
Convenience studies are logistically simpler than probability-based stud-
ies and can be less expensive (Newton et al., 2009). Although useful for
identifying problems, results of these studies cannot accurately estimate the
population prevalence of poor-quality drugs. They do, however, suggest
signals for further research (Newton et al., 2009). This section presents the
results of some key convenience studies and review papers.
Antimicrobial Drugs
Antimicrobial drugs treat bacterial, viral, fungal, and parasitic diseases.
There are considerable data to suggest that antimicrobial drug quality, par-
ticularly the quality of antibiotics and antimalarials, is a problem in low-
and middle-income countries. In 2007 Kelesidis and colleagues conducted
a comprehensive literature review on antimicrobial drug quality, reviewing
3 Including, but not limited to, amoxicillin, ampicillin, chloroquine, rifampicin, and
co-trimoxazole.
13%
Registered
Unregistered
8%
24%
convenient vendor. For these reasons, they are often the target of criminals
and unscrupulous manufacturers.
A WHO study of antimalarial drug quality in six African countries4
used a stratified convenience sample (WHO, 2011a). In the spring of 2008,
regulatory agency staff in the six countries collected samples from central
stores, licensed outlets, and unlicensed markets (WHO, 2011a). Investi-
gators screened all samples in the field and sent a subset for full quality-
control testing (WHO, 2011a). Investigators found unregistered medicines
least often at the central distribution level (see Figure 3-4). Quality analysis
on a subset of products found no evidence that the unregistered drugs were
of lower quality than the registered ones (see Figure 3-5) (WHO, 2011a).
Field screening of 893 samples detected ingredient failures in 12 percent
(WHO, 2011a). Of the 267 samples selected for full quality-control testing,
28 percent failed (see Figure 3-6) (WHO, 2011a).
The WHO study findings are cause for concern, especially as they sam-
pled heavily from national central medicine stores, the most controlled set-
ting. A recent review paper includes some higher estimates of poor-quality
antimalarial drugs (Nayyar et al., 2012). The review included 28 published
and unpublished studies, mostly (n = 22) from convenience samples, but
also 7 that included some type of randomized design (Nayyar et al., 2012).
90
90
(%)
testing(%) 80
80 Domestic
Domestic
laboratorytesting
Imported
Imported
70
70
30%
30% 20%
20%
QClaboratory
60
60
50
50
ratesininQC
40
40
Failurerates
30
30
Failure
20
20
10
10
0
0
Registered
Registered Unregistered
Unregistered
(n=227)
(n=227) (n=40)
(n=40)
FIGURE
FIGURE 3–5
3–5 Quality
Quality failures
failures of
of registered
registered and
and unregistered
unregistered
antimalarials
antimalarials in
in 267
267 samples.
samples.
NOTE: QC = quality control.
NOTE: QC = quality control.
SOURCE: WHO, 2011a.
SOURCE: WHO, 2011a.
31
31
Compliant
Compliant with
with
45 specifications
specifications
45
Noncompliant
Noncompliant
with specifi
with specifications—
cations—
nonextreme
nonextreme deviations
deviations
Noncompliant
Noncompliant
with
with specifi
specifications—
cations—
extreme
extreme deviations*
deviations*
191
191
FIGURE
FIGURE 3–6 3–6 Results
Results of
of full
full quality-control
quality-control analysis
analysis inin 267
267 samples.
samples.
** In this survey, extreme deviations were defined as a deviation by at least 20% from
In this survey, extreme deviations were defined as a deviation by at least 20% from
the declared content of one or more active ingredients, and/or dissolved percentage
the declared content of one or more active ingredients, and/or dissolved percentage
of one or more active ingredients less than the pharmacopoeial limit (Q) minus 25%.
of one or more active ingredients less than the pharmacopoeial limit (Q) minus 25%.
SOURCE: WHO, 2011a.
SOURCE: WHO, 2011a.
resentative cross section of the market. Such studies are uncommon (Seear,
2012). The expense of required assays, discussed further in Chapter 6, is
one barrier, but a large part of the problem is logistical. The first step in
drawing a systematic random sample of drugs is identifying the sampling
frame, the list of every drug vendor in a given area. In developed coun-
tries, registered pharmacies and dispensaries are the only place most of the
population gets medicine. In low- and middle-income countries, however,
there is often an extensive pharmaceutical gray market. Identifying all the
vendors is difficult and can be further complicated by the blurry lines be-
tween licit and illicit commerce (Seear et al., 2011). Health workers may
supplement their incomes by selling medicine informally (Peters and Bloom,
2012); peddlers may trade medicines occasionally, along with any number
of dry goods, at bazaars and flea markets. Without formative research to
catalogue the sampling frame, research on medicines quality is vulnerable
to bias.
There is also opportunity for bias in sample collection. Samples should
be bought by patient actors, local study staff posing as shoppers who
conceal from the vendor that they are working on an epidemiological
investigation.
Without taking steps to protect study validity, the researchers risk wast-
ing time and money on a study that does not produce reliable estimates.
For example, in 2009 the Indian government conducted a massive survey of
drug quality across the country, estimating that only 0.04 percent of drugs
are substandard (CDSCO, 2009). Questions about the methodological rigor
of the survey, particularly the choice of sampling frame and methods for
sample collection, have called these results into question both within India
and internationally (Bate, 2009, 2010; Pandeya, 2009).
The committee supports the guidelines on field surveys of medicine
quality that Newton and colleagues proposed in March 2009 (Newton
et al., 2009). They provide a standard protocol for collecting medicines
samples and concrete advice on sampling techniques (Newton et al., 2009).
More research adhering to the checklist in Table 3-8 would allow for a
better understanding of the burden of falsified and substandard drugs, and
it would facilitate valid comparisons of the problem among countries and
over time.
in countries where there are not sufficient systems to monitor drug safety
and adverse events. A 2012 IOM report called for greater international
investment in low- and middle-income-country surveillance systems (IOM,
2012). The committee agrees with this report, especially the call for tech-
nical support for surveillance tools and protocols (IOM, 2012). National
surveillance systems should work to detect signals of substandard and falsi-
fied drugs. Incorporating pharmacovigilance into the broader public health
surveillance system will help ensure the system’s survival.
Methods
continued
Results
Missing samples 15 The reasons why any outlets chosen for sampling
did not furnish a sample. Do these outlets differ
systematically from those in which samples were
obtained?
State company and 18 If the names of companies and addresses are not
address as given on given, give a reason as to why this information is
packaging not provided.
Sharing data with the 19 Whether the data shared with the appropriate
regulatory authority regulatory agency
Discussion
Other information
5
Cambodia, Croatia, Georgia, Indonesia, Kyrgyzstan, Malaysia, the Philippines, Russia,
Ukraine, and Vietnam.
Suspect product details Full details of all suspect products (up to 30 per report)
who finds an illegitimate drug completes the form and sends it to WHO
headquarters, copying regional and country offices. At the WHO, the
spreadsheet data populate a master database. Receipt of the form triggers
a follow-up contact from a WHO investigator, who queries the reporter on
the drug’s packaging, registration, and physical and chemical attributes and
if the incident might be connected to other criminal activity (personal com-
munication, Michael Deats, WHO, October 12, 2012). Table 3-9 shows
more detail about the data collected in the rapid alert form.
Regulators from the 10 pilot countries testing the rapid alert form
and incident investigation system had training on the system in September
2012. The pilot testing ran until December 2012 (personal communication,
Michael Deats, WHO, October 12, 2012). Already the system has allowed
investigators to link incidents in multiple countries. Eventually, regulators
may link falsified and substandard reporting to national pharmacovigilance
systems, which would give more depth to information about lack of efficacy
(personal communication, Michael Deats, WHO, October 12, 2012).
The lack of consensus on how to define falsified and substandard
medicines has held back all public action on the topic, even surveillance.
The WHO project gets around this problem by recording problems with
medicines and by not attempting to make the observed problems fit the
confusing and contradictory national definitions of substandard, falsely
labeled, spurious, counterfeit, etc.
Patient reporting triggers most investigations in the pilot countries.
This depends on motivated and knowledgeable patients, and a longer-term
improvement to the project might aim to increase reporting from health
workers. However, Michael Deats, the WHO technical officer in charge of
the program, explained that while he was working as a regulator in Brit-
ain, some of his best leads came from patients, especially ones who take
medication for chronic disease. One informant was a patient in the habit of
rubbing his pill in his hand before taking it and was immediately suspicious
when the color rubbed off (personal communication, Michael Deats, WHO,
November 11, 2012). A similar signal came from a patient accustomed to
cutting his pills in half, who noticed irregular friability when he cut them
(personal communication, Michael Deats, WHO, November 11, 2012).
The committee recognizes that building surveillance systems will be
challenging in many countries. Nevertheless, taking steps to establish a
system or to strengthen the existing system is a reasonable first step in
most of the world. Developing countries may benefit from momentum for
building surveillance among donor governments and international orga-
nizations (IOM, 2012). The WHO is also encouraging action in the same
direction. At a meeting in November 2012, representatives of 200 member
states agreed to develop instruments to more accurately measure the burden
of substandard and falsified drugs (WHO, 2012c). To this end, the WHO
capacity-building project is testing and developing surveillance tools spe-
cifically for use in low- and middle-income countries. As the pilot project
goes on, the regulators and WHO staff may identify revisions that make
the protocol more accessible in poor countries.
The WHO monitoring format advances understanding of the scope
of the problem without depending on common variable names. The com-
mittee sees the WHO rapid alert system as an uncommonly thorough and
precise tool for data collection. These data will inform tailored drug quality
programs. For example, if the data indicate that substandard medicines are
the main drug quality problem in one part of the world, then better regula-
tion of manufacturers can do much to improve the problem. Similarly, if
it becomes clear that a country has a problem with diverted medicines in
commerce, then some of the distribution chain improvements presented in
Chapter 5 would enhance the national drug safety program. Consistent
use of this rapid alert form and eventually linking it to national pharmaco-
vigilance systems would advance international discourse and give a more
nuanced understanding of the extent and type of falsified, substandard, and
unregistered medicines that circulate around the world.
Therapeutic
Country Action Justificationa Quantity Category Drugs Value Arrests
Afghanistan Destroyed Substandard & 32 tons Unknown Unknown $- 0
expired
continued
111
TABLE 3–5 Continued
112
Therapeutic
Country Action Justificationa Quantity Category Drugs Value Arrests
China Seized Counterfeit 800 boxes, 121 types of Unknown Unknown $253,440 0
drugs
Colombia Seized-arrests Counterfeit 150,000 dosage units Cytostatic, Cancer & HIV/AIDS $- 22
anti-infective
contraband
Israel Seized-arrests Counterfeit 230,000 dosage units Metabolism, Weight loss & ED $- 1
genitourinary
continued
TABLE 3–5 Continued
114
Therapeutic
Country Action Justificationa Quantity Category Drugs Value Arrests
Mozambique Seized-arrests Diverted Anti-infective HIV/AIDS $5,200 9
Portugal Seized Unknown 133,000 dosage units Genitourinary, ED & weight loss $- 0
metabolism
Spain Seized-arrests Counterfeit & 710,000 dosage units Hormones, Steroids & ED $- 26
illegal genitourinary
continued
115
TABLE 3–5 Continued
116
Therapeutic
Country Action Justificationa Quantity Category Drugs Value Arrests
Thailand Seized-arrests Counterfeit Metabolism Weight loss $969,932 10
West Africa Seized-arrests Counterfeit & illicit 10 tons Unknown Unknown $- 100
NOTES: $- = value unknown; CNS = central nervous system; ED = erectile dysfunction; HGH = human growth hormone; UAE = United Arab Emirates; UK = United Kingdom.
a As noted in footnote a under Table 3-2, PSI uses the term counterfeit broadly, in accordance with a WHO definition. See page 89. The organization’s open-source review
Characteristics Pharmaceutical
of Counterfeit or Companies Involved or
Results Substandard Drugs Country of Manufacture
Two lots of capsules and No active ingredient, wrong China
one lot of granule had no ingredient, no color change in
active ingredients imitating sulphuric acid reaction
erythromycin ethylsuccinate
capsule and azithromycin
granule, respectively, one lot of
erythromycin tablets imitating
roxithromycin tablets, and two
lots of meleumycin capsule
imitating midecamycin capsule.
At the time of purchase, Poor in vitro drug release TPI (metronidazole), Holden
the drug content of all the profiles and dissolution Medica (metronidazole),
formulations was within the (four formulations [three Labophar (TMP–SMX, sulfadoxine
limits recommended by the sulfamethoxazole/trimethoprim and pyrimethamine), Shalina
USP 24, but after 6-month and one sulfadoxine/ (sulfamethoxazole), ACE (TMP–
storage, the drug content pyrimethamine combination]). SMX). Rwanda and Tanzania.
of one sulfamethoxazole/ Some of the formulations tested
trimethoprim was found to be were not stable in terms of drug
substandard. Immediately after content (one sulfamethoxazole/
purchase, four formulations trimethoprim) and dissolution
(three sulfamethoxazole/ (three metronidazole
trimethoprim and one formulations), upon storage
sulfadoxine/pyrimethamine under simulated tropical
combination) failed the USP 24 conditions.
dissolution test. Except for three
metronidazole, dissolution tests
performed after 6 months of
storage under simulated tropical
conditions showed that drug
release remained within the USP
24 recommended values. In total,
24% of the sampled formulations
(8/33) failed the dissolution test.
The percentage of substandard No active ingredient (ampicillin 24% (23 out of 97) of the drugs
drugs decreased significantly and tetracycline), too little from Lao factories, 17% (24 out of
from 46% to 22% (66 out of (ampicillin) or too much 143) of the drugs from Thailand
300) between 1997 and 1999 (tetracycline) active ingredient, and 47% (17 out of 36) of the
(P < 0.001). Substandard weight variation outside drugs of unknown origin were
ampicillin and tetracycline were pharmacopoeial limits substandard.
reduced significantly from 67%
to 9% and from 38% to 12%,
respectively (P < 0.001). In total,
3% versus 1% contained no active
ingredient, 12% versus 4% had
too little or too much active
ingredient and 35% versus 14%
had weight variation outside
pharmacopoeial limits.
continued
Characteristics Pharmaceutical
of Counterfeit or Companies Involved or
Results Substandard Drugs Country of Manufacture
Among the 21 different specialty Inappropriate labeling, expired Lombisin, Unicorn, China
products, only 3 displayed the drug, no active ingredient (chlortetracycline), Yong Fong,
official “registered” label. Three (chlortetracycline), reduced Myanmar (co-trimoxazole),
drugs were expired and the active ingredient China (benzylpenicillin),
expiration date was not available Helm Pharmaceutical GMBH,
for six others. One product did Hamburg, Germany (benzathine
not contain the active drug benzylpenicillin), Cadila
declared (chlortetracycline) and Lab, Ahmedabad, India, Dr.
did not show any in vitro activity Reddy’s Lab, Bollaram, India
against bacteria. Seven of 21 (ciprofloxacin), Remedica Ltd,
products (33%) did not contain Limassol, Cyprus (erythromycin
the stated dosage (one more and doxycycline), ICPA Lab Ltd,
than stated dosage and six less Bombay, India (TMP–SMX)
than stated dosage). The highest
deficit observed was 48% in two
products (co-trimoxazole and
benzylpenicillin). The dosage
was not available for five drugs.
As a result, only 8 of 21 products
(38%) did not contain the stated
dosage of active drug.
For all groups of drugs, Zero (metronidazole Most drugs that failed to pass
antibacterial and antituberculosis suspension), or very low the test were manufactured in
agents, more than 50% failed (ampicillin [syrup and countries such as China, Holland,
to comply with specifications. capsules], amoxicillin [syrup], India, Malaysia, Nigeria, Pakistan,
For some individual drug pyrimethamine and sulfadoxine Romania, Switzerland, and the
preparations, all samples [syrup], cloxacillin [syrup and United Kingdom or were of
assayed were within capsules], and ampicillin and unknown origin.
pharmacopoeial limits. These cloxacillin [syrup and capsules])
included trimethoprim and quantities of active ingredient
sulfamethoxazole tablets. No
metronidazole suspension met
pharmacopoeial specifications.
Several antibacterial preparations
contained very low quantities of
active ingredient (ampicillin and
amoxicillin 24% to 40%), and for
five metronidazole suspension
preparations, no active ingredient
was detected.
continued
Stenson et al. 366 Laos Ampicillin (tablets Three tests were used: identity,
(1998) and capsules) and assay, and measurement of
tetracycline (tablets weight variation. The identity
and capsules) was confirmed by TLC, UV, and
color reactions. Titrimetric,
UV, and HPLC methods were
used for assay. Potentiometric
titration method.
Nazerali and 789 samples Zimbabwe Injectable Not reported. Drug quality was
Hogerzeil of 26 benzylpenicillin, measured by level of active
(1998) brands of amoxicillin, ampicillin, ingredient as percentage
13 essential doxycycline, phenyl- of stated content and by
drugs methoxypenicillin, and compliance (pass/fail) with
tetracycline assay standards of the British
Pharmacopoeia. Drug stability
was measured by comparing
mean assay values at central
and rural levels and by paired
analysis of central and rural
samples of the same batch.
Characteristics Pharmaceutical
of Counterfeit or Companies Involved or
Results Substandard Drugs Country of Manufacture
All 13 FDCs contained the stated Reduced (rifampicin) or excess Not reported
drugs. However, four (31%) were (rifampicin and pyrazinamide)
substandard, including two (15%) content of active ingredient
with low rifampicin content, one
(8%) with excessive rifampicin
and one (8%) with excessive
pyrazinamide. Both FDCs with
low rifampicin contained four
drugs and failed TLC screening.
The FDC with excessive
rifampicin was not detected by
TLC screening. Using UV as the
gold standard, the sensitivity of
TLC for low rifampicin was 2/2
(100%) and the specificity was
9/10 (90%).
12 (3.3%) out of the 366 drugs No active ingredient (ampicillin), Out of the 61 samples that were
contained no active ingredient, low concentration of active found to contain no active
42 (11.5%) had levels of active ingredient (ampicillin and ingredient or to be substandard
ingredient outside acceptable tetracycline), weight variation according to the assay, only
limits in assay, 128 (35.0%) had outside pharmacopoieal limits 37 were labeled. Of these, 20
excessive weight variation and (all), bad retail management originated from Laos, 5 from
4 (1.1%) were managed badly in (ampicillin) Thailand and 3 from Vietnam,
the pharmacy, 67% of ampicillin whereas 9 were of unknown
samples and 38% of tetracycline origin.
had bad quality.
Poor initial quality accounted for Reduced level of active Not reported
problems in injectable ampicillin ingredient
(2/10 central samples failed,
with 87% and 91% content). An
aqueous formulation of injectable
procaine benzylpenicillin showed
moderate instability with 4%
(1% to 6%) loss after 4.3 months
but the assay remained within
pharmacopoeial limits.
36% of samples from Nigeria and Zero (amoxicillin) or very The countries of origin were
40% of samples from Thailand low (amoxicillin, TMP–SMX, India, Italy, Nigeria, Pakistan,
were substandard with respect ampicillin–cloxacillin) quantities Thailand, and the United
to British Pharmacopoeia limits. of active ingredient Kingdom, but no patterns
One amoxicillin sample from emerged with respect to quality
Nigeria contained no active of product and country of origin.
ingredient at all.
continued
NOTE: The complete references for the studies cited in this table can be found in Kelesidis et al., 2007.
Kelesidis and colleagues use the term counterfeit broadly, the way this report uses the term falsified. See
page 23. FCIS = fast chemical identification test; FDC = fixed-dose combination; HCI = ondansetron hydro-
chloride; HPLC = high-performance liquid chromatography; INH = isoniazid; LC = liquid chromatography;
MIC = minimum inhibitory concentration; RIF = rifampicin; TLC = thin layer chromatography; TMP-SMX =
trimethoprim-sulfamethoxazole; USP = U.S. Pharmacopeia; UV = ultraviolet spectrophotometry.
SOURCE: Kelesidis et al., 2007. Reprinted with permission from Oxford University Press.
Characteristics Pharmaceutical
of Counterfeit or Companies Involved or
Results Substandard Drugs Country of Manufacture
A significant proportion of a It appeared that active Not reported
variety of drug preparations was ingredients had been
substandard (27%). Ten brands deliberately kept below the
of ampicillin were found to be required levels.
substandard in this study and
8 of them had already been
assessed as substandard by the
regulatory authorities. This was
also true of the two brands of
co-trimoxazole suspension found
to be substandard.
Date of Sample
Location Collection Drug Tested Method of Testing
Southeast Asia
Sub-Saharan Africa
Public and Convenience 303 103/303 (34%); 99/303 (33%) 99/303 (33%)
private 99/103 (96)†
pharmacies
and outlets
and facilities
Public and Convenience 451 122/451 (27%); 72/111 (65%) 88/111 (79%)§
private 30/122 (25%)†
pharmacies,
and outlets
and facilities
continued
Date of Sample
Location Collection Drug Tested Method of Testing
continued
Date of Sample
Location Collection Drug Tested Method of Testing
continued
Date of Sample
Location Collection Drug Tested Method of Testing
NOTE: DFID = Data are n/N (%), unless otherwise indicated. Samples failing chemical assay analysis might
have failed packaging analysis; HPLC = high-perfomance liquid chromatography; NA = not applicable;
NS = not specified.
* Falsified is used as a synonym for counterfeit.
† Samples with no active pharmaceutical ingredient.
REFERENCES
Amin, A. A., R. W. Snow, and G. O. Kokwaro. 2005. The quality of sulphadoxine-
pyrimethamine and amodiaquine products in the Kenyan retail sector. Journal of Clinical
Pharmacy and Therapeutics 30(6):559-565.
Bate, R. 2009. India’s counterfeit claims on counterfeit drugs. http://www.american.com/
archive/2009/september/indias-counterfeit-claims-on-counterfeit-drugs (accessed Septem-
ber 24, 2012).
———. 2010. Delhi’s fake drug whitewash. Wall Street Journal, September 2.
Bate, R., L. M. K. Hess, and J. M. A. Attaran. 2012. Anti-infective medicine quality: Analysis
of basic product quality by approval status and country of manufacture. Research and
Reports in Tropical Medicine 3:57-61.
Bate, R., L. Mooney, and K. Hess. 2010. Medicine registration and medicine quality: A pre-
liminary analysis of key cities in emerging markets. Research and Reports in Tropical
Medicine 1:89-93.
BMI (British Monitor International). 2012. Global pharmaceutical sales. Data available
from http://www.citibank.com/transactionservices/home/about_us/online_academy/docs/
JDavis.pdf (accessed April 10, 2013).
Bogdanich, W. 2007. F.D.A. tracked poisoned drugs, but trail went cold in China. New York
Times, June 17.
CDSCO (Central Drugs Standard Control Organization). 2009. Report on countrywide survey
for spurious drugs. New Delhi: CDSCO.
Cockburn, R., P. N. Newton, E. K. Agyarko, D. Akunyili, and N. J. White. 2005. The global
threat of counterfeit drugs: Why industry and governments must communicate the dan-
gers. PLoS Medicine 2(4):e100.
Devine, J., and C. Jung. 2012. FDA efforts to build supply chain integrity. Paper presented
at Committee on Understanding the Global Public Health Implications of Substandard,
Falsified, and Counterfeit Medical Products: Meeting 2, Washington, DC, May 10.
Evans, L., V. Coignez, A. Barojas, D. Bempong, S. Bradby, Y. Dijiba, M. James, G. Bretas,
M. Adhin, N. Ceron, A. Hinds-Semple, K. Chibwe, P. Lukulay, and V. Pribluda. 2012.
Quality of anti-malarials collected in the private and informal sectors in Guyana and
Suriname. Malaria Journal 11(1):203.
FDA (U.S. Food and Drug Administration). 2009a. Inspections, compliance, enforcement,
and criminal investigations: Mission. http://www.fda.gov/ICECI/CriminalInvestigations/
ucm123027.htm (accessed November 12, 2012).
———. 2009b. Inspections, compliance, enforcement, and criminal investigations: What
OCI investigates. http://www.fda.gov/ICECI/CriminalInvestigations/ucm123062.htm (ac-
cessed November 12, 2012).
———. 2011. FDA conducts preliminary review of agency’s diversion and counterfeit criminal
case information. Washington, DC: FDA Office of Criminal Investigations.
Interpol. 2010a. Operation Mamba. http://www.interpol.int/Crime-areas/Pharmaceutical-
crime/Operations/Operation-Mamba (accessed November 12, 2012).
———. 2010b. Operation Storm. http://www.interpol.int/Crime-areas/Pharmaceutical-crime/
Operations/Operation-Storm (accessed November 6, 2012).
———. 2011a. Interpol launches global campaign against fake medicines with powerful Af-
rican voices. http://www.interpol.int/News-and-media/News-media-releases/2011/PR090
(accessed August 20, 2012).
———. 2011b. Operation Cobra. http://www.interpol.int/Crime-areas/Pharmaceutical-crime/
Operations/Operation-Cobra (accessed November 12, 2012).
———. 2012a. Operations. http://www.interpol.int/Crime-areas/Pharmaceutical-crime/
Operations (accessed November 6, 2012).
———. 2012b. Operations: Operation Pangea. http://www.interpol.int/Crime-areas/
Pharmaceutical-crime/Operations/Operation-Pangea (accessed October 22, 2012).
The committee recognizes that the factors that encourage the prolifera-
tion of substandard and falsified medicines are different but overlapping.
In general, neglect of good manufacturing practices, both accidental and
deliberate, drives the circulation of substandard drugs, while falsification
of medicines has its roots in crime and corruption. Both types of products
circulate because of the erratic supply and constant demand for medicines
and weaknesses in the regulatory system. An inaccurate or inadequate
understanding of the problem among health workers and the public con-
tributes to the problem.
137
run in accordance with best practices does not need to be the most tech-
nologically advanced or use state-of-the-art equipment, but there are costs
to bring a factory up to standard, train staff on appropriate protocols, and
observe them consistently. There are many exemplary manufacturers in de-
veloping countries that observe international best practices. There are also
many that do not, but they operate anyway, either because the regulatory
authority is unaware of the problem, or because regulators are under pres-
sure to ignore it in the name of promoting industry.
Quality control is a part of good manufacturing practices sometimes
neglected in developing countries. The WHO compendium on pharma-
ceutical manufacture describes the importance of having quality-control
staff who are separate from production staff, working in an independent
department (WHO, 2007b). A manager trained in quality control should
supervise this department and run an equipped quality-control laboratory
(WHO, 2007b). Quality-control staff should verify that everything that
is a part of the factory’s product, including packaging, starting materials,
intermediates, and finished products, meets requirements (WHO, 2007b).
They may also do internal inspections and quality audits and evaluate the
quality controls used by their suppliers (WHO, 2007b). The majority of the
pharmaceutical industry in the poorest countries only formulates and re-
BOX 4-1
The Medicines Manufacturing Process
Drugs are made with four or five main steps between the raw ma-
terials and the packaged final formulation (Figure 4-1). Medicines manu-
facture in the poorest countries is generally limited to the last steps in
this process: formulation and packaging (Bumpas and Betsch, 2009; IFC,
2007). Of the 46 countries in sub-Saharan Africa, about 80 percent have
local pharmaceutical industries, but only South Africa produces active
ingredients (Bumpas and Betsch, 2009). South Africa alone accounts for
70 percent of the region’s medicines production (Bumpas and Betsch,
2009).
The firms that make final formulations in developing countries buy
excipients and active ingredient from chemical suppliers abroad, mostly
from China and India. China supplies about 43 percent of the world’s
active ingredients for anti-infective medicines and exports 77 percent of
the active ingredient made in the country, a $4.4 billion industry. India
exports 75 percent of the $2 billion worth of active ingredients it pro-
duces (Bumpas and Betsch, 2009).
COMPONENTS
Sterile
API
API
innovator and generic, operate on a scale that allows them to recover the
costs of running high-quality factories. This is not true for many smaller
manufacturers in developing countries. In India, for example, large pharma-
ceutical companies supply medicines and vaccines of the highest quality to
every country in the world, but thousands of small manufacturers struggle
to implement quality-assurance and quality-control procedures (Kaplan and
Laing, 2005). A World Bank study found that one-tenth of Indian regis-
tered pharmacies report substandard medicines, most of them coming from
small- and medium-sized producers (Kaplan and Laing, 2005). Because the
registered pharmacy is the most strictly regulated medicines outlet in India,
the proportion of substandard medicines sold in the informal market is
presumably much higher. The problem is not limited to India. In a survey
of antibiotic quality in Indonesia, investigators found 89 percent of samples
of one local company’s cotrimoxazole were substandard (Hadi et al., 2010).
Critics of local manufacture have cited these problems as reasons
against pharmaceutical manufacturing in low- and middle-income coun-
tries (Ahmed, 2012; Bate, 2008). This may be a short-sighted argument.
Domestic manufacture of medicines is an important part of health and
industrial policy in many countries. Governments are understandably eager
to ensure a safe drug supply for their population. In theory, locally made
products could be cheaper because of lower shipping costs incorporated
into the final price (Kaplan et al., 2012). Manufacturing medicines also
gives people jobs and facilitates technology transfer (Wilson et al., 2012).
Companies that start out packaging only finished drugs will slowly develop
the trained workforce needed for more complicated secondary and primary
manufacturing.
Initial capital investments and infrastructure problems stand between
quality medicines and many small- and medium-sized medicine manufactur-
ers. There are companies in developing countries that want to meet inter-
national quality standards and buy from reliable suppliers, but they fail to
do so for reasons beyond their control. Governments alone cannot supply
the technical depth or money to fix these problems (Wilson et al., 2012).
The private sector must be involved. The International Finance Corporation
(IFC) and the Overseas Private Investment Corporation (OPIC) can work
to encourage private sector growth in developing countries. With the initial
investments made, governments can take on the more manageable role of
encouraging partnerships with foreign manufacturers.
Tiered Production
In practice it is difficult to distinguish the quality failures that are to
blame on a manufacturer’s inability to meet international best practices
from those which come from a decision to cut corners and produce inferior
products for poorly regulated markets. When a producer capable of meet-
ing international standards fails to do so consistently and only in product
lines sold to the poor, one may conclude that the noncompliance is part of
a more insidious system.
Rich countries enforce high quality standards for medicines, and manu-
facturers recognize the need to use good-quality ingredients and good
manufacturing practices to sell in these markets. United Nations (UN)
agencies and the larger international aid organizations will also refuse to
do business with companies that cannot meet stringent regulatory author-
ity quality standards. Manufacturers are aware, however, that low- and
middle-income countries are less likely to enforce these standards. Some
companies exploit this and produce drugs of lower quality for the loosely
regulated markets (Caudron et al., 2008). When a manufacturer produces
medicines of inferior quality for less exacting markets, it is known as tiered
or parallel production (Caudron et al., 2008; World Bank, 2007).
Tiered production is a complicated problem, in part because some
kinds of tiered production are legal. International manufacturers may sup-
ply to multiple markets which use different legal product quality standards.
For example, the British Pharmacopoeia monograph for amoxicillin gives
no dissolution standard (British Pharmacopoeia, 2012); the U.S. Pharma-
copeia does (USP-NF, 2010). Assay limits may also be different, making a
product illegal by one pharmacopeia but legal by another. A manufacturer
may supply to one country that stipulates a uniformity of dosage at 90-120
percent of declared dosage and to another country that stipulates 85-115
percent, for example. Both these standards aim to correct for the fact that
drugs such as antibiotics degrade quickly, making a high initial dose accept-
able. However, manufacturers could technically aim to fill only the lower
bound of the dosage requirements and be within the letter of the law. A
study of amoxicillin samples in Arab countries found that most samples’ ac-
tive ingredient concentrations were bordering the U.S. Pharmacopeia lower
limit (Kyriacos et al., 2008). The authors admitted, however, that many of
the problematic samples would have been judged acceptable by the wider
British Pharmacopoeia standard (Kyriacos et al., 2008).
Participants at the public meetings for this study mentioned concerns
with parallel production, but evidence for it is largely anecdotal. There is
reason to suspect tiered manufacturing when the dose of active ingredient
is consistently lower, never higher, than the label claim (Bate et al., 2009b).
Drugs, especially tablets, of less than half the labeled potency before the ex-
piry date are particularly dubious. In a hospital dispensary in rural Nepal,
a bottle of pediatric amoxicillin from a WHO-certified producer with
many obvious labeling and packaging defects also suggests either parallel
manufacturing or diversion, a problem discussed in Chapter 5 (Brhlikova
et al., 2007).
Tiered manufacturing is a rising problem in emerging manufacturing
nations. A 2006 Lancet report described a shift in Russia from most bad
medicines being falsified drugs made “in basements and small backroom
enterprises” to ones coming from legitimate companies running “a ‘night
shift’ to produce extra quantities of a certified drug that does not pass
quality control, or sophisticated copies of well-known drugs . . . often with
reduced levels of expensive active ingredients” (Parfitt, 2006, p. 1481). The
United Nations Office on Drugs and Crime (UNODC) described a similar
case in India. The U.S. Food and Drug Administration (FDA) revoked
market authorization from an Indian drug manufacturer found to be pro-
ducing antibiotics with no active ingredients (UNODC, 2010). After losing
its license, “the factory continued to operate at night, until an evening
raid by police uncovered an underground cellar in the factory, where exact
look-alikes of several popular, fast-moving, high-cost medicines were being
manufactured, most of which contained no active ingredient” (UNODC,
2010, p. 187).
Jiben Roy reported on a similar case: A Bangladeshi company delib-
erately kept the active ingredients in paracetemol, ampicillin, and cotri-
moxazole below the labeled concentrations after repeated warnings from
the regulatory authority (Roy, 1994). In the same paper he attributed the
manufacturer’s quality failures in their cheaper product lines to negligence
alone. Their B-vitamins, for example, contained the proper ingredients,
but in erratic doses (Roy, 1994). This paper was able to make distinctions
between the deliberate quality failures and negligence because the author
had close knowledge of the manufacturer and its history. Usually only the
national regulatory authority could have the information needed to make
this distinction. In many countries, even the regulatory authority would
not have that information or the political will to act on it (Christian et al.,
2012b).
Pinpointing cases of deliberate tiered manufacturing is difficult to do,
though it is easier to see practices that allow it happen. Poor oversight of
ance System for procurement lays out the steps necessary for efficient and
open procurement of the best-quality medicines possible (WHO, 2007a).
WHO Stringent regulatory authority WHO Good Manufacturing Dossier reviews and site inspections;
approval; WHO prequalification; Practices (GMP) and WHO recognition of approval by stringent
or expert review panel Model Quality Assurance System regulatory agencies (FDA, EMA,
recommendation (MQAS) for procurement etc.)
agencies
Global Fund WHO prequalification; stringent WHO GMP and WHO MQAS Expert review panel dossier reviews
regulatory authority approval; or (Global Fund, 2012) and inspections (Daviaud and Saleh,
expert review panel 2010)
recommendation (Daviaud and
Saleh, 2010)
European Commission’s Quality-assurance guidelines are “Every activity in the procurement None
Humanitarian Aid based on WHO prequalification and process should be carried out
Department (ECHO) model quality assurance system according the WHO standards
Countering the Problem of Falsified and Substandard Drugs
USAID FDA or other stringent regulatory WHO GMP; WHO MQAS; U.S. Dossier reviews and inspections and
approval; WHO manufacturer Pharmacopeia; and International product quality assessments
and product prequalification; Pharmacopoeia
and Supply Chain Management
System procurement agency
prequalification
Department for International Procurement of commodities are Stringent regulatory approval; Third-party compliance is monitored
Development (DFID) done through third parties such national drug regulatory authority through normal channels of
as multilateral organizations, approval; WHO prequalification; oversight such as boards, steering
partnerships, or procurement and expert review panel approval committees and program reviews
agencies. DFID relies on the quality- (DFID, 2012) (personal communication, James
assurance policies of those third Droop, DFID, October 17, 2012).
parties (personal communication,
James Droop, DFID, October 17,
2012).
Countering the Problem of Falsified and Substandard Drugs
The Bill & Melinda Gates WHO prequalification of Grantees and partners use Funds WHO to conduct dossier
Foundation grantees and partners (personal WHO GMP and the WHO MQAS reviews and required inspections
communication, Vincent (personal communication, Vincent through its prequalification process.
Ahonkhai, The Bill & Melinda Gates Ahonkhai, The Bill & Melinda The foundation also reviews grantee
Foundation, October 3, 2012). Gates Foundation, October 3, and partner prequalification
2012). performance reports provided by
WHO (personal communication,
Vincent Ahonkhai, The Bill & Melinda
Gates Foundation, October 3, 2012).
Clinton Health Access Stringent regulatory approval Stringent regulatory authority Independent evaluations of product
Initiative (CHAI) of product dossiers and good approval and participation in dossiers (personal communication,
manufacturing approval for the Pharmaceutical Inspection Kelly Catlin, CHAI, October 3, 2012).
manufacturing sites (personal Cooperation Scheme (personal
UNITAID, UNAIDS, and Stringent regulatory authority WHO GMP; WHO MQAS Dossier reviews and inspections
UNICEF approval; WHO prequalification; (Unicef, 2011)
expert review panel
recommendation; and WHO/PAHO
pooled procurement (Unicef, 2011)
Doctors Without Borders MSF Qualification Scheme, WHO WHO GMP; WHO International Product dossier and manufacturing
(MSF) prequalification and stringent Pharmacopoeia; European site audits (MSF, 2006)
regulatory authority approval Pharmacopoeia;
(MSF, 2006) British Pharmacopoeia;
U.S. Pharmacopeia; and MSF
specifications for pharmaceutical
products (MSF, 2006)
NOTE: DFID = Department for International Development; ECHO = European Commission’s Humanitarian Aid Department; EMA = European Medicines Agency;
FDA = U.S. Food and Drug Administration; GMP = Good Manufacturing Practice; MQAS = Model Quality Assurance System for procurement agencies; PAHO = Pan Ameri-
can Health Organization; UNAIDS = Joint United Nations Programme on HIV/AIDS; Unicef = United Nations Children’s Fund; USAID = United States Agency for International
Countering the Problem of Falsified and Substandard Drugs
Pre-Procurement Stage
Prequalification must include quality assurance and quality testing through product and
manufacture assessments, including testing of batches.
Have management information systems in place to monitor actual supply and payment of
drugs as well as post-supply quality.
Estimates of medicines needed should be based on data like past use, morbidity records,
and consumption predictions.
Procurement Stage
Procurement should be transparent, following formal written procedures and clear public
selection criteria.
Quality assessment of drugs upon receipt, including lab testing, inspection of shipments,
and certificate of analysis of delivered products.
Ensure technical specifications are right (e.g., dosage, storage, shelf life, delivery
expectations, etc.) in bidding documents.
Post-Procurement Stage
this is not possible. The regulator can only confirm that the producer is un-
known and turn the case over to law enforcement. The police and detectives
who inherit these cases have a difficult job gathering sufficient evidence for
a prosecution; there is usually little if anything to tie the falsified drug in
the market to the culprit (see Box 4-2). It is also hard to convince agents to
investigate pharmaceutical crime when they are under immediate pressure
to prosecute murders and other violent felonies. For all these reasons, falsi-
fying medicines has been called the perfect crime (Dobert, 2012; Kontnik,
2004; Nelson et al., 2006).
BOX 4-2
Fatal Falsified Iron
When a drug that had been on the market for 40 years killed a
young, generally healthy woman in 2004 despite her six previous doses
with no side effects, the technical director of the AstraZeneca subsidiary
in Río Negra, Argentina, was alarmed and suspected impropriety. The
drug was Yectafer, an injectable iron supplement given to the patient for
her anemia. She died of liver failure within weeks of receiving the fatal in-
jection, unable to undergo transplant surgery quickly enough to save her
life (Loewy, 2007). A sample of the drug was sent for testing at the plant
and was immediately identified as a fake: the package labels were ap-
plied incorrectly, the name of the drug written in a different font, and the
color of the liquid significantly altered. Chemical analysis confirmed that
the bottle did not contain iron sorbitol, the active ingredient in Yectafer,
but a different form of iron at three times the stated dosage (Loewy,
2007). Despite an attempted recall, one more woman died in the ensuing
months, and at least eight women undergoing the same treatment were
hospitalized for liver damage, including a 22-year-old pregnant woman
whose condition caused her to deliver her baby prematurely at 26 weeks
(Loewy, 2007; WHO, 2006b).
Although some of the people involved in distributing the danger-
ous fake were charged for their crimes, lack of an effective paper trail
prevented Argentine authorities from tracking down the manufacturer.
The victims’ youth lent an emotional appeal to this incident, making it
the public face of drug regulation agenda, but Argentina was no stranger
to tragedy of this sort. Fake drugs for treating Parkinson’s disease circu-
lated in 1997 and exacerbated the symptoms they were taken to prevent
(Loewy, 2007). Weak regulation and the legal confusion made Argen-
tina’s drug supply vulnerable and hampered efforts to prosecute those
involved (WHO, 2006b).
tunistic crime, emerging where regulatory capacity is low, not where profits
would be highest” (UNODC, 2010, p. vi).
This is not to say that profits generated from falsifying drugs are
insignificant. In a study of fake malaria medications in Southeast Asia,
Dondrop and colleagues found the falsified artesenuate to be cheaper, but
only somewhat, than the authentic one (Dondorp et al., 2004). By pricing
their product just slightly under the legitimate drug, criminals can guaran-
tee market share, but they avoid pricing it so low as to arouse suspicion.
Falsified medicines can be priced less cautiously in the wholesale market,
however, because these markets are less regulated and customers are not the
general public but buyers for retail who are sometimes complicit. Tempo,
an Indonesian news magazine, reported on “astonishingly low” prices
BOX 4-3
Adulterated Cancer Drugs
2 Brazil 36 74 50 2 1 0 163
4 Colombia 4 10 30 7 0 3 54
4 India 22 1 3 28 0 0 54
6 Pakistan 3 0 1 47 0 0 51
7 Thailand 37 0 3 10 0 0 50
8 South Korea 42 1 1 4 0 0 48
9 Israel 19 4 0 0 0 20 43
10 Poland 3 1 16 0 0 17 37
11 Spain 0 0 27 0 0 0 27
NOTES: Dist = distributing; Mfg = manufacturing; POS = point of sale; Trans = transporting; Unk = unknown.
SOURCE: PSI data shared with the committee, Thomas Kubic, PSI-Inc., July 11, 2012.
BOX 4-4
Pharmaceutical Security Institute Crime and Arrest Data
400
380 2010
350 355
2011
300 308
288
Number of arrests
250
239
231
200
150
131 126
100
50
32
20
0
Point of sale Manufacturing Distributor Transporting Theft
Type of activity
Number of
Country Seizures
1 Russia 93
2 China 87
3 South Korea 66
4 Peru 54
5 Colombia 50
6 United States 42
7 United Kingdom 39
8 Ukraine 28
9 Germany 25
10 Israel 25
against falsified medicines in 2011. Table 4-3 presents the number of ar-
rests in the PSI incident database by country. The countries with the most
serious problems might have no arrests in a year, as arrests depend on
government motivation to marshal the police. The momentum for labor-
intensive police raids is difficult to sustain. Only half of the countries on
PSI’s 2006 arrests list appear on the same list in 2011 (see Table 4-4). In
2006 Russia led in arrests for pharmaceutical crime after a series of raids
reported in the Lancet (Parfitt, 2006). At the time, Gennady Shirshov,
director of a Russian pharmaceutical industry association, predicted that
other criminal manufacturers would quickly replace the closed ones (Parfitt,
2006). Mr. Shirshov mentioned insufficient law enforcement interest in the
problem but concluded, “The legislation is inadequate. It’s a civil liability,
not a criminal one . . . and the fines are negligible” (Parfitt, 2006, p. 2).
As Box 4-5 mentions, perpetrators who are caught falsifying medicine
are punished leniently in some countries (Kyriacos et al., 2008; WHO,
2012a). In the United States, the Food, Drug, and Cosmetic Act dictates
a penalty of 1 year in prison, a fine of no more than $1,000, or both
(Donaldson, 2010). Even repeat offenders are punished with no more than
3 years in prison or a fine of $10,000 (Donaldson, 2010). Considering that
the profit margin for falsified drugs runs in the billions, the risk-to-profit
BOX 4-5
Manuel Calvelo
analysis favors the crime. Table 4-5 shows the penalties for falsifying medi-
cines in a selection of countries. The leniency in many countries may be a
function of outdated laws. Tables 4-6 and 4-7 show penalties for patent
and trademark infringement, which are dealt with more severely in some
countries.
Stricter and more consistent penalties could do much to fight the public
health crime of producing and trading fake medicines. Chapter 7 discusses
this solution in more detail, describing how a global code of practice could
encourage consistent strict minimum punishments for these offenses.
$2,000 per person per year after age 45 (Paez et al., 2009). The cost of
medicine is even more of a burden in low- and middle-income countries,
where it accounts for 20-60 percent of health spending, and 90 percent
of the population pays for medicine out-of-pocket (Cameron et al., 2008;
WHO, 2004a,b).
The drug market is not stable; both price and supply fluctuate. Some-
times the supply falters because of shortages in the raw materials, as in 2004
when increased demand for artemisinin, combined with a poor Artemesia
annua harvest, drove up the price and led to stock-outs (Kindermans et al.,
2007; Newton et al., 2006b; Pilloy, 2009). More generally, drug supply
problems are driven by the economy. In the United States, for example,
manufacturers sometimes stop producing products with low profit margins,
such as sterile injectables—inexpensive products that are complicated to
make (Hoffman, 2012). Manufacturers also can lose interest in a drug after
its patent expires, when revenues from the product drop (Hoffman, 2012).
Although the United States has a more stable drug supply than most devel-
oping countries, there have been regular shortages for the past 15 years, es-
pecially among injectables, cancer drugs, and antibiotics (Hoffman, 2012).
Drug shortages are more common in developing countries (MDG Gap
Task Force, 2008). Survey data from the WHO and Health Action Inter-
national suggest that although medicines may be available free or cheaply
in public health centers, these centers often do not have the medicines
needed; availability is generally better in the private sector but for a much
higher price (Cameron et al., 2008; MDG Gap Task Force, 2008). Figure
4-3 shows that although private-sector outlets have a higher percentage of
drugs available than public-sector ones, there is still a great deal of unmet
need. A month’s course of the lowest-priced generic ulcer medication, for
example, is still more than 3 days’ wages for a low-paid government worker
in much of Africa, Eastern Europe, and the Middle East (Cameron et al.,
2008).
Reducing the costs and increasing the availability of medicines would
remove some of the financial incentive to produce and procure falsified
and substandard medicines. If patients had a plentiful supply of reliable,
affordable medicines, there would be less need to shop at unregulated gray
markets.
The WHO has recommended generic substitution as a way to keep
medicines costs down (MDG Gap Task Force, 2008), but this depends
on a supply of high-quality generic medicines on the market. For generic
manufacturers, companies that generally run on low margins, the costs of
proving bioequivalence and preparing a manufacturer’s dossier for regula-
tory review can be prohibitive to market entry (Lionberger, 2008). Different
regulatory authorities have different, often widely divergent, requirements
for establishing bioequivalence (Mastan et al., 2011). To complicate the
90
80
76.3
70
64.3 65.8
60.7 61.8
60
50
44.3 43.1
40
37.8
36.1 36.1
20
10
0
Low Other Lower– Upper– Low Other Lower– Upper– Low Other Lower– Upper–
income: low middle middle income: low middle middle income: low middle middle
India income income income India income income income India income income income
(n=7) (n=15) (n=9) (n=3) (n=7) (n=17) (n=11) (n=3) (n=7) (n=17) (n=11) (n=4)
Public sector, generics Private sector, generics Private sector, originator brands
from the common format which allows them to prepare submissions for
several countries at once (Poh, 2011).
The cost of bioequivalence testing runs from $50,000 to $200,000
(GIZ, 2012). Bioequivalence testing also requires sophisticated laborato-
ries that are not available in many countries. This baseline cost to generic
companies does not include several person-months of staff costs for revis-
ing registration application data into a new dossier. The costs of market
authorization are prohibitively expensive, especially for entry into a small
country’s market. When the overwhelmed regulatory authority will allow
it, companies avoid the expense by submitting no proof of bioavailability;
others falsify bioavailability data (Silverman, 2011).
Evidence suggests that these high costs keep generics companies out
of the market and increase costs to the consumer (Mastan et al., 2011;
Rawlins, 2004). Even multinational, innovator pharmaceutical companies
struggle to convert applications between FDA and EMA formats. A 1996
industry study estimated that converting applications took between 2 and
10 months and significant staff time and expense (Molzon, 2009). Differ-
ent standards for bioequivalence assessment also encourage the problem
of widely divergent national drug quality standards (Mastan et al., 2011).
If the application and registration process were more straightforward
then more good-faith companies could enter the market, increasing the sup-
ply of reliable drugs and controlling costs. The committee also believes that
a consistent use of the common registration format could further the cause
of regulatory harmonization, which would improve the drug regulatory
systems in low- and middle-income countries. Harmonization also controls
the burdens regulation puts on manufacturers; shared inspections are more
efficient and less disruptive to industry. Generics companies, which gener-
ally have fewer staff than innovator companies, are disproportionately
disturbed by frequent inspections.
25
20
Number of NMRAs
15
10
0
Marketing Licensing Inspection Quality Pharmaco- Control of Control of
authorization control vigilance promotion clinical trials
(WHO, 2010a). The same study found that several regulatory authorities
grant licenses and renewals with no inspections, that operating procedures
for conducting inspections were woefully weak, and that 35 percent of
the regulatory authorities have no legal authority for inspections (WHO,
2010a). Figure 4-4 shows the number of agencies out of the 26 surveyed
that can perform drug regulatory functions. All of these weaknesses allow
for falsified and substandard drugs to circulate. As one of the participants
in the WHO study explained, “The illicit medicines market has become a
real plague. . . . All therapeutic classes can be found, including psychotro-
pic medicines, and there is no national strategy to combat this situation”
(WHO, 2010a, p. 16).
Governments in low- and middle-income countries need a strategy to
act against falsified and substandard medicines. Any viable solution will
include strengthening the drug regulatory system, including building the
inspectorate, enforcing quality standards, and licensing in accordance with
international standards. Without a competent regulatory authority to in-
spect wholesalers, distributors, and manufacturers, opportunities to corrupt
the drug supply abound. Box 4-6 describes a patient safety disaster follow-
ing the disbanding of the Pakistani national regulatory authority.
A 2012 Institute of Medicine report called for greater international
investment in building food and drug regulatory systems in developing
countries and for an international training and credentialing system for
BOX 4-6
Dissolution of the Pakistani Drug Regulatory Authority
Over the course of several weeks in January 2012, more than 120
patients in Lahore, Pakistan, died of drug overdoses and hundreds more
suffered adverse reactions after being treated with contaminated heart
medicine at the Punjab Institute of Cardiology (Arie, 2012). The drug
responsible was Isotab (isosorbide mononitrate, 20 mg), manufactured
by Efroze Chemical in Karachi, Pakistan (Arie, 2012). Each Isotab tablet
contained isosorbide mononitrate, as well as 14 times the normal dose
of the antimalarial drug pyrimethamine. The overdose caused rapid bone
marrow, white blood cell, and platelet depletion (BBC, 2012). The drug’s
packaging did not contain dates of manufacture or expiration, and the
drugs were given to patients for free (Arie, 2012). Drug pricing was a
concern at Punjab Institute of Cardiology. Anonymous sources at the
hospital reported significant pressure to buy the lowest cost drugs avail-
able. Under Pakistani law, when the lowest bidder does not win a sale,
rejected firms can bring lawsuits against the hospital (BBC, 2012).
Pharmaceutical regulation in Pakistan is particularly weak. Though
the government approved an independent drug regulatory authority in
2005, political tensions prevented action (Arie, 2012). In 2010, a con-
stitutional amendment further debilitated regulation by abolishing the
ministry of health. Provincial governments, many with weak infrastruc-
tures, were given sole responsibility for drug regulation. Manufacturers
exploited the confused system by rapidly registering thousands of drugs
(Khan, 2012).
Following the Isotab scandal, the Pakistan Supreme Court ordered
action on the independent agency. Doctors have expressed doubts, fear-
ing that insufficient regulatory expertise and ineffective execution will
impede the new agency’s success (Khan, 2012). Their concerns appear
to be well founded. The new agency’s board includes only one position
for an expert in medicine or pharmacy (Khan, 2012).
would agree to work toward the priorities identified on the strategic plan,
and all work would be directly related to the plan, an openly shared docu-
ment (Tominaga, 2012). For many smaller countries the plan should include
a strategy for sharing work and pooling resources. At the regional level,
the New Partnership for Africa’s Development recently published a 5-year
strategic plan for regulatory harmonization (NEPAD, 2011). This docu-
ment identified the technical barriers facing African regulators, clarified
the mission of the African Medicines Regulatory Harmonization (AMRH)
project, and identified objectives for 2011-2015 (NEPAD, 2011).
Multilateral agencies, such as development banks, should support the
development and implementation of strategic plans for compliance with
international standards. The pharmaceutical market is international, and
everyone has an interest in promoting global standards. There is precedent
for such investment. The Bill & Melinda Gates Foundation, the British De-
partment for International Development, the World Bank, and the WHO
all support the AMRH program (AMRH, 2012). Donor agencies can do
similar work, as USAID has in support of postmarket surveillance in Latin
America, Southeast Asia, and Africa (Miarlles, 2011).
Regulators will welcome the strategic investments this planning would
bring. Governments need to support these investments as well. Compliance
with international standards will demand a wide range of activities, includ-
ing research, education, supply chain management, and incentives for the
private sector. The regulatory agency alone cannot effect change and will
need government support to marshal the involvement of all stakeholders.
Developed country governments also need to improve support for their
regulatory agencies. At the time this report was prepared, substandard
injectable drugs caused a fungal meningitis outbreak in the United States,
bringing the topic of drug regulatory oversight to the forefront of the U.S.
political discourse.
FDA’s MedWatch system had identified drug quality problems with meth-
ylprednisolone acetate, the steroid that caused the 2012 outbreak, at New
England Compounding Center in 2002 and 2004 (Energy and Commerce
Committee, 2012). The FDA and Massachusetts state inspectors uncov-
ered sanitary violations in a joint inspection and issued the manufacturer a
warning in 2006 (Energy and Commerce Committee, 2012). The problem
is not confined to New England Compounding Center. In 2002, nonsterile
practices at a South Carolina compounding pharmacy caused a similar,
though smaller, outbreak (CDC, 2002). Since 2001, the FDA has issued
67 warning letters to various compounding pharmacies (Markey, 2012),
but the FDA’s authority over these organizations is unclear and has been
for some time. In 1996, David Kessler, then FDA commissioner, testified
that compounding pharmacies threatened to create “a shadow industry” of
unregulated drug manufacture (Kessler, 1996).
In the United States, professional practice, including the practice of
medicine and pharmacy, is regulated by the states. Compounding phar-
macies, which were traditionally small operations that prepared custom
drugs for individual patients, fall under state jurisdiction (Burton et al.,
2012). Pharmacy councils have long resisted federal interference in their
practice, including oversight of compounding pharmacies (Calvan, 2012;
Markey, 2012). At the same time, enforcement of the Food, Drug, and
Cosmetic Act, which controls the marketing and manufacture of medicines,
is the FDA’s responsibility. Large compounding pharmacies are in practice
much closer to small manufacturers than pharmacies (Burton et al., 2012),
though compounding pharmacies do not register with the FDA as manu-
facturers (Outterson, 2012). A 2007 bill aimed to increase FDA oversight
of compounding pharmacies, but met the vociferous opposition of the
International Association of Compounding Pharmacists and died in com-
mittee (Burton et al., 2012). Confusion over the regulation of compounding
pharmacies was evident at congressional hearings on November 14, 2012
(Grady, 2012). New York Times reporter Denise Grady observed, “The
hearing was titled ‘The Fungal Meningitis Outbreak: Could It Have Been
Prevented?,’ but the question was never really answered” (Grady, 2012).
Disagreement over what authority the FDA has promotes a degree of
paralysis. Neither the state of Massachusetts nor the FDA had clear control
over the New England Compounding Center. Confusion about their re-
sponsibilities created a regulatory gap that the company exploited. Similar
confusion causes regulatory gaps in other countries where national and
local governments share responsibilities for drug regulation. In 2003, the
Mashelkar Report raised concerns with Indian states’ uneven implementa-
tion of drug regulations (Government of India, 2003). More recent testing
and sampling confirms that drug quality is still more reliable in states with
stricter regulations (Bate et al., 2009a). Brazil, China, Russia, and many
other large countries face similar problems (Mooney, 2010; Vashisth et al.,
2012).
Uneven Awareness
Starting in the early 2000s, medicines counterfeiting (as it was then
called) has been the topic of some media attention. General awareness of
the problem was still poor, however (Cockburn et al., 2005; Newton et al.,
2006a). Reporting was “alarming[ly] low”: between 2002 and 2004 the
WHO received no reports of fake drugs from any member states (Newton
et al., 2006a). This began to change in 2006 when the International Medical
Products Anti-Counterfeiting Task Force (IMPACT) made raising aware-
ness one of its main goals (Liberman, 2012).
IMPACT, and the larger debate about pharmaceutical fraud that it was
a part of, appears to have had success in raising awareness of the problem
in some parts of the world. A 2010 Gallup poll in sub-Saharan African
countries found that the majority of the public in 15 of the 17 countries
surveyed were aware that fake medicines were a problem (see Table 4-8)
(Ogisi, 2011). The leadership of drugs regulators in Nigeria, one of the
largest and most influential African countries, might have contributed to
the public consciousness in Africa (see Box 4-7). More recently, Interpol
launched an awareness campaign featuring South Africa’s Yvonne Chaka
Chaka and Senegal’s Youssou N’Dour, two of the continent’s biggest ce-
lebrities (Interpol, 2011). Awareness of the problem is also growing in
Southeast Asia (Christian et al., 2012a; Gleeson, 2012).
Other research suggests gaps in awareness, especially among the poor-
est people in society. A qualitative study of Sudanese policy makers and
pharmacists suggested that awareness of counterfeit products is lowest
Country % Yes
Cameroon 91
Sierra Leone 83
Nigeria 83
Liberia 79
Ghana 74
Mali 74
Burkina Faso 71
Uganda 70
Zimbabwe 69
Tanzania 66
Senegal 65
Kenya 63
Niger 62
Chad 58
Botswana 32
South Africa 25
among the poor and people living in remote areas (Alfadl et al., 2012).
Participants at overseas site visits for this study mentioned similar patterns
in many developing countries. Often, well-educated urban consumers un-
derstand the threat of fake drugs and take precautions to avoid them. The
poorest patients, and those living in areas with few to no reliable pharma-
cies, are often the least aware. Moreover, as Chapter 5 will discuss, they
often have no choice but to buy medicines in the open market or have no
money to buy from a registered pharmacy.
It is not clear how well informed populations in other parts of the
BOX 4-7
A National Awareness Campaign in Nigeria
world are about falsified and substandard drugs. People in developed coun-
tries, who have long taken medicines regulation for granted, are among
the least knowledgeable. An Inter-Press Service story reported that 20
percent of Western Europeans did not consider it dangerous to circumvent
traditional pharmacies to buy medicine (Stracansky, 2010). The same be-
havior has long been normal in the United States, where pharmacy tourism
to Canada and Mexico has been common since the 1970s (Rabinovitch,
2005). Chapter 5 will discuss the internet pharmacies that have largely
replaced in-person cross-border shopping.
Public Action
Educating the public on the problems of falsified and substandard
medicines is important, but only insomuch as education empowers people
to act. In an international site visit for this report, a procurement agency
informed the IOM delegation that when they uncover manufacturers mak-
ing substandard drugs they do not report the offense to the authorities.
The reasons they gave included doubt that the regulator would act on their
information and fear of litigation.
Similar attitudes may underlie a lack of reporting of adverse drug reac-
tions among health workers in developing countries. Health workers are
the first line for monitoring the safety of medicines. Their role in surveil-
lance is important in low- and middle-income countries, where falsified
and substandard drugs are common, and less than 27 percent have func-
tional pharmacovigilance systems (Pirmohamed et al., 2007). Reporting of
adverse drug events is generally low in these countries (Chedi and Musa,
2011; Fernandopulle and Weerasuriya, 2003; WHO, 2002b). Few staff are
trained in pharmacovigilance, a practice sometimes seen as adding to the
responsibilities of already overworked health professionals (Olsson et al.,
2010; Sharma and Ahuja, 2010).
The increasing awareness of falsified and substandard medicines could
drive improved pharmacovigilance in developing countries. Awareness cam-
paigns and investigative reporting reach health workers as well as they
reach the rest of the public. There is also a need for targeted health worker
education on falsified and substandard medicines, emphasizing the correct
reporting channels health workers can use to confirm suspected cases of
falsified and substandard drugs. Much useful work has been done on the
first steps of this process; clinicians struggling to broach the topic with their
patients can consult the World Health Professionals Alliance guidelines on
how to inquire about suspicious medicines (see Box 4-8).
Chapter 3 describes governments’ and drug companies’ reluctance to
share information on substandard and falsified drugs (Cockburn et al.,
2005). Pharmaceutical companies fear damage to their branding from
BOX 4-8
Health Worker Guidelines
1. Where patients will or did buy the medicine. Emphasis can be placed
on the importance of buying medicine from a pharmacy or other
known and reliable sources.
For example: “Did you purchase the medicine from a known and reli-
able source?”
2. W
hat patients should look out for when they buy medicines. It can be
suggested that patients check the packaging, the product, and the
patient leaflet when they purchase medicine.
For example: “Was the packaging of the product intact, properly
sealed, clearly labeled with dosing, manufacturer, batch number, and
expiry date?”
3. H
ow the medicine is expected to take effect. By explaining what
should happen when patients take medicine, health professionals can
help patients identify anything unusual.
For example: “Did the medicine cause any unexpected side effects?”
For example: “Has the medicine taken longer than anticipated to have
an effect?”
Falsified and substandard drugs are a potential threat around the world,
though risk varies widely from country to country. Awareness of the prob-
lem also varies and may be most limited in countries with strong regulatory
systems but where, because of the global drug supply chain, substandard
and falsified drugs still reach consumers. An effective communication cam-
paign should present accurate information in a way that empowers patients
to protect their own health. For example, the FDA website discourages buy-
ing drugs from foreign websites (see Figure 4-5) (FDA, 2012c). The CDC
website gives similar guidance, discussing poor-quality antimalarials and
alerting prospective travelers to avoid buying drugs abroad (CDC, 2010).
Education and communication are feasible in rich and poor countries
alike. Representatives of 200 WHO member states stressed the importance
of educational initiatives for consumers and health workers at the first
meeting of the WHO global mechanism against falsified and substandard
drugs (WHO, 2012c). Many developing countries have already made head-
way in consumer education. Figure 4-6, for example, shows a Cambodian
health education poster promoting licensed pharmacies. Similarly, as Box
4-7 explains, the Nigerian drugs regulatory authority improved public
understanding of the problem with relatively simple steps: public service
announcements, newspaper ads, and school essay contests. This kind of
campaign is realistic in many low- and middle-income countries.
While information about the problem is important, it is also important
to link this information to action. The messages communicated and the ac-
tion promoted will vary by country or region. In many countries, the most
useful messages will be about specific drugs and vendors. Buying antima-
larials from street markets, for example, is a dangerous behavior in most of
Africa and Southeast Asia. Chapter 5 discusses some of the safe medicine
outlets that the communication campaigns could promote.
The most wide-reaching communication strategies make use of many
channels, including print media, television, radio, the internet, mobile de-
vices, and social media. Governments and NGOs have made good progress
using these channels to promote understanding of the problem (Besançon,
2008, 2012; Elliot, 2012; FIP, 2011). Educated consumers may now be
more receptive to messages about the correct appearance or taste of medi-
cines, the normal responses to it, and possible side effects. Patients who
Counterfeit
medicines
filled with empty promises.
Don’t be a victim!
Buy medicines only from state-licensed
pharmacies that are located in the United
States. Find your state’s contact information
from the National Association of Boards of
Pharmacy (NABP) at www.nabp.info
or
www.fda.gov 1-888-INFO-FDA
E
TM
DE P AR
FIGURE 4-5 An FDA public service announcement that promotes the Verified Internet
Pharmacy Practice certification discussed in Chapter 5. This is an example of an em-
powering consumer education message.
NOTE: The poster uses the term counterfeit broadly, the way this report uses falsified. See page 23.
SOURCE: FDA, 2012.
FIGURE 4-6 The English translation of a Cambodian poster encouraging consumers to buy
medicines only from licensed pharmacies and to examine the drug’s color, shape, and taste
for abnormalities.
NOTE: The poster uses the term counterfeit broadly, the way this report uses falsified. See page 23.
SOURCE: U.S. Embassy, Phnom Penh, Cambodia.
Canada e Up to $5,000
Lebanon f Up to $30,000
Singapore g Up to $100,000
France, i
South Africa, j $100,000 or more
Switzerland k,l
Uganda p Up to $2,000
Pakistan q Up to $5,000
continued
Brazil w, x Up to $98,000
Thailand gg Up to $1,700
m Bate, R. 2012. Phake: The deadly world of falsified and substandard medicines. Washington, DC: AEI Press.
n Medicinal Products Act. (Germany). (2010). Chap. 17, Sec. 95 (3) 3.
o AEI. 2012. The deadly world of fake drugs. AEI.
p The National Drug Policy and Authority Act of 2003. (Uganda). Chap. 206, Part IV, Sec. 30.
q The Drugs Act, 1976. (Pakistan). Chap. IV, Sec. 27 (1);(2).
r AEI. 2012. The deadly world of fake drugs. AEI.
s Phana, C. 2007. Country presentation: Cambodia. Presented at First ASEAN–China Conference on combat-
ing counterfeit medicinal products. Jakarta, Indonesia.
t WHPA. 2011. Background document on counterfeit medicines in Asia. Paper read at WHPA Regional Work-
shop on Counterfeit Medical Products, Taipei, Taiwan.
u WHPA. 2011. Background document on counterfeit medicines in Asia. Paper read at WHPA Regional Work-
shop on Counterfeit Medical Products, Taipei, Taiwan.
v Counterfeit and Fake Drugs and Unwholesome Processed Food (miscellaneous provisions) Act
of 1999. (Nigeria). Sec. 3 (1).
w Capell, K., S. Timmons, J. Wheatley, and H. Dawley. 2001. What’s in that pill? Bloomberg Businessweek
Magazine.
x Lei Nº 6.437 De 20 De Agosto De 1977. (Brazil). Tit. 1, Art. 2, §1º.
y The Anti-Counterfeit Bill, 2008. (Kenya). Part VI, Sec. 35 (a); (b).
z AEI. 2012. The deadly world of fake drugs. AEI.
aa Ley General De Salud, 2012. (Mexico). Titulo Decimo Octavo, Capitulo VI, Artículo 464 Ter. (I); (II).
bb Drug Administration Law of the People’s Republic of China. (China). 2001, No. 45. 20th meeting, 9th Cong.,
Chap. IX, Art. 74.
cc Jailing, D. 2011. China broadens scope of counterfeit drugs criminal prosecution, but definition still murky.
Elsevier Business Intelligence.
dd Sinha, K. 2009. From Monday, spurious drug sellers can be jailed. Times of India.
ee Special Law on Counterfeit Drugs. (Philippines). 1996. Republic Act No. 8203, Cong. of the Philippines
Metro Manila, 2nd sess., Sec. 8 (b); (e); (f).
ff Counterfeit Drug Penalty Enhancement Act of 2011, HR 3468. 112th Cong., 1st Sess., Sec. 2 (a); (b).
gg Thailand Drug Act, B.E. 2510 (1967). (Thailand). Chap. X, Sec. 117.
Canada p Up to $500
Singapore q Up to $10,000
Lebanon u Up to $33,000
France x Up to $650,000
Kenya z Up to $6,000
Tanzania aa Up to $300
q Singapore Patents Act as amended by Act No. 2 of 2007. (Singapore). (April 1, 2007). Part XVIII, Sec.
99 (1).
r Loi fédérale sur les brevets d’invention. (Switzerland). (June 25, 1954). Tit. 3, Chap. 3, Art. 81 (1).
s Patents Act Consolidation. (Thailand). No. 3. (1999). Part VI, Chap. VI, Art. 85.
t Germany Patent Act. (Germany). (July 30, 2009). Part 9, Sec. 142 (1).
u Patents Law of Lebanon, Law No. 240. (Lebanon). (August 7, 2000). Chap. 2, Sec. 1, Art. 42.
v Law of the Republic of Indonesia Regarding Patents. (Indonesia). No. 14. 2001. Chap. XV, Art. 130.
w Law on the Patents, Utility Model Certificates and Industrial Designs. (Cambodia). 8th sess., 1st legis.
(December 31, 2002). Chap. 7, Art. 133.
x Intellectual Property Code. (France). (July 1, 1992). Chap. V, Sec. II, Art. L615-14 (1).
y Patent Act (Act No. 121 of 1959). (Japan). Chap. XI, Art. 196-2.
z The Industrial Property Act, 2001. (Kenya). Part XVI, Sec. 109 (1); (2).
aa The Patents (Registration) Act. (Tanzania). Part XV, Sec. 70 (1).
bb Penal Code of Argentina. (Argentina). Law 11,179 (1984). Chap. IV, Art. 172.
cc Legal Intellectual Property Regime (Argentina). Law No. 11.723, Art. 71.
dd Patent Act (Act No. 950 of December 31, 1961, as last amended by Act No. 9985 of January 30, 2009).
(Republic of Korea). Chap. XII, Art. 225 (1).
Korea j Up to $47,000
Jordan m Up to $8,500
Indonesia t Up to $105,000
REFERENCES
Ahmed, R. 2012. India’s plan to distribute free medicines raises questions. Wall Street Journal
India, July 6.
Akunyili, D. 2005. Counterfeit drugs and pharmacovigilance. Paper presented at 10th Phar-
macovigilance Training Course held at Uppsala Monitoring Centre, Uppsala, Sweden,
May 25.
———. 2006. Women leadership in emerging democracy—my NAFDAC experience. Address
by the director general of the National Agency for Food and Drug Administration and
Control, Dora Nkem Akunyili. Wilson Center, April 29.
Alfadl, A. A., M. A. Hassali, and M. I. M. Ibrahim. 2012. Counterfeit drug demand: Percep-
tions of policy makers and community pharmacists in Sudan. Research in Social and
Administrative Pharmacy. [Epub ahead of print].
Ames, J., and D. Z. Souza. 2012. Counterfeiting of drugs in Brazil. Revista de Saúde Pública
46(1). DOI: 10.1590/S0034-89102012005000005.
AMRH (African Medicines Regulatory Harmonization). 2012. Partners. http://www.amrh.
org/partners (accessed January 9, 2013).
Anderson, T. 2010. Tide turns for drug manufacturing in Africa. Lancet 375(9726):1597-1598.
APIC (Association for Professionals in Infection Control and Epidemiology). 1999. Cleaning
validation in active pharmaceutical ingredient manufacturing plants. Washington, DC:
APIC.
Arie, S. 2012. Contaminated drugs are held responsible for 120 deaths in Pakistan. British
Medical Journal 344:e951.
Arnold, R. 2008. Economics. 9th ed. Mason, OH: Cenage Learning.
Bate, R. 2007. On the trail of a cure: Reality and rhetoric on treating malaria. Washington,
DC: American Enterprise Institute for Public Policy Research.
———. 2008. Local pharmaceutical production in developing countries: How economic
protectionism undermines access to quality medicines. London: Campaign for Fighting
Diseases.
Bate, R., R. Tren, K. Hess, L. Mooney, and K. Porter. 2009a. Pilot study comparing technolo-
gies to test for substandard drugs in field settings. African Journal of Pharmacy and
Pharmacology 3(4):165-170.
Bate, R., R. Tren, L. Mooney, K. Hess, B. Mitra, B. Debroy, and A. Attaran. 2009b. Pilot study
of essential drug quality in two major cities in India. PLoS ONE 4(6):e6003.
BBC (British Broadcasting Company). 2012. Police investigate Pakistan heart drug deaths.
BBC News, January 24.
Beken, T. V., and A. Balcaen. 2006. Crime oppotunities provided by legislation in market
sectors: Mobile phones, waste disposal, banking, pharmaceuticals. European Journal on
Criminal Policy Research 12:299-323.
Belluck, P. 2001. Prosecuters say greed drove pharmacist to dilute drugs. http://www.
nytimes.com/2001/08/18/us/prosecutors-say-greed-drove-pharmacist-to-dilute-drugs.
html?pagewanted=all&src=pm (accessed July 12, 2012).
Besançon, L. 2008. Country specific case studies—best practices to combat counterfeit medi-
cines and to protect public health. The Hague, The Netherlands: International Pharma-
ceutical Federation.
———. 2012. Health professionals in the risk communication process on counterfeit medi-
cines. Generics and Biosimilars Initiative Journal 1(3-4):135-137.
Brhlikova, P., I. Harper, and A. Pollock. 2007. Good manufacturing practice in the pharma-
ceutical industry. Workshop on Tracing Pharmaceuticals in South Asia, University of
Endinburgh, Centre for International Public Health Policy.
British Pharmacopoeia. 2012. Amoxicillin capsules. In British Pharmacopoeia Vol. III: For-
mulated preparations—specific monographs. http://bp2012.infostar.com.cn/Bp2012.
aspx?tab=browse&a=display&n=53&id=6349 (accessed March 5, 2013).
Brousseau, Z. 2012. Mastering the ECTD format critical for regulatory submissions pros.
http://www.raps.org/focus-online/news/news-article-view/article/1191/mastering-the-
ectd-format-critical-for-regulatory-submissions-pros.aspx (accessed November 5, 2012).
Bumpas, J., and E. Betsch. 2009. Exploratory study on active pharmaceutical ingredient
manufacturing for essential medicines. Washington, DC: The International Bank for
Reconstruction and Development/The World Bank.
Burkitt, L. 2012. Beijing says counterfeit drugs seized. Wall Street Journal, August 5.
Burton, T., J. Grimaldi, and T. Martin. 2012. Pharmacies fought controls. Wall Street Journal,
October 14.
Calvan, B. 2012. Compounding pharmacies have long evaded the tight oversight governing
established drug makers. Boston Globe, October 29.
Cameron, A., M. Ewen, D. Ross-Degnan, D. Ball, and R. Laing. 2008. Medicines prices,
availability, and affordability in 36 developing and middle-income countries: A secondary
analysis. Lancet 373(9659):240-249.
Caudron, J. M., N. Ford, M. Henkens, C. Macé, R. Kiddle-Monroe, and J. Pinel. 2008. Sub-
standard medicines in resource-poor settings: A problem that can no longer be ignored.
Tropical Medicine & International Health 13(8):1062-1072.
CDC (Centers for Disease Control and Prevention). 2002. Exophiala infection from contami-
nated injectable steroids prepared by a compounding pharmacy. Morbidity and Mortality
Weekly Report 51(49):1109-1112.
———. 2010. Counterfeit and substandard antimalarial drugs. http://www.cdc.gov/malaria/
travelers/counterfeit_drugs.html (accessed November 6, 2012).
———. 2012. CDC and FDA joint telebriefing on investigation of meningitis outbreak. http://
www.cdc.gov/media/releases/2012/t1004_meningitis_outbreak.html (accessed November
26, 2012).
———. 2013. Multistate fungal meningitis outbreak—current case count. http://www.cdc.gov/
hai/outbreaks/meningitis-map-large.html (accessed January 28, 2013).
Chaudhury, R. R., R. Parameswar, U. Gupta, S. Sharma, U. Tekur, and J. Bapna. 2005. Qual-
ity medicines for the poor: Experience of the Delhi programme on rational use of drugs.
Health Policy and Planning 20(2):124-136.
Chedi, J. F. O. E. A. A. B., and A. Musa. 2011. Knowledge, attitude and practice of adverse
drug reaction reporting among healthcare workers in a tertiary centre in northern Nige-
ria. Tropical Journal of Pharmaceutical Research 10(3):235-242.
Cho, Y., D. Margolis, D. Newhouse, and D. Robalino. 2012. Labor markets in low- and
middle-income countries. Washington, DC: World Bank.
Christian, L., L. Collins, M. Kiatgrajai, A. Merle, N. Mukherji, and A. Quade. 2012a. The
problem of substandard medicines in developing countries. Madison, WI: La Follette
School of Public Affairs, University of Wisconsin–Madison.
———. 2012b. The problem of substandard medicines in developing countries. Paper pre-
sented at Workshop in International Public Affairs, La Follette School of Public Affaris,
University of Wisconsin–Madison, May 21.
Chua, G. N., M. A. Hassali, A. A. Shafie, and A. Awaisu. 2010. A survey exploring knowl-
edge and perceptions of general practitioners towards the use of generic medicines in the
northern state of Malaysia. Health Policy 95(2-3):229-235.
Clark, E. 2008. Counterfeit medicines: The pills that kill. Telegraph, April 5.
Cockburn, R., P. N. Newton, E. K. Agyarko, D. Akunyili, and N. J. White. 2005. The global
threat of counterfeit drugs: Why industry and governments must communicate the dan-
gers. PLoS Medicine 2(4):e100.
Daviaud, J., and A. Saleh. 2010. Quality assurance policy for pharmaceutical products. Joint
WHO and UNAIDS consultation meeting with pharmaceutical companies, December
9-10, Geneva, Switzerland.
DFID (Department for International Development). 2012. DFID quality assurance policy for
reproductive health commodities. London: DFID.
Dickens, T. 2011. The world medicines situation 2011: Procurement of medicines. Geneva:
WHO.
Dobert, D. n.d. Pharmaceutical counterfeiting: A clear and present danger. http://www.atlco.
com/pharma_counter.html (accessed August 15, 2012).
DOJ (U.S. Department of Justice). 2011. Press release: Belgian citizen sentenced for selling
counterfeit, misbranded drugs. Washington, DC: DOJ and U.S. Food and Drug Admin-
istration Office of Criminal Investigations.
Donaldson, A. 2010. “And the ones that mother gives you don’t do anything at all,” combat-
ing counterfeit pharmaceuticals: The American and British perspectives. New England
Journal of International and Comparative Law 16(1):145-168.
Dondorp, A. M., P. N. Newton, M. Mayxay, W. Van Damme, F. M. Smithuis, S. Yeung, A.
Petit, A. J. Lynam, A. Johnson, T. T. Hien, R. McGready, J. J. Farrar, S. Looareesuwan,
N. P. J. Day, M. D. Green, and N. J. White. 2004. Fake antimalarials in Southeast
Asia are a major impediment to malaria control: Multinational cross-sectional survey
on the prevalence of fake antimalarials. Tropical Medicine & International Health
9(12):1241-1246.
Draper, R. 2003. The toxic pharmacist. http://www.nytimes.com/2003/06/08/magazine/the-
toxic-pharmacist.html?pagewanted=print&src=pm (accessed July 12, 2012).
Droop, J. 2012. DFID quality assurance/ procurement policies for medicines, October 17.
Economist. 2012a. Bad medicine, October 13.
———. 2012b. Bulging in the middle, October 20.
Elliot, J. 2012. Global interagency efforts stem counterfeit drugs in Greater Mekong Asia—
update. http://casestudiesforglobalhealth.org/post.cfm/global-interagency-efforts-stem-
counterfeit-drugs-in-greater-mekong-asia-update (accessed November 6, 2012).
Energy and Commerce Committee. 2012. Committee leaders request documents related to
deadly meningitis outbreak. http://energycommerce.house.gov/press-release/committee-
leaders-request-documents-related-deadly-meningitis-outbreak (accessed November 26,
2012).
European Commission. 2011. Guidelines for the award of procurement contracts within the
framework of humanitarian aid actions financed by the European Union. European
Commission.
FDA (U.S. Food and Drug Administration). 2012a. Counterfeit medicines: Filled with empty
promises (print public service announcement). http://www.fda.gov/Drugs/Resources
ForYou/ucm079306.htm (accessed November 6, 2012).
———. 2012b. Department of Health and Human Services, Food and Drug Administration,
inspection form for New England Compounding Pharmacy, Inc. http://www.fda.gov/
downloads/AboutFDA/CentersOffices/OfficeofGlobalRegulatoryOperationsandPolicy/
ORA/ORAElectronicReadingRoom/UCM325980.pdf (accessed November 26, 2012).
———. 2012c. Information for consumers (drugs): Buying medicines over the Internet. http://
www.fda.gov/Drugs/ResourcesForYou/Consumers/BuyingUsingMedicineSafely/Buying
MedicinesOvertheInternet/default.htm (accessed November 6, 2012).
Fernandopulle, R. B. M., and K. Weerasuriya. 2003. What can consumer adverse drug reaction
reporting add to existing health professional-based systems?: Focus on the developing
world. Drug Safety 26(4):219-225.
———. 2011b. Peddling poison: The counterfeit drug problem in America. http://www.safe
medicines.org/2012/01/peddling-poison-the-counterfeit-drug-problem-in-america-397.
html (accessed September 24, 2012).
PWC (PriceWaterhouseCooper). 2010. Global pharma looks to India: Prospects for growth.
http://www.pwc.com/gx/en/pharma-life-sciences/publications/india-growth.jhtml (ac-
cessed March 5, 2013).
Quingyun, W. 2012. Major crackdown in fake medicine scam. China Daily, August 6.
Rabinovitch, S. 2005. On the legitimacy of cross-border pharmacy. Alberta Law Review
43(2):327-368.
Rägo, L. 2011. Practical use of ICH CTD in facilitating approval of products by prequalifi-
cation programme and beyond. Paper presented at APEC Asia Regulatory Conference:
Asia’s Role in Global Drug Development, Seoul, Korea, April 26-28.
Rao, R., P. Mellon, and D. Sareley. 2006. Procurement strategies for health commodities: An
examination of options and mechanisms within the commodity security context. Arling-
ton, VA: DELIVER, for the U.S. Agency for International Development.
Raufu, A. 2006. Nigeria leads fight against “killer” counterfeit drugs. Bulletin of the World
Health Organization 84(9). http://www.who.int/bulletin/volumes/84/9/06-020906/en (ac-
cessed May 1, 2013).
Rawlins, M. 2004. Cutting the cost of drug development? Nature Reviews Drug Discovery
3:360-364.
Reuters. 2008. Currently China has about 3,500 drug companies falling from more than 5,000
in 2004, March 18.
Roy, J. 1994. The menace of substandard drugs. World Health Forum 14:406-407.
Russo, G., and B. McPake. 2010. Medicine prices in urban mozambique: A public health and
economic study of pharmaceutical markets and price determinants in low-income set-
tings. Health Policy and Planning 25(1):70-84.
Sahoo, A. 2008. Drug approval trends at the FDA and EMEA. Business Insights. http://www.
pharmatree.in/pdf/reports/Drug%20Approval%20Trends%20at%20the%20FDA%20
and%20EMEA_Process%20improvements,%20heightened%20scrutiny%20and%20
industry%20response.pdf (accessed May 1, 2013).
Shaji, J., and S. Lodha. 2010. Regulatory status of banned drugs in India. Indian Journal of
Pharmaceutical Education and Research 44(1):86-94.
Sharma, V., and V. Ahuja. 2010. Training in post-authorization pharmacovigilance. Perspec-
tives in Clinical Research 1(2):70-75.
Silverman, E. 2011. Will Teva sell a generic Lipitor in the US? Forbes, November 3.
Silverman, M., M. Lydecker, and P. Lee. 1992. Bad medicine: The prescription drug industry
in the third world. Stanford, California: Stanford University Press.
Siminski, P. 2011. The price elasticity of demand for pharmaceuticals amongst high-income
older Australians: A natural experiment. Applied Economics 43(30):4835-4846.
Stracansky, P. 2010. Fake medicines may kill a million a year. Global Issues, October 28.
Sun, Q., M. A. Santoro, Q. Meng, C. Liu, and K. Eggleston. 2008. Pharmaceutical policy in
China. Health Affairs 27(4):1042-1050.
TIPC (Therapeutic Information and Pharmacovigilance Center). 2010. The Namibia Medicines
Watch 2(2):5-8. Nambian Medicines Regulatory Council.
Tominaga, T. 2012. Pharmaceuticals and Medical Devices Agency. APEC MRCT roadmap:
Regulatory authorities’ efforts to promote multi-regional clinical trials (MRCTs). Paper
presented at 24th Annul EuroMeeting, Copenhagen, Denmark.
Torstensson, D., and M. Pugatch. 2012. What lies within? Procurement processes and the risk
of substandard medicines. London: The Stockholm Network.
UNDP (United Nations Development Programme). 2004. Constraints on the private sector
in developing countries. In Unleashing entrepreneurship: Making business work for the
poor. New York: UNDP.
Unicef (United Nations Children’s Fund). 2011. Supplies and logistics. http://www.unicef.org/
supply/index_39994.html (accessed October 3, 2012).
UNODC (United Nations Office on Drugs and Crime). 2010. The globalization of crime.
New York: UNODC.
USP-NF (U.S. Pharmacopeia and The National Formulary). 2010. Amoxicillin capsules. USP
Monographs. Rockville, MD: USP-NF.
Vaidyanathan, G. 2012. Failings exposed at India’s drug regulator. Nature News, May 18.
van Zyl, A., J. Daviaud, and S. Logez. 2012. Model quality assurance system for procurement
agencies: Harmonized assessment tool. WHO Drug Information 26(3):5.
Vashisth, S., G. Singh, and A. Nanda. 2012. A comparative study of regulatory trends of
pharmaceuticals in Brazil, Russia, India and China (BRIC) countries. Journal of Generic
Medicines: The Business Journal for the Generic Medicines Sector 9(3):128-143.
WHO (World Health Organization). 1998. Drug supply choices: What works best? WHO
Essential Drugs Monitor (25-26). http://apps.who.int/medicinedocs/en/d/Jwhozip10e/1.
4.html#Jwhozip10e.1.4 (accessed May 1, 2013).
———. 1999. Operational principles for good pharmaceutical procurement. Geneva: WHO.
———. 2002a. Practical guidelines on pharmaceutical procurement for countries with small
procurement agencies. WHO Regional Office for the Western Pacific, Manila, Philippines.
———. 2002b. Safety of medicines: A guide to detecting and reporting adverse drug reactions:
Why health professionals need to take action. Geneva: WHO.
———. 2004a. Equitable access to essential medicines: A framework for collective action.
Geneva: WHO.
———. 2004b. The world medicines situation. Geneva: WHO.
———. 2006a. Annex 6: A model quality assurance system for procurement agencies (rec-
ommendations for quality assurance systems focusing on prequalification of products
and manufacturers, purchasing, storage and distribution of pharmaceutical products).
Geneva: WHO.
———. 2006b. Counterfeit medicines. http://www.who.int/medicines/services/counterfeit/
impact/ImpactF_S/en/index.html (accessed July 12, 2012).
———. 2007a. A model quality assurance system for procurement agencies. Geneva: WHO.
———. 2007b. Quality assurance of pharmaceuticals: A compendium of guideines and related
materials (volume 2, 2nd updated edition). Geneva: WHO.
———. 2010a. Assessment of medicines regulatory systems in sub-Saharan African countries:
An overview of findings from 26 assessment reports. Geneva: WHO.
———. 2010b. Fact sheet n°278: Prequalification of medicines by WHO. http://www.who.int/
mediacentre/factsheets/fs278/en/index.html (accessed November 5, 2012).
———. 2011. Annex 15: Guidelines on submission of documentation for a multisource
(generic) finshed product. General format: Preparation of product dossiers in common
technical document format. Geneva: WHO.
———. 2012a. General information on counterfeit medicines. http://www.who.int/medicines/
services/counterfeit/overview/en (accessed April 25, 2012).
———. 2012b. Medicines regulatory support. http://www.who.int/medicines/areas/quality_
safety/regulation_legislation/en/index.html (accessed August 16, 2012).
———. 2012c. New global mechanism to combat substandard/spurious/falsely-labelled/
falsified/counterfeit medical products. Geneva: WHO.
WHPA (World Health Professions Alliance). n.d. Be aware: Helping to fight counterfeit medi-
cines, keeping patients safer. France: WHPA.
Wilson, K. R., J. C. Kohler, and N. Ovtcharenko. 2012. The make or buy debate: Considering
the limitations of domestic production in Tanzania. Globalization and Health 8(1):20.
World Bank. 2007. Policy note: Improving the competitiveness of the pharmaceutical sector in
Bangladesh—draft. http://www.scribd.com/doc/97075636/Pharmaceutical-Bangladesh-
Report (accessed March 5, 2013).
Yu, J., and H. v. Hindenburg. 2012. IFC invests in China’s Fosun Pharma, helping increase
global supply of affordable drugs. IFC: News and Multimedia. http://www.ifc.org/
IFCExt/pressroom/IFCPressRoom.nsf/0/A4EE3BD6C275AB0385257A69001C66A4?
OpenDocument (accessed January 3, 2013).
197
pharmacies, about half of which are national chains or food stores with an
internal pharmacy (Yadav and Smith, 2012). These vendors handle a wide
variety of products sold in an even wider variety of packaging. Retailers in
developed countries would find it logistically impossible to buy their stock,
in its many different packages, directly from manufacturers (Yadav et al.,
2012). Therefore, most vendors buy their inventory from pre-wholesalers
and wholesalers.
The drug distribution system in low- and middle-income countries
has the same basic steps as that described in Figure 5-1, but with more
intermediaries between the manufacturer and patient (Yadav and Smith,
2012). Instead of having one coordinated distribution chain that reaches
the whole country, there are many small chains and many small companies
at every step (Yadav and Smith, 2012). Figure 5-2 describes the drug flow
for public, private, and nongovernmental organizations, and their separate,
but sometimes overlapping, intermediaries.
A comparison of Figures 5-1 and 5-2 and Table 5-1 illustrate some
important differences in drug distribution in developing and developed
countries. For example, a few large firms generally control the national
wholesale market in developed countries. Cardinal Health, McKesson, and
AmerisourceBergen distribute 90 percent of drugs sold in the United States;
four or five major firms distribute to 90 percent of the market in West-
ern Europe and Japan (Yadav and Smith, 2012). In developing countries,
hundreds, even thousands, of companies control tiny shares of the drug
wholesale market (Yadav and Smith, 2012).
Excessive fragmentation is an important difference between developed
and developing countries’ drug distribution systems. In developed countries,
comparatively few large firms control the market and regulatory authorities
require some chain of custody documentation. In low- and middle-income
countries, the system is vastly more complicated. Sometimes multiple par-
allel distribution systems of varying efficiency run in the same country.
NGO international
International Manufacturers Procurement agents
warehouse
Importers and
wholesalers
Health center
Small town or
rural community
Second-tier pharmacies Community
NGO clinics
and chemical sellers health worker
PATIENTS
Patented, generic The market for prescription drugs Poor regulatory structure
vs. branded generic consists of patented drugs and creates a strong market for
generics. branded generics (brand is
used as a signal of quality by
the patient).
Box 5-1 describes the confusing drug distribution systems often found in
humanitarian emergencies.
In OECD countries, private companies ship and transport almost all
pharmaceuticals, but in developing countries, despite their vastly smaller
tax base, the government does (Yadav, 2010). In sub-Saharan Africa, a
government-owned-and-operated central medical store manages the distri-
bution of drugs, transporting goods around the country in a government-
owned fleet. Donors and developing-country governments favor this system,
wherein the central store manager can neither hire people with business
experience nor fire incompetent workers (Yadav, 2010). Inefficient supply
chain management directly drives up costs and causes drug stock-outs in
BOX 5-1
Drug Distribution in Humanitarian Emergencies
required when dealing with many suppliers and their generally poorer bar-
gaining power give the secondary wholesalers weak incentives to stock a
wide variety of products or brands (Yadav, 2009).
Wholesalers may sell and resell medicines repeatedly among themselves
before filling a pharmacy order. Wholesalers often repackage products with
every sale, or at least repackage individual containers for final sale (Catizone,
2006; Laven, 2006). Through a process called salting, legitimate and fake
drugs are mixed at wholesale, and in the wholesale repackaging, the fake
products gain authentic labels (Donaldson, 2010; Liang, 2006). Salting
can be done unknowingly, such as when primary wholesalers buy from
other intermediaries, accidentally launder fake products, package them in
authentic labels, and send them to pharmacies (Spies and VanDusen, 2003).
In repackaging the manufacturer’s expensive fraud-protection packaging
can be removed, and batch numbers reprinted (Satchwell, 2004). Not only
does this interfere with tracking requirements, but it leaves the wholesaler
repackagers with clean, unused packaging that is not always destroyed
(Satchwell, 2004).
Manufacturers usually have no distribution agreements with secondary
wholesalers (Ziance, 2008). The firms may trade in many kinds of prod-
ucts other than pharmaceuticals. Their staff are not required to show skills
BOX 5-2
Falsified Avastin’s Circuitous Path to the United States
2012). The same forces that lead patients to buy unreliable drugs can
lead doctors to do the same, creating yet another vulnerable link in the
medicines supply chain.
Corporate Headquarters
Supplier
Gainesboro, TN Marketing
Not in Canada Drugs’ network
In Canada Drugs’ network
DRUG DIVERSION
More stringent licensing requirements can improve the wholesale sys-
tem, but drugs will still need to move from the factory to the vendor, pass-
ing through many hands before reaching the patient. With every transaction
on the chain, there is a risk of the drug supply’s being compromised. Crimi-
nals take advantage of places where the distribution chain breaks down and
medicines depart from documented chain of custody. Drugs that leave the
proper distribution system are called diverted drugs; the markets that trade
diverted drugs, or more generally, markets that trade with little authorized
oversight, are called gray markets.
Drug diversion is the means through which medicines approved for
sale in one country are sold in others, where they may not be registered.
These schemes depend on false statements, forged customs declarations, or
smuggling (PSI data shared with the committee, Thomas Kubic, PSI-Inc.,
July 11, 2012). On the surface, drug diversion is not the public health threat
that falsified and substandard medicines are (Bate, 2012). Some countries
have made legal provisions for importation of unregistered lifesaving drugs
that are not available in local markets (Zaza, 2012). Others argue that
thieves bring good-quality drugs to otherwise neglected markets, and that,
issues of fraud aside, the end consumer is no worse off (Bate et al., 2010a).
If thieves trafficked solely in quality-assured medicines, then this point
might be valid. Once a medicine leaves the responsible chain of custody,
there is no way to ensure that it has been properly stored. As Chapter 3
explains, drug quality research indicates that unregistered medicines are
sometimes dangerous (Bate et al., 2010b; Lon et al., 2006; Stanton et al.,
2012; Wondemagegnehu, 1999). By chance, drug diversion may bring good
products to some patients, but it hurts many more, not only by defrauding
the official channels.
Drug diversion is roughly synonymous with theft, and trade in diverted
drugs is an indicator of the relative ease with which criminals exploit weak-
nesses on the distribution chain. Figure 5-3 shows common diversion points
in the distribution chain. In the United States, for example, the resale of
prescription drugs is a common problem, but illicit vendors also circumvent
the regulated distribution chain at other points. In developing countries, the
sale of donated drugs for profit is a common type of diversion (World Bank,
2005). Small-scale theft, also called pilfering, happens mostly between the
vendor and patients; larger cargo heists tend to happen to bulk drug pack-
ages, generally between the manufacturer and the vendor.
Prescription
Hospitals VVVVVVVVVVV
resellers
Secondary Patients
wholesalers
Countering the Problem of Falsified and Substandard Drugs
DispensingVVVVVVVVVVV
VVVVVVVVVVVVVVVVVVVVVVVVVVVVVVVVVVVVVV
clinics
Illegitimate
manufacturers
V VV
VVVVVVVVVVVVVV
Internet
VVVVVVVVVVVVVVVVVVVVVVVVVVVVVVVVVVVVVV
pharmacies
Number
Event Type of Thefts
Hijacking 12
Facility burglary 9
Facility robbery 1
Barcodes
Mass-produced items such as packaged foods and electronics use
machine-readable barcodes to store product information. Some countries
require the pharmaceutical industry to mark drugs with unique product
codes that contain the product’s tracking and identification number. The
FDA, for example, requires all human drugs to carry a 10-digit universal
identifier called a national drug code (FDA, 2012b). The first digits of the
number identify the firm that manufactures, repackages, or relabels the
product; the second segment identifies the product, its dosage form, and
formulation, and the last digits identify the packaging (FDA, 2012b). The
use of national drug codes predated the widespread use of electronic readers
(HIMSS, 2003; Simonaitis and McDonald, 2009). In 2004 the FDA issued
a rule requiring some human drugs and biologics to carry the national drug
code in a linear barcode (FDA, 2011b).1
Machine-readable barcodes have many advantages. When used in the
hospital or at the point of dispensing medication, these codes can verify
that the drug is of the correct dose and dosage form (Pedersen et al., 2003).
There is a limit to how much data a simple linear barcode can hold, how-
ever. Two-dimensional barcodes can encode more information in a small
space and are therefore gaining popularity for supply chain management
(McCathie and Michael, 2005).
1 Bar
Code Label Requirement for Human Drug Products and Biological Products, 69 Fed.
Reg. 9120 (Feb. 26, 2004).
Mobile Verification
For the time being, the poorest countries are not likely to use electronic
tracking systems below the tertiary or bulk packaging at the warehouse
level. Mobile phone verification, an ingenious form of mass serialization,
can fill in for an electronic pedigree at a drug’s last step to the consumer.
Mobile verification companies such as Sproxil take subscriptions from drug
companies and wholesalers. Sproxil provides labels to their clients; each
label is marked with a visible serial number and secret code hidden under
the scratch-off surface. When the label is attached to the final package,
the manufacturer enters the visible serial number in the Sproxil database
through a secure web portal. The visible serial number links the product
manufacturer, batch number, manufacture, and expiry dates to the secret
scratch-off code.
At the point of purchase, the consumer sends a text message or, in
some systems, an e-mail to the verification company, the company that
makes the scratch-off labels and manages the linked database. The mes-
sage is sent to a secure server, usually for no charge. An immediate text
message response confirms if the secret code number is registered with the
manufacturer, or if it is from a shipment reported to have left the legitimate
supply chain. Mobile verification of pharmaceuticals is gaining users in 17
sub-Saharan African countries and India (Mukherjee, 2012; Sproxil, 2012;
Versel, 2012). An elegant system for assigning unique product numbers,
mobile verification empowers consumers to act for their own safety.
Mobile verification cannot prevent fraud, nor is it a substitute for phar-
macovigilance and postmarket surveillance. A product could be substan-
dard at the factory but still gain a valid mobile verification label. Mobile
verification, however, appeals to good-quality manufacturers, who see the
service as an investment in their brand or as a way for consumers to have
Sproxil standard labels with visible serial number and scratch-off covering the secret
code number.
SOURCE: Ashifi Gogo.
confidence in the quality their internal records already show. A more likely
problem would be a wholesaler assigning a legitimate label to a falsified
drug. Also, the verification service only confirms a product’s identity at the
end of the distribution chain, at purchase. These systems cannot track the
chain of custody or monitor if the product has been stored and transported
properly.
A reliable system for tracking and tracing drugs through the distribu-
tion chain would greatly reduce the likelihood of falsified and substandard
medicines reaching patients. Recent technological advances, such as the use
of radio frequency identification and the expansion of mobile phones in de-
veloping countries, hold promise for supply chain security. The committee
believes that manufacturers and governments should use these technologies
to integrate all records of a drug’s chain of custody.
trace will require changes to drug primary pack labels and changes to the
packaging and repackaging practices at wholesale. These changes have
delayed acceptance of full track-and-trace (Yukhananov, 2012).
Nevertheless, consumers and governments have demanded a stronger
chain of custody (DeCardenas, 2007). This problem has been lingering for
years and should be addressed promptly (Palmer, 2012). Without a clear
federal mandate on the problem, companies and state governments work
in a state of uncertainty, not knowing where and how to make the neces-
sary investments that track-and-trace will require. If Congress does not
set a mandatory requirement, then the competing demands of state track-
and-trace systems will create an unmanageable burden for manufacturers,
wholesalers, and retailers. For example, in 2015 California will require
unique serial numbers on pill bottles and drug vials (GPhA, 2011; Norman,
2012).
In 2011, the FDA held a workshop on tracking and tracing prescription
drugs. Stakeholder comments on the workshop mentioned the importance
of track-and-trace and “the need for one standard, without variations
imposed, for example, by individual states” (Ducca, 2011). There is risk
to allowing a piecemeal approach to pharmaceutical track-and-trace. Any
track-and-trace system will be an expense to manufacturers and industry,
but the expense can be contained by making one national requirement.
Other stakeholders commented on the expense of implementing a na-
tional track-and-trace system (GPhA, 2011). Generic manufacturers and
drug wholesalers operate on lean margins (Berndt and Newhouse, 2010;
CBO, 2007). An increased track-and-trace requirement will put a financial
burden on these companies, even if the added cost is low. There are also
costs to pharmacies, between $84,000 and $110,000, about 0.88 percent of
annual sales (RFID Update, 2008). Therefore, the committee recommends
that the FDA bring all industry stakeholders together to work toward vol-
untary use of track-and-trace technology. This can help control the burden
an inevitable shift to drug tracking will put on these businesses.
Tracking primary packages through the drug distribution chain with
unique serial numbers is a good defense against criminal infiltration
(Ludwig, 2012; Pellek, 2009; Power, 2008). A method of tracking medi-
cines from the factory to the consumer could greatly reduce the chances of
a dangerous product being sold at a reputable pharmacy. These solutions
are of limited value in the vast pharmaceutical gray markets, however. Ig-
norance, convenience, and desperation, or some combination thereof, drive
patients to unlicensed pharmacies in street bazaars and on the internet.
Medicines retail, the last leg of the drug distribution system, is often the
most chaotic.
MEDICINES RETAIL
The drug distribution system becomes more disordered as the products
leak out of regulated distribution chains. The risk increases as drugs move
farther from the manufacturer en route to the vendor. Licensed pharmacies
and dispensaries can control the quality of their stock, at least insomuch
as they can trust their wholesalers. There are no such efforts at quality
control in the unlicensed market. Unlicensed vendors are often minimally
educated. They may approach medicines dispensing as any other sales job
and not want a customer to leave without making a purchase. In general,
these vendors exploit the chaos inherent to street markets and dry goods
shops in low- and middle-income countries and to online drug stores in
middle- and high-income ones. Their stock is poor because the stockists are
either unable or unwilling to judge quality.
Their customers are similarly ill-equipped to evaluate the dangers of
buying medicine outside of controlled chains. Unlicensed medicine vendors
fill a need, especially in poor countries, when time, expense, and distance
impede access to registered pharmacies. Internet pharmacies can fill a simi-
lar void, appealing to customers eager to save time and money or to pur-
chase discretely. Both types of market are dangerous and more similar than
they may appear at first glance. A Chinese military pharmacist described
the appeal of unlicensed medicine shops: “There are people who choose
to seek medical help from these places, possibly because of lower prices or
privacy concerns, which may increase their chances of getting counterfeit
products” (Quingyun, 2012). The observation is true of all unregulated
pharmacies. Street markets and the internet are a main source of falsified
and substandard medicines for patients around the world (WHPA, 2011).
The committee believes some changes to medicines retail could improve the
world’s vast and disorganized pharmaceutical bazaars.
nations tend to use highly regulated services. The poor, by contrast, usually
seek care elsewhere” (Peters and Bloom, 2012, p. 164).
the first line of postmarketing surveillance (Ziance, 2008). Too few people
are trained to do this job in the parts of the world where falsified and sub-
standard medicines are a systemic problem. As Figure 5-4 shows, poorer
countries often have more pharmacies than pharmacists, sometimes many
times more; in some counties even these estimates may be inflated (FIP,
2009). The International Pharmaceutical Federation (known by the French
acronym FIP) estimates that only slightly more than half of all pharmacists
are active in the workforce (FIP, 2009).
The WHO commented on this problem in a 2010 report, observing
that many types of medicines outlets in sub-Saharan Africa are not man-
aged by pharmacists (WHO, 2010). In general, the region has a pharmacist
for every 23,375 people; 75 percent of these pharmacists live in Nigeria
or South Africa (Kome and Fieno, 2006). After excluding these countries,
the ratio is closer to 1:64,640 (Kome and Fieno, 2006). Though the short-
age is especially acute in sub-Saharan Africa, the ratio of pharmacists to
population in most low- and middle-income countries falls far short of the
WHO-recommended 1:2,000 (Azhar et al., 2009). National estimates in
Malaysia (1:6,207) and Pakistan (≈ 1:19,748) also suggest serious problems
(Azhar et al., 2009).
The world distribution of pharmacists shown in Figure 5-5 indicates
a dearth of pharmacy professionals in sub-Saharan Africa and Southeast
Asia. This map fails to capture the relative privation of rural areas, where
far fewer pharmacists per person work (Hawthorne and Anderson, 2009).
In India, for example, most pharmacists work in the country’s drug manu-
facturing sector (Mohanta et al., 2001). So, although the national average
ratio of pharmacists to population is 1:1,785, this number masks regional
disparities (Basak et al., 2009). In states with less manufacturing the ratio
hovers around 1:4,000 (Basak et al., 2009). All states struggle with a pro-
nounced rural-urban imbalance. Few pharmacists work outside of cities,
and almost none works in remote areas (Basak et al., 2009). This problem
is not unique to India. Survey data indicate that managers around the world
find it difficult to fill pharmacy positions in the public sector and outside of
urban areas (FIP, 2006).
Rural disadvantage starts with education. Pharmacy schools are in cit-
ies and therefore attract urban students who have little interest in working
in the countryside or reason to move there after graduation (Anderson et
al., 2009). Furthermore, pharmacy training in many low- and middle-
income countries, especially in Asia, qualifies people to work in industry
(Azhar et al., 2009; Mohanta et al., 2001). A critic of the Indian pharmacy
education system observed, “Community pharmacy practice does not exist
in its true sense, only drug selling” (Mohanta et al., 2001, p. 810).
Improvements to the practice of community pharmacy would curtail
0 5 10 15 20
Malta
Japan
Jordan
Spain
Kuwait
Iceland
Republic of Korea
Germany
Belgium
France
Italy
Portugal
Canada
United States
Australia
Switzerland
Great Britain
Syria
Macedonia
Czech Republic
Denmark
Israel
Hungary
Brazil
Austria
Costa Rica
India
Finland
Mexico
Argentina
Paraguay
Colombia
Turkey
Uruguay
Singapore
Albania
Sudan
Vietnam
Zimbabwe
Ghana Density of pharmacists per 10,000
Indonesia Density of pharmacies per 10,000
Kenya
Pakistan
Mali
Nigeria
Nepal
Tanzania
Rwanda
Ethiopia
Uganda
FIGURE 5–5 The world’s pharmacists, pharmacy technicians, and pharmacy assistants,
2006 data.
SOURCE: Worldmapper, 2006. © Copyright SASI Group (University of Sheffield) and Mark Newman (Univer-
sity of Michigan).
Improving Retail
Providing safe, affordable medicine to the population is not within
the budget of many countries. The private sector, however, will invest in
medicines retail if there is a good business reason to do so. Governments
can take steps that would encourage private-sector investment and create
an environment where responsible private drug sellers will thrive.
One promising example of government and private-sector investment in
medicine retail is the Accredited Drug Dispensing Outlet (ADDO) program
in Tanzania (MSH, 2012). The Tanzanian regulatory authority was eager
to discourage illegal stocking and improve dispensing practices at unreg-
istered drug stores through an accreditation program (MSH, 2012; Rutta
et al., 2011). The Bill & Melinda Gates Foundation funded the program,
which used a combination of training, incentives, and creation of consumer
demand to drive changes in the private sector (MSH, 2005; Rutta et al.,
2011). Trainers from the Tanzanian Ministry of Health educated drug
shopkeepers on proper dispensing techniques, medicines storage, national
regulations, business skills, and ethics (MSH, 2005; Rutta et al., 2011).
The government offered low-interest loans for improving drug shops, many
of which had been stuffy, hot, humid (therefore unsuitable for medicine
storage), and not properly secured against theft (MSH, 2012). Participants
who met the program’s standards were rewarded with legal authority to sell
some controlled drugs (MSH, 2005). The government has made efforts to
increase the ADDO customer base, allowing ADDOs in some districts to
dispense subsidized artemisinin combination therapies (Rutta et al., 2011).
The subsidy also ensures that good-quality antimalarials are as affordable
to poor customers as the ubiquitous falsified ones.
The ADDO certification program conferred a competitive advantage
on participating shopkeepers. A widespread social marketing campaign on
access to malaria drugs promoted the outlets as reliable vendors (Hetzel et
al., 2007). This publicity helps build consumer confidence in the program
and create demand for the outlet’s services. An emphasis on customer ser-
vice and good management in the accreditation process gave the shops a
professional quality that enhanced consumer satisfaction.
BOX 5-3
CareShop Franchisee Profiles
Kofi Asiam
assignment for better paying jobs in places with a higher standard of living
and better opportunities for their children. This pattern can undermine the
best efforts to improve rural-urban equity and should be discouraged, while
respecting the individual right to emigrate.
Governments should reward service in underserved areas and attempt
to mitigate the hardships of these posts. The people who take advantage
of training programs are bright and ambitious. They naturally want their
children to be at least as educated as they are. Scholarships for the children
of pharmacy staff in underserved areas could assuage fears that a rural
posting puts their children at a disadvantage. Efforts to guarantee good
schooling for children, possibly through boarding schools or scholarships,
could remove a barrier to rural service (Rao et al., 2010). Better salaries can
draw trained staff to cities, but tax breaks and hardship pay can alleviate
this obstacle (CSG, 2008; Rao et al., 2010).
Health workers also have concerns about quality of life and physical
hardships in rural posts (Rao et al., 2010). Subsidized housing or provi-
sion of modern living quarters could help in places where this is a common
concern. It is also possible to recruit pharmacy technicians and pharmacy
assistants from underserved communities. Training students from rural and
remote areas is a known way to reduce attrition in these posts (Rabinowitz
et al., 1999). The Australian Rural and Remote Pharmacy program has
successfully increased service to rural and isolated communities, in part
through giving scholarships to students from rural backgrounds (see Box
5-4).
BOX 5-4
The Australian Rural Pharmacy Workforce Program
(WHO, 2011). Of those countries that have legislation about internet phar-
macies, more disallow (19 percent) than allow them (7 percent) (WHO,
2011). Figure 5-6 shows the geographic breakdown of the 30 countries that
have legislation on the operation of internet pharmacies.
One of the most common tools for governing internet pharmacy is ac-
creditation and certification, though only 7 percent of responding countries
have a national process for certifying, accrediting, or regulating internet
pharmacies (WHO, 2011). Most of these countries are in Europe, and all
are either in the World Bank’s high-income or upper-middle-income groups
(WHO, 2011). A few countries have an accreditation process for their own
internet pharmacies, but internet commerce is transnational. Legislation on
internet pharmacies outside of a country’s jurisdiction is even less common;
only 25 percent of the 144 countries surveyed have legislation governing the
purchase of medicine from foreign countries (WHO, 2011). The countries
that allow purchasing from online pharmacies abroad are mostly in Europe
(WHO, 2011), where the single market has hastened consideration of trans-
60
Prohibits
50
Allows
40
Percent of countries
30
20
10
0
Africa Eastern Europe Americas Southeast Western
Mediterranean Asia Pacific
Legislation by WHO region
Available 24 hours per day, 7 days per week Limited participation by third-party payers
websites, Levaggi and colleagues found that one-third of their drug orders
never arrived, though no company issued refunds (Levaggi et al., 2012).
On average, the investigators paid more for the drugs that never arrived
than for those that arrived (€0.83 per pill for those that arrived, €1.27 for
those that did not). Investigators cited other hidden costs, including ship-
ping and customs fees, as well as the cost of time spent waiting for the slow
transactions to process (Levaggi et al., 2012). More importantly, of the 13
pharmacies that filled orders, only two were not of substandard quality
(Levaggi et al., 2012).
Levaggi and colleagues are an Italian research group. They criticized
the false economy of online drug sellers in part because the products they
bought sell for less in Italian regulated, storefront pharmacies (Levaggi et
al., 2012). For consumers in the United States, the cost-to-benefit analysis
is not as clear. A 2001 study of Parkinson’s disease medications found on-
line drug stores offered substantial savings off U.S. list prices; brand-name
drugs were 7 to 58 percent cheaper, generics 31 to 76 percent less expensive
(Wagner et al., 2001). A study of American shoppers at internet pharmacies
found that 37.6 percent perceive the costs as one of the main advantages of
internet drug sellers (Crawford, 2003).
REFERENCES
Aleccia, J. 2011. Half of hospitals buy back-door drugs, new survey shows. NBC News,
August 26. http://www.nbcnews.com/id/44280296/ns/health-health_care/#.uvmyshzvtdr
(accessed March 27, 2013).
Altunkan, S. M., A. Yasemin, I. T. Aykac, and E. Akpinar. 2012. Turkish pharmaceuticals track
& trace system. Paper read at Health Informatics and Bioinformatics (HIBIT), 2012 7th
International Symposium, April 19-22, 2012.
Amin, A., and R. Snow. 2005. Brands, costs and registration status of antimalarial drugs in the
Kenyan retail sector. Malaria Journal 4(36). DOI: 10.1186/1475-2875-4-36.
Anderson, C., I. Bates, D. Beck, T. P. Brock, B. Futter, H. Mercer, M. Rouse, S. Whitmarsh, T.
Wuliji, and A. Yonemura. 2009. The WHO UNESCO FIP pharmacy education taskforce.
Human Resources for Health 7(45). DOI: 10.1186/1478-4491-7-45.
Appleby, J. 2003. Fake drugs show up in U.S. pharmacies. USA Today, May 14.
Attanasio, O. P., A. D. Kugler, and C. Meghir. 2009. Subsidizing vocational training for
disadvantaged youth in developing countries: Evidence from a randomized trial. Bonn,
Germany: Institute for the Study of Labor.
Australian Government. 1999. Looking after pharmacy in rural and remote Australia. https://
www.health.gov.au/internet/main/publishing.nsf/Content/health-mediarel-yr1999-mw-
mw99010.htm (accessed October 5, 2012).
Azhar, S., M. Hassali, M. Ibrahim, M. Ahmad, I. Masood, and A. Shafie. 2009. The role of
pharmacists in developing countries: The current scenario in Pakistan. Human Resources
for Health 7(1):54.
Baert, B., and B. De Spiegeleer. 2010. Quality analytics of internet pharmaceuticals. Analytical
and Bioanalytical Chemistry 398(1):125-136.
Barlas, S. 2005. Drug companies on different RFID frequencies. http://www.packworld.com/
package-type/thermoformed-packaging/drug-companies-different-rfid-frequencies (ac-
cessed September 28, 2012).
———. 2011a. FDA’s track-and-trace workshop talks Turkey about item-level pharmaceutical
packaging. Healthcare Packaging, March 15.
———. 2011b. Track-and-trace drug verification: FDA plans new national standards, phar-
macies tread with trepidation. Pharmacy and Therapeutics 36(4):203-204, 208, 231.
Basak, S., J. van Mil, and D. Sathyanarayana. 2009. The changing roles of pharmacists
in community pharmacies: Perception of reality in India. Pharmacy World & Science
31(6):612-618.
Basco, L. K. 2004. Molecular epidemiology of malaria in Cameroon. XIX. Quality of an-
timalarial drugs used for self-medication. American Journal of Tropical Medicine and
Hygiene 70(3):245-250.
Bate, R. 2012. Phake: The deadly world of falsified and substandard medicines. Washington,
DC: AEI Press.
Bate, R., K. Hess, and L. Mooney. 2010a. Antimalarial medicine diversion: Stock-outs and
other public health problems. Research and Reports in Tropical Medicine 1:19-24.
Bate, R., L. Mooney, and K. Hess. 2010b. Medicine registration and medicine quality: A
preliminary analysis of key cities in emerging markets. Research and Reports in Tropical
Medicine 1:89-93.
Bate, R., G. Z. Jin, and A. Mathur. 2012. Unveiling the mystery of online pharmacies: An
audit study. Cambridge, MA: National Bureau of Economic Reasearch.
Bendavid, Y., E. Lefebvre, L. Lefebvre, and S. F. Wamba. 2007. B-to-B e-commerce: Assessing
the impacts of RFID technology in a five layer supply chain. Proceedings of the 40th
Hawaii International Conference on System Sciences. http://www.computer.org/csdl/
proceedings/hicss/2007/2755/00/27550143c.pdf (accessed December 12, 2013).
deKieffer, D. E. 2006. The internet and the globalization of counterfeit drugs. Journal of
Pharmacy Practice 19(3):171-177.
Donaldson, A. 2010. Current development: “And the ones that mother gives you don’t do
anything at all” combating counterfeit pharmaceuticals: The American and British per-
spectives. New England Journal of International and Comparative Law 16(1):145-168.
Dondorp, A. M., P. N. Newton, M. Mayxay, W. Van Damme, F. M. Smithuis, S. Yeung, A.
Petit, A. J. Lynam, A. Johnson, T. T. Hien, R. McGready, J. J. Farrar, S. Looareesuwan,
N. P. J. Day, M. D. Green, and N. J. White. 2004. Fake antimalarials in Southeast
Asia are a major impediment to malaria control: Multinational cross-sectional survey
on the prevalence of fake antimalarials. Tropical Medicine & International Health
9(12):1241-1246.
DrugTopics. 2012. Strategies to combat online counterfeit drug supply. http://drugtopics.
modernmedicine.com/drugtopics/article/articleDetail.jsp?id=782394 (accessed October
22, 2012).
Ducca, A. 2011. Re: Determination of system attributes for the tracking and tracing of presrip-
tion drugs; [docket no. FDA-2010-n-0633]. 76 Fed. Reg. 1182 (January 7, 2011). http://
www.regulations.gov/#!documentDetail;D=FDA-2010-N-0633-0012 (accessed October
12, 2012).
DuPont, R. 2010. Prescription drug abuse: An epidemic dilemma. Journal of Psychoactive
Drugs 42(2):127-132.
EAASM (European Alliance for Access to Safe Medicines). 2008. The counterfeiting super-
highway. Surrey, UK: EAASM.
Efrati, A., and P. Loftus. 2010. Lilly drugs stolen in warehouse heist. Wall Street Journal,
March 17.
EMA (European Medicines Agency). 2011. Falsified medicines. http://www.ema.europa.
eu/ema/index.jsp?curl=pages/special_topics/general/general_content_000186.
jsp&mid=WC0b01ac058002d4e8 (accessed November 7, 2012).
Engelberg, A. B., A. S. Kesselheim, and J. Avorn. 2009. Balancing innovation, access, and
profits—market exclusivity for biologics. New England Journal of Medicine 361(20):
1917-1919.
EPCglobal. 2007. Electronic Product Code (EPC): An overview. http://www.gs1.org/docs/
epcglobal/an_overview_of_EPC.pdf (accessed March 7, 2013).
Erhun, W. O., O. O. Babalola, and M. O. Erhun. 2001. Drug regulation and control in
Nigeria: The challenge of counterfeit drugs. Journal of Health and Population in Devel-
oping Countries 4(2):23-34.
Exel. 2003. Role of pre-wholesalers in generic pharmaceutical manufacturers’ demand chain
management strategy. http://www.touchbriefings.com/pdf/15/pg031_t_exel.pdf (accessed
March 7, 2013).
Faucon, B., and J. Whalen. 2012. Fake Avastin took murky path to U.S. Wall Street Journal,
April 5.
FDA (U.S. Food and Drug Administration). 2005. The possible dangers of buying medicines
over the Internet. Washington, DC: FDA.
———. 2011a. CPG sec. 160.900 Prescription Drug Marketing Act—pedigree requirements
under 21 CFR Part 203. http://www.fda.gov/ICECI/ComplianceManuals/Compliance
PolicyGuidanceManual/ucm073857.htm (accessed October 2, 2012).
———. 2011b. Guidance for industry: Bar code label requirements—questions and answers.
Washington, DC: FDA.
———. 2012a. FDA news release: FDA takes action against thousands of illegal Internet phar-
macies. http://www.fda.gov/NewsEvents/Newsroom/PressAnnouncements/ucm322492.
htm#.UG2Q840J6nY.email (accessed October 22, 2012).
McCain, J. 2011. Doing more with less? Past and present challenges for the FDA. Pharmacy
and Therapeutics 36(3):145-155.
McCathie, L., and K. Michael. 2005. Is it the end of barcodes in supply chain management?
Paper read at Collaborative Electronic Commerce Technology and Research Conference
LatAm, October 3-5, 2005, University of Talca, Chile.
Medco Health. 2012. Find out about us. http://www.medcohealth.com/medco/consumer/
helpcenter/help_article.jsp?accessLink=SHP_ANON_01&faq=HelpCenterUR_Find
AboutMedcoHealth-VIPPS (accessed January 9, 2013).
Mohanta, G., P. Manna, K. Valliappan, and R. Manavalan. 2001. Achieving good pharmacy
practice in community pharmacies in India. American Journal of Health-System Phar-
macy 58(9):809-810.
MSH (Management Sciences for Health). 2005. Tanzania: Accredited drug dispensing
outlets—Duka la Dawa Muhimu. Cambridge, MA: MSH.
———. 2012. Drug seller initiatives. Cambridge, MA: MSH.
Mukherjee, R. 2012. Now send SMS to find if pill is genuine. Times of India, September 23.
Muskal, M. 2012. $80-million mystery solved? Brothers held in pharmaceutical heist. Los
Angeles Times, May 3.
NABP (National Association of Boards of Pharmacy). 2011. VIPPS information and veri-
fication site of the National Association of Boards of Pharmacy. http://vipps.nabp.net
(accessed October 22, 2012).
———. 2012a. Accreditation process. www.nabp.net/programs/accreditation/vawd/
accreditation-process (accessed October 24, 2012).
———. 2012b. Buying medicine online: Online pharmacies and you. http://www.nabp.net/
programs/consumer-protection/buying-medicine-online (accessed October 22, 2012).
———. 2012c. Find a VIPPS online pharmacy. http://www.nabp.net/programs/accreditation/
vipps/find-a-vipps-online-pharmacy (accessed October 22, 2012).
———. 2012d. Not recommended sites. http://www.nabp.net/programs/consumer-protection/
buying-medicine-online/not-recommended-sites (accessed October 22, 2012).
———. 2012e. VIPPS: Set yourself apart from rogue sites! http://www.nabp.net/programs/
accreditation/vipps (accessed October 22, 2012).
Nkamnebe, A. 2007. Attitude of Nigerian consumers toward made-in-India pharmaceutical
products. 2nd IIMA Conference on Research in Marketing. Ahmedabad, India, January
3-5.
Norman, B. 2012. User fee bill may hinge on drug tracking system. Politico, May 17.
O’Neil, M., Z. Jarrah, L. Nkosi, D. Collins, C. Perry, J. Jackson, H. Kuchande, and A.
Mlambala. 2010. Evaluation of Malawi’s emergency human resources program. Depart-
ment for International Development, Management Sciences for Health, and Management
Solutions Consulting, Ltd.
OFT (Office of Fair Trading). 2007. Medicines distribution: An OFT market study. London:
OFT.
Onwujekwe, O., H. Kaur, N. Dike, E. Shu, B. Uzochukwu, K. Hanson, V. Okoye, and P.
Okonkwo. 2009. Quality of anti-malarial drugs provided by public and private healthcare
providers in south-east Nigeria. Malaria Journal 8(22). DOI: 10.1186/1475-2875-8-22.
Orizio, G., A. Merla, P. Schulz, and U. Gelatti. 2011. Quality of online pharmacies and web-
sites selling prescription drugs: A systematic review. Journal of Medical Internet Research
13(3). http://www.jmir.org/2011/3/e74 (accessed May 1, 2013).
Palmer, E. 2012. FiercePharma: It is RIP for track and trace. http://www.fiercepharma.com/
story/it-rip-track-and-trace/2012-06-19 (accessed January 9, 2013).
Palmer, R. 2010. FDA strengthens its stance against unethical researchers. Nature Medicine
16(7):728.
Segrè, J., and J. Tran. 2008. What worls: CareShop Ghana. Washington, DC: World Resources
Institute.
Shelton, D. 2012. Fake medicine poses growing threat to consumers. Chicago Tribune, Oc-
tober 16.
Shepherd, M. 2007a. Drug importation and safety of drugs obtained from Canada. Annals of
Pharmacotherapy 41(7):1288-1291.
———. 2007b. Impact of drug importation on community pharmacy and patient care. Journal
of the American Pharmacists Association 47:319-327.
Shulman, D., K. Jobarteh, E. Makani, M. Mtwea, A. Mapwelemwe, A. Manjomo, J. Crocker,
and K. Joseph. 2009. Task-shifting in the pharmacy: A framework for expanding and
strengthening services in rural Malawi. 5th IAS Conference on HIV Pathogenesis and
Treatment.
Simonaitis, L., and C. J. McDonald. 2009. Using national drug codes and drug knowl-
edge bases to organize prescription records from multiple sources. American Journal of
Health-System Pharmacy 66(19):1743-1753.
Spies, A., and V. VanDusen. 2003. Counterfeit drugs: A menace keeps growing. U.S. Phar-
macist 28(1). http://legacy.uspharmacist.com/index.asp?show=article&page=8_1014.htm
(accessed May 1, 2013).
Sproxil. 2012. Countries. http://sproxil.com/countries.html (accessed October 1, 2012).
Srivastava, B. 2004. Radio frequency ID technology: The next revolution in SCM. Business
Horizons 47(6):60-68.
Stanton, C., A. Koski, P. Cofie, E. Mirzabagi, B. L. Grady, and S. Brooke. 2012. Uterotonic
drug quality: An assessment of the potency of injectable uterotonic drugs purchased by
simulated clients in three districts in Ghana. BMJ Open 2(3):1-7.
Stringer, J., I. Zulu, J. Levy, E. Stringer, A. Mwango, B. Chi, V. Mtonga, S. Reid, R. Cantrell,
M. Bulterys, M. Saag, R. Marlink, A. Mwinga, T. Ellerbrock, and M. Sinkala. 2006.
Rapid scale-up of antiretroviral therapy at primary care sites in Zambia: Feasibility and
early outcomes. JAMA 296(7):782-793.
Tavernise, S., and A. Pollack. 2012. FDA details contamination at pharmacy. New York Times,
October 26.
Tavrow, P., Y.-M. Kim, and L. Malianga. 2002. Measuring the quality of supervisor–provider
interactions in health care facilities in Zimbabwe. International Journal for Quality in
Health Care 14(Suppl 1):57-66.
Tavrow, P., and J. Shabahang. 2002. Vendor-to-vendor education to improve malaria treat-
ment by the private sector: A “how-to” manual for district managers. Bethesda, MD:
Quality Assurance Project.
Taylor, L. 2011. Pfizer’s DTP system “threatens Australia’s drugs supply.” Pharma Times
Online, July 11.
Taylor, P. 2010. Safety in numbers: Brazil’s medicine serialisation initiative. http://www.
securingindustry.com/pharmaceuticals/safety-in-numbers-brazil-s-medicine-serialisation-
initiative-/s40/a440 (accessed October 4, 2012).
TechNewsDaily. 2012. Internet pharmacy crackdown shutters 18,000 sites. October 5.
Tipke, M., S. Diallo, B. Coulibaly, D. Störzinger, T. Hoppe-Tichy, A. Sie, and O. Müller.
2008. Substandard anti-malarial drugs in Burkina Faso. Malaria Journal 7(95). DOI:
10.1186/1475-2875-7-95.
UNODC (United Nations Office on Drugs and Crime). 2010. The globalization of crime.
New York: UNODC.
USAID (United States Agency for International Development). 2011. Using last mile distribu-
tion to increase access to health commodities. Arlington, VA: USAID DELIVER Project.
van Dijk, D. P. J., G.-J. Dinant, and J. A. Jacobs. 2011. Inappropriate drug donations: What
has happened since the 1999 WHO guidelines? Education for Health 24(2):462.
Versel, N. 2012. Sproxil deal offers free mobile drug authentication in 17 African countries.
MobiHealthNews, August 29.
Vian, T. 2008. Review of corruption in the health sector: Theory, methods and interventions.
Health Policy and Planning 23:83-94.
Villacorta-Linaza, R. 2009. Bridging the gap: The role of pharmacists in managing the drug
supply cycle within non-governmental organizations. International Journal of Health
Planning and Management 24:73-86.
Wagner, M., J. Alonso, and A. Mehlhorn. 2001. Comparison of Internet and community
pharmacies. Annals of Pharmacotherapy 35(1):116-119.
Wasik, J. 2012. Fake medicine bought online: How to avoid counterfeits. Forbes, October 18.
Weaver, C., and J. Whalen. 2012. How fake cancer drugs entered U.S. Wall Street Journal,
July 20.
White, S., and J. Bothma. 2009. Pharmintercom: 7-11 September 2008 (part 3). South
African Pharmaceutical Journal 76(2). http://www.sapj.co.za/index.php/SAPJ/article/
view/521/478 (accessed May 1, 2013).
WHO (World Health Organization). 2008. First global conference on task shifting. http://
www.who.int/mediacentre/events/meetings/task_shifting/en/index.html (accessed October
11, 2012).
———. 2010. Assessment of medicines regulatory systems in sub-Saharan African countries:
An overview of findings from 26 assessment reports. Geneva: WHO.
———. 2011. Safety and security on the Internet: Challenges and advances in member states
(executive summary). Geneva: WHO.
Whoriskey, P. 2012. Medicare overspending on anemia drug. Washington Post, August 9.
WHPA (World Health Professions Alliance). 2011. World Health Professions Alliance (WHPA)
brief to WHO member states on spurious/falsely-labelled/falsified/counterfeit medical
products (SFFC). France: WHPA.
Wolinsky, H. 2006. Tagging products and people. EMBO Reports 7(10):965-968.
Wondemagegnehu, E. 1999. Counterfeit and substandard drugs in Myanmar and Viet Nam:
Report of a study carried out in cooperation with the governments of Myanmar and Viet
Nam. Geneva: WHO.
World Bank. 2005. HNP brief #2: Pharmaceuticals: Counterfeits, substandard drugs, and drug
diversion. Washington, DC: World Bank.
Worldmapper. 2006. Pharmacists working (map no. 17). http://www.worldmapper.org/display.
php?selected=217 (accessed November 28, 2012).
Wunder, G., and B. Roach. 2008. Electronic pedigrees and counterfeit pharmaceuticals: The
U.S. Experience with RFID. Washburn University School of Business Working Paper
Series 104.
Yadav, P. 2009. Countering drug resistance in the developing world: An assessment of incen-
tives across the value chain and recommendations for policy intervention. Working Paper
No. 183. Washington, DC: Center for Global Development.
———. 2010. In-country supply chains: The weakest link in the health system. Global Health
Magazine 5:18-19.
Yadav, P., and L. Smith. 2012. Pharmaceutical company strategies and distribution systems in
emerging markets. Elsevier Encyclopedia on Health Economics.
Yadav, P., R. Smith, and K. Hanson. 2012. Pharmaceuticals and the health sector. In Health
systems in low- and middle-income countries: An economic and policy perspective, edited
by R. Smtih and K. Hanson. Oxford, NY: Oxford University Press.
Yadav, P., O. Stapleton, and L. v. Wassenhove. 2013. Learning from Coca-Cola. Stanford So-
cial Innovation Review. http://www.ssireview.org/articles/entry/learning_from_coca_cola
(accessed March 7, 2013).
Yadav, P., H. L. Tata, and M. Babaley. 2011. The world’s medicines situation 2011: Storage
and supply chain management. Geneva: WHO.
Yao, W., C.-H. Chu, and Z. Li. 2010 June 17-19. The use of RFID in healthcare: Benefits and
barriers. Paper presented at IEEE International Conference on RFID-Technology and
Applications, Guangzhou, China.
Yukhananov, A. 2012. Congress fails to agree on national drug trace plan. Reuters, June 18.
Zaza, A. 2012. Importation of unregistered drugs in the UAE. http://www.tamimi.com/
en/publication/publications/section-3/may/importation-of-unregistered-drugs-in-the-uae.
html (accessed October 17, 2012).
Ziance, R. 2008. Roles for pharmacy in combatting counterfeit drugs. Journal of the American
Pharmacists Association 48(4):e71-e91.
Detection Technology
255
•
As criminals become more sophisticated, there will be an increased
need for expensive technologies to detect falsified medicines.
•
There are several categories of techniques to analyze pharmaceuti-
cals. They include visual inspection of product and packaging; tests
for physical properties such as disintegration, reflectance spectros-
copy, and refractive index; chemical tests including colorimetry and
dissolution; chromatography; spectroscopic techniques; and mass
spectrometry.
•
Novel technologies are constantly being developed to detect falsified
and substandard medicines.
how that content will be absorbed in the body. Qualitative assays may be
used to quickly detect the least sophisticated falsified drugs, such as those
with the wrong or no active ingredient. Quantitative deficiencies, such as
an unacceptably high level of impurities or an unacceptably low or high
dosage of active ingredient, are more common among substandard drugs.
Tests for drug quality use both qualitative data (e.g., the identity of ingredi-
ents, the presence and nature of any packaging and inserts, the presence or
absence of impurities, and any data referring to the drug’s appearance) and
quantitative data (e.g., the amount of an ingredient present, tablet hardness,
the rate and extent of disintegration and dissolution, and measured levels
of impurities).
A full evaluation of drug quality requires a range of qualitative and
quantitative testing to verify the identities and amounts of active ingredi-
ents, check for impurities, and ensure acceptable disintegration, dissolu-
tion, stability over time, and sterility (USP, 2007). Identifying falsified and
substandard drugs does not always follow the same process as a rigorous
quality evaluation. A few simple tests can identify a product with no active
ingredient or one made under gross manufacturing negligence. More so-
phisticated fakes resist easy detection. Appearance, content, and therapeutic
effect can all be considered in classifying falsified drugs. Box 6-1 outlines
one method for making categories.
Criminals in the business of making falsified drugs can buy crude active
ingredients, chemicals that have not undergone the appropriate purification
steps required to meet pharmacopeial standards or manufacturer’s dossier
requirements, for example. The drugs made from such chemicals would
pass most tests. Only highly sophisticated and expensive assays could de-
BOX 6-1
Classifying Falsified Medicines
tect trace contaminates. Figure 6-1 gives an overview of the different levels
of technology needed to catch progressively more complex falsified drugs.
TLC and colorimetry can Some falsified drugs HPLC can detect falsified
SOPHISTICATION OF DRUGS
sometimes detect falsified lacking active ingredients drugs containing an alter-
drugs containing an or containing the wrong nate drug therapy (Box
alternate drug therapy active ingredients are 6–1, category 4) and drugs
(Box 6–1, category 4) detectable with near- with the wrong amount of
and drugs with the infrared, Raman, or UV- active ingredient.
wrong amount of active visible spectroscopy
Dissolution tests can
ingredient. (Box 6–1, category 4).
detect substandard and
Physical tests such as falsified drugs with poor
Countering the Problem of Falsified and Substandard Drugs
the authentic product is the standard first step in any drug quality analysis
(Martino et al., 2010). These visual inspections provide qualitative data
about drugs’ identities. Differences from the authentic materials in color,
size, shape, tablet quality, and packaging indicate a possible falsified or
substandard drug. These differences range from subtle to obvious. An
educated consumer could probably identify a very-poor-quality fake, such
as a pill of entirely the wrong color or shape, if they knew some proper-
ties of the authentic product, but even experts struggle to recognize more
subtle inconsistencies. The Global Pharma Health Fund’s Minilab toolkit
promotes visual inspection as the first step to identifying falsified and sub-
standard drugs but admits that this is challenging even for experts (Jähnke
et al., 2008; Sherma, 2007). In recent years, criminals have produced very
accurate reproductions of legitimate packaging. And, as Chapter 4 men-
tions, poor-quality drugs can sometimes be hidden in legitimate packaging
(Sherma, 2007).
Visual inspection of a product can yield useful information, however.
Some substandard drugs are of visibly low quality. Tablets that are cracked
or falling apart are products of poor manufacturing practices (Kaur et al.,
2010). Falsified drugs’ packaging may have missing or misplaced expiry
dates, lack instructions or manufacturing information, not have a batch
number, or differ from the genuine packaging in many other ways. Some-
times poorly written instructions and spelling errors expose fake medicines;
poor-quality inks may dissolve in water (Kaur et al., 2010). Similarly, the
drugs may be the wrong color, size, or shape, have the wrong markings on
them, have a different coating or texture, or be otherwise different from
what is expected (Kaur et al., 2010). Sometimes the differences are obvi-
ous: fake Viagra seized in Hungary was pink instead of the well-known
blue color of the genuine product. Further analysis revealed that the tablets
contained 15 milligrams of amphetamine instead of the correct active ingre-
dient (U.S. Drug Enforcement Administration Office of Forensic Sciences,
2004).
Visual inspections are often unreliable because substandard and falsi-
fied drugs and their packaging often appear identical or very similar to the
genuine products. Criminals have copied holograms, barcodes, packaging
styles, and tablet colors and markings with astonishing accuracy (Lim,
2012). Microscopic packaging analysis can identify some of these very care-
ful copies. Under magnification, fine differences in printing, imprints, and
alignment become clear. Figure 6-2 shows a high-magnification comparison
of the lettering on a legitimate and fake blister pack. As this illustration sug-
gests, visual inspection alone is not adequate to test for drug quality (Lim,
2012; Martino et al., 2010). Though a trained inspector can draw conclu-
sions about drug quality by visual inspection, physical analysis is generally
a more reliable way to identify fakes.
FIGURE 6-2 The printing on a fake Cialis blister pack is less crisp at 32× magnification.
SOURCE: Lim, 2012.
Chromatography
Chromatography separates mixtures into their constituent parts based
on a variety of chemical and physical properties. It can be used to separate
drug ingredients for further testing and, when used with appropriate detec-
tors, provides both qualitative and quantitative information about active
ingredients and impurities (Kaale et al., 2011). Chromatography is there-
fore the most common analytical method used in drug evaluations (Martino
et al., 2010). Chromatographic techniques range from basic techniques,
such as thin layer chromatography (TLC) with visual inspection, to more
specialized laboratory methods, such as high-performance liquid chroma-
1,000
800
Mefloquine
Absorbance (mAU)
600
Chloroquine Quinine
400
200
0
1 2 3 4 5 6 7 8 9 10
Time (minutes)
CF3
N N CF3
HN HO
N
O
H H
Cl N N
N HO
and countries most affected by falsified and substandard drugs have lim-
ited access to such facilities (IOM, 2012). HPLC and gas chromatography
are time-consuming, especially considering the time spent preparing the
samples for analysis. The return on the time investment is mixed, as chro-
matography separates a minimum number of components present in a
sample. A peak assumed to represent one compound may be hiding several
other compounds.
Spectroscopy
Spectroscopy is a class of analytical techniques that measures the in-
teraction of matter and radiation, thereby giving insight into chemical
structure and contents. These techniques all provide qualitative data, and
some provide significant quantitative data as well. Often referred to as the
chemical fingerprints of drugs, the various spectra produced using these
techniques elucidate different aspects of drug composition; characteristic
absorption or emission peaks correspond to aspects of chemical composi-
tion and molecular structure. A chemist can extract detailed chemical and
structural information from a spectrum, and an inspector with minimal
training can also identify falsified and substandard medicines by comparing
the drug spectra to reference materials in drug spectra databases (Kaur et
al., 2010). The WHO maintains a digital version of the International Phar-
macopoeia with drug quality determination protocols for many common
medicines (WHO, 2011). This guide includes a reference infrared spectrum
for each drug.
Molecular vibration and rotation energies occur in the infrared regions
of the electromagnetic spectrum, and these movements may be observed
with infrared, near-infrared, or Raman spectrometers. These techniques
are relatively straightforward to use and moderately expensive, and routine
comparative applications do not require extensive training. Chemists ana-
lyze the absorption peaks in these spectra primarily to identify molecular
functional groups; most active pharmaceutical ingredients and some or-
ganic excipients and impurities have characteristic spectral peaks or spectral
fingerprints that can be used to help identify them.
80
CH3
60 O O
H O
%T
H3 C
O
40 H
O
H
CH3
20
0
3600 3200 2800 2400 2000 1800 1600 1400 1200 1000 800 600 400
cm-1
100
80
Countering the Problem of Falsified and Substandard Drugs
CH3
H
O
60 H3 C
O
O
%T
H H
40 O
CH3
O
20 H3C
0
3600 3200 2800 2400 2000 1800 1600 1400 1200 1000 800 600 400
cm-1
FIGURE 6–4 Infrared Spectra for artemisinin (top) and artemether, an artemisinin derivative (bottom)
FIGURE 6-5 Use of FT-IR spectroscopy to distinguish between real (left) and falsified
(right) packaging in Singapore.
SOURCE: Lim, 2012.
FIGURE 6-6 Image (a) is a pain relief tablet, image (b) its near-infrared spectra. The
red spots indicate active ingredient and other colors indicate other ingredients.
SOURCE: Adapted from Koehler et al., 2002. Reprinted with permission from John Wiley & Sons, Ltd.
14 12 10 8 6 4 2 0
ppm
fuller, clearer molecular picture can be developed. Using these methods, sci-
entists have successfully differentiated between many authentic and falsified
versions of antimalarials, erectile dysfunction drugs, and antidepressants
(Martino et al., 2010).
X-ray diffraction and X-ray fluorescence are other techniques that can
give substantial information about drug contents. X-ray diffraction can be
used to analyze active ingredients and excipients, while X-ray fluorescence
is used for elemental analyses that can often distinguish real from falsified
drugs (Kaur et al., 2010; Martino et al., 2010).
Mass Spectrometry
Mass spectrometry, generally called mass spec, is a sophisticated ana-
lytical technique that requires extensive training and expertise to use. It
provides abundant structural information and the precise molecular weight
of the compound under investigation. Mass spec can identify many ac-
tive ingredients and excipients, as well as some impurities (Kaur et al.,
2010; Martino et al., 2010). This technique successfully detected falsified
halofantrine syrup, an antimalarial, in West Africa that instead contained
a sulphonamide antibiotic (Wolff et al., 2003). When mass spectrometers
were the size of a dishwasher (Stroh, 2007), their value in the poorest
countries was hard to realize, but newer, portable machines can take this
sophisticated technology into the field (Yang et al., 2008). However, mass
spectrometers require a stable electrical power source, which may be dif-
ficult to obtain in some developing countries.
An isotope ratio mass spectrometer provides detailed information
about the abundance of various elemental isotopes. Many elements have
naturally occurring isotopes that are present in minute quantities in any
sample. The exact ratio of isotopes varies over time and space and with dif-
ferent production techniques. Isotopic ratios have been able to distinguish
different sources of drugs and therefore may be useful for combating highly
sophisticated copies (Lim, 2012). Regulators and law enforcement can use
isotopic ratios to connect seemingly disparate events and build evidence
that separate drug seizures have a common source. Documenting the iso-
topic ratios of a selection of common elements, such as carbon, hydrogen,
oxygen, or nitrogen, can help identify these patterns (Lim, 2012).
Other kinds of mass spectrometry (e.g., direct ionization, tandem,
time-of-flight, secondary ion, and electrospray ionization [ESI]) can be used
alone and in combination with other analyses to detect illegitimate drugs
(Deisingh, 2005; Martino et al., 2010; Wolff et al., 2003). Direct ionization
mass spec, for one, is a relatively new class of mass spectrometric analysis
that does not require lengthy sample preparation. Other techniques, such as
direct analysis in real time (DART) mass spec and desorption ESI mass spec,
can identify correct and incorrect active ingredients and some excipients.
Desorption ESI mass spec in particular provides information about tablet
surface homogeneity and the distribution of active ingredients and excipi-
ents in or on the surface of a tablet (Martino et al., 2010). For example,
an artesunate sample with homogeneous surface distribution of lactose
and paracetamol, a fever reducer, is illegitimate; an authentic, good-quality
sample should have homogeneous distribution of artesunate and scattered
distribution of lactose (Martino et al., 2010).
The most sophisticated drug copies may resist identification with any
technology other than mass spectrometry. Among these are very close ana-
logues of genuine active ingredients. These analogues can be so chemically
and structurally similar that they behave the same under nearly any analy-
sis. Mass spectrometry’s ability to precisely measure molecular weight and
compare fragmentation patterns can help distinguish between compounds
that differ by only one or two atoms. For example, the erectile dysfunction
drug Cialis is often copied with varying degrees of sophistication (Putze et
al., 2012; Trefi et al., 2008). U.S. Food and Drug Administration (FDA)
forensic chemists have discovered several analogues of the active ingredi-
ent, tadalafil, in so-called herbal remedies (Gamble et al., 2008). Figure 6-8
compares the molecular structure of one such analogue, aminotadalafil, to
tadalafil. The two differ only by the substitution of an amino (-NH2) group
for a methyl (-CH3) group, making aminotadalafil slightly heavier. The
health threats posed by such products have led researchers to investigate
ways of reliably detecting and identifying these illicit drug compounds;
other sophisticated techniques have been shown to detect some analogues,
but the high specificity and sensitivity of mass spec makes it the most popu-
lar method (Singh et al., 2009; Venhuis and Kaste, 2012).
CH3 NH2
N O N O
O N O N
O O
O HN O HN
Emerging Technologies
Innovative technologies to detect falsified and substandard drugs are
constantly emerging. Many of the most promising examples draw from
a range of scientific disciplines. Researchers in Pakistan found that the
relative susceptibility of ofloxacin-sensitive bacteria to various samples of
ofloxacin is a good indicator of drug quality (Iqbal et al., 2004). A team of
researchers from U.S. Pharmacopeia and Boston University is developing
another new technology called PharmaCheck (see Box 6-2). PharmaCheck
uses microfluidics, the control of fluids at a sub-millimeter scale, for rapid
field drug testing (EurekAlert, 2012). PharmaCheck, which will weigh less
than 10 pounds and fit in a shoebox, promises to greatly reduce the need
for confirmatory laboratory testing (Barlow, 2012; Gaffney, 2012).
Capillary electrophoresis, a separation technique, has recently been
BOX 6-2
PharmaCheck
USING TECHNOLOGY
The previous section outlined the main categories of techniques for
detecting falsified and substandard drugs. A summary of a selection of the
techniques discussed is presented in Table 6-1. Understanding when, where,
and why to use the various techniques can be difficult. The information a
technique provides, as well as its reliability, cost, required expertise, speed,
and portability make it more or less appropriate in any given situation.
In order to conclude that a drug is of good quality, an inspector must
test a sample for all of the main deficiencies of substandard and falsified
drugs: fake packaging, incorrect color, shape, or markings, absent or incor-
rect active ingredients, incorrect quantities of ingredients, impurities, and
reduced dissolution or disintegration. Table 6-2 outlines which classes of
analytical techniques can test for these problems and how well they can be
used in the field. In general, field use describes a relatively straightforward
assay or technique that depends on portable or sturdy equipment. Most
field methods can be used by professionals such as regulators, pharmacists,
or health workers, but some, like mobile verification, are accessible to a
layperson.
The Counterfeit Drug Forensic Investigation Network (CODFIN) uses
a systematic analytical process to detect and classify substandard and falsi-
fied drugs (Fernandez et al., 2011). Figure 6-9 shows how investigators in
the network test samples in national drug quality surveys (Fernandez et
al., 2011). The steps shown in green can be done in the field, but samples
are generally sent to a central laboratory for the steps show in brown
(Fernandez et al., 2011). Fernandez and colleagues have used this system
Detection Techniques
Chemical or Spectroscopy Physical
Visual Package Microbiological and Mass Measurements
Problems Inspection Technologies Tests Spectrometry or Examination Chromatography Reflectance
Absent or incorrect
active ingredient
Wrong color
Wrong shape or
markings
Countering the Problem of Falsified and Substandard Drugs
Fake packaging
Incorrect quantities of
ingredients
Impurities
Dissolution
Disintegration
Uniformity of dosage
units
Microbial
contamination
Subset 3
(if available)
Wrong AIs,
Ambient MS analysis
excipients, coatings, Y Identify sample Quantify by
Forward or store (accurate mass Any toxic?
or packaging ingredients HPLC
subset 3 for fingerprints)
Countering the Problem of Falsified and Substandard Drugs
defects
national medicine
regulatory agency N
Ambient MS analysis
Degradation
AI amount N (selective reaction Y
products Degraded
within +15% monitoring of
present?
degradation products)
Y
N
Dissolution
Substandard FIGURE 6–9 CODFIN analysis flowchart.
testing
NOTE: AI = active ingredient; CODFIN = Counterfeit
Drug Forensic Investigation Network; DOSY NMR = diffu-
sion ordered spectroscopy nuclear magnetic resonance;
HPLC = high-performance liquid chromatography; IRMS =
isotope-ratio mass spectrometry; MS = mass spectrometry;
Combining Techniques
Although any one test may suffice to label a drug substandard or falsi-
fied, no single analytical technique provides enough information to con-
firm that a drug is genuine. Similarly, while colorimetry and TLC are field
techniques for testing for the presence of a particular ingredient, knowing
a sample’s full content requires more testing. Spectroscopic techniques are
useful for identifying active ingredients but cannot rule out the presence
of countless possible impurities. Chromatographic techniques may suggest
that the drug contains sufficient active ingredient, but they do not provide
any information about how much of that active ingredient will reach the
patient. Time and budget allowing, the best understanding of drug quality
comes from the several complementary experiments.
Even combinations of techniques from within a class, such as spec-
troscopy, can be helpful. One study illustrated how, due to differences in
the ranges of their spectral regions, infrared spectroscopy may at times be
better at identifying organic substances in tablet coatings, whereas Raman
spectroscopy may better identify the inorganic components (Witkowski,
2005). Experiments that looked at the coating on Cialis tablets found that
Klaus Boehm of Merck presents minilabs to Hiti Sillo, head of the Tanzanian Food and
Drugs Authority.
SOURCE: GPHF, 2012e.
Raman spectroscopy did not distinguish between the real coating and falsi-
fied coating, but infrared spectroscopy did (Lim, 2012).
Chemists typically pair mass spec with separation techniques, such as
HPLC, to achieve a more definitive analysis. These hyphenated techniques
have broad capabilities. For example, liquid chromatography-mass spec-
trometry is a highly reliable separation technique, but does not directly
provide quantitative data about the amount of active ingredient present;
analysts must compare results to standards to determine content (Kaur
et al., 2010). A type of combined gas chromatography and mass spec
(GC-MS) can provide information missing in HPLC-MS analysis. Mulligan
and colleagues found that automated equilibrium headspace sampling with
capillary gas chromatography provides information about volatile impuri-
ties, but adding mass spec analysis provides extra qualitative information
about the identity of any impurities present (Mulligan et al., 1996).
When Captagon, a stimulant drug popular in the Middle East, was out-
lawed, illegal manufacturers began selling the drug (Alabdalla, 2005). The
copies were generally falsified drugs containing amphetamines and caffeine
meant to mimic Captagon’s therapeutic effects. Early investigations primar-
ily relied on ultraviolet, infrared, and TLC analysis to determine the active
ingredients in suspect tablets (Alabdalla, 2005). In 2005, Alabdalla and
Field Technologies
Technologies for field detection of falsified and substandard drugs in
developing countries must be portable, relatively simple to use, sturdy, and
inexpensive to buy, use, and maintain. They must also provide reliable,
useful data. Field techniques (including visual inspection, colorimetry, disin-
tegration tests, TLC, and handheld spectrometry) can detect many falsified
and substandard drugs. As the previous section explains, these techniques
are durable, fast, relatively inexpensive, and fairly easy to use, making them
attractive to regulators interested in monitoring drug quality. Box 6-3 de-
scribes the Chinese regulatory authority’s mobile verification labs. Package
verification technologies can also aid in field detection of falsified drugs,
although these methods are useful more to the patient at the point of use
than to the regulator.
The more reliable field analytic tools are also more expensive. Although
fairly inexpensive TLC and disintegration testing are useful field techniques,
BOX 6-3
Chinese Mobile Laboratories
The Chinese drug regulatory authority uses mobile labs for drug
surveillance (Jin, 2007). First used in Henan province in March 2006,
mobile labs quickly spread to 29 provinces (Jin, 2007). The mobile lab
program also trained 760 technicians to operate the labs, which bring
drug screening technology to rural areas (Jin, 2007).
The mobile labs, housed in vans, can carry out rapid on-site screen-
ing of suspicious drugs (NICPBP, 2012). Each van carries chemical analy-
sis technologies, including TLC systems, a near-infrared spectrometer,
and portable computers (NICPBP, 2012). The vans also house information
on 200,000 manufacturers, including names, addresses, and licensed
products, as well as a provincial “Drug Quality Bulletin” with annually
updated information on known poor-quality drugs (Jin, 2007). The labs’
operations are designed to be simple, fast, and easily executed (NICPBP,
2012). A mobile lab can test the quality of more than 800 drugs, includ-
ing antimalarials, antiretroviral, tuberculosis medication, other essential
drugs, and traditional Chinese herbal medicines (Jin, 2007; NICPBP,
2012).
In the first 6 months of operation, mobile labs screened 110,426
batches of drugs and confirmed 3,122 of them to be substandard (Jin,
2007). The project’s success has inspired the regulatory authority to
help other countries develop similar mobile labs and to use them in drug
procurement (Jin, 2007). Members of the Thai FDA, for example, visited
Chinese mobile labs in 2006 (Jin, 2007).
according to Bate they are less reliable than handheld spectrometric devices
(Bate et al., 2009a). Of 78 samples tested in one study, 17 passed both TLC
and disintegration tests but did not pass either Raman or near-infrared
spectroscopic analysis (Bate et al., 2009a). Field tests are no substitute for
definitive laboratory techniques and cannot test all aspects of a product’s
quality, including its drug content, impurity profile, and dissolution profile.
Noting the cost of laboratory pharmaceutical testing and the dearth
of qualified laboratories in developing countries, the German Pharma
Health Fund (now known as the Global Pharma Health Fund) devel-
oped the Minilab, a portable quality-analysis laboratory described in Box
6-4 (Jähnke et al., 2001; Kaale et al., 2011). During a November 2012
Minilab training session in Angola, trainees tested an illegal shipment of
various pharmaceuticals seized by customs officials along the African coast
(Minilab Saves Lives, 2012; World Customs Organization, 2012). Using
the TLC and visual inspection techniques, they identified many drugs with
no or little active ingredient (Minilab Saves Lives, 2012). Merck S.A. in
Portugal provided 10 Minilabs to Angola, which has no drug testing labs
(Minilab Saves Lives, 2012). Other similar field kits also exist, such as the
Thermo Scientific FirstDefender and TruDefender field laboratory devices
used by the Singaporean regulatory authority (Lim, 2012).
BOX 6-4
The Global Pharma Health Fund Minilab
Each Minilab fits into two suitcases for durable portability. Price and
simplicity guided the kit’s design; the solvents and reagents used in the
assessments are safe for use with very little training and are widely avail-
able and inexpensive. Each Minilab quality test costs no more than $3 to
run (GPHF, 2012c; Kaale et al., 2011).
The kit includes equipment and instructions for thin layer chroma-
tography (TLC), chemical colorimetry, and disintegration tests, as well
as a visual inspection protocol. Testing and inspection protocols and
materials are included for more than 50 World Health Organization es-
sential medicines, including reference standards for 63 drug compounds
(GPHF, 2012a; Kaale et al., 2011). By using colorimetry, which tests for the
identity of active ingredients, and TLC, which provides information about
potency, the kit is capable of testing for the top three kinds of substan-
dard and falsified drugs: those that contain no active ingredient, those
that contain too little active ingredient, and those that contain the wrong
active ingredient (GPHF, 2012c; Jähnke et al., 2001). Since the reliability
of TLC is based in large part on the tester’s level of training, the Minilab
attempts to simplify the analysis by providing reference tablets that can
be used to prepare 100 percent and 80 percent dosage strengths for
comparison (Kaale et al., 2011).
Currently, there are more than 500 Minilabs in 80 countries, and
many prominent national and international organizations recommend the
kits for field testing (GPHF, 2012c). The U.S. Pharmacopeial Convention
distributes and administers trainings for Minilabs in developing countries
through its Drug Quality and Information program, in collaboration with
USAID (Smine and Hajjou, 2009). The lab has also been used by the
WHO’s Roll Back Malaria program and by several local nongovernmental
organizations in countries such as Tanzania and Ghana (Jähnke et al.,
2001).
tion technology can replace stringent drug regulation in the fight against
falsified and substandard drugs. The sentiment that no one can test quality
into drugs is true to a certain extent. It is important to be able to test drug
quality, but also important to impose good manufacturing practices on
companies to prevent quality problems before they arise. However, effective
use of technology can help improve drug quality. A study on drug quality
in Nigerian pharmacies before and after handheld spectrometers were dis-
tributed indicated that drug quality improved when testing became more
reliable and convenient (Bate and Mathur, 2011).
Making detection technology more accessible in low- and middle-
income countries is invaluable to controlling the trade in falsified and sub-
FIGURE 6-10 Genuine (left) and falsified (right) holograms on artesunate blister packs
found in Southeast Asia.
SOURCE: Newton et al., 2008.
standard drugs. Technologies can protect consumers and also help generate
accurate estimates of the magnitude of the problem. An understanding of
the technological landscape, the range and gaps in available technologies,
and the likely improvements in the near future is necessary for using tech-
nologies in developing countries.
At a Minilab training session in Angola, field inspectors learn how to test drug quality.
SOURCE: Minilab Saves Lives, 2012.
Physical and bulk property Varies, but usually identifying the active Varies
testing (e.g., density, ingredient
solubility, refractive index)
and analysis. All of the methods described in this chapter, for example, are
relevant to small molecules, but hormones, oral contraceptives, low-dose
vaccines, and biologics are also vulnerable to quality failures, failures that
are much harder to detect. Even the existing technologies to detect falsi-
fied and substandard small molecules could be improved. For example, the
Minilab, a useful and elegant kit, can test only 63 drugs (GPHF, 2012b).
The Global Pharma Health Fund should expand this inventory; the WHO
should help identify which products are the first priority for inclusion.
Similarly, expansion of the Raman active ingredient database would
make handheld Raman spectrometers more useful in detecting falsified
drugs. All drug detection technologies would be more powerful if there
were a full authentication database with information about drug color,
shape, size, weight, Raman and near-infrared reflectance, and a TLC proce-
dure for assay. Drug companies may balk at releasing this information, but
the committee believes that stringent regulatory agencies should require it.
Sharing all drug authentication information in a drug quality library would
vastly improve the power of existing drug detection technologies.
Level of Used in
Training Speed Field? Example
Low Fast Yes A sample of falsified Viagra in Hungary was pink
instead of the correct blue color. Further analysis
revealed that the tablets contained 15 mg amphetamine
instead of the correct active ingredient (U.S. Drug
Enforcement Administration Office of Forensic
Sciences, 2004).
Low Fast Yes Fast Red TR dye turns yellow in the presence of
artesunate (Green et al., 2001).
Low Fast Yes Close to 12% of drugs sampled from Delhi in a study of
drug quality in India failed disintegration testing (Bate
et al., 2009b).
continued
Level of Used in
Training Speed Field? Example
High Slow No Organic volatile impurities detected by GC can help link
different batches of falsified drugs back to common
manufacturers (Mulligan et al., 1996).
Moderate Fast Yes Was able to distinguish real from falsified artesunate
tablets with 100% accuracy in an analysis of samples
from Southeast Asia (Dowell et al., 2008).
Low Fast Yes Falsified artesunate samples did not produce the strong
fluorescence characteristic of artesunate when scanned
with a portable device (Ricci et al., 2008).
REFERENCES
Alabdalla, M. A. 2005. Chemical characterization of counterfeit Captagon tablets seized in
Jordan. Forensic Science International 152:185-188.
Barlow, R. 2012. A new counterfeit problem: Anti-malarial drugs. BU Today, July 26.
Barras, J., K. Althoefer, M. D. Rowe, I. J. Poplett, and J. A. S. Smith. 2012. The emerging
field of medicines authentication by nuclear quadrupole resonance spectroscopy. Applied
Magnetic Resonance 43(4):511-529.
Bate, R., and A. Mathur. 2011. Working paper: The impact of improved detection technology
on drug quality: A case study of Lagos, Nigeria. Washington, DC: American Enterprise
Institute.
Bate, R., R. Tren, K. Hess, L. Mooney, and K. Porter. 2009a. Pilot study comparing technolo-
gies to test for substandard drugs in field settings. African Journal of Pharmacy and
Pharmacology 3(4):165-170.
Bate, R., R. Tren, L. Mooney, K. Hess, B. Mitra, B. Debroy, and A. Attaran. 2009b. Pilot study
of essential drug quality in two major cities in India. PLoS ONE 4(6):e6003.
Brown, T., H. E. LeMay, B. Bursten, C. Murphy, and P. Woodward. 2011. Chemistry: The
central science. 12th ed. Boston, MA: Prentice Hall.
Deisingh, A. K. 2005. Pharmaceutical counterfeiting. Analyst 130(3):271-279.
Dowell, F. E., E. B. Maghirang, F. M. Fernandez, P. N. Newton, and M. D. Green. 2008.
Detecting counterfeit antimalarial tablets by near-infrared spectroscopy. Journal of Phar-
maceutical and Biomedical Analysis 48:1011-1014.
EurekAlert. 2012. New technology represents next-generation tool for detecting substan-
dard and counterfeit medicines. http://www.eurekalert.org/pub_releases/2012-07/up-
ntr072612.php (accessed January 31, 2013).
Fernandez, F. M., M. D. Green, and P. N. Newton. 2008. Prevalence and detection of coun-
terfeit pharmaceuticals: A mini review. Industrial and Engineering Chemistry Research
47:585-590.
Fernandez, F. M., D. Hostetler, K. Powell, H. Kaur, M. D. Green, D. C. Mildenhall, and P. N.
Newton. 2011. Poor quality drugs: Grand challenges in high throughput detection, coun-
trywide sampling, and forensics in developing countries. Analyst 136(15):3073-3082.
Filho, A. F. M., P. Blanco, and L. E. Ferreira. 2010. Pharmaceutical products traceability sys-
tem pilot project in Brazil. 2010/2011 GS1 Healthcare Reference Book. http://www.gs1.
org/docs/healthcare/case_studies/Case_study_Brazil_pharma_traceability.pdf (accessed
March 7, 2013).
Gaffney, A. 2012. New portable anti-counterfeiting technology promises “paradigm shift.”
http://www.raps.org/focus-online/news/news-article-view/article/2020/new-portable-anti-
counterfeiting-technology-promises-paradigm-shift.aspx (accessed March 7, 2013).
Gamble, B. M., S. R. Gratz, J. J. Litzau, K. J. Mulligan, and R. A. Flurer. 2008. FDA forensic
investigations using mass spectrometry. Paper presented at Conference on Small Molecule
Science, San Jose, CA.
Gaudiano, M. C., E. Antoniella, P. Bertocchi, and L. Valvo. 2006. Development and valida-
tion of a reversed-phase lc method for analysing potentially counterfeit antimalarial
medicines. Journal of Pharmaceutical and Biomedical Analysis 42:132-135.
Gottlieb, H. E., V. Kotlyar, and A. Nudelman. 1997. NMR chemical shifts of common labora-
tory solvents as trace impurities. Journal of Organic Chemistry 62:7512-7515.
GPHF (Global Pharma Health Fund). 2012a. GPHF-Minilab®—fact sheet. http://www.gphf.
org/web/en/minilab/factsheet.htm (accessed July 30, 2012).
———. 2012b. The GPHF-Minilab®—focusing on prevalent medicines against infectious dis-
eases. http://www.gphf.org/web/en/minilab/wirkstoffe.htm (accessed November 5, 2012).
Wilkinson, N. 2012. Briefcase encounter: An invention to detect fake drugs. Wellcome Trust.
http://wellcometrust.wordpress.com/2012/11/12/briefcase-encounter-an-invention-to-
detect-fake-drugs (accessed March 7, 2013).
Witkowski, M. R. 2005. The use of Raman spectroscopy in the detection of counterfeit and
adulterated pharmaceutical products. American Pharmaceutical Review 8:56-62.
Wolff, J.-C., L. A. Thomson, and C. Eckers. 2003. Identification of the “wrong” active
pharmaceutical ingredient in a counterfeit Halfan™ drug product using accurate mass
electrospray ionisation mass spectrometry, accurate mass tandem mass spectrometry and
liquid chromatography/mass spectrometry. Rapid Communications in Mass Spectrometry
17:215-221.
World Customs Organization. 2012. High-impact customs operation tackles illicit medi-
cines in Africa. http://www.wcoomd.org/en/media/newsroom/2012/october/high-impact-
customs-operation-tackles-illicit-medicines-in-africa.aspx (accessed November 14, 2012).
Yang, M., T.-Y. Kim, H.-C. Hwang, S.-K. Yi, and D.-H. Kim. 2008. Development of a palm
portable mass spectrometer. Journal of the American Society for Mass Spectrometry
19:1442-1448.
Zook, A. 2012. Detection of counterfeit pharmaceuticals: Merck case study. Presentation at
Committee on Understanding the Global Public Health Implications of Substandard,
Falsified, and Counterfeit Medical Products: Meeting One, Institute of Medicine, Wash-
ington, DC.
295
quality control errors as crimes (Attaran and Bate, 2010). And, though
Susanne Keitel, the director of the European Directorate for the Quality of
Medicines, explained to the committee in March that the Medicrime Con-
vention does not cover infringement of intellectual property rights, some see
hostility to generics companies in the treaty (Attaran and Bate, 2010). This
impression is fueled by the recent memory of European Union (EU) customs
officials seizing as counterfeit generic drug shipments produced in India
and bound for Africa or Latin America (EUbusiness, 2010; Reuters, 2011).
The Anti-Counterfeiting Trade Agreement (ACTA) is the other treaty
relevant to falsified medicines. ACTA sets international standards for in-
tellectual property protection and creates a regime outside of the World
Trade Organization (WTO) and the World Intellectual Property Organiza-
tion (WIPO) to protect intellectual property (Ilias, 2012). Eight countries3
signed ACTA in October 2011, but Japan is the only country that has for-
mally ratified the treaty (MOFA of Japan, 2012; USTR, 2011). ACTA will
come into force only when six countries ratify it (USTR, 2011).
As Chapter 1 explains, this report is not concerned with intellectual
property rights. The committee believes that the real or perceived mixing
of public health and intellectual property concerns only holds back action
on the problem of falsified and substandard drugs.
3 Australia, Canada, Japan, Morocco, New Zealand, Singapore, South Korea, and the
United States have signed ACTA (USTR, 2011).
4 World Health Organization, Basic Documents, Constitution of the World Health Organi-
nization, 45th ed., Supplement, October 2006. Chapter V, Article 18(a), (c), (d), (f), (h), (j).
Engaging Stakeholders
In developing the proposed code of practice, the WHO should engage
all major stakeholders; the inclusion of scientific experts and civil society
6 World Health Organization, Basic Documents, Constitution of the World Health Organi-
zation, 45th ed., Supplement, October 2006. Chapter V, Article 20, Article 23.
groups is essential. The WHO Essential Medicines division can bring great
technical depth to the discussion, especially the public health aspects of the
problem. Because the problem has legal dimensions, it will also be crucial
to include experts in law enforcement, criminal justice, and customs. In
order to assure the proper range of expertise in the drafting of the proposed
code of practice, the committee recommends that the WHO work with the
UNODC and the WCO.
The UNODC helps member states fight organized crime, trafficking,
corruption, and terrorism (UNODC, 2012a). Its previous work has de-
scribed the trade in illegitimate medicines as the business of terrorist orga-
nizations and criminal cartels (UNODC, 2009, 2011, 2012b). The agency’s
2012-2015 strategy emphasizes that responding to transnational, organized
crime is a priority, and it highlights their work against new kinds of drug
trafficking (UN, 2012). Contributing to the law enforcement and criminal
justice sections of an international code on falsified and substandard medi-
cines would draw on the agency’s strengths and complement the goals set
out in its 3-year strategy.
The WCO, the only international organization dedicated to policing
flows of goods into and out of countries, is the other stakeholder orga-
nization that should contribute to the proposed code. The WCO works
on supply chain security and on the harmonization of simplified customs
procedures (WCO, 2012a). National customs offices are under pressure to
facilitate international trade and to monitor the safety of products enter-
ing the country; they have a unique understanding of the circumstances
through which illegitimate medicines enter commerce. The inclusion of
the WCO in the development of an international code on falsified and
substandard drugs could help ensure the code’s validity to stakeholders
in customs.
The committee recognizes that some stakeholders might object to the
inclusion of the WCO in this process, given the organization’s professed
commitment to protecting intellectual property rights (WCO, 2012b).
Monitoring the trade in illegitimate medicines and enforcing laws against
them depend on customs bureaus, however. Failing to include them in the
development of the code would risk its being unacceptable or impractical
for customs officers, one of the main groups that would need to adhere to
it. While some stakeholders might not approve of the WCO, the committee
sees no value in excluding them from the discussion.
Global governance for health increasingly requires health organiza-
tions such as the WHO to work with other international agencies. There
is precedent for the WHO’s forming partnerships in the development of a
code of practice. The WHO and Unicef collaborated on the International
Code of Marketing of Breast-Milk Substitutes, and, since its release in
1981, 84 countries have enacted legislation implementing all or many of
its provisions (Unicef, 2012; WHO, 1981). Unicef continues to work with
legislators and lawyers to implement maternity protection laws in more
countries (Unicef, 2012). Given the clear relationship between maternal and
child health and Unicef’s mission, it was only appropriate for the WHO
to engage this organization. Similarly, a partnership with the UNODC and
WCO would benefit the development and implementation of the proposed
code of practice.
International Surveillance
As Chapter 3 explains, surveillance for substandard and falsified drugs
is uncoordinated, largely voluntary, and highly variable. The modern drug
Medicines Regulation
The proposed code of practice should give guidelines on the quality,
safety, and efficacy of medicines that all countries can work toward. The
code could suggest national minimum standards for licensing of importers,
distributors, and wholesalers and guidelines on retail and dispensing of
medicines. The WHO has already collected most of this information; the
Medicines Regulatory Package will be a useful reference on how to organize
regulatory authorities and monitor their performance (WHO, 2011).
The code should direct countries to enact comprehensive medicines
legislation that provides for all the drug regulatory functions, including
the licensing of manufacturers and distributors, the issuing of market au-
Law Enforcement
Guidelines for surveillance and drug regulation will be central to a
code of practice on substandard and falsified drugs. This report makes
clear, however, that the problem cannot be solved without input from law
enforcement, a broad category that includes disparate agencies with limited
budgets and competing priorities.
The nature of pharmaceutical crimes and the constraints on law en-
forcement agencies pose challenges to prosecuting and punishing offenders.
The illegitimate drug business is a global industry that mirrors legitimate
business in many ways: it sources materials from around the world and
bases manufacturing in countries with the cheapest labor and most favor-
able regulatory regimes. Criminals and unscrupulous manufacturers use
the internet to identify suppliers and customers. They may also sell drugs
over the internet, on the black market, or even through legitimate distribu-
tion channels. Thorough investigation and successful prosecution of those
responsible is difficult and expensive because of clandestine manufacturing
and distribution networks.
Pharmaceutical crime covers a spectrum of low-risk, high-reward of-
fenses. Many countries have not enacted laws making these acts crimes or
set out terms for international cooperation on investigations (Attaran et al.,
2011). The code of practice on falsified and substandard medicines could
give guidelines on how to investigate and punish the illegitimate medicines
trade, as well as standard minimum punishments for different crimes. The
code could also establish common definitions for different criminal acts
such as the manufacture of an illegitimate drug, the unauthorized reuse of
packaging, tampering with any documents or receipts necessary to recreate
the chain of custody, and knowingly selling or distributing an illegitimate
product.
A code of practice would build momentum for international coop-
eration on the investigation of pharmaceutical crimes. The national police
agencies’ authority stops at the border. Investigating transnational crimes
sometimes requires mutual legal assistance treaties (Attaran et al., 2011;
Palmer, 2012). Pharmaceutical crimes are particularly time-consuming and
expensive to investigate. They put novel demands on the detectives and
prosecutors who are expected to work on homicides and other violent
crimes. The code of practice could suggest guidelines for police agencies on
how to balance priorities. It could also give political cover to police agen-
cies looking to direct more staff time to investigating crimes against the
drug supply. The code would also establish guidelines for both choosing
the venue to prosecute and the terms for extradition.
Chapter 4 describes how police and customs officers may channel their
work against falsified drugs in brief, intense campaigns and not in sus-
REFERENCES
Adbul-Rahman, L. 1996. Financing of a drug regulatory authority and user fees. Paper read
at Proceedings of the Eighth International Conference of Drug Regulatory Authorities
(ICDRA), Bahrain.
Attaran, A., and R. Bate. 2010. A counterfeit drug treaty: Great idea, wrong implementation.
Lancet 376(July):1446-1448.
Attaran, A., R. Bate, and M. Kendall. 2011. Why and how to make an international crime of
medicine counterfeiting. Journal of International Criminal Justice 9(2):325-354.
Chan, M. 2012. Opening remarks at the first meeting of the member state mechanism on
substandard/spurious/falsely-labelled/falsified/counterfeit medical products. Buenos Ar-
ies, Argentina: WHO.
Council of Europe. 2011. Council of Europe Convention on the Counterfeiting of Medical
Products and Similar Crimes Involving Threats to Public Health. Moscow.
———. 2012. Council of Europe Convention on the Counterfeiting of Medical Products
and Similar Crimes Involving Threats to Public Health, CETS no.: 211: Status as
of 26/11/2012. http://conventions.coe.int/Treaty/Commun/ChercheSig.asp?NT=211&
CM= 8&DF=&CL=ENG (accessed November 26, 2012).
EUbusiness. 2010. WHO raps EU for seizing generic drug shipments from India. http://www.
eubusiness.com/news-eu/who-health-drugs.4u5 (accessed April 27, 2012).
Gostin, L. 2013. International law, global institutions, and world health. Cambridge, MA:
Harvard University Press.
Gostin, L. O., and A. L. Taylor. 2008. Global health law: A definition and grand challenges.
Public Health Ethics 1(1):53-63.
ICDRA (International Conference of Drug Regulatory Authorities). 2010 (Novemer
30-December 3). 14 ICDRA recommendations. Paper presented at the Fourteenth Inter-
national Conference of Drug Regulatory Authorities, Singapore.
Ilias, S. 2012. The proposed Anti-Counterfeiting Trade Agreement: Background and key issues.
Congressional Research Service 7-5700(R41107).
IOM (Institute of Medicine). 2012. Ensuring safe foods and medical products through stronger
regulatory systems abroad. Washington, DC: The National Academies Press.
Jessamine, S. 2010. Field of dreams: The New Zealand experience with recognition of other
regulator’s decisions. Paper presented at Pre-ICDRA Meeting, Singapore.
Lourenco, C. 2008. Good regulatory practices: Presentation to APEC preliminary workshop
on review of drug development in clinical trials. Therapeutic Products Directorate,
Health Canada. Ottawa, Canada.
———. 2010. Managing health workforce migration—the Global Code of Practice. http://
www.who.int/hrh/migration/code/practice/en (accessed November 26, 2012).
———. 2011. Regulatory information and practical manuals: WHO medicines regulatory
package. http://www.who.int/medicines/areas/quality_safety/regulation_legislation/
regulatory_package/en/index.html (accessed November 26, 2012).
———. 2012a. International Conference of Drug Regulatory Authorities. http://www.who.
int/medicines/areas/quality_safety/regulation_legislation/icdra/en (accessed November 26,
2012).
———. 2012b. New global mechanism to combat substandard/spurious/falsely-labelled/
falsified/counterfeit medical products. Geneva: WHO.
Appendix A
Glossary
309
Antiretroviral drugs: Drugs used to treat people infected with the human
immunodeficiency virus.
Artemisinin: A drug used to treat malaria derived from the Artemisia An-
nua plant family. It and its derivatives are a group of drugs that possess the
most rapid action against the disease.
APPENDIX A 311
Black market: A market of goods or services that operates outside the for-
mal market, not supported by an established state power.
Blister packet: Perforated packaging used for drugs and other consumer
products.
Bulk property: A property that does not depend on the size or amount of a
sample. For example, density is a bulk property because it does not depend
on the amount of substance tested. A bulk property may also be called an
intensive property.
the separation. This method is used to separate mixtures such as drugs for
accurate and precise analysis.
APPENDIX A 313
Crude active ingredients: Chemicals that have not undergone the appropri-
ate purification steps required to meet pharmacopeial standards or manu-
facturer’s dossier requirements.
Direct ionization: The impulses alpha and beta particles apply to orbital
electrons to ionize, or completely remove an electron from an atom follow-
ing the transfer of energy from a passing charged particle. Specific ioniza-
tion, the number of ion pairs formed per unit path length for a given type
of radiation, is a measure of the intensity of ionization. Because of their
double charge and relatively slow velocity, alpha particles have a high spe-
cific ionization and a relatively short range in matter (a few centimeters in
air and only fractions of a millimeter in tissue). Beta particles have a much
lower specific ionization than alpha particles and, generally, a greater range.
Drug pedigree: A statement of origin that identifies each prior sale, pur-
chase, or trade of a drug, including the date of those transactions and the
names and addresses of all parties to them.
Drug potency: The extent to which a drug product contains the specified
amount of active ingredient.
APPENDIX A 315
Expert review panel: A panel of independent experts, who review the po-
tential risks and benefits associated with the use of finished pharmaceutical
products or diagnostic products. The panel makes recommendations as to
whether the products may be procured.
Fake: Widely used as a synonym for falsified in this report and by other
scholars.
APPENDIX A 317
mass and charge. The GC-MS spectrometer helps separate and determine
the individual elements and molecules in a sample. It is used for the quan-
titation of drugs and provides forensic investigators the ability to identify
individual substances that may be found within a very small test sample.
Infrared spectroscopy: The spectroscopy that deals with the infrared region
of the electromagnetic spectrum, that is, light with a longer wavelength and
lower frequency than visible light. It covers a range of techniques, mostly
based on absorption spectroscopy. As with all spectroscopic techniques,
it can be used to identify and study chemicals. A common laboratory in-
strument that uses this technique is a Fourier transform infrared (FTIR)
spectrometer.
Innovator drug: Generally the pharmaceutical product that was first au-
thorized for marketing (normally as a patented product) on the basis of
documentation of efficacy, safety, and quality according to requirements at
the time of the authorization.
APPENDIX A 319
Marginal cost: The change in total cost that arises when the quantity pro-
duced changes by one unit.
APPENDIX A 321
APPENDIX A 323
Salting: The process by which legitimate and fake drugs are mixed at
wholesale.
APPENDIX A 325
Spectroscopy: The study of the absorption and emission of light and other
radiation by matter.
APPENDIX A 327
Tiered pricing: The concept that different classes of buyers are charged dif-
ferent prices for the same product.
seller or provider from those of others, and to indicate the source of the
goods and services. Trademarks are registered with the sovereign state and
that registration may be used to protect it.
APPENDIX A 329
Visual inspection: The standard first step in any drug quality assessment.
It is the inspection of a suspected substandard or falsified pharmaceutical
product: looking for differences in color, size, shape, tablet quality, and
packaging, and comparing it to an authentic product.
Appendix B
Committee Biographies
331
American Society of Law, Medicine & Ethics and adjunct professor of law
and public health at Harvard University. In the United Kingdom, Professor
Gostin was the chief executive of the National Council for Civil Liberties,
legal director of the National Association of Mental Health, and faculty
member of Oxford University. Professor Gostin received the Rosemary Del-
bridge Memorial Award from the National Consumer Council (UK) for the
person “who has most influenced Parliament and government to act for the
welfare of society.” He also received the key to Tohoku University in Japan
for distinguished contributions to human rights in mental health. Professor
Gostin’s latest books are both published by the University of California
Press and the Milbank Memorial Fund: Public Health Law: Power, Duty,
Restraint (2000) and Public Health Law and Ethics: A Reader (2002).
APPENDIX B 333
ner of both the 2011 Herbert Simon Award of the Midwest Political Science
Association for a scholar “who has made a significant career contribution
to the scientific study of bureaucracy,” as well as the 2011 David Collier
Award of the American Political Science Association for career contribu-
tions to qualitative and multi-method research. In addition to his ongoing
teaching and scholarship on the political economy of government regula-
tion and health, Professor Carpenter has recently launched a long-term
project on petitioning in North American political development, examining
comparisons and connections to petitioning histories in Europe and India.
He hopes to draw upon the millions of petitions in local, state, and federal
archives to create an educational, genealogical, and scholarly resource for
citizens, students, and scholars.
Patrick Lukulay, Ph.D., is currently the director of the United States Agency
for International Development–funded program Promoting the Quality of
Medicines, implemented by U.S. Pharmacopeia. He oversees the work of
about 20 staff to provide technical assistance to developing countries to
strengthen quality assurance and quality systems for pharmaceuticals. Dr.
Lukulay has a Ph.D. in analytical chemistry from Michigan State Univer-
sity. He worked in the pharmaceutical industry for Wyeth and Pfizer for a
combined 12 years as senior principal scientist. He is the author of several
APPENDIX B 335
Arti K. Rai, J.D., is the Elvin R. Latty Professor of Law at Duke University
Law School and a member of the Duke Institute for Genome Science and
Policy. She is an authority in patent law, administrative law, and innovation
policy. Ms. Rai has also taught at Harvard, Yale, the University of Penn-
sylvania, and the University of San Diego law schools. Ms. Rai’s academic
research on innovation policy in areas such as synthetic biology, green tech-
nology, drug development, and software has been funded by the National
Institutes of Health (NIH), the Kauffman Foundation, and Chatham House.
She has published widely in both peer-reviewed journals and law reviews,
including Nature Biotechnology, PLoS Biology, PLoS Medicine, the Annals
of Internal Medicine, and the Columbia, Georgetown, and Northwestern
law reviews. She is the editor of Intellectual Property Law and Biotechnol-
ogy: Critical Concepts (Edward Elgar, 2011) and has also co-authored a
casebook on law and the mental health system.
From 2009 to 2010, Ms. Rai took a leave of absence from Duke Law
School to serve as the administrator of the Office of External Affairs at the
U.S. Patent and Trademark Office. Prior to that, she served on President-
Elect Obama’s transition team reviewing the Patent and Trademark Office
and as an expert advisor to the Department of Commerce’s Office of Gen-
eral Counsel. Prior to entering academia, Ms. Rai clerked for the Honor-
able Marilyn Hall Patel of the U.S. District Court for the Northern District
of California; was a litigation associate at Jenner & Block (doing patent
litigation as well as other litigation); and was a litigator at the Federal Pro-
grams Branch of the U.S. Department of Justice’s Civil Division. Ms. Rai
has served as a peer reviewer for Science, Research Policy, the Journal of
Legal Studies, various National Academy of Sciences reports on intellec-
tual property, and various NIH study sections. She has also testified before
Congress on innovation policy issues and regularly advises federal agencies
on policy issues (including intellectual property policy issues) raised by the
research that they fund. Recently, her work has focused on advising the
Defense Advanced Research Projects Agency. Ms. Rai is currently the chair
of the Intellectual Property Committee of the Administrative Law Section
of the American Bar Association. She is also a fellow of the American Bar
Foundation. In 2011, Ms. Rai won the World Technology Network Award
for Law.
Ms. Rai graduated from Harvard College, magna cum laude, with a
B.A. in biochemistry and history (history and science), attended Harvard
Medical School for the 1987-1988 academic year, and received her J.D.,
cum laude, from Harvard Law School in 1991.
John Theriault, M.B.A., has had senior leadership roles with the Federal
Bureau of Investigation, Pfizer, and Apple. Today, he is recognized interna-
tionally as an expert on the subject of product counterfeiting and the devel-
opment of effective programs to combat its far-ranging impact on society.
Mr. Theriault served as a special agent of the Federal Bureau of Investiga-
tion (FBI) for more than 25 years, rising to the Bureau’s Senior Executive
Service. For 7 years he was diplomatically accredited as the legal attaché at
various U.S. embassies and was responsible for managing all of the FBI’s
law enforcement, counterintelligence, and counterterrorism relationships
with senior government officials of the host countries. In 1996 he joined
Pfizer as vice president and chief security officer. In response to widespread
counterfeiting of their medicines, he created, staffed and led the company’s
global anti-counterfeiting program, which became a model for the industry.
He has testified before the U.S. Senate Special Committee on Aging, the
U.S. House of Representatives Committee on Energy and Commerce, and
APPENDIX B 337
the U.S. Surgeon General’s Drug Importation Task Force. He has briefed
the U.S. Secretary of Commerce and other senior government officials on
the dangers of counterfeit medicines, and he has appeared on a number
of broadcasts including 60 Minutes, Larry King Live, CNN Late Edition,
BBC Radio, and others. In 2007 Mr. Theriault was recruited by Apple to
create a global security organization with a strong focus on combating
their growing counterfeiting problem. In November 2011 he retired from
Apple to return to the East Coast and pursue other interests. Mr. Theriault
earned a B.A. from the University of Memphis and an M.B.A. degree from
Emory University.
in 2008. She joined the Board of Trustees for the International Centre for
Diarrheal Disease Research, Bangladesh and is a member of the Board of
Scientific Counselors for the Centers for Disease Control and Prevention,
the FXB-USA board, and the Alliance for Prudent Use of Antibiotics Board
of Directors.
Appendix C
Meeting Agendas
Session 1 – Closed
IOM Committee Process and Charge to Committee
Room 204
Objective: To review the National Academies’ study process that
includes a bias and conflict of interest discussion; to discuss the role of
the committee in addressing the statement of task; and to ensure the
committee understands its statement of task.
Session 2 – Open
Questions on Statement of Task
Room 100
11:10-11:30 Project Timeline and Statement of Task
Sponsor Representative Introductions
Larry Gostin, Committee Chair
11:45-12:15 Questions
339
12:15-1:15 Lunch
Session 3 – Open
Technologies for Detecting Unsafe Drugs
Room 100
1:15-1:25 Welcome and Introductions
Larry Gostin, Committee Chair
Session 4 – Closed
Committee Planning
APPENDIX C 341
Session 5 – Open
Framing and Defining the Problem
8:15-8:30 Opening Remarks
Mary Lou Valdez, Associate Commissioner,
Office of International Programs, FDA
10:15-10:30 Break
Session 6 – Open
National and International Collaboration
1:15-2:15 Enforcement in Pharmaceutical Fraud
Susanne Keitel, Director, European Directorate for
Quality of Medicines and Healthcare, Council of
Europe
Aline Plançon, Manager, Interpol Medical Products
Counterfeiting and Pharmaceutical Crime Unit
(by video conference)
Sebastian Mollo, Intelligence Director, Pharmaceutical
Security Institute
Sameer Barde, Federation Indian Chambers of
Commerce and Industry
Daniel Carpenter, Moderator
3:30-3:45 Break
Session 7 – Open
Scope of Work on the Problem
3:45-4:30 Investigating Trends and Analyzing Policy in
Pharmaceutical Fraud
Laurie Garrett, Senior Fellow for Global Health,
Council on Foreign Relations
Alan Coukell, Director Medical Programs,
Pew Health Group
Judit Rius, U.S. Manager of the Campaign for Access
to Essential Medicines, Doctors Without Borders
Ann Marie Kimball, Moderator
APPENDIX C 343
Session 5 – Open
FDA Data on Fake Drugs
11:30-12:00 Questions
12:30-1:45 Lunch
APPENDIX C 345
1:30-2:15 Lunch
12:00-1:30 Lunch
12:30-1:30 Lunch
APPENDIX C 347
12:00-1:00 Lunch
APPENDIX C 349
11:30-1:00 Lunch
1:45 Adjourn
10:45-11:00 Break
12:15-1:15 Lunch
2:30-3:00 Break
4:00 Adjourn
APPENDIX C 351