British Journal of Clinical
Pharmacology
DOI:10.1111/bcp.12298
Correspondence
Substandard drugs:
a potential crisis for
public health
Professor Atholl Johnston, Clinical
Pharmacology, Barts and The London,
Charterhouse Square, London EC1M 6BQ,
UK.
Tel.: +44 20 7882 6055
Fax: +44 20 7882 3408
E-mail:
[email protected]
-----------------------------------------------------------------------
Keywords
Atholl Johnston1 & David W. Holt2
drug quality, falsification, inspection,
regulation, substandard
1
-----------------------------------------------------------------------
Clinical Pharmacology, Barts and The London School of Medicine and Dentistry, Queen Mary
2
University of London, London, UK and St George’s – University of London, London, UK
Received
13 August 2013
Accepted
1 November 2013
Accepted Article
Published Online
29 November 2013
Poor-quality medicines present a serious public health problem, particularly in emerging economies and developing countries, and
may have a significant impact on the national clinical and economic burden. Attention has largely focused on the increasing
availability of deliberately falsified drugs, but substandard medicines are also reaching patients because of poor manufacturing and
quality-control practices in the production of genuine drugs (either branded or generic). Substandard medicines are widespread and
represent a threat to health because they can inadvertently lead to healthcare failures, such as antibiotic resistance and the spread
of disease within a community, as well as death or additional illness in individuals. This article reviews the different aspects of
substandard drug formulation that can occur (for example, pharmacological variability between drug batches or between generic and
originator drugs, incorrect drug quantity and presence of impurities). The possible means of addressing substandard manufacturing
practices are also discussed. A concerted effort is required on the part of governments, drug manufacturers, charities and healthcare
providers to ensure that only drugs of acceptable quality reach the patient.
Introduction
Poor-quality medicines can reach the market through substandard production of legitimate drugs due to inadequate quality-control processes during manufacture, as
well as by deliberately fraudulent practices. The relative
contribution of the two sources is unknown; however,
genuine but low-quality drugs are likely to account for the
majority of cases [1]. To date, legislation has focused on
the control of deliberately falsified drugs, but poor-quality
legitimate drugs, i.e. those that have gone through some
sort of regulatory procedure, are more commonly seen
and pose a greater threat to patient health, so both issues
need to be tackled. In recent years, there have been a
number of high-profile recalls; for example, the European
Medicines Agency (EMA) recommended the recall of
eight generic clopidogrel-containing medicines for which
the active pharmaceutical ingredient (API) was produced in India, following an inspection of the manufacturing site [2], and in the USA all products compounded
at New England Compounding Pharmacy’s facility in
Framingham, Massachusetts were recalled following a
218
/
Br J Clin Pharmacol
/ 78:2
/
218–243
fatal outbreak of fungal meningitis associated with injectable steroids [3]. In some cases, there have been import
bans on drugs from companies suspected of having
substandard production practices (e.g. Ranbaxy [4, 5]).
However, more must be done to improve the manufacturing practices and registration of drugs, so that poorquality drugs (albeit approved by a regulatory authority)
do not reach the market.
In this article, we aim to raise awareness of the problem
of substandard drugs; a problem that we regard as having
the potential to be a public-health crisis. We highlight the
types of formulation defects that can occur and the consequences of substandard drugs. We also review some of
the efforts being made to ensure that manufacturing and
quality-control processes comply with internationally
accepted practices.
Definitions
In 2009, the World Health Organization (WHO) defined
‘substandard’ drugs (also called ‘out of specification products’) as ‘genuine medicines produced by manufacturers
authorized by the NMRA [national medicines regulatory
© 2013 The Authors. British Journal of Clinical Pharmacology published by
John Wiley & Sons Ltd on behalf of The British Pharmacological Society.
This is an open access article under the terms of the Creative Commons Attribution-NonCommercial-NoDerivs License, which permits use and distribution in any
medium, provided the original work is properly cited, the use is non-commercial and no modifications or adaptations are made.
Substandard drugs
authority] which do not meet quality specifications set for
them by national standards’ [6]. A new definition was proposed by the WHO in May 2010 [7]: ‘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 NMRA
before the product is authorized for marketing. Substandard medicines are pharmaceutical products that do not
meet their quality standards and specifications.’
The WHO defines ‘counterfeit’ drugs as ‘medicines that
are deliberately and fraudulently mislabelled with respect
to identity and/or source’ [8]. It also states that both
branded and generic products may be counterfeited and
that ‘counterfeit medicines may include products with the
correct ingredients or with the wrong ingredients, without
active ingredients, with insufficient or too much active
ingredient, or with fake packaging’. However, because of
the potential misunderstanding of the term ‘counterfeit’ –
which, in the context of intellectual property, refers specifically to trademark infringement – the phrase ‘falsified
medicines’ is used by some authorities, particularly in
Europe. The Commission of the European Communities
defines these as ‘medicinal products which are falsified in
relation to their identity, history or source. These products
. . . usually contain sub-standard or false ingredients, or no
ingredients or ingredients in the wrong dosage, including
active ingredients’ [9].
Thus, falsified drugs are highly likely to be of substandard quality, possibly containing no API. However, only a
small proportion of substandard drugs are falsified; the
rest reach the market as a result of poor manufacturing
practices, inadequate quality-control processes, incorrect
storage or inappropriate packaging, or a combination of
these factors. This can affect both branded and generic
drugs. In many cases, the reason why a drug product is
substandard (i.e. deliberate falsification or poor manufacturing practice) is not stated or is not known. Whether or
not a drug product is substandard because of criminal
intent or because of failures in manufacturing, storage, etc.
is immaterial to the patient because the impact on their
health will be the same, regardless of cause [10]. In this
article, we consider the term ‘substandard’ to apply both
to legally approved but poor-quality drugs and to falsified
drugs, but we focus on the former with regard to reviewing potential solutions.
The terms ‘medicine’, ‘medications’ and ‘drugs’ are
used interchangeably in this article.
Literature search
This narrative review was based on literature searches conducted using PubMed to identify English-language articles
giving relevant examples of substandard drugs; identified
articles were hand searched for further relevant examples.
A systematic review of all publications was not intended
and thus a search for published literature using PubMed
alone was deemed to be adequate. Search terms included
combinations of the following: substandard, quality, resistance, (drug OR drugs OR medication OR medications OR
medicine OR medicines), (impurity OR impurities OR contamination OR contaminant OR contaminants). Searches
were generally limited to recent publications (since 2000).
Data from publications were included and tabulated if
they provided examples of the marketing of substandard
drugs, based on analytical or clinical evidence. Internet
searches using similar terms were also performed using
the Internet search engine Google, and specific searches
were conducted of relevant websites [e.g. the US Food and
Drug Administration (FDA) and the UK Medicines and
Healthcare products Regulatory Agency (MHRA) websites].
Using examples identified through this search, the
article will review information on substandard medicines,
including the following: inappropriate API content, impurities and inconsistent pharmacological response due to
variable drug content; the prevalence of and potential
adverse health effects associated with use of substandard
drugs; and methods by which drug quality might be
improved.
Defective drug formulation
In cases of substandard medication that arise through
inadequate production processes, rather than through
deliberate falsification of drugs, the lack of quality may be
the result of a variety of factors, including the following:
inadvertent use of substandard or incorrect APIs or
excipients; poor control of drug quantity; manufacturing
processes that cause contamination or do not adequately
ensure sterility; and inadequate packaging design or
quality. In addition, ineffective quality-control measures,
either on the part of the manufacturer or the NMRA, allow
such faults to remain undetected.
Drug content
Any formulation of a medication may be regarded as substandard if it has either too much or too little of the API
compared with the formulation specifications. Official
national pharmacopoeias, such as the British Pharmacopoeia (BP) and United States Pharmacopeia (USP), publish
the quality standards for medicinal substances and preparations manufactured or sold in the country. The information given specifies the acceptable limits for the amount of
the API that should be present in a given formulation.
However, many examples from a range of drug classes
have been published of over/underconcentration of APIs
in marketed drugs (see Table 1 [11–39]). In some cases, all
sampled antibiotics or antimalarials were found to contain
API concentrations outside the officially specified limits
[11].
Br J Clin Pharmacol
/ 78:2
/
219
A. Johnston & D. W. Holt
Table 1
Substandard drugs: drug content
Drug(s)
Issue
Region/countries
Reference
Chloroquine phosphate, chloroquine
sulphate and quinine sulphate
Chloroquine phosphate: 70% of capsules (n = 29), 100% of syrup
samples (n = 20) and 94% of tablets (n = 18) outside BP limits.
Chloroquine sulphate: 79% of tablet samples (n = 19) and 73% of
syrup samples (n = 20) outside BP limits
Quinine sulphate: 24% of tablet samples (n = 17) outside BP limits
Nigeria
Taylor et al. (2001)
[11]
Amodiaquine and
sulfadoxine-pyrimethamine
11% of sulfadoxine samples (n = 18) failed the test for content of APIs
Tanzania
Minzi et al. (2003)
[12]
Ceftriaxone
9% of generics (n = 35) failed to achieve minimum 97% content
specified in EP
Brazil, Pakistan and
Philippines
Lambert and
Conway (2003)
[13]
Chloroquine, quinine and antifolates
Drugs purchased from unofficial vendors: 38% of chloroquine
(n = 133), 74% of quinine (n = 70) and 12% (n = 81) of antifolate
samples had no API or insufficient, incorrect or unknown ingredients
Tablets collected from patients who self-medicated before consultation:
of 15 quinine or chloroquine samples collected, six (40%) contained
no API, one had an insufficient dose and two contained quinine
instead of chloroquine
Cameroon
Basco (2004) [14]
Artesunate
53% of samples (n = 188) contained no artesunate
Burma, Lao People’s
Democratic Republic,
Vietnam, Cambodia
and Thailand
Dondorp et al.
(2004) [15]
Chloroquine phosphate tablets and
chloroquine syrup
6.7% of syrup samples (n = 25) and 20% of tablets (n = 25) failed to
meet content specifications
Yemen
Abdo-Rabbo et al.
(2005) [16]
Sulfadoxine-pyrimethamine and
amodiaquine
45.3% of sulfadoxine-pyrimethamine and 33.0% of amodiaquine
samples (n = 116) were substandard; 40.5% of samples did not meet
USP specifications for content and/or dissolution
Kenya
Amin et al. (2005)
[17]
Artemisinin-derivatives: artemether,
arteether, artesunate or DHA
Seven of 24 samples were underdosed and two of two samples were
overdosed (i.e. outside of allowed range of 95–105%) according to
European requirements
DHA was the API in 57% of the underdosed samples
Kenya and Democratic
Republic of the Congo
Atemnkeng et al.
(2007) [18]
Sulfadoxine-pyrimethamine,
amodiaquine, mefloquine,
artesunate, artemether, DHA and
artemether-lumefantrine
fixed-dose combination
35% of samples (n = 210) failed tests for the concentration of APIs
compared with internationally accepted standards
Africa
Bate et al. (2008)
[19]
Chloroquine, sulfadoxinepyrimethamine, quinine,
amodiaquine, artesunate and
artemether-lumefantrine
Nine of 77 (12%) samples had substandard concentrations of API
Burkina Faso
Tipke et al. (2008)
[20]
Artesunate, DHA, sulfadoxinepyrimethamine, quinine and
chloroquine
37% of the samples tested (n = 225) did not meet USP limits for the
amount of API
46% of quinine samples and 39% of sulfadoxine-pyrimethamine
samples did not meet the criteria
Nigeria
Onwujekwe et al.
(2009) [21]
Artemisinin-based drugs
Thirteen of 14 (93%) contained either too low or too high a dose of
the specified drug
Ghana
El Duah and
Ofori-Kwakye
(2012) [22]
10% (n = 40) of samples contained <85% of stated content
21% of fixed-dose combinations were substandard vs. 13% of
single-drug samples
Isoniazid: 100% of tablet samples (n = 4) were outside BP limits
Rifampicin: 33% of capsule samples (n = 15) were outside BP limits
Colombia, Estonia, India,
Latvia, Russia and
Vietnam
Nigeria
Laserson et al. (2001)
[23]
Amoxicillin: 25% of capsule formulations (n = 32) and 40% of dry
syrup formulations (n = 5) were outside BP limits
Ampicillin: 59% of capsules (n = 39) and 71% of dry syrup formulations
(n = 7) were outside BP limits
Ketoconazole: 80% of cream preparations (n = 5) were outside BP limits
Metronidazole: 100% of suspension formulations (n = 5) and 72% of
tablets (n = 36) were outside BP limits
Nigeria
Antimalarials
Antituberculosis drugs
Isoniazid and rifampicin
Isoniazid and rifampicin
Taylor et al. (2001)
[11]
Other antibiotics
Amoxicillin, ampicillin, ketoconazole
and metronidazole
220
/ 78:2
/
Br J Clin Pharmacol
Taylor et al. (2001)
[11]
Substandard drugs
Table 1
Continued
Drug(s)
Issue
Region/countries
Reference
Benzathine benzylpenicillin,
ceftriaxone, chlortetracycline,
ciprofloxacin, clotrimazole,
co-trimoxazole, doxycycline and
erythromycin
33% of 21 products for STDs did not contain the stated dose of API
The highest deficit was 48% (co-trimoxazole and benzylpenicillin)
Burma
Prazuck et al. (2002)
[24]
Ampicillin, tetracycline and
chloroquine. Acetylsalicylic acid
also included in study
The proportion of substandard drugs decreased significantly from 46%
(n = 366) to 22% (n = 300) between 1997 and 1999 (P < 0.001)
4% of drugs had too little or too much API; 1% had no API (1999
data)
Lao People’s Democratic
Republic
Syhakhang et al.
(2004) [25]
Clarithromycin
9% (n = 65) of samples did not contain 95–105% of the dose of API
specified in the label of the reference (manufactured by Abbott)
17% of samples from Latin America did not meet criteria for content
Multinational
(18 countries)
Nightingale (2005)
[26]
Ciprofloxacin
Six of 30 samples of generic eye drops had <95% of the stated drug
content (range −36 to −16%); 24 of 30 had >105% of stated
content (37% had >120%)
India
Weir et al. (2005)
[27]
Ciprofloxacin
Three of 16 samples of generic tablets had 90–95% and one of 16 had
>105% of the dose of API stated in the label
Multinational
Trefi et al. (2007)
[28]
Amoxicillin
56% (n = 72) of capsules and 8% (n = 39) of suspensions had API
levels outside of pharmacopoeial limits
38% of all samples (n = 111) had API levels outside of pharmacopoeial
limits after storage for 38 days
Lebanon, Jordan, Egypt
and Saudi Arabia
Kyriacos et al. (2008)
[29]
Antibiotics (ciprofloxacin,
erythromycin), antimalarials
(chloroquine) and
antimycobacterials (isoniazid,
rifampicin)
12% (n = 281) of samples from Delhi failed tests for the concentration
of APIs compared with internationally acceptable standards
India
Bate et al. (2009)
[30]
Amoxicillin, chloramphenicol,
tetracycline, co-trimoxazole and
ciprofloxacin
18% (n = 104) of samples had small deviations in the dose of API (less
than BP standards)
Indonesia
Hadi et al. (2010)
[31]
Amoxicillin, ampicillin and
cephalexin
10% (19 of 185) failed quantitative tests according to USP 30
Purchased in Cambodia
(31% domestically
manufactured)
Okumura et al.
(2010) [32]
The standard deviation of content for the generic versions of Prograf
Tenacrine, Tacrobell and T-Inmun (up to 30 of each) was 29.3, 6.9
and 5.6, respectively
Mexico
Petan et al. (2008)
[33]
Streptokinase
Thirteen of 16 products exhibited only 20.8–86.6% of the activity
stated in the label
Multinational
Hermentin et al.
(2005) [34]
Carvedilol
48.6% (n = 35) of generic samples failed to meet EP and Roche
specifications for content (95–105%)
Multinational
(19 countries)
Smith et al. (2006)
[35]
Antihypertensives
20% (n = 10) of drugs tested were substandard in terms of API
content at time of purchase
70% were substandard after 6 months in accelerated storage
conditions
Rwanda
Twagirumukiza et al.
(2009) [36]
Ramipril
24% (n = 17) of generic copies of Tritace failed to meet Sanofi-Aventis
specifications (90–105% of label claim) for the amount of API
This increased to 47% after storage for 3 months
Italy
Angeli and Trezza
(2009) [37]
Twenty-one of 31 commercially available generic versions of Taxotere
had <90% of expected mass of docetaxel, and 11 of these contained
<80% of expected mass
Multinational
(14 countries in Asia,
Africa, the Middle East
and Latin America)
Vial et al. (2008) [38]
Four of 14 generics failed tests for content following accelerated
shelf-life tests, based on Roche criteria for content
Two of 14 samples and three of 14 samples failed tests for content
according to EP and USP specifications, respectively
Not specified
Taylor and Keenan
(2006) [39]
Immunosuppressants
Tacrolimus
Cardiovascular
Oncology
Docetaxel
Dermatology
Isotretinoin (acne)
Abbreviations are as follows: API, active pharmaceutical ingredient; BP, British Pharmacopoeia; DHA, dihydroartemisinin; EP, European Pharmacopoeia; STD, sexually transmitted
disease; USP, United States Pharmacopeia.
Br J Clin Pharmacol
/ 78:2
/
221
A. Johnston & D. W. Holt
Inappropriate packaging can affect formulation content in certain storage conditions. For example, a study of
generic versions of ramipril tablets found that, on initial
inspection, four of 17 samples (24%) did not meet the label
specifications for drug content. After 3 months of storage
in temperature-stressed conditions (40°C and 75% relative
humidity), a further four samples (total 47%) failed to meet
the content specifications [37].
In some cases, a product may contain no API or the
drug content may be completely different to that stated
on the label. This may occur through deliberate falsification, but as the examples shown below demonstrate, accidental mislabelling may also occur.
• One batch of the antibiotic Rofact® (rifampicin) was mislabelled; bottles actually contained the anti-epileptic
clonazepam (Canada, May 2009) [40].
• One lot of minocycline was mislabelled as amlodipine
(Canada, March 2011) [41].
• One lot of bottles containing finasteride was labelled as
containing citalopram (USA, March 2011) [42].
• Zopiclone was substituted for furosemide in a possible
packaging mix-up (France, June 2013) [43].
Impurities
An impurity may be defined as any substance in the
product that is neither the chemical entity defined as the
drug nor an excipient [44]. Impurity profiling is required as
part of the registration process by many regulatory
authorities, including the FDA and the European Union’s
Committee for Medicinal Products for Human Use (CHMP).
Impurities fall into one of three categories – organic
substances, inorganic substances and residual solvents
[45] – and may include starting materials, intermediate
compounds, reagents and catalysts, heavy metals, degradation products, polymorphic forms (alternative crystal
forms with potentially different dissolution profiles) and
enantiomeric impurities, as well as extraneous contaminants. Impurities can arise in formulations due to poor
manufacturing procedures and storage conditions
(Table 2 [13, 23, 26, 28, 34, 35, 37–39, 46–59]). Impurities
can alter medication properties or be toxic. Probably the
best known cases of contamination involve diethylene
glycol (DEG); at least 14 mass poisonings have been documented that were due to the presence of DEG in pharmaceutical preparations [47, 48]. Indeed, it was an incidence
of mass poisoning with DEG (in this case, due to the use of
DEG as a diluent for a liquid preparation of sulfanilimide)
that brought about the 1938 Federal Food, Drug and Cosmetic Act in the USA and created the regulatory role of the
FDA [60].
Many cases of DEG poisoning are likely to have
occurred as a result of DEG being substituted for the commonly used diluents propylene glycol or glycerine [47].
The substitution was probably deliberate at the level of
222
/ 78:2
/
Br J Clin Pharmacol
provision of an ingredient, although it is possible that contamination could have occurred during the manufacture
of glycerine [47]. Subsequent use of the contaminated
ingredient may not have been deliberate. However, whatever the cause of contamination of the raw materials, the
responsibility for the safety of the drug product rests with
the drug manufacturer, and appropriate testing of materials should be in place to prevent such occurrences.
In many other cases, contamination has clearly
occurred due to poor manufacturing and/or qualitycontrol processes, or unsuitable packaging. Contaminants
have included the following: particulate matter in injectable cefotaxime [50]; small glass particles in bottles of
generic atorvastatin tablets [61]; degradation products in
docetaxel, streptokinase and clopidogrel [34, 38, 56]; and
potentially genotoxic impurities in batches of nelfinavir
due to incomplete removal of ethanol following the cleaning of manufacturing equipment [54]. In 1990, unacceptably high levels of benzene were detected in bottled
Perrier water [62], which highlighted the need to understand the impact of ingredient interactions and packaging.
The presence of benzene was thought to arise from the
reaction of the preservative benzoic acid with ascorbic
acid, possibly accelerated by the presence of citric acid,
heat and light [63, 64]. In other cases, bacterial contamination in several different drug products has been attributed
to unhygienic practices [46].
In addition to the official recall notices and studies published in peer-reviewed journals, there are numerous
examples in the press of contamination in marketed drugs,
such as the incidents described below.
• Albupax (a paclitaxel formulation produced in India) was
found to contain excessive endotoxin levels and was
withdrawn from the market (April 2009) [65].
• Batches of Tylenol, Motrin, Rolaids and Benadryl were
recalled in the USA due to the presence of 2,4,6tribromoanisole (January 2010) [66].
• Generic formulations of clopidogrel marketed in India
and Europe were found to contain methyl chloride, which
can cause hepatic, renal and nervous system damage
[67].
• Methyldopa (Dopamet) 250 mg tablets produced in
Cyprus were banned by the Tanzania Food and Drugs
Authority. It was found that drug identification labels
could be detached easily from the packaging, and there
was ‘vivid fungal growth’ on the tablets (January 2011)
[68].
Pharmacological variability and stability
Generic drugs can aid the provision of healthcare to a wide
patient population, particularly in developing countries.
However, generic formulations should only be marketed if
their quality is equivalent to that of the originator drug. At
present, to gain marketing approval, a generic drug only
Substandard drugs
Table 2
Substandard drugs: contamination
Drug(s)
Contaminant
Source (if known)
Countries
Reference
Ampicillin, tetracycline,
paracetamol,
chloroquine and
metronidazole
Bacteria
Ampicillin: S. aureus in six of 16
Tetracycline: B. subtilis in five of 15, P. Mirabilis in 5 of 15
Paracetamol: S. aureus in five of 15, A. aerogenes in four
of 15, P. mirabilis in three of 15
Choloquine: B. subtilis in 11 of 23, A. aerogenes in three
of 23
Metronidazole: B. subtilis in three of 15, S. aureus in 13
of 15
The authors concluded the
source to be unhygienic
manufacturing practices
and poor adherence to
good manufacturing
practice
Nigeria, USA and
Panama
Itah et al. (2004) [46]
Paracetamol, cough
expectorant, propolis,
teething syrup,
armillarisin,
sulfanilamide, sedatives
and silver sulfadiazine
Diethylene glycol
Contaminated propylene
glycol or other glycols
Nigeria, USA,
Panama, South
Africa, India,
Spain, Bangladesh,
Argentina, Haiti
and China
Schier et al. (2009)
[47]; Schep et al.
(2009) [48]
Orlistat
Nine of nine generic products contained higher levels of
impurities than permitted by branded drug specifications
Side-chain homologues and
unidentified impurities
India, Malaysia,
Argentina,
Philippines,
Uruguay and
Taiwan
Taylor et al. (2010) [49]
Germany
Lehr et al. (2002) [50]
Brazil, India, Pakistan
and Philippines
Lambert and Conway
(2003) [13]
Multinational
(18 countries)
Multinational
Nightingale (2005) [26]
Miscellaneous
Antibiotics
Cefotaxime
Ceftriaxone
Clarithromycin
Ciprofloxacin
Particulate matter present in injectable antibiotics
Injection of particles from two of three generic
formulations into hamsters reduced capillary perfusion in
muscle previously exposed to ischaemia and reperfusion
12% of generic formulations (n = 35) were not sterile;
15% contained impurities; 97% contained thiotriazinone
(drug degradation product)
19% (n = 65) of generics did not meet Abbott criteria for
≤3% impurities
None of 16 generic samples analysed met the European
Pharmacopoeia limits for levels of impurities.
Samples contained fluorinated and nonfluorinated
impurities
Trefi et al. (2007) [28]
Anticoagulants
Heparin
Oversulphated chondroitin sulphate
Contaminant not removed
in production process
USA, Germany
Blossom et al. (2008)
[51]
Heparin
Serratia marcescens
Prefilled syringes; the
manufacturer was found
to have inadequate
controls to ensure
sterility
USA
Blossom et al. (2009)
[52]
Colombia, Estonia,
India, Latvia,
Russia and
Vietnam
Laserson et al. (2001)
[23]
Multinational
(14 countries in
Asia, Africa, the
Middle East and
Latin America)
Vial et al. (2008) [38]
USA
Kutty et al. (2008) [53]
Global
Pozniak et al. (2009)
[54]
Antituberculosis drugs
Isoniazid and
rifampicin
Two of 40 samples contained an additional unidentified
chemical component
Oncology
Docetaxel
Ophthalmology
Endosol (balanced
salt solution)
Twenty-three of 31 generic formulations of Taxotere had an
impurity content >3%
Thirty-three unknown impurities were present at >0.05% in
31 generic versions of Taxotere
Included 7-epidocetaxel,
a product of docetaxel
degradation; others
unidentified
Endotoxin was present at concentrations exceeding the
specified limit in 35% of samples
Resulted in toxic anterior segment syndrome
Protease inhibitors
Nelfinavir mesylate
Elevated levels of ethyl methanesulphonate
Ethanol cleaning of
manufacturing
equipment
Br J Clin Pharmacol
/ 78:2
/
223
A. Johnston & D. W. Holt
Table 2
Continued
Drug(s)
Contaminant
Antidiabetics
Glimepiride
Source (if known)
Countries
Reference
Italy
Attorrese and
Massi-Benedetti
(2007) [55]
Degradation
Multinational
(five countries)
Gomez et al. (2004)
[56]
Degradation suspected
Multinational
Not specified in abstracts
Multinational
(19 countries)
Hermentin et al.
(2005) [34]
Smith et al. (2006) [35]
Determined as major
metabolite
Italy
Angeli and Trezza
(2009) [37]
Manufacturing error:
pyrimethamine added
instead of pregelatinized
starch
Pakistan
Attaran et al. (2012)
[57]; Choudary
(2012) [58]; World
Health Organization
(2012) [59]
Not specified
Not specified
Taylor and Keenan
(2006) [39]
In stressed conditions (storage at 60°C for up to 21 days),
levels of glimepiride degradation products were higher
vs. the reference (Amaryl) in two of 23 generic samples
on day 7and four of 24 samples on day 21
Other impurities and solvents were above reference
specifications (≤1%) in two samples
Cardiovascular
Clopidogrel
>60% of 18 generics studied contained >4 times the
amount of hydrolysis products or the R-enantiomer
compared with Plavix
After storage for 3 months at 40°C and 75% humidity, the
differences were more pronounced
Biochemical analysis by electrophoresis revealed additional
bands on SDS-PAGE gels, suggesting impurities
Three of 35 samples of generics had excessive impurities
(>3%) according to European Pharmacopoeia and Roche
specifications
32% (n = 22) of samples had impurities above the
reference specifications (≤5%)
This increased to 68% after storage for 3 months
Antimalarial agent pyrimethamine
Streptokinase
Carvedilol
Ramipril
Isosorbide-5mononitrate
Dermatology
Isotretinoin (acne)
Eight of 14 generic samples failed criteria for total
impurities
Six of 14 samples contained five or more unknown
impurities
Abbreviation is as follows: SDS-PAGE, sodium dodecyl sulphate polyacrylamide gel electrophoresis.
needs to demonstrate equivalent average pharmacokinetic properties compared with the originator drug.
Bioequivalence is achieved, and hence therapeutic
equivalence is presumed, if the mean ratios for key
pharmacokinetic parameters (maximal plasma concentration and area under the concentration–time curve) of the
generic drug vs. the originator have a 90% confidence
interval (CI) within 0.80 and 1.25 of the originator in
healthy volunteers; for some NMRAs, narrower ranges may
apply for some drugs with a narrow therapeutic index.
Even bioequivalence may not be met in some developing
countries that do not have the necessary quality-assurance
resources or regulations. A case reported in Greece highlighted the fact that problems may also occur in the manufacturing of generics in developed countries. Following an
increase in postoperative infections in patients receiving
generic cefuroxime compared with patients receiving the
originator drug, analysis of the generic version (manufactured in Greece) revealed a substandard formulation [69].
It should also be noted that in some cases, a ‘generic’
might in fact be a ‘copy drug’, i.e. a version of the drug that,
although it is available on the market, has not undergone
224
/ 78:2
/
Br J Clin Pharmacol
any bioequivalence testing. This was thought to be the
case for a substandard version of imatinib manufactured in
India, for which no regulatory documentation was evident
[70].
Excipients used in a generic formulation are not
required to be identical to those in the originator formulation. Small changes in excipients can alter the properties of
a formulation (e.g. lead to differences in particle size, or
modify the shelf-life) and hence affect drug efficacy
and safety. For example, although excipients such as
polysorbate 80 and polyoxyethylated castor oil are considered inert, some investigators have reported evidence of
altered drug metabolism with changes in the source of
such compounds [71, 72].
The testing of a drug’s dissolution properties is a relatively simple in vitro test used as an indicator of in vivo
bioavailability. Drug-specific limits for dissolution times
are defined in pharmacopoeias, against which samples can
be compared. This might be used, for example, to check
batch samples of a branded drug or to compare generic
versions against originator drugs. There are many examples in the published literature in which studies have
Substandard drugs
shown marked variation in dissolution times between supposedly bioequivalent drugs (Table 3 [12, 17, 19, 26, 33,
35–37, 55, 56, 73–87]). For example, testing of five generic
versions of tacrolimus available in Mexico demonstrated
considerable differences in their dissolution profiles compared with that of the branded originator drug [33]. In
another study, approximately one-third of samples of a
variety of antimalarial drugs acquired in Africa failed to
meet the USP dissolution specifications [19].
Stability of a formulation in a variety of storage conditions is also an important issue, particularly in tropical settings. Dissolution tests performed after simulation of
tropical conditions can reveal drug deterioration. For
example, an analysis of chloroquine phosphate tablets
marketed in Tanzania found that the reference formulation and one of the six generic products tested failed
to meet the USP dissolution specifications after 6 months
of stability testing (Table 3) [74]. Some drugs appear
to be inherently unstable and potentially unsuitable for
use in tropical conditions. For example, concerns have
been raised regarding the WHO’s recommendation of
dihydroartemisinin-containing antimalarial combination
drugs [88]. Although dihydroartemisinin is an effective
antimalarial agent, it has been shown to fail accelerated
stability tests [88]. The WHO has issued recommendations
for appropriate stability testing when drugs are to be distributed in countries with tropical climates [89]. Appropriate storage facilities may also be required; for instance, a
study in Brazil found that some state storerooms and basic
health units in the north of the country provided inadequate storage for antimalarials, given the area’s conditions of high temperature and high humidity [90].
Drug stability can be influenced by packaging, and
appropriate packaging is particularly important in conditions such as high humidity, heat or strong light. A study
by Singh and Mohan found that, under accelerated stability testing, blister-packed fixed-combination antituberculosis products were prone to physical and chemical
changes not observed with strip-packed counterparts [81].
Analysis of the unpacked products showed decomposition
of the rifampicin component, highlighting the potential
consequences if the packaging was not sufficiently robust.
Unfortunately, issues such as packaging are not as tightly
controlled as drug content, and packaging of generic
products in particular may be less able to withstand certain
storage conditions.
Even in developed countries, there is a lack of appreciation of the importance of packaging of generic products. For example, in a recent review, Zore et al. note the
influence of the bottle tip design on the dose of topical
ophthalmic preparations delivered to the eye, with drop
size varying substantially between generic manufacturers
of eye drops and causing different amounts of drug to be
delivered to the eye [91].
The advent of biosimilars [92] has also necessitated
the development of new guidelines specifically for such
products and will be another area requiring careful regulation and monitoring. The FDA has published draft
guidelines on biosimilar product development [93], while
the EMA has issued general guidance and guidelines for
specific drugs [94]. Other countries, including Japan,
India, Canada, Mexico, Brazil, Malaysia and Australia, have
also issued guidelines for registration of biosimilars.
Potential consequences of
substandard drugs
Substandard drugs pose a serious health concern from
several perspectives (Table 4 [47, 48, 51, 52, 57, 59, 70,
95–104]). Although falsified drugs have perhaps received
most of the attention with respect to causing unnecessary deaths, substandard drug manufacture also leads to
morbidity and mortality. A formulation with insufficient
API may lead to a lack of clinical response, and possibly,
death. For example, there are reports of patients failing to
respond to antimalarial treatment [95, 96] because the
drugs contained less than the stated dose of API and, in
one reported case, contained more paracetamol than
antimalarial agent [95]. In other cases, a reduced therapeutic response has been associated with generic/copy
versions of drugs compared with the originator drugs,
including antibiotics, tacrolimus and imatinib [70, 97,
99–102].
Adverse events also occur due to drug–drug interactions with contaminants, the presence of excess API,
contamination with poisonous substances, or allergic reactions to contaminants or substituted excipients. As mentioned above, some of the most extreme cases involve
the (possibly deliberate) contamination of medicines with
DEG [47, 48]. In another case, heparin was found to be
contaminated with oversulphated chondroitin sulphate,
which was thought to be responsible for the allergic or
hypersensitivity-type reactions experienced by a number
of patients, some of which proved fatal [51]. At the time of
the heparin incident, the oversulphated chondroitin sulphate could not be distinguished from heparin by the
standard quality-control tests used. However, the FDA has
since implemented changes to the USP standards for
heparin, including a new test method that is able to detect
such impurities [105].
There are also adverse societal effects arising from the
use of substandard drugs. The inadvertent use of suboptimal doses of drugs is likely to be one of the key factors
contributing to antimicrobial resistance and thereby
leading to the wider spread of disease. This has been most
widely discussed with regard to malaria [106–108]; the
repeated administration of subtherapeutic doses of antimalarials will promote the selection and spread of resistant
parasites [95, 106]. Indeed, artemisinin-resistant malaria
has been reported in Cambodia and Thailand [109, 110],
Br J Clin Pharmacol
/ 78:2
/
225
A. Johnston & D. W. Holt
Table 3
Substandard drugs: pharmacological variability
Drug(s)
Issue
Region/countries
Reference
Antimalarials
Chloroquine phosphate
Significant differences in bioavailability between two different formulations
Tanzania
Rimoy et al. (2002) [73]
Chloroquine phosphate
One locally sourced formulation and the reference formulation failed to
meet the USP dissolution specifications after 6 months in simulated
tropical conditions
Drugs from China,
Tanzania and India
available in Tanzania;
reference drug from
Belgium
Risha et al. (2002) [74]
Sulfadoxine-pyrimethamine
Two of three locally available drugs failed USP requirements for dissolution
at time of purchase
Drugs from Kenya,
Tanzania and India;
reference drug from
Switzerland
Risha et al. (2002) [74]
Amodiaquine and
sulfadoxine-pyrimethamine
13% (n = 15) of amodiaquine and 44% (n = 18) of
sulfadoxine-pyrimethamine samples failed the dissolution test, based on
USP criteria
Tanzania
Minzi et al. (2003) [12]
Sulfadoxine-pyrimethamine and
amodiaquine
40.5% (n = 116) of samples did not meet USP specifications for content
and/or dissolution
Kenya
Amin et al. (2005) [17]
Artesunate, chloroquine, quinine,
mefloquine, sulfadoxinepyrimethamine and tetracycline
1.9% (n = 53) of artesunate, 10.5% (n = 86) of chloroquine, 4.5% (n = 88)
of quinine and 1.1% (n = 93) of tetracycline samples failed disintegration
tests
Thailand
Vijaykadga et al. (2006)
[75]
Quinine, chloroquine, sulfadoxinepyrimethamine and mefloquine
46% (n = 28) failed to meet the USP dissolution standards
Congo, Burundi and
Angola
Gaudiano et al. (2007)
[76]
Sulfadoxine-pyrimethamine,
amodiaquine, mefloquine,
artesunate, artemether,
dihydroartemisinin and
artemether-lumefantrine
fixed-dose combination
35% (n = 210) of samples tested failed dissolution tests
Africa
Bate et al. (2008) [19]
12.2% (n = 304) of formulations analysed failed to meet USP specifications
for dissolution; this included 23.8% of quinine tablets
Amodiaquine samples were generally of better quality
Tanzania
Kaur et al. (2008) [77]
Generic formulation was not bioequivalent to branded version
Malawi
Hosseinipour et al. (2007)
[78]
Rifampicin
One of three locally manufactured drugs had lower bioavailability in 12
patients (ratio 0.86) compared with the reference standard
Indonesia
van Crevel et al. (2004)
[79]
Rifampicin in FDC products
Rifampicin bioavailability reduced in seven of 10 FDC products vs. individual
formulation
Subsequent study showed that FDCs were inadequately packaged, leading
to drug deterioration
South Africa/India
Pillai et al. (1999) [80];
Singh and Mohan
(2003) [81]
Generic formulation unstable; rapidly hydrolysed into two ineffective
molecules
34% (n = 50) of generics tested released less drug in 30 min than the
reference (Abbott)
Only one generic did not meet Abbott’s specification (80% must dissolve
within 30 min)
Antibacterial disc diffusion zone diameters varied significantly between
different products (n = 34)
Greece
Mastoraki et al. (2008)
[69]
Nightingale (2005) [26]
Pakistan
Iqbal et al. (2004) [82]
Tacrolimus
Compared with branded Prograf, generic Tacrobell and T-Inmun exhibited
faster dissolution; Tenacrine, Framebin and Talgraf showed slower and
incomplete drug dissolution, releasing 24–51% of tacrolimus within 2 h
Solubility of generics was decreased relative to Prograf
Mexico
Petan et al. (2008) [33]
Mycophenolate sodium
At pH 6.8, mean mycophenolate sodium release with Myfortic (reference)
was 104.9% compared with 62.3% for the generic Femulan (P = 0.04)
Six samples were tested from a single batch of each formulation
There was intratablet variability with Femulan
Mexico
Esquivel et al. (2010) [83]
Antifolates (sulfadoxinepyrimethamine,
sulfamethoxypyrazinepyrimethamine), amodiaquine,
quinine and artemisinin
derivative samples
Antiretrovirals
Triomune-40 (fixed-dose
combination of stavudine,
lamivudine and nevirapine)
Antituberculosis drugs
Other antibiotics
Cefuroxime
Clarithromycin
Ofloxacin
Multinational
(18 countries)
Immunosuppressants
226
/ 78:2
/
Br J Clin Pharmacol
Substandard drugs
Table 3
Continued
Drug(s)
Issue
Region/countries
Reference
Two of 18 generics failed the dissolution test according to USP and
Sanofi-Aventis specifications
Eleven of 35 generics did not meet dissolution criteria (<75% in 30 min)
specified by the European Pharmacopoeia and Roche
Five of 19 generics failed to meet USP specifications for dissolution
There was significantly more variability among generics produced outside
the USA
Eight of 10 test formulations were substandard for combined tests of
content/dissolution
24% (n = 21) of generics/copies failed to meet the reference ramipril
product specifications for dissolution (≥80% dissolved in 30 min)
The failure rate increased to 57% after storage for 3 months
Multinational
(five countries)
Multinational
(19 countries)
Multinational; based on
Internet availability of
generics
Rwanda
Gomez et al. (2004) [56]
Acetylsalicylic acid tablets
Three of three samples failed to meet USP dissolution specifications
Drugs from Tanzania and
Kenya available in
Tanzania
Risha et al. (2002) [74]
Paracetamol tablets
Two of nine samples failed to meet USP dissolution specifications at
purchase; five of nine failed after 3 months in simulated tropical
conditions
Drugs from India or
Tanzania, available in
Tanzania
Risha et al. (2002) [74]
Diclofenac sodium
In simulated intestinal medium, four of 16 national brands did not meet USP
specifications of 80% drug release in 8 h
Five national brands, defined as substandard, demonstrated first-order
release kinetics as opposed to zero-order kinetics specified by USP
Bangladesh
Abdullah et al. (2008)
[85]
52% of generics (n = 23) failed to meet branded Amaryl specifications for
dissolution (≥85% dissolved in 15 min)
Italy
Attorrese and
Massi-Benedetti (2007)
[55]
β-Blocker and α-agonist eye drops
for glaucoma
Generics (five samples of six products) varied significantly from branded
equivalents (five samples of five products) in drop volume, viscosity,
surface tension and bottle orifice diameter; for example, generic timolol
products delivered 37–40% less drop volume and daily prescribed dosage
than branded versions
USA and Canada
Mammo et al. (2012)
[86]
Octreotide
Comparison of Sandostatin® LAR® with three other versions of depot
octreotide formulations showed variations in microparticle size, shape,
molecular weight and acid:base ratio, suggesting different drug-release
patterns
Not stated
Petersen et al. (2011)
[87]
Cardiovascular
Clopidogrel
Carvedilol
Simvastatin
Antihypertensives
Ramipril
Italy
Smith et al. (2006) [35]
Veronin and Nguyen
(2008) [84]
Twagirumukiza et al.
(2009) [36]
Angeli and Trezza (2009)
[37]
Analgesics
Antidiabetics
Glimepiride
Other
Abbreviations are as follows: FDC, fixed-dose combination; USP, United States Pharmacopeia.
although the extent to which this can be attributed to the
use of substandard drugs is unknown. Likewise, poorquality antibiotics may contribute to the resistance
and spread of diseases such as tuberculosis [23, 111,
112]. The use and subsequent failure of substandard
narrow-spectrum antibiotics may lead to the unnecessary
administration of broad-spectrum antibiotics, thus potentially creating further resistance [113]. Substandard
antihelminthics have been implicated in the development
of drug-resistant human helminths [114], and substandard
antiviral drugs are likely to contribute to the evolution of
drug-resistant viruses, including human immunodeficiency virus (HIV) [115].
In developed countries, the impact of poor-quality
antimicrobials on drug resistance may be seen as being
minimal, with overuse considered to be the key factor. This
is exemplified by the recently published ‘threat report’
from the US Centers for Disease Control and Prevention,
which highlights the urgent issue of antibiotic resistance
and presents ‘four core actions to prevent antibiotic resistance’; the potential contribution made by substandard
drugs is not discussed [116]. However, in the modern
world, what is initially geographically localized drug resistance can rapidly become a global issue and should be
taken into account in strategies to limit drug resistance.
The potential for administration of substandard drugs to
contribute to antimicrobial resistance has been recognized by the WHO, which lists ‘inadequate systems to
ensure quality . . . of medicines’ as one of the underlying
factors that hasten the emergence and dissemination of
antimicrobial resistance [117]. A greater understanding of
the prevalence, distribution and type of quality issues
Br J Clin Pharmacol
/ 78:2
/
227
A. Johnston & D. W. Holt
Table 4
Examples of adverse outcomes associated with substandard drugs
Drug(s)
Adverse outcome
Cause
Region/countries
Reference
Diethylene glycol contamination
Nigeria, USA, Panama,
South Africa, India,
Spain, Bangladesh,
Argentina, Haiti and
China
Schep et al. (2009) [48];
Schier et al. (2009)
[47]
Miscellaneous
Paracetamol, cough
Gastrointestinal symptoms, metabolic
expectorant, propolis,
acidosis, renal injury, neuropathies and
teething syrup,
death
armillarisin, sulfanilamide,
sedatives
and silver sulfadiazine
Antimalarials
Artesunate
One patient died due to cerebral malaria
despite treatment with oral artesunate
Artemether
Sulfadoxine-pyrimethamine
No clinical response was observed after
5 days of treatment with artemether
(one patient)
An in vivo failure rate of 28.5% was
documented at a refugee camp with an
outbreak of malaria
Subsequent analysis of the drug found
Burma
Newton et al. (2006) [95]
that although artesunate was present,
the main API was paracetamol. The
drug was traced to a fake batch
purchased in good faith by the hospital
The used vial contained only 74% of the Lao People’s Democratic Keoluangkhot et al.
manufacturer’s stated dose
Republic
(2008) [96]
Locally manufactured
sulfadoxine-pyrimethamine was
substandard, i.e. it did not meet
tolerance limits for dissolution
Pakistan
Leslie et al. (2009) [97]
Generic cefuroxime (n = 305 patients) was Unstable formulation
compared with original cefuroxime
(n = 313 patients) as antimicrobial
prophylaxis in patients undergoing
coronary artery bypass grafting surgery;
12.8 vs. 2.5% of patients experienced
postoperative infections with generic vs.
original cefuroxime, respectively
(P < 0.001)
Greece
Mastoraki et al. (2008)
[69]
Allergic or hypersensitivity-type reactions
(some fatal) in patients treated with
heparin that was imported from China
and subsequently found to be
contaminated with oversulphated
chondroitin sulphate
162 cases of Serratia marcescens
bloodstream infection
Contaminant not removed in production
process
USA and Germany
Blossom et al. (2008)
[51]; Food and Drug
Administration (2009)
[98]
The infection was traced to prefilled
syringes
USA
Blossom et al. (2009) [52]
Antibiotics
Cefuroxime
Anticoagulants
Heparin
Heparin
Cardiovascular
Isosorbide-5-mononitrate
tablets
Immunosuppressants
Tacrolimus
More than 120 deaths
Contamination with antimalarial drug
Pakistan
pyrimethamine due to a manufacturing
error
Attaran et al. (2012) [57];
World Health
Organization (2012)
[59]
Use of generic version associated with
higher rate of acute kidney rejection
than use of original Prograf formulation
(20.8 vs. 11.8%; P = 0.08)
Mexico
Holm and Hernandez
(2008) [99]
Several case reports of patients with CML
who achieved a complete or partial
haematological response on Glivec, lost
the response when switched to a copy
version, but regained response when
switched back to Glivec
India, Egypt and
Morocco
Goubran (2009) [70];
Mattar (2010) [100];
Asfour and Elshazly
(2009) [101]; Chouffai
(2010) [102]
USA
Centers for Disease
Control and Prevention
(2012) [103]; Kainer
et al. (2012) [104]
Oncology
Imatinib
Steroids
Methylprednisolone
acetate
478 cases of fungal meningitis (including
34 deaths) and 12 peripheral joint
infections
Exserohilum rostratum contamination in
steroid injections
Abbreviations are as follows: API, active pharmaceutical ingredient; CML, chronic myeloid leukaemia.
228
/ 78:2
/
Br J Clin Pharmacol
Substandard drugs
affecting antimalarial drugs is also a key aspect of the
WorldWide Antimalarial Resistance Network’s efforts to
monitor the development and spread of malaria drug
resistance [118].
Some of the most obvious examples of potential societal, as well as individual, harm come from the use of substandard vaccines. As documented by Kelesidis and
colleagues, there have been incidences in which ‘vaccines’
have contained only water. A notorious case occurred in
Niger, where over 50 000 people received falsified meningitis vaccine during a meningitis epidemic in 1995; 2500
people died, and many were permanently handicapped
[119].
In addition to these clinical outcomes, being treated
with substandard drugs is likely to result in a loss of confidence in medication by both the prescribing physician
and the patient. Effective drug classes may be perceived
to be ineffective due to inadvertent suboptimal dosing,
potentially leading to unnecessary testing for suspected
resistance [97] and to unnecessary drug switching or
augmentation.
All of the above clinical and humanistic factors contribute to an increased economic burden, both on a national
scale and to individuals. In some developing countries, up
to 90% of the population have to pay for their medicines,
and these costs can account for a large proportion of
household income [120]. Paying for replacement or additional drugs, or for repeated courses of inadequate ones,
may impose a severe economic burden on a household,
especially if combined with loss of income due to illness. At
a national level, the costs associated with inadequate
or contaminated drugs may include those for lost productivity, in addition to increased direct healthcare costs if
these are at least in part met by the state. As noted by
Wertheimer and Norris, development of resistance secondary to the use of commonly available (often generic)
drugs will necessitate the development of new, probably
more expensive alternatives, thus further aggravating the
economic burden of treating infectious diseases [115].
How prevalent are
substandard drugs?
The true extent of the problem is unknown but can occur
worldwide (Figure 1). A few published reviews by investigators and reports by international bodies, such as the
WHO, have collated information from individual studies or
a range of countries to try to gain an overview of the prevalence of substandard drugs, particularly antimicrobials, in
developing countries [1, 119, 121, 122]. For example,
a comprehensive review published by Kelesidis and
colleagues highlighted the extent of the problem of
‘counterfeit/substandard’ antimicrobial drugs, particularly
in southeastern Asia and Africa [119]. This apparent geographical bias reflects the poorer regulatory control in
these areas. The authors also highlight the fact (as also
shown in Tables 1–3 in this article) that although there
have been reports of falsified or substandard drugs
in many pharmacotherapeutic groups, antibiotics and
antiparasitic drugs appear far more likely to be falsified
than other drugs [119].
In 2006, the WHO’s International Medical Products
Anti-Counterfeiting Taskforce (IMPACT) estimated that in
parts of Africa, Asia and Latin America, more than 30% of
drugs on sale could be falsified [123], although the sources
for this statistic are unclear. The UK charity Oxfam has
branded such statistics as ‘dubious’, as they appear to be
based on anecdotal reports [124].
Much of the attention in the past has focused on the
problem of deliberately falsified drugs, but even in this
area the figures are vague. Governments and the pharmaceutical industry have been criticised previously for being
reticent to make knowledge about falsified drugs public
[125]. The figure of 10% of marketed drugs being falsified
is frequently quoted and has been attributed to the WHO
and to the FDA [124, 126, 127]. However, the origin of this
statistic (as pointed out in a Wall Street Journal article [128])
may be in a 2002 British Medical Journal editorial [126] that
misquoted an earlier WHO report. Although the WHO has
used the figure in factsheets, it has since refuted it via
IMPACT [123].
The problem of patient exposure to substandard drugs
is not confined to developing countries, although IMPACT
estimated that the prevalence of falsified medicines would
be less than 1% of sales in most developed countries with
adequate regulatory systems and market control [123]. A
search of the UK’s MHRA website revealed that between
December 2005 and October 2012, 211 drug alerts were
issued, including 22 Class 1 (critical) alerts for problems
such as falsified drugs, incorrect drug quantities, packaging issues and contamination [129]. Between March 2011
and November 2012, approximately 40 company-led
recalls were also issued for problems including contamination, inadequate stability and labelling or packaging concerns. The country of origin of the drugs concerned was
not stated.
Substandard medicines are also sold via the Internet,
thus potentially affecting a wide patient population. The
WHO has estimated that over 50% of medicines purchased
via the Internet from sites that conceal their physical
address are falsified [8] although, again, the evidence for
this is not clear. However, an analysis of selected drugs
purchased from a range of website pharmacies found
that, with the exception of Viagra®, the drugs met quality
standards [130]. Phosphodiesterase type 5 inhibitors (i.e.
sildenafil, tadalafil and vardenafil) are the drugs most likely
to be falsified [131] and so are probably the most common
falsified drugs to be sold via the Internet. Contaminants in
falsified sildenafil have included talcum powder, paint and
a range of APIs [131]. Conversely, it has been reported that
a ‘natural’ herbal product sold as a remedy for impotence
Br J Clin Pharmacol
/ 78:2
/
229
A. Johnston & D. W. Holt
M. Russia
November 2012
A. Concorde, USA
December 2010
E. UK
June 2011
B. Framingham, USA
F. Switzerland
September 2012
June 2007
L. Jodhpur, India
February 2011
N. Beijing, China
January 2011
O. China,
May 2012
G. Senegal
C. Angier, USA
June 2007
December 2007
K. India
November 2012
D. Brazil
H. Makurdi, Nigeria
November 2011
January 2011
I. Malawi
J. Dar-es-Salaam,Tanzania
January 2011
October 2012
P. Australia
October 2012
Figure 1
Examples of recent accounts of substandard drugs around the world. (A) GlaxoSmithKline fined because four drugs it marketed did not meet US Food and
Drug Administration’s strength and purity standards (Boston.com). (B) Outbreak of fungal meningitis traced to contaminated methylprednisolone injections produced by the New England Compounding Center [103]. (C) Five deaths and 300 people infected due to distribution of contaminated prefilled
heparin and saline syringes by AM2PAT (The Institute for Southern Studies, 2009). (D) Defects in quality of antimalarial drugs held at national and state
facilities [90]. (E) Plasticizers, including diisodecyl phthalate and diisononyl phthalate, found in Augmentin formulations manufactured in the UK; products
recalled in Hong Kong and China (The Telegraph/FiercePharma). (F) Ethyl mesylate contamination of nelfinavir at Roche’s Swiss manufacturing plant
(National AIDS Manual, 2007). (G) Ghana’s medicines regulatory authority ordered Pfizer to recall substandard Camoquine-plus suspension manufactured
in Senegal and distributed in Ghana (Ghana News Agency). (H) Nigeria’s National Agency for Food and Drug Administration raided pharmacies and other
outlets and impounded a variety of drugs believed to be falsified (Nigerian Tribune). (I) Pharmacy, Medicines and Poisons Board recalled aspirin tablets on
suspicion of being substandard (Malawi News). (J) The Tanzania Food and Drugs Authority suspended importation, distribution, sale and use of nine types
of medicines found to be substandard (The Citizen, Tanzania). (K) As of November 2012, almost 300 drugs have been listed as being found to be substandard
in 2012 (DrugsControl.org, India). (L) Thirteen women died in little over a week due to contaminated intravenous fluids. No system was in place for testing
or checking medicines (The New Indian Express). (M) Russia’s Federal Service on Surveillance in Healthcare (Roszdravnadzor) discovered and removed 530
series of ‘low-quality’ drugs from circulation in 2012; over 70% of these were produced by domestic manufacturers (HIS Global Insight/Rossiyskaya Gazeta).
(N) Six hundred and sixty-nine batches of gel capsules from 254 drug manufacturers were found to have high chromium levels (30 May 2012;
FiercePharmaManufacturing). (O) More than 60 tonnes of fake and substandard pharmaceuticals and medical equipment were destroyed by Beijing’s
municipal drug supervision bureau (People’s Daily Online). (P) Batches of Typhim Vi vaccine were recalled because they may have lower than expected
antigen content (Therapeutic Goods Administration)
actually contained sildenafil [132]. The standards of packaging and labelling of drugs sold via the Internet has also
raised concerns [133].
With regard to the distribution of poor-quality genuine
medicines that have resulted from deficiencies in the
manufacturing process, there are isolated published
reports, press articles, as well as recall information made
available by national authorities, but there are no definitive statistics regarding the scale of the overall problem. If
systems are not in place to make manufacturers assess and
report such defects and to make the regulatory bodies
manage the drug recall, there is considerable scope for
harm. The scale of such incidents in countries with poor or
non-existent fail-safes is, of course, unknown.
230
/ 78:2
/
Br J Clin Pharmacol
How can drug quality be
improved?
There are two separate, albeit related, issues in preventing
the dissemination of substandard drugs. One is combating
the trade in falsified drugs from a legal perspective. The
other is ensuring adequate quality at the drug manufacture
and quality control levels for both branded and generic
drugs alike. While many steps are being taken to ensure the
authenticity of marketed drugs, including the approval of
new legislation by the European Parliament [134], these do
not necessarily address the wider issue of controlling drug
quality. There are many levels at which drug quality monitoring and regulation can be improved. A full discussion of
Substandard drugs
the potential research activities and possible solutions that
could be implemented by academics, regulators and
manufacturers is beyond the scope of this article, and
readers are encouraged to consult recent comprehensive
reviews of these topics [10, 135–137]. Some of the key
requirements and initiatives for combating the distribution
of substandard drugs will be discussed below.
Better understanding of the problem
There is an urgent need for greater understanding of the
problem, in particular through better systematic collection
and accurate, transparent documentation of information
on substandard drug manufacture and dissemination. This
would help inform national authorities about the scale of
the problem and provide a database against which
batches of drugs could be checked. In addition, it may help
in assessing the true degree of local drug resistance to, for
example, antimalarials. The paucity of accurate data is
highlighted by the oft-quoted but rather doubtful ‘statistics’ on substandard drugs, as discussed earlier. While
some information is documented in peer-reviewed journals (as shown above) or official reports, much of our
current awareness of the problem of substandard drugs is
due to press reports, Internet articles and anecdotal
accounts. An interesting contrast can be made with the
effort expended on trying to combat ‘illicit’ drug distribution (where such drugs usually comprise narcotics, stimulants, sedatives, hallucinogens and cannabis, i.e. drugs of
abuse for the want of a better term). National governments commit substantial amounts of money to investigate and prevent illicit drug production. Each year, the
United Nations issues a large ‘World Drug Report’ that
includes extensive statistics on production and consumption of such drugs and analyses of the economic burden of
illicit drug use [138]. The issue of substandard drugs is
probably a much larger problem, and affects more people,
therefore surely warranting similar attention.
Carefully conducted surveys with precise targets and
incorporating standardized testing could be used to help
define the extent of the problem. For example, an ongoing
study has been established by the WHO and the Drug
Quality and Information Program on the quality of antimalarial medicines in 10 Sub-Saharan countries (the QAMSA
study). Antimalarial drug samples are obtained from both
the regulated and ‘informal’ market and tested using the
Global Pharma Health Fund Minilab® kit and/or full USP
laboratory analysis. Initial findings have shown that 44% of
sampled antimalarial medicines in Senegal failed to meet
USP quality standards. The failure rates in Madagascar and
Uganda were 30 and 26%, respectively [139]. Minilab
testing (444 samples) showed that 43, 12 and 6% of
samples in Senegal, Uganda and Madagascar, respectively,
failed quality assessments. A similar QAMSA survey in
Cameroon, Ethiopia, Ghana, Kenya, Nigeria and Tanzania
found that of 267 tested antimalarial drug samples, 28.5%
failed to comply with specifications, with the proportions
ranging from 0% in Ethiopia to 64% in Nigeria [140].
The International Medical Products Anti-Counterfeiting
Taskforce was launched by the WHO in 2006 with the aim
of detecting and preventing the production and sale of
‘counterfeit’ medicines. However, this project has been
criticised by member states, and most notably in a recent
report issued by Oxfam, for apparently focusing on enforcing intellectual property rights in order to prevent criminal
trademark-infringement activities (i.e. prevent the distribution of falsified drugs), rather than trying to combat the
wider issues of falsified drugs or the substandard manufacture of legally produced and marketed drugs [124].
Oxfam has also criticised IMPACT for causing ‘unnecessary
confusion’ by misusing the term ‘counterfeit’ to refer to
‘substandard and falsified medicines that are unrelated to
criminal trademark infringement’. Oxfam has called for
IMPACT to be disbanded [124]. At the 63rd World
Health Assembly in May 2010, it was decided to create a
‘time-limited and results-oriented working group on
substandard/spurious/falsely labelled/falsified/counterfeit
medical products (SSFFC) comprised of and open to all
member states’. The remit of the working group would
include investigating IMPACT. Progress has been slow,
much to the concern of several countries, particularly India
[141]. However, in May 2012 a Member State Mechanism
was approved for international collaboration to prevent
and control SSFFC [142], and the first formal meeting was
held in November 2012.
In 2009, the US Agency for International Development
(USAID) and USP jointly set up the Promoting the Quality
of Medicines programme, aimed at ensuring the quality,
safety and efficacy of medicines in USAID’s health programmes. In April 2011, in collaboration with authorities in
Africa, South America and Southeast Asia, the Promoting
the Quality of Medicines programme launched the Medicines Quality Database [143], which reports on the quality
of a variety of medicines collected in these regions.
The WorldWide Antimalarial Resistance Network has
recognized the paucity of high-quality published data on
the topic of substandard drugs and has established a pilot
database of reports on antimalarial drug quality [118].
Most recently, as part of its ongoing aim to collate information on the prevalence and distribution of poor-quality
antimalarials around the world, the Antimalarial Quality
Scientific Group of the WorldWide Antimalarial Resistance
Network launched an online interactive antimalarialquality surveyor, which maps reports of antimalarial
quality from a variety of sources [144, 145].
Improved regulatory control and monitoring
Key to the improvement and maintenance of drug quality
is the implementation of strong regulatory control. There
is a need for pharmacovigilance programmes to be in
place in order to monitor the safety of marketed drugs
Br J Clin Pharmacol
/ 78:2
/
231
A. Johnston & D. W. Holt
constantly and to communicate any safety issues to manufacturers, healthcare providers and patients.
The level of such control varies widely. In the USA, for
example, minimal Good Manufacturing Practice standards
are defined by the FDA and upheld by law. In many other
countries, there is negligible control. Caudron and colleagues even noted that manufacturers may run parallel
production processes, adjusting their standard according
to that of the recipient country [1]. The WHO estimates
that only approximately 20% of its 191 member states
have well-developed drug regulation; approximately 30%
are thought to have no or minimal drug regulation in place
[146]. It should also be borne in mind that the standards
set by different NMRAs may vary and may not meet international standards. The survey conducted by the WHO in
six Sub-Saharan countries found that 14% of collected
antimalarial drug samples were not registered by the
NMRA [140]. Worryingly, in Pakistan the pharmaceutical
industry has been operating without formal control since
the national Ministry of Health was abolished in June 2011
and its power devolved to the provinces. However, following the deaths in January 2012 of over 100 patients in
Lahore due to contamination of the antianginal agent
Isotab® (isosorbide mononitrate 20 mg) with the antimalarial pyrimethamine, the government of Pakistan established the Drug Regulatory Agency to regulate the
country’s pharmaceutical industry [59, 147].
The WHO has implemented a number of programmes
to try and address the problem of substandard drugs. One
is the prequalification programme, established in 2001 on
behalf of the United Nations, which aims to ensure access
to medicines that meet specified standards of quality,
safety and efficacy, mainly for HIV/AIDS, malaria, tuberculosis and reproductive health [148]. Candidate products
and their manufacturing sites are assessed by stringent
regulatory bodies. Those that pass are placed on a list that
can be used by international agencies such as UNICEF, as
well as by national agencies requiring guidance. The
prequalification programme regularly re-inspects manufacturing sites of prequalified products to ensure the continued safety of such products. The WHO survey of
antimalarial drug quality in six Sub-Saharan countries
noted that, while the overall failure rate was almost 30%,
only 4% of samples of prequalified medicines failed testing
and, in each case, the deviation was considered minor
[140]. However, as has been pointed out by others, the list
of prequalified drugs for HIV/AIDS issued by the WHO
includes a disclaimer stating that ‘Inclusion in the list does
not constitute an endorsement, or warranty of the fitness,
by WHO of any product for a particular purpose, including
in regard of its safety and/or efficacy in the treatment of
HIV/AIDS’ [149], which raises questions about the value of
the programme. It is also worth noting that the WHO has
approved a ‘biowaiver’ procedure, whereby it accepts evidence of equivalence other than in vivo equivalence
testing [150].
232
/ 78:2
/
Br J Clin Pharmacol
The Good Governance for Medicine programme was
set up by the WHO in 2004 to help combat corruption in
the pharmaceutical sector and is currently operating in 26
countries [151]. The programme comprises a three-phase
approach, covering assessment, development and implementation of national Good Governance for Medicine programmes. Using a standardized assessment instrument
[152], the first phase of the programme in any country
involves a national assessment of transparency and potential vulnerability to corruption in different aspects of drug
production and marketing. These aspects include the
following: registration of medicines and control of their
promotion; inspection and licensing of establishments;
selection, procurement and distribution of essential medicines; and control of clinical trials. A recent report into five
countries noted deficiencies in the areas of ‘management
of conflict of interest with regard to inspection activities’
and ‘written procedures or mechanisms to prevent personal relations between an inspector and the manufacturers or distributors’, among others [153]. However, these
findings are only of value if action is taken by the individual
countries to revise their existing policies and enforce good
governance. The WHO reports a number of successes for
the programme but emphasizes that high-level government commitment is required [154].
In addition to monitoring of drug products, more stringent control is needed for the quality of APIs. A press
report from India noted that, despite regulations being
introduced in 2003 to curb the import of substandard raw
materials, customs authorities seized several consignments of substandard materials imported by traders. The
ingredients were believed to have entered the country
through smaller ports, where ‘customs scrutiny was less
stringent’ [155]. In 2010, the WHO’s prequalification programme was extended to include a pilot scheme for
prequalification of selected APIs for products for treating
HIV and related diseases, for antimalarial medicinal products and for antituberculosis products [156], but wider
regulation is still required.
In Africa, Nigeria’s National Agency for Food and Drug
Administration (NAFDAC) has provided a model for how to
improve the provision of safe medicines. Since 2001, it has
implemented a series of measures to combat falsified and
poor-quality drugs and to ensure drug safety. In 2001, a
survey estimated that 68% of the drugs available in Nigeria
were not registered with NAFDAC; by 2004, this level had
been reduced by 80% [157]. The actions of NAFDAC
include recommendations for changes in the law, dismissal of corrupt NAFDAC personnel, guidelines for staff
behaviour and use of incentive schemes for NAFDAC staff,
destruction of large quantities of falsified and expired
drugs, strict enforcement of registration guidelines, implementation of new guidelines to ensure that imported
drugs are genuine, and raising public awareness of falsified drugs, including publication of lists of identified fake/
substandard products in newspapers [157–159]. A review
Substandard drugs
conducted in 2007 using the WHO’s Good Governance for
Medicine assessment tool found that the pharmaceutical
sector in Nigeria was still vulnerable to corruption and that
the greatest weakness was the lack of conflict-of-interest
guidelines [160].
The NAFDAC, in partnership with some pharmaceutical
companies, has also tested and adopted an innovative
new mobile authentication service developed by a US
company (Sproxil) to help ensure that only genuine drugs
reach the consumer. This comprises a scratch panel added
to the drug package; the consumer scratches the panel to
reveal a code, which can then be sent in a text message to
the authentication service. A message is sent back confirming whether or not the drug is genuine. The service is
free for consumers [161, 162]. In January 2012, Sproxil
announced that the verification service had been used to
check medicines more than one million times in Africa
[163]. The system is also being adopted in India and Kenya.
Other regions and countries have implemented initiatives to prevent the distribution of substandard drugs,
although these are mainly aimed at detecting falsified
drugs. For example, the Directorate General of Foreign
Trade in India is currently implementing a barcode-based
‘track and trace’ system, to help prevent the export of substandard drugs [164, 165].
As mentioned above, the European Parliament will call
for new safety features to be applied to individual packs of
drugs in order to ‘identify them, guarantee their authenticity, and enable pharmacists to check whether the outer
packaging has been tampered with’ [134]. Member states
will also be required to implement a system to prevent
medicinal products that are ‘falsified and with quality
defects’ from reaching the patient. The European Falsified
Medicines Directive [134] came into force in January 2013,
with the aim of improving the monitoring of the supply
chain for medicinal products and active substances. The
Directive includes the use of unique identifiers on packs of
medicines. Tracking systems are being developed and
tested by the European Federation of Pharmaceutical
Industries and Associations [166] and the European Directorate for the Quality of Medicines and Healthcare [167].
In the USA, the FDA has been in discussion with pharmaceutical companies regarding the instigation of a ‘track
and trace’ system, but this is likely to be delayed by cost
concerns.
The European Falsified Medicines Directive also
includes development of guidelines for good distribution
practices for medical products and APIs. The European
Commission [168] has revised its good distribution practice guidelines for medical products and established a new
database (EudraGMDP) to facilitate the checking of information on manufacturers, importers and distributors of
APIs. Draft guidelines covering APIs have also been issued
recently by the European Commission [169]. Even in developed countries, the issue of poor-quality APIs has caused
problems and is being addressed by tighter control and
international agency cooperation. A pilot programme was
initiated in 2008 to share information and arrange joint
inspections of API manufacturers. The pilot programme
included the EMA, France, Germany, Ireland, Italy, UK, the
European Directorate for the Quality of Medicines and
Healthcare, the FDA and the Australian Therapeutic Goods
Administration and proved successful over a 2-year period.
The programme will be continued and other NMRAs
encouraged to participate [170].
Developed countries import large quantities of generic
drugs and APIs from abroad. It has been estimated that
approximately 40% of drugs and 80% of the APIs in pharmaceutical products available in the USA are manufactured elsewhere [171]. Key source countries are China and
India. In recognition of the need to ensure regulatory oversight, the FDA has established offices in several countries,
including China, India, Costa Rica, Chile, Mexico and South
Africa [172]. Activities run from these offices include
inspections of facilities and products, training of local
regulators and aiding with investigation of adverse incidents, such as suspected contamination.
Parallel importation also carries a risk of substandard
drugs being made available as the drugs may be imported
via a complex supply chain that is difficult to track. This was
highlighted by the recent case of falsified Avastin, thought
to have originated in Turkey, which was purchased by
medical practices in the USA from foreign or unlicensed
suppliers. The purchased drugs were found to contain no
API [173].
Most recently, a group of experts in aspects of
healthcare provision have proposed the establishment of
a global treaty to address the issues of both falsified and
substandard medicines. The actions of the treaty would
include a legal definition of substandard drugs, establishing track and trace technologies to authenticate medicines, and provision of financial and technical assistance to
poorer countries in order to strengthen their medicines
regulatory authorities [57].
Human and material resources
A lack of resources to test drug quality is a central issue. A
review of occurrences of DEG poisoning noted that in one
incident in Haiti, although the pharmaceutical company
had the necessary equipment for purity testing, it was not
in use and the staff did not know how to use it [47]. A
telling press article from Bangladesh reported that the
high court had ordered the government to collect and test
samples of all the medicines available in 20 specific areas
[174]. The authorities had duly collected samples of 174
medicines, but could only test 48 items ‘due to shortage of
lab equipment’.
Of course, one of the simplest tests is visual inspection
of the drugs and their packaging. For example, a case was
recently reported in the UK of a patient with malaria that
had not resolved with administration of artesunat purchased in Equatorial Guinea. Examination of the outer
Br J Clin Pharmacol
/ 78:2
/
233
A. Johnston & D. W. Holt
packaging of the purchased medication revealed spelling
mistakes and design differences, compared with that of
the genuine drug, as well as an incorrect company registration number [175]. The tablets were found to contain no
API. Reference standards are one of the essential requirements for routine assessment of drug quality and can be
purchased from several pharmacopoeias. However, as
noted by the USP, limited resources may restrict the use of
such standards, and quality testing is then compromised
[176]. As part of the Promoting the Quality of Medicines
programme, the USP has launched a pilot Technical Assistance Program, which aims to provide practical assistance
to regulators in developing countries [176]. Participating
countries (initially, Ethiopia, Ghana, Kenya, Senegal and
Sierra Leone) will receive, free of charge, a set of pharmaceutical reference standards, documentary standards and
technical training to aid in assessing drug quality. The
success of the programme will be evaluated after
12 months.
A number of relatively simple and inexpensive tests
have been developed for drug testing. Although not
necessarily as sensitive or specific as pharmacopoeial
methods, they can be used to screen samples quickly. Such
techniques include thin-layer chromatography, which has
been used as a screening method for a range of suspected
substandard drugs, including antituberculosis and antimalarial drugs [23, 177, 178]. Simple colorimetric assays have
also been established for some drugs, including the Fast
Red TR test that can distinguish the presence of the
artemisinin-derived compounds artemether, artesunate
and dihydroartemisinin [14, 179, 180]. Dissolution and disintegration tests are also widely used (see Table 3). The
Global Pharma Health Fund has developed Minilab®,
which incorporates four tests, namely visual inspection,
disintegration, colour reaction and thin-layer chromatography tests. A Minilab fits into two suitcases and includes
supplies for approximately 1000 assays [181]. The kit has
been used for testing the identity of a variety of drugs and
may be suitable for initial screening of the quality of
imported medicines, as piloted by the Tanzania Food and
Drugs Authority [182]. Hand-held Raman and near-infrared
instruments are also available for testing of drugs in the
field in developing countries, although there are some
drawbacks with these approaches [183–186].
The activity of an NMRA or other body assessing drug
quality is only as reliable as its staff and, thus, is vulnerable
to deficiencies in training and to corruption. There are
many levels in the drug production and marketing processes that may be influenced by corruption and lead to
substandard drugs entering the market. This may occur,
for example, during the construction or equipping of
manufacturing facilities, during drug registration or certification, during quality-control checks, including drug
testing and site inspections, and during drug procurement
[187–189]. One such case was highlighted when the FDA
uncovered evidence that a facility in India owned by
234
/ 78:2
/
Br J Clin Pharmacol
Ranbaxy Laboratories had falsified data and test results in
approved and pending drug applications on at least two
occasions [5, 190]. Provision of adequate training, pay and
incentives for staff, as well as greater transparency in all
these processes, are required to help alleviate such problems. In Nigeria, for example, as part of a drive to eradicate
corruption and to support staff, NAFDAC introduced
incentives for its personnel, including training abroad,
improved facilities and a better working environment
[159].
Improvements in equipment, staff training and
implementation of testing and procedures obviously
require funding. One possible source is increased drugregistration fees. These fees are substantially lower in
developing countries than in developed countries. For
example, a review of fees in Latin American countries
found that the charges were generally of the order of a few
hundred US dollars, but were as low as just US$6 in Guatemala (cost in 2002) [191]. Other developing countries
also charge fees substantially lower than those in developed countries (Table 5). Following a review of NMRA fees
in 2002, Kaplan and Laing concluded that for new drug
registrations, developing countries could charge between
one and five times their per capita gross national product
or between US$17 000 and US$80 000 for each US$1000
spent per capita on healthcare, with lower fees for generic
drugs [192]. Higher fees could contribute to a more effective regulatory service. However, it is important to strike
a balance so that higher costs are not passed on to the
consumer, perpetuating the vicious circle in which the
production of falsified or low-quality drugs is promoted
because of the need for cheaper medicines. Kaplan and
Laing emphasize the need for significant additional government funding.
Conclusions
Effective drugs are now available for some of the most
prevalent and destructive diseases in the developing
world, including tuberculosis, malaria and HIV/AIDS.
However, the effectiveness of drugs in treating these diseases, as well as many other illnesses, is compromised by
the distribution of substandard drugs. Both branded and
generic drugs are affected. Generic formulations offer lowcost options for many drugs, and generic substitution may
be mandatory in some countries, but the quality of these
drugs must be regulated. In parallel with the resources
invested in tackling the problem of deliberately falsified
drugs, global effort is required to combat the distribution
of low-quality medicines arising through poor manufacturing processes and poor regulatory oversight. Having
strong, suitably empowered and well-funded national
drug regulatory agencies is essential, and many countries
will need help in achieving this goal. Not funding
resources to address substandard drug production is a
Substandard drugs
Table 5
Example drug-registration fees
Country/area Authority
Date
Drug registration fee
Source
Australia
Therapeutic Goods Administration 2012
Prescription medicines (application http://www.tga.gov.au/about/fees-current.htm
+ evaluation)
New chemical entity:
AU$210 100
New generic product: AU$80 200
€267 400
http://www.ema.europa.eu/docs/en_GB/document
_library/Other/2012/03/WC500124904.pdf
Europe
European Medicines Agency
2012
India
Central Drugs Standard Control
Organization
http://cdsco.nic.in/html/Sec_122_A.htm
Kenya
Pharmacy and Poisons Board,
Ministry of Health
Not known; information currently Imported drug: INR50 000
available on website dated
(approximately US$913/£576)
2007
Current information on website
Imported: US$1000.
dated 2010
Fully manufactured in Kenya:
US$500
(US$4000 for Good
Manufacturing Practice
inspection)
Nigeria
National Agency for Food, Drug
Administration and Control
2011
Tanzania
Food and Drugs Authority
2011/2012
NGN700 000 (approx.
US$4500/£2700) every 5 years
+ 5% VAT
Foreign manufactured products:
application fee US$500;
registration certificate US$100
per product
http://www.unido.org/fileadmin/user_media/Services/
PSD/BEP/Nigeria_Pharma%20Sector%20Profile
_032011_Ebook.pdf
http://pharmabiz.com/Services/ExportImport/
Countries/Tanzania.aspx
Uganda
National Drug Authority
2010
USA
Food and Drug Administration
Published in 2012, applicable in
2012/2013
Foreign manufactured products:
US$500.
Application requiring clinical data:
US$1 958 800
http://pharmabiz.com/Services/ExportImport/
Countries/Uganda.aspx
http://www.gpo.gov/fdsys/pkg/FR-2012-08-01/pdf/
2012-18711.pdf
http://pharmacyboardkenya.org/?page_id=401
Abbreviations are as follows: INR, Indian rupee; NGN, Nigerian naira; PKR, Pakistani rupee.
false economy; in addition to causing human suffering, the
use of substandard drugs can have an enormous economic impact on individuals, families, health providers
and states. Governments, drug manufacturers, charities,
care providers and patients alike must play their part in
ensuring that only drugs of sufficient quality are available
for use.
Competing Interests
Both authors have completed the Unified Competing
Interest form at http://www.icmje.org/coi_disclosure.pdf
(available on request from the corresponding author) and
declare: Novartis Pharmaceuticals Corporation funded
the editorial assistance from Anthemis Consulting Ltd
for the submitted work; AJ has received reimbursements
and funding from pharmaceutical companies including
Astellas, AstraZeneca, Baxter, Novartis, Roche and Sanofi
and owns stock in Abbott, AstraZeneca, Pfizer, Roche and
Sanofi; DWH has been a speaker for Novartis and Sanofi on
issues relating to the impact of drug quality on safety and
efficacy.
Financial support for medical editorial assistance was
provided by Novartis Pharmaceuticals Corporation. We
thank Dr Julie Ponting of Anthemis Consulting Ltd for medical
editorial assistance with this manuscript.
REFERENCES
1 Caudron JM, Ford N, Henkens M, Mace C, Kiddle-Monroe R,
Pinel J. Substandard medicines in resource-poor settings: a
problem that can no longer be ignored. Trop Med Int
Health 2008; 13: 1062–72.
2 European Medicines Agency. European Medicines Agency
recommends precautionary recall of batches of
clopidogrel-containing medicines from Acino Pharma
GmbH. March 2010. Available at http://www.ema.europa
.eu/ema/index.jsp?curl=pages/news_and_events/
news/2010/03/news_detail_001010.jsp&mid=
WC0b01ac058004d5c1 (last accessed 18 December 2013).
3 Food and Drug Administration. New England
Compounding Center issues voluntary nationwide recall of
all products. October 6, 2012. Available at
http://www.fda.gov/Safety/Recalls/ucm322901.htm (last
accessed 18 December 2013).
Br J Clin Pharmacol
/ 78:2
/
235
A. Johnston & D. W. Holt
4 Food and Drug Administration. FDA issues warning letters
to Ranbaxy Laboratories Ltd., and an import alert for drugs
from two Ranbaxy plants in India, 2008. Available at
http://www.fda.gov/newsevents/newsroom/
pressannouncements/2008/ucm116949.htm (last accessed
18 December 2013).
5 Food and Drug Administration. Department of Justice files
consent decree of permanent injunction against Ranbaxy.
January 25, 2012. 2012. Available at http://dineshthakur
.com/wp-content/uploads/2013/05/2012.01.25-FDA
-Consent-Decree-Press-Release.pdf (last accessed 18
December 2013).
6 World Health Organization. Counterfeit medicines:
frequently asked questions, 2009. Available at
http://www.who.int/medicines/services/counterfeit/faqs/
QACounterfeit-October2009.pdf (last accessed 18
December 2013).
7 United Nations Office on Drugs and Crime. World Drug
Report 2010, 2010. Available at http://www.unodc.org/
documents/wdr/WDR_2010/World_Drug_Report_2010
_lo-res.pdf (last accessed 18 December 2013).
8 World Health Organization. Substandard and counterfeit
medicines. Fact sheet No. 275, 2003. Available at
http://www.who.int/mediacentre/factsheets/2003/fs275/
en/ (last accessed 18 December 2013).
9 Commission of the European Communities. Executive
summary. Proposal for a directive of the European
parliament and of the council amending Directive
2001/83/EC as regards the prevention of the entry into the
legal supply chain of medicinal products which are falsified
in relation to their identity, history or source, 2008.
Available at http://eur-lex.europa.eu/LexUriServ/
LexUriServ.do?uri=SEC:2008:2675:FIN:en:PDF (last accessed
18 December 2013).
10 Bate R. Phake: The Deadly World of Falsified and
Substandard Medicines. Washington, DC: AEI Press, 2012.
11 Taylor RB, Shakoor O, Behrens RH, Everard M, Low AS,
Wangboonskul J, Reid RG, Kolawole JA. Pharmacopoeial
quality of drugs supplied by Nigerian pharmacies. Lancet
2001; 357: 1933–6.
12 Minzi OM, Moshi MJ, Hipolite D, Massele AY, Tomson G,
Ericsson O, Gustafsson LL. Evaluation of the quality of
amodiaquine and sulphadoxine/pyrimethamine tablets
sold by private wholesale pharmacies in Dar Es Salaam
Tanzania. J Clin Pharm Ther 2003; 28: 117–22.
13 Lambert PA, Conway BR. Pharmaceutical quality of
ceftriaxone generic drug products compared with
Rocephin®. J Chemother 2003; 15: 357–68.
14 Basco LK. Molecular epidemiology of malaria in Cameroon.
XIX. Quality of antimalarial drugs used for self-medication.
Am J Trop Med Hyg 2004; 70: 245–50.
15 Dondorp AM, Newton PN, Mayxay M, Van Damme W,
Smithuis FM, Yeung S, Petit A, Lynam AJ, Johnson A, Hien
TT, McGready R, Farrar JJ, Looareesuwan S, Day NP, Green
MD, White NJ. Fake antimalarials in Southeast Asia are a
major impediment to malaria control: multinational
236
/ 78:2
/
Br J Clin Pharmacol
cross-sectional survey on the prevalence of fake
antimalarials. Trop Med Int Health 2004; 9: 1241–6.
16 Abdo-Rabbo A, Bassili A, Atta H. The quality of
antimalarials available in Yemen. Malar J 2005; 4: 28.
17 Amin AA, Snow RW, Kokwaro GO. The quality of
sulphadoxine-pyrimethamine and amodiaquine products
in the Kenyan retail sector. J Clin Pharm Ther 2005; 30:
559–65.
18 Atemnkeng MA, De Cock K, Plaizier-Vercammen J. Quality
control of active ingredients in artemisinin-derivative
antimalarials within Kenya and DR Congo. Trop Med Int
Health 2007; 12: 68–74.
19 Bate R, Coticelli P, Tren R, Attaran A. Antimalarial drug
quality in the most severely malarious parts of Africa – a six
country study. PLoS ONE 2008; 3: e2132.
20 Tipke M, Diallo S, Coulibaly B, Störzinger D, Hoppe-Tichy T,
Sie A, Müller O. Substandard anti-malarial drugs in Burkina
Faso. Malar J 2008; 7: 95.
21 Onwujekwe O, Kaur H, Dike N, Shu E, Uzochukwu B,
Hanson K, Okoye V, Okonkwo P. Quality of anti-malarial
drugs provided by public and private healthcare providers
in south-east Nigeria. Malar J 2009; 8: 22.
22 El-Duah M, Ofori-Kwakye K. Substandard artemisinin-based
antimalarial medicines in licensed retail pharmaceutical
outlets in Ghana. J Vector Borne Dis 2012; 49: 131–9.
23 Laserson KF, Kenyon AS, Kenyon TA, Layloff T, Binkin NJ.
Substandard tuberculosis drugs on the global market and
their simple detection. Int J Tuberc Lung Dis 2001; 5:
448–54.
24 Prazuck T, Falconi I, Morineau G, Bricard-Pacaud V,
Lecomte A, Ballereau F. Quality control of antibiotics
before the implementation of an STD program in Northern
Myanmar. Sex Transm Dis 2002; 29: 624–7.
25 Syhakhang L, Lundborg CS, Lindgren B, Tomson G. The
quality of drugs in private pharmacies in Lao PDR: a repeat
study in 1997 and 1999. Pharm World Sci 2004; 26: 333–8.
26 Nightingale CH. A survey of the quality of generic
clarithromycin products from 18 countries. Clin Drug
Investig 2005; 25: 135–52.
27 Weir RE, Zaidi FH, Charteris DG, Bunce C, Soltani M,
Lovering AM. Variability in the content of Indian generic
ciprofloxacin eye drops. Br J Ophthalmol 2005; 89: 1094–6.
28 Trefi S, Gilard V, Malet-Martino M, Martino R. Generic
ciprofloxacin tablets contain the stated amount of drug
and different impurity profiles: a 19F, 1H and DOSY NMR
analysis. J Pharm Biomed Anal 2007; 44: 743–54.
29 Kyriacos S, Mroueh M, Chahine RP, Khouzam O. Quality of
amoxicillin formulations in some Arab countries. J Clin
Pharm Ther 2008; 33: 375–9.
30 Bate R, Tren R, Mooney L, Hess K, Mitra B, Debroy B,
Attaran A. Pilot study of essential drug quality in two major
cities in India. PLoS ONE 2009; 4: e6003.
31 Hadi U, van den Broek P, Kolopaking EP, Zairina N, Gardjito
W, Gyssens IC. Cross-sectional study of availability and
Substandard drugs
pharmaceutical quality of antibiotics requested with or
without prescription (over the counter) in Surabaya,
Indonesia. BMC Infect Dis 2010; 10: 203.
32 Okumura J, Taga M, Tey S, Kataoka Y, Nam N, Kimura K.
High failure rate of the dissolution tests for 500-mg
amoxicillin capsules sold in Cambodia: is it because of the
product or the test method? Trop Med Int Health 2010; 15:
1340–6.
33 Petan JA, Undre N, First MR, Saito K, Ohara T, Iwabe O,
Mimura H, Suzuki M, Kitamura S. Physiochemical properties
of generic formulations of tacrolimus in Mexico. Transplant
Proc 2008; 40: 1439–42.
34 Hermentin P, Cuesta-Linker T, Weisse J, Schmidt KH, Knorst
M, Scheld M, Thimme M. Comparative analysis of the
activity and content of different streptokinase
preparations. Eur Heart J 2005; 26: 933–40.
35 Smith JC, Tarocco G, Merazzi F, Salzmann U. Are generic
formulations of carvedilol of inferior pharmaceutical
quality compared with the branded formulation? Curr Med
Res Opin 2006; 22: 709–20.
44 Health Canada. Adoption of ICH Guidance: impurities in
new drug substances – ICH Topic Q3A(R), 2003. Available
at http://www.hc-sc.gc.ca/dhp-mps/alt_formats/
hpfb-dgpsa/pdf/prodpharma/q3a(r)-eng.pdf (last accessed
18 December 2013).
45 International Conference on Harmonisation Expert
Working Group. Impurities in new drug substances
Q3A(R2), 2006. Available at http://private.ich.org/
LOB/media/MEDIA422.pdf (last accessed 18 December
2013).
46 Itah AY, Udokpoh AE, Ofum MU. Bacteriological quality of
some pharmaceutical products marketed by drug vendors
in Uyo, Nigeria. Afr J Health Sci 2004; 11: 128–33.
47 Schier JG, Rubin CS, Miller D, Barr D, McGeehin MA.
Medication-associated diethylene glycol mass poisoning: a
review and discussion on the origin of contamination.
J Public Health Policy 2009; 30: 127–43.
48 Schep LJ, Slaughter RJ, Temple WA, Beasley DM.
Diethylene glycol poisoning. Clin Toxicol (Phila) 2009; 47:
525–35.
36 Twagirumukiza M, Cosijns A, Pringels E, Remon JP, Vervaet
C, Van Bortel L. Influence of tropical climate conditions on
the quality of antihypertensive drugs from Rwandan
pharmacies. Am J Trop Med Hyg 2009; 81: 776–81.
49 Taylor PW, Arnet I, Fischer A, Simpson IN. Pharmaceutical
quality of nine generic orlistat products compared with
Xenical®. Obes Facts 2010; 3: 231–7.
37 Angeli DG, Trezza C. Quality and stability of ramipril
generics/copies versus reference ramipril (Tritace): a
3-month stability comparative study. Clin Drug Investig
2009; 29: 667–76.
50 Lehr HA, Brunner J, Rangoonwala R, Kirkpatrick CJ.
Particulate matter contamination of intravenous antibiotics
aggravates loss of functional capillary density in
postischemic striated muscle. Am J Respir Crit Care Med
2002; 165: 514–20.
38 Vial J, Cohen M, Sassiat P, Thiébaut D. Pharmaceutical
quality of docetaxel generics versus originator drug
product: a comparative analysis. Curr Med Res Opin 2008;
24: 2019–33.
39 Taylor PW, Keenan MH. Pharmaceutical quality of generic
isotretinoin products, compared with Roaccutane. Curr
Med Res Opin 2006; 22: 603–15.
40 Health Canada. A small number of bottles of the antibiotic
Rofact® (rifampin) may contain a different drug. 2009.
Available at http://www.healthycanadians.gc.ca/recallalert-rappel-avis/hc-sc/2009/13384a-eng.php (last accessed
18 December 2013).
41 Health Canada. Immediate recall by Mylan
Pharmaceuticals: potential serious risk due to mislabelling
of products. 2011. Available at http://www
.healthycanadians.gc.ca/recall-alert-rappel-avis/hc-sc/
2011/13584a-eng.php (last accessed 18 December 2013).
42 Food and Drug Administration. Greenstone announces
voluntary nationwide recall of citalopram and finasteride
due to possible mislabelling. 2011. Available at
http://www.fda.gov/Safety/Recalls/ucm248552.htm (last
accessed 18 December 2013).
43 L’Agence nationale de sécurité du médicament et des
produits de santé. Furosémide Teva 40 mg, comprimé
sécable. 2013. Available at http://ansm.sante.fr/Dossiers
-thematiques/Furosemide/Furosemide-Teva-40-mg
-comprime/%28offset%29/0 (last accessed 18 December
2013).
51 Blossom DB, Kallen AJ, Patel PR, Elward A, Robinson L, Gao
G, Langer R, Perkins KM, Jaeger JL, Kurkjian KM, Jones M,
Schillie SF, Shehab N, Ketterer D, Venkataraman G,
Kishimoto TK, Shriver Z, McMahon AW, Austen KF,
Kozlowski S, Srinivasan A, Turabelidze G, Gould CV,
Arduino MJ, Sasisekharan R. Outbreak of adverse reactions
associated with contaminated heparin. N Engl J Med 2008;
359: 2674–84. Erratum in: N Engl J Med 2010; 362: 1056.
52 Blossom D, Noble-Wang J, Su J, Pur S, Chemaly R, Shams A,
Jensen B, Pascoe N, Gullion J, Casey E, Hayden M, Arduino
M, Budnitz DS, Raad I, Trenholme G, Srinivasan A.
Multistate outbreak of Serratia marcescens bloodstream
infections caused by contamination of prefilled heparin
and isotonic sodium chloride solution syringes. Arch Intern
Med 2009; 169: 1705–11.
53 Kutty PK, Forster TS, Wood-Koob C, Thayer N, Nelson RB,
Berke SJ, Pontacolone L, Beardsley TL, Edelhauser HF,
Arduino MJ, Mamalis N, Srinivasan A. Multistate outbreak
of toxic anterior segment syndrome, 2005. J Cataract
Refract Surg 2008; 34: 585–90.
54 Pozniak A, Müller L, Salgo M, Jones JK, Larson P, Tweats D.
Elevated ethyl methanesulfonate (EMS) in nelfinavir
mesylate (Viracept, Roche): overview. AIDS Res Ther 2009;
6: 18.
55 Attorrese G, Massi-Benedetti M. Quality and behavior of
glimepiride generics versus amaryl under stressed
conditions. Diabetes Technol Ther 2007; 9: 287–96. Erratum
in: Diabetes Technol Ther 2007; 9: 482.
Br J Clin Pharmacol
/ 78:2
/
237
A. Johnston & D. W. Holt
56 Gomez Y, Adams E, Hoogmartens J. Analysis of purity in 19
drug product tablets containing clopidogrel: 18 copies
versus the original brand. J Pharm Biomed Anal 2004; 34:
341–8.
57 Attaran A, Barry D, Basheer S, Bate R, Benton D, Chauvin J,
Garrett L, Kickbusch I, Kohler JC, Midha K, Newton PN,
Nishtar S, Orhii P, McKee M. How to achieve international
action on falsified and substandard medicines. BMJ 2012;
345: e7381.
58 Choudary A. Police body recommends action against 17
people. Dawn (Pakistan). 2012. Available at http://www
.dawn.com/news/695377/police-body-recommends-action
-against-17-people (last accessed 18 December 2013).
59 World Health Organization. Alert No. 125. Contaminated
Isotab® (isosorbide mononitrate) incident in Lahore
Pakistan. 2012. Available at http://www.who.int/
medicines/publications/drugalerts/DrugSafetyAlert125.pdf
(last accessed 18 December 2013).
69 Mastoraki E, Michalopoulos A, Kriaras I, Mouchtouri E,
Falagas ME, Karatza D, Geroulanos S. Incidence of
postoperative infections in patients undergoing coronary
artery bypass grafting surgery receiving antimicrobial
prophylaxis with original and generic cefuroxime. J Infect
2008; 56: 35–9.
70 Goubran HA. Failure of a non-authorized copy product to
maintain response achieved with imatinib in a patient with
chronic phase chronic myeloid leukemia: a case report.
J Med Case Rep 2009; 3: 7112.
71 Tayrouz Y, Ding R, Burhenne J, Riedel KD, Weiss J,
Hoppe-Tichy T, Haefeli WE, Mikus G. Pharmacokinetic and
pharmaceutic interaction between digoxin and Cremophor
RH40. Clin Pharmacol Ther 2003; 73: 397–405.
72 Wandel C, Kim RB, Stein CM. ‘Inactive’ excipients such as
Cremophor can affect in vivo drug disposition. Clin
Pharmacol Ther 2003; 73: 394–96.
60 Wax PM. Elixirs, diluents, and the passage of the 1938
Federal Food, Drug and Cosmetic Act. Ann Intern Med
1995; 122: 456–61.
73 Rimoy GH, Moshi MJ, Massele AY. Comparative
bioavailability of oral sugar-coated and plain formulation
of chloroquine phosphate marketed in Tanzania. Trop Doct
2002; 32: 15–7.
61 Food and Drug Administration. FDA statement on the
Ranbaxy atorvastatin recall. November 30, 2012. Available
at http://www.fda.gov/Drugs/DrugSafety/ucm329951.htm
(last accessed 18 December 2013).
74 Risha PG, Shewiyo D, Msami A, Masuki G, Vergote G,
Vervaet C, Remon JP. In vitro evaluation of the quality of
essential drugs on the Tanzanian market. Trop Med Int
Health 2002; 7: 701–7.
62 James G. Perrier recalls its water in U.S. after benzene is
found in bottles. The New York Times 1990. Available at
http://www.nytimes.com/1990/02/10/us/perrier-recalls
-its-water-in-us-after-benzene-is-found-in-bottles.html (last
accessed 18 December 2013).
75 Vijaykadga S, Cholpol S, Sitthimongkol S, Pawaphutanan A,
Pinyoratanachot A, Rojanawatsirivet C, Kovithvattanapong
R, Thimasarn K. Strengthening of national capacity in
implementation of antimalarial drug quality assurance in
Thailand. Southeast Asian J Trop Med Public Health 2006;
37 (Suppl. 3): 5–10.
63 Gardner LK, Lawrence GD. Benzene production from
decarboxylation of benzoic acid in the presence of ascorbic
acid and a transition-metal catalyst. J Agric Food Chem
1993; 41: 693–5.
76 Gaudiano MC, Di Maggio A, Cocchieri E, Antoniella E,
Bertocchi P, Alimonti S, Valvo L. Medicines informal market
in Congo, Burundi and Angola: counterfeit and
sub-standard antimalarials. Malar J 2007; 6: 22.
64 Food and Drug Administration. Questions and answers on
the occurrence of benzene in soft drinks and other
beverages. 2009. Available at http://www.fda.gov/Food/
FoodborneIllnessContaminants/ChemicalContaminants/
ucm055131.htm# (last accessed 18 December 2013).
77 Kaur H, Goodman C, Thompson E, Thompson KA, Masanja
I, Kachur SP, Abdulla S. A nationwide survey of the quality
of antimalarials in retail outlets in Tanzania. PLoS ONE
2008; 3: e3403.
65 Deccan Herald. Govt declares breast cancer drug Albupax
sub-standard. 2009. Available at http://www
.deccanherald.com/content/66392/govt-declares-breast
-cancer-drug.html (last accessed 18 December 2013).
78 Hosseinipour MC, Corbett AH, Kanyama C, Mshali I, Phakati
S, Rezk NL, van der Horst C, Kashuba AD. Pharmacokinetic
comparison of generic and trade formulations of
lamivudine, stavudine and nevirapine in HIV-infected
Malawian adults. AIDS 2007; 21: 59–64.
66 Kavilanz P. Tylenol recall: FDA slams company. CNN Money
2010. Available at http://money.cnn.com/2010/01/15/
news/companies/over_the_counter_medicine_recall/ (last
accessed 18 December 2013).
67 Zoler ML. Methyl chloride contamination found in generic
clopidogrel. Internal Medicine News 6 December 2010.
Available at http://www.internalmedicinenews.com/
single-view/methyl-chloride-contamination-found-in
-generic-clopidogrel/970bc96188.html (last accessed 18
December 2013).
68 Ernest S. Tanzania: TFDA stops sale of nine types of drugs,
2011. Available at: http://allafrica.com/stories/
201101120547.html (last accessed 3 March 2011).
238
/ 78:2
/
Br J Clin Pharmacol
79 van Crevel R, Nelwan RH, Borst F, Sahiratmadja E, Cox J,
van der Meij W, de Graaff M, Alisjahbana B, de Lange WC,
Burger D. Bioavailability of rifampicin in Indonesian
subjects: a comparison of different local drug
manufacturers. Int J Tuberc Lung Dis 2004; 8: 500–3.
80 Pillai G, Fourie PB, Padayatchi N, Onyebujoh PC, McIlleron
H, Smith PJ, Gabriels G. Recent bioequivalence studies on
fixed-dose combination anti-tuberculosis drug
formulations available on the global market. Int J Tuberc
Lung Dis 1999; 3 (11 Suppl. 3): S309–16.
81 Singh S, Mohan B. A pilot stability study on four-drug
fixed-dose combination anti-tuberculosis products. Int J
Tuberc Lung Dis 2003; 7: 298–303.
Substandard drugs
82 Iqbal M, Hakim ST, Hussain A. Iqbal SP, Abbasi K. Ofloxacin:
laboratory evaluation of the antibacterial effectiveness of
34 brands representing 31 manufacturers available in
Pakistan. Pak J Med Sci 2004; 20: 349–56.
83 Esquivel A, González-Ramírez R, Alberú J, Gracida C,
Medeiros M, Castañeda-Hernández G. Comparison of
dissolution properties of 2 enteric-coated formulations
containing mycophenolate sodium: Myfortic vs Femulan.
Transplant Proc 2010; 42: 353–6.
84 Veronin MA, Nguyen NT. Comparison of simvastatin tablets
from the US and international markets obtained via the
Internet. Ann Pharmacother 2008; 42: 613–20.
85 Abdullah MD, Bepary S, Rouf AS. Report: in vitro
dissolution studies of different brands of sustained release
diclofenac sodium matrix tablet available in Bangladesh.
Pak J Pharm Sci 2008; 21: 70–7.
86 Mammo ZN, Flanagan JG, James DF, Trope GE. Generic
versus brand-name North American topical glaucoma
drops. Can J Ophthalmol 2012; 47: 55–61.
87 Petersen H, Bizec J-C, Schuetz H, Delporte M-L.
Pharmacokinetic and technical comparison of Sandostatin®
LAR® and other formulations of long-acting octreotide.
BMC Res Notes 2011; 4: 344.
88 Jansen FH. The pharmaceutical death-ride of
dihydroartemisinin. Malar J 2010; 9: 212.
89 World Health Organization. WHO technical report series
No. 953. WHO expert committee on specifications for
pharmaceutical preparations. Forty-third report. 2009.
90 Nogueira FH, Moreira-Campos LM, Santos RL, Pianetti GA.
Quality of essential drugs in tropical countries: evaluation
of antimalarial drugs in the Brazilian health system. Rev
Soc Bras Med Trop 2011; 44: 582–6.
91 Zore M, Harris A, Tobe LA, Siesky B, Januleviciene I, Behzadi
J, Amireskandari A, Egan P, Garff K, Wirostko B. Generic
medications in ophthalmology. Br J Ophthalmol 2013; 97:
253–7.
92 European Medicines Agency. Biosimilar medicines. 2012.
Available at http://www.ema.europa.eu/ema/index
.jsp?curl=pages/special_topics/document_listing/
document_listing_000318.jsp&mid=WC0b01ac0580281bf0
(last accessed 18 December 2013).
93 Food and Drug Administration. FDA issues draft guidance
on biosimilar product development. February 9, 2012.
Available at http://www.fda.gov/NewsEvents/
Newsroom/PressAnnouncements/ucm291232.htm (last
accessed 18 December 2013).
94 European Medicines Agency. Biosimilars. 2013. Available at
http://www.ema.europa.eu/ema/index.jsp?curl=pages/
regulation/general/general_content_000408.jsp&mid
=WC0b01ac058002958c#Overarchingbiosimilarguidelines
(last accessed 18 December 2013).
95 Newton PN, McGready R, Fernandez F, Green MD, Sunjio
M, Bruneton C, Phanouvong S, Millet P, Whitty CJ, Talisuna
AO, Proux S, Christophel EM, Malenga G, Singhasivanon P,
Bojang K, Kaur H, Palmer K, Day NP, Greenwood BM,
Nosten F, White NJ. Manslaughter by fake artesunate in
Asia – will Africa be next? PLoS Med 2006; 3: e197.
96 Keoluangkhot V, Green MD, Nyadong L, Fernández FM,
Mayxay M, Newton PN. Impaired clinical response in a
patient with uncomplicated falciparum malaria who
received poor-quality and underdosed intramuscular
artemether. Am J Trop Med Hyg 2008; 78: 552–5.
97 Leslie T, Kaur H, Mohammed N, Kolaczinski K, Ord RL,
Rowland M. Epidemic of Plasmodium falciparum malaria
involving substandard antimalarial drugs, Pakistan, 2003.
Emerg Infect Dis 2009; 15: 1753–9.
98 Food and Drug Administration. Information on adverse
event reports and heparin, 2009. Available at
http://www.fda.gov/Drugs/DrugSafety/
PostmarketDrugSafetyInformationforPatientsandProviders/
UCM112669 (last accessed 18 December 2013).
99 Holm A, Hernandez M. A post-hoc analysis of the safety
and efficacy of tacrolimus (Prograf) versus a ‘generic
formulation’ of tacrolimus (Tenacrine) as primary
immunosuppressive therapy in LRD and CAD, adults and
pediatric renal transplant recipients. Abstract presented at
XXII International Congress of the Transplantation Society,
Sydney, Australia, 10–14 August 2008.
100 Mattar M. Failure of copy Imatib (CIPLA, India) to maintain
hematologic and cytogenetic responses in chronic myeloid
leukemia in chronic phase. Int J Hematol 2010; 91: 104–6.
101 Asfour IA, Elshazly SA. Changing therapy from Glivec® to a
‘copy’ imatinib results in a worsening of chronic myeloid
leukemia disease status: two case reports. Cases J 2009; 2:
9342.
102 Chouffai Z. Hematologic relapse after 2 years on a
non-authorized copy version of imatinib in a patient with
chronic myeloid leukemia in chronic phase: a case report.
Case Rep Oncol 2010; 3: 272–6.
103 Centers for Disease Control and Prevention. Multistate
fungal meningitis outbreak – current case count. 19
November 2012. Available at http://www.cdc.gov/hai/
outbreaks/meningitis-map.html (last accessed 19
November 2012).
104 Kainer MA, Reagan DR, Nguyen DB, Wiese AD, Wise ME,
Ward J, Park BJ, Kanago ML, Baumblatt J, Schaefer MK,
Berger BE, Marder EP, Min JY, Dunn JR, Smith RM,
Dreyzehner J, Jones TF, Tennessee Fungal Meningitis
Investigation Team. Fungal infections associated with
contaminated methylprednisolone in Tennessee. N Engl J
Med 2012; 367: 2194–203.
105 Food and Drug Administration. FDA Public Health Alert:
change in heparin USP monograph. 2010. Available
at http://www.fda.gov/Drugs/DrugSafety/
PostmarketDrugSafetyInformationforPatientsandProviders/
ucm184502.htm (last accessed 18 December 2013).
106 White NJ, Pongtavornpinyo W, Maude RJ, Saralamba S,
Aguas R, Stepniewska K, Lee SJ, Dondorp AM, White LJ,
Day NP. Hyperparasitaemia and low dosing are an
important source of anti-malarial drug resistance. Malar J
2009; 8: 253.
Br J Clin Pharmacol
/ 78:2
/
239
A. Johnston & D. W. Holt
107 Newton PN, Green MD, Mildenhall DC, Plançon A, Nettey H,
Nyadong L, Hostetler DM, Swamidoss I, Harris GA, Powell K,
Timmermans AE, Amin AA, Opuni SK, Barbereau S, Faurant
C, Soong RC, Faure K, Thevanayagam J, Fernandes P, Kaur
H, Angus B, Stepniewska K, Guerin PJ, Fernández FM. Poor
quality vital anti-malarials in Africa – an urgent neglected
public health priority. Malar J 2011; 10: 352.
108 Sengaloundeth S, Green MD, Fernandez FM, Manolin O,
Phommavong K, Insixiengmay V, Hampton CY, Nyadong L,
Mildenhall DC, Hostetler D, Khounsaknalath L, Vongsack L,
Phompida S, Vanisaveth V, Syhakhang L, Newton PN. A
stratified random survey of the proportion of poor quality
oral artesunate sold at medicine outlets in the Lao PDR –
implications for therapeutic failure and drug resistance.
Malar J 2009; 8: 172.
109 Noedl H, Se Y, Schaecher K, Smith BL, Socheat D, Fukuda
MM, Artemisinin Resistance in Cambodia 1 (ARC1) Study
Consortium. Evidence of artemisinin-resistant malaria in
western Cambodia. N Engl J Med 2008; 359: 2619–20.
110 Phyo AP, Nkhoma S, Stepniewska K, Ashley EA, Nair S,
McGready R, ler Moo C, Al-Saai S, Dondorp AM, Lwin KM,
Singhasivanon P, Day NP, White NJ, Anderson TJ, Nosten F.
Emergence of artemisinin-resistant malaria on the western
border of Thailand: a longitudinal study. Lancet 2012; 379:
1960–6.
111 Tadeg H, Berhane Y. Substandard and counterfeit
antimicrobials: recent trends and implications to key public
health interventions in developing countries. East Afr J
Public Health 2012; 9: 85–9.
112 Caminero JA. Multidrug-resistant tuberculosis:
epidemiology, risk factors and case finding. Int J Tuberc
Lung Dis 2010; 14: 382–90.
113 Issack MI. Substandard drugs. Lancet 2001; 358: 1463.
114 Geerts S, Gryseels B. Anthelmintic resistance in human
helminths: a review. Trop Med Int Health 2001; 6: 915–21.
115 Wertheimer AI, Norris J. Safeguarding against
substandard/counterfeit drugs: mitigating a
macroeconomic pandemic. Res Social Adm Pharm 2009; 5:
4–16.
116 Centers for Disease Control and Prevention. Antibiotic
resistance threats in the United States, 2013. 2013.
Available at http://www.cdc.gov/drugresistance/threat
-report-2013/pdf/ar-threats-2013-508.pdf#page=5 (last
accessed 18 December 2013).
117 World Health Organization. Antimicrobial resistance. Fact
sheet no. 194. 2013. Available at http://www.who.int/
mediacentre/factsheets/fs194/en/# (last accessed 18
December 2013).
118 WorldWide Antimalarial Resistance Network. Drug quality
and the fight against malaria. 2010. Available at
http://www.wwarn.org/sites/default/files/
WWARNAntimalarialQualityBrochure.pdf (last accessed 18
December 2013).
119 Kelesidis T, Kelesidis I, Rafailidis PI, Falagas ME. Counterfeit
or substandard antimicrobial drugs: a review of the
scientific evidence. J Antimicrob Chemother 2007; 60:
214–36.
240
/ 78:2
/
Br J Clin Pharmacol
120 Kanavos P, Das P, Durairaj V, Laing R, Abegunde DO.
Options for financing and optimizing medicines in
resource-poor countries. World Health Report Background
Paper 34, 2010. Available at http://www.who.int/
healthsystems/topics/financing/healthreport/
34Medicinesrev.pdf (last accessed 18 December 2013).
121 Amin AA, Kokwaro GO. Antimalarial drug quality in Africa.
J Clin Pharm Ther 2007; 32: 429–40.
122 Maponga C, Ondari C. The quality of antimalarials. A
study in selected African countries, 2003. Available at
http://whqlibdoc.who.int/hq/2003/WHO_EDM_PAR
_2003.4.pdf (last accessed 18 December 2013).
123 IMPACT. Counterfeit medicines: an update on estimates.
2006. Available at http://www.who.int/medicines/services/
counterfeit/impact/TheNewEstimatesCounterfeit.pdf (last
accessed 18 December 2013).
124 Oxfam. Eye on the ball. Medicine regulation – not IP
enforcement – can best deliver quality medicines, 2011.
Available at http://www.oxfam.org/sites/www
.oxfam.org/files/eye-on-the-ball-medicine-regulation
-020211-en.pdf (last accessed 18 December 2013).
125 Cockburn R, Newton PN, Agyarko EK, Akunyili D, White NJ.
The global threat of counterfeit drugs: why industry and
governments must communicate the dangers. PLoS Med
2005; 2: e100. Erratum in: PLoS Med 2007; 4: e289.
126 Newton PN, White NJ, Rozendaal JA, Green MD. Murder by
fake drugs. BMJ 2002; 324: 800–1.
127 World Health Organization. Substandard and counterfeit
medicines. Fact sheet No. 275, 2003. Available at
http://www.who.int/mediacentre/factsheets/2003/fs275/
en/print.html (last accessed 18 December 2013).
128 Bialik C. Counterfeit drug count is tough to swallow. The
Wall Street Journal, 11 September 2010.
129 Medicines and Healthcare Products Regulatory Agency.
Drug alerts. Available at http://www.mhra.gov.uk/
Safetyinformation/Safetywarningsalertsandrecalls/
DrugAlerts/index.htm (last accessed 22 November 2012).
130 Bate R, Hess K. Assessing website pharmacy drug quality:
safer than you think? PLoS ONE 2010; 5: e12199.
131 Jackson G, Arver S, Banks I, Stecher VJ. Counterfeit
phosphodiesterase type 5 inhibitors pose significant safety
risks. Int J Clin Pract 2010; 64: 497–504.
132 Kenyon SL, Button J, Perella P, McKeown DA, Holt DW. An
herbal remedy for impotence: more than was bargained
for. J Clin Pharmacol 2006; 46: 1379–81.
133 Veronin M. Packaging and labeling of pharmaceutical
products obtained from the internet. J Med Internet Res
2011; 13: e22.
134 European Parliament. Directive 2011/62/eu of the
European Parliament and of the Council of 8 June 2011.
2011. Available at http://ec.europa.eu/health/files/
eudralex/vol-1/dir_2011_62/dir_2011_62_en.pdf (last
accessed 18 December 2013).
135 Davison M. Pharmaceutical Anti-Counterfeiting: Combating
the Real Danger from Fake Drugs. Hoboken, New Jersey:
John Wiley & Sons, Inc, 2011.
Substandard drugs
136 Riviere JE, Buckley GJ, eds. Ensuring Safe Foods and
Medical Products through Stronger Regulatory Systems
Abroad. Washington, DC: The National Academies Press,
2012.
149 World Health Organization. Access to HIV/AIDS drugs and
diagnostics of acceptable quality, 63rd Edition, 2008.
Available at http://apps.who.int/prequal/lists/hiv
_suppliers.pdf (last accessed 18 December 2013).
137 Gostin LO, Buckley GJ, eds. Countering the Problem of
Falsified and Substandard Drugs. Washington, DC: The
National Academies Press, 2013.
150 World Health Organization. Multisource (generic)
pharmaceutical products: guidelines on registration
requirements to establish interchangeability, 2006.
Available at http://apps.who.int/prequal/info_general/
documents/TRS937/WHO_TRS_937_eng.pdf#page=359
(last accessed 18 December 2013).
138 United Nations Office on Drugs and Crime. World Drug
Report 2013, 2013. Available at http://www.unodc.org/
unodc/secured/wdr/wdr2013/World_Drug_Report_2013
.pdf (last accessed 18 December 2013).
139 United States Pharmacopeia and United States Agency for
International Development. Survey of the quality of
selected antimalarial medicines circulating in Madagascar,
Senegal, and Uganda, November 2009. Available at
http://apps.who.int/medicinedocs/documents/s17069e/
s17069e.pdf (last accessed 18 December 2013).
140 World Health Organization. Survey of the quality of
selected antimalarial medicines circulating in six
countries of sub-Saharan Africa. 2011. Available at
http://www.who.int/medicines/publications/
WHO_QAMSA_report.pdf (last accessed 18 December
2013).
141 Saez C. WHO members show dismay at delay on
counterfeit medicines group. Intellectual Property Watch.
2011. Available at http://www.ip-watch.org/2011/01/19/
who-members-show-dismay-at-delay-on-counterfeit
-medicines-group/ (last accessed 18 December 2013).
142 World Health Organization. 65th World Health Assembly
closes with new global health measures. Published 26 May
2012. 2012. Available at http://www.who.int/mediacentre/
news/releases/2012/wha65_closes_20120526/en/index
.html (last accessed 18 December 2013).
143 United States Pharmacopeial Convention. Medicines
quality database (MQDB). Available at http://www
.usp.org/around-world/pqm-uspusaid/medicines-quality
-database-mqdb (last accessed 18 December 2013).
144 Tabernero P, Newton PN. The WWARN antimalarial quality
surveyor. Pathog Glob Health 2012; 106: 77–8.
145 WorldWide Antimalarial Resistance Network. Antimalarial
quality literature surveyor. 2013. Available at
http://www.wwarn.org/aqsurveyor/ (last accessed 18
December 2013).
146 World Health Organization. General information on
counterfeit medicines, 2011. Available at http://www.who
.int/medicines/services/counterfeit/overview/en/
index2.html# (last accessed 18 December 2013).
147 Khan AA. Pakistani drug regulator ‘destined to be a failure’.
Published 4 April 2012. Nature News 2012. Available at
http://www.nature.com/news/pakistani-drug-regulator
-destined-to-be-a-failure-1.10380 (last accessed 18
December 2013).
148 World Health Organization. Prequalification programme.
A United Nations Programme managed by WHO, 2010.
Available at http://apps.who.int/prequal/ (last accessed 18
December 2013).
151 World Health Organization. Good governance for
medicines. Curbing corruption in medicines regulation
and supply, 2010. Available at http://www.who.int/
medicines/areas/policy/goodgovernance/
ENCurbingCorruption4May2010.pdf (last accessed 18
December 2013).
152 World Health Organization. Measuring transparency in the
public pharmaceutical sector. Assessment instrument,
2009. Available at http://www.who.int/medicines/
areas/policy/goodgovernance/AssessmentInstrument
MeastranspENG.PDF (last accessed 18 December 2013).
153 World Health Organization. Measuring transparency to
improve good governance in the public pharmaceutical
sector. A comparative analysis of five country assessment
studies, 2009. Available at http://www.who.int/medicines/
areas/policy/goodgovernance/5countryAssessment.pdf
(last accessed 18 December 2013).
154 World Health Organization. Good governance for
medicines. Progress report 2010, 2010. Available at
http://www.who.int/medicines/areas/policy/
goodgovernance/GGM2010ProgressReport.pdf (last
accessed 18 December 2013).
155 Francis PA. Editorial: overseas inspections. 2011. Available
at http://www.pharmabiz.com/ArticleDetails.aspx?aid
=61038&sid=3 (last accessed 18 December 2013).
156 World Health Organization. WHO initiates pilot
prequalification of active pharmaceutical ingredients, 2010.
Available at http://apps.who.int/prequal/info_applicants/
API/API_communique.pdf (last accessed 18 December
2013).
157 Akunyili D. The fight against counterfeit drugs in Nigeria.
In: Global Corruption Report 2006, eds Kotalik J, Rodriguez
D. London, UK: Pluto Press, 2006; 96–100.
158 Chinwendu O. The fight against fake drugs by NAFDAC in
Nigeria, 2008. Available at http://apps.who.int/
medicinedocs/documents/s18405en/s18405en.pdf (last
accessed 18 December 2013).
159 Raufu A. Nigeria leads fight against ‘killer’ counterfeit
drugs. Bull World Health Organ 2006; 84: 690.
160 Garuba HA, Kohler JC, Huisman AM. Transparency in
Nigeria’s public pharmaceutical sector: perceptions from
policy makers. Global Health 2009; 5: 14.
161 Taylor N. NAFDAC launches mobile anti-counterfeiting
service. February 2010. Available at http://www
.in-pharmatechnologist.com/Drug-Delivery/NAFDAC
-launches-mobile-anti-counterfeiting-service (last accessed
18 December 2013).
Br J Clin Pharmacol
/ 78:2
/
241
A. Johnston & D. W. Holt
162 Byrne C. Sproxil lands $1.8 million to fight fake drugs.
March 2011. Available at http://venturebeat.com/2011/03/
22/sproxil-lands-1-8-million-to-fight-fake-drugs/ (last
accessed 18 December 2013).
163 Sproxil. Drug anti-counterfeiting technology reaches 1
million users. January 25 2012. Available at http://sproxil
.com/download/for-the-press/2012/PressRelease
-SproxilReaches1Mil2012-01-25.pdf (last accessed 18
December 2013).
164 Government of India Directorate General of Foreign Trade.
Public notice No. 21 (RE-2010)/2009-14. Procedure relating
to tracing and tracking of export consignment of
pharmaceuticals and drugs. January 2011. Available at
http://dgft.gov.in/exim/2000/pn/pn10/
pn2110.htm (last accessed 18 December 2013).
165 Government of India Directorate General of Foreign Trade.
Public Notice No. 10 (RE-2012)/2009-2014. Deferment in
the date of effect for implementation of bar-coding on
Primary and Secondary level packaging on export
consignment of pharmaceuticals and drugs for tracing
and tracking purpose. July 2012. Available at
http://164.100.9.245/Exim/2000/PN/PN12/pn1012.htm (last
accessed 18 December 2013).
166 European Federation of Pharmaceutical Industries and
Associations. ESM. European Stakeholder Model. Ensuring
patients have access to safe medicines. 2012. Available at
http://www.efpia.eu/uploads/Modules/Documents/
european_stakeholder_model_esm_publication_june
_2012-20120622-007-en-v1.pdf (last accessed 18
December 2013).
167 European Directorate for the Quality of Medicines and
Healthcare. The EDQM anti-counterfeiting strategy. 2013.
Available at http://www.edqm.eu/en/eTACT-1466.html (last
accessed 18 December 2013).
168 European Commission. Guidelines of 7 March 2013 on
good distribution practice of medicinal products for
human use (2013/C 68/01). 2013. Available at http://eur-lex
.europa.eu/LexUriServ/LexUriServ.do?uri=OJ:C:2013:068
:0001:0014:EN:PDF (last accessed 18 December 2013).
169 European Commission. Guidelines on the principles of
good distribution practices for active substances for
medicinal products for human use. 2013. Available at
http://ec.europa.eu/health/files/gmp/2013-02_gdp_for
_api_cons.pdf (last accessed 18 December 2013).
170 European Compliance Academy. Regulatory monitoring of
API manufacturers - FDA, TGA, EMA and EDQM express
common objectives for inspections collaboration. March
2012. Available at http://www.gmp-compliance.org/
eca_news_3030_7186,7225,7218.html (last accessed 18
December 2013).
171 United States Government Accountability Office. Report to
congressional Committees. High-risk series: an update.
February 2013. Available at http://www.gao.gov/assets/
660/652133.pdf (last accessed 18 December 2013).
172 Food and Drug Administration. Global engagement. April
2012. 2012 Available at http://www.fda.gov/downloads/
AboutFDA/ReportsManualsForms/Reports/UCM298578.pdf
(last accessed 18 December 2013).
242
/ 78:2
/
Br J Clin Pharmacol
173 Food and Drug Administration. Another counterfeit cancer
medicine found in U.S. – illegal practice puts patients at
risk. April 3, 2012. 2012. Available at http://www.fda.gov/
Drugs/DrugSafety/DrugIntegrityandSupplyChainSecurity/
ucm298047.htm (last accessed 18 December 2013).
174 The Daily Star. Stop sale, production of 8 drugs, 28 January
2011. Available at http://www.thedailystar.net/newDesign/
news-details.php?nid=171868 (last accessed 18 December
2013).
175 Chaccour CJ, Kaur H, Mabey D, Del Pozo JL. Travel and fake
artesunate: a risky business. Lancet 2012; 380: 1120.
176 United States Pharmacopeia. Medicines quality in five
sub-Saharan African countries to benefit from USP pilot
program, 2011. Available at http://us.vocuspr.com/
ViewAttachment.aspx?EID=bK0ke822q8svy24Psob15tx9
Qclx9ZG5aZ6ZqbADRA4%3D (last accessed 18 December
2013).
177 Kenyon TA, Kenyon AS, Sibiya T. Drug quality screening in
developing countries: establishment of an appropriate
laboratory in Swaziland. Bull World Health Organ 1994; 72:
615–20.
178 Lon CT, Tsuyuoka R, Phanouvong S, Nivanna N, Socheat D,
Sokhan C, Blum N, Christophel EM, Smine A. Counterfeit
and substandard antimalarial drugs in Cambodia. Trans R
Soc Trop Med Hyg 2006; 100: 1019–24.
179 Green MD, Mount DL, Wirtz RA. Authentication of
artemether, artesunate and dihydroartemisinin antimalarial
tablets using a simple colorimetric method. Trop Med Int
Health 2001; 6: 980–2.
180 Hu CQ, Zou WB, Hu WS, Ma XK, Yang MZ, Zhou SL, Sheng
JF, Li Y, Cheng SH, Xue J. Establishment of a fast chemical
identification system for screening of counterfeit drugs of
macrolide antibiotics. J Pharm Biomed Anal 2006; 40:
68–74.
181 Global Pharma Health Fund. The GPHF-Minilab® –
Protection against counterfeit medicines, 2011. Available at
http://www.gphf.org/web/en/minilab/index.htm (last
accessed 18 December 2013).
182 Risha PG, Msuya Z, Clark M, Johnson K,
Ndomondo-Sigonda M, Layloff T. The use of Minilabs to
improve the testing capacity of regulatory authorities in
resource limited settings: Tanzanian experience. Health
Policy 2008; 87: 217–22.
183 Dowell FE, Maghirang EB, Fernandez FM, Newton PN,
Green MD. Detecting counterfeit antimalarial tablets by
near-infrared spectroscopy. J Pharm Biomed Anal 2008; 48:
1011–4.
184 Ricci C, Nyadong L, Yang F, Fernandez FM, Brown CD,
Newton PN, Kazarian SG. Assessment of hand-held Raman
instrumentation for in situ screening for potentially
counterfeit artesunate antimalarial tablets by FT-Raman
spectroscopy and direct ionization mass spectrometry.
Anal Chim Acta 2008; 623: 178–86.
185 Fernandez FM, Hostetler D, Powell K, Kaur H, Green MD,
Mildenhall DC, Newton PN. Poor quality drugs: grand
Substandard drugs
challenges in high throughput detection, countrywide
sampling, and forensics in developing countries. Analyst
2011; 136: 3073–82.
186 Hajjou M, Qin Y, Bradby S, Bempong D, Lukulay P.
Assessment of the performance of a handheld Raman
device for potential use as a screening tool in evaluating
medicines quality. J Pharm Biomed Anal 2013; 74: 47–55.
187 Bate R, Mooney L, Harris J, Mitra B. A safe medicines chest
for the world. Preventing substandard products from
tainting India’s pharmaceuticals, 2010. Available at http://
www.aei.org/files/2010/05/18/SafeMedicinesChest2010.pdf
(last accessed 18 December 2013).
188 Transparency International. Corruption in the procurement
of medicines and equipment, 2011. Available at
http://archive.transparency.org/global_priorities/other
_thematic_issues/health/service_delivery/procurement
(last accessed 18 December 2013).
189 Vian T. Review of corruption in the health sector: theory,
methods and interventions. Health Policy Plan 2008; 23:
83–94
190 Food and Drug Administration. FDA takes new regulatory
action against Ranbaxy’s Paonta Sahib plant in India. 2009.
Available at http://www.fda.gov/NewsEvents/Newsroom/
PressAnnouncements/2009/ucm149532.htm (last accessed
18 December 2013).
191 Homedes N, Ugalde A. Multisource drug policies in Latin
America: survey of 10 countries. Bull World Health Organ
2005; 83: 64–70.
192 Kaplan WA, Laing R. Paying for pharmaceutical registration
in developing countries. Health Policy Plan 2003; 18:
237–48.
Br J Clin Pharmacol
View publication stats
/ 78:2
/
243