You are on page 1of 13

Chapter 6

Drug Regulation: History, Present and


Future1
Lembit Rägo, Budiono Santoso
I. History of medicines regulation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 65
II. Why regulating drugs? . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 66
III. What is medicines regulation? . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 67
IV. Drug registration . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 68
V. Role of WHO in drug regulation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 74
VI. Future of medicines regulation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 75
Bibliography . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 76

I. HISTORY OF MEDICINES REGULATION was issued in 1581. The standards for the manufac-
ture of Mithridatum were established in England in
Medicines are perhaps as old as mankind and the The London Pharmacopoeia only in 1618.
concepts how their quality has to be ensured has The modern medicines regulation started only af-
evolved gradually over the time. For example, Mith- ter breakthrough progress in the 19th century life sci-
ridates VI (120 BC), King of Pontus, concocted a ences, especially in chemistry, physiology and phar-
compound preparation called “Mithridatium” which macology, which laid a solid foundation for the mod-
included 41 individual components and was held as a ern drug research and development and started to
panacea for almost all diseases until as late as 1780s. flourish after the second World War.
It took until 1540 when in England the manufac- Unfortunate events have catalysed the develop-
ture of Mithridatium and other medicines was sub- ment of medicines regulation more than the evolu-
jected to supervision under the Apothecaries Wares, tion of a knowledge base. In 1937 over 100 people
Drugs and Stuffs Act. The Act was one of the earli- in the United States died of diethylene glycol poi-
soning following the use of a sulfanilamide elixir,
est British statutes on the control of medicines and
which used the chemical as a solvent without any
it established the appointment of four inspectors of
safety testing. This facilitated introduction of The
“Apothecary Wares, Drugs and Stuffs”. This could
Federal Food, Drug and Cosmetic Act with the pre-
be seen as the start of pharmaceutical inspections.
market notification requirement for new drugs in
History of Pharmacopoeias, the official books of
1938. However, in countries with poor regulatory
drug quality standards, probably dates back to one
environment even recently medicines contaminated
of the proclamations of the Salerno Medical Edict
with diethylene glycol have killed patients.
issued by Fredrick II of Sicily (1240), and ordered
The second catastrophe that influenced the de-
apothecaries to prepare remedies always in the same velopment of medicines regulation far more than
way – forma curiae. The first Pharmacopoeias as we any event in history was the thalidomide disaster.
know them today stared to appear in Europe from Thalidomide was a sedative and hypnotic that first
16th century e.g. the first Spanish Pharmacopoeia went on sale in Western Germany in 1956. Be-
tween 1958 and 1960 it was introduced in 46 dif-
1 The views stated in this chapter reflect the views of the authors ferent countries worldwide resulting in an estimated
and not necessarily those of the World Health Organization. 10,000 babies being born with phocomelia and other

Drug Benefits and Risks: International Textbook of Clinical Pharmacology, revised 2nd edition
Edited by C.J. van Boxtel, B. Santoso and I.R. Edwards. IOS Press and Uppsala Monitoring Centre, 2008. © 2008 The authors. All rights reserved.
66 Drug Benefits and Risks

deformities. The role of this disaster in shaping the Somewhat parallel with the ongoing harmoniza-
medicines regulatory systems is not hard to underes- tion and movement towards creating a common mar-
timate. ket for medicines inside the EU, the need for wider
As a result the whole regulatory system was re- harmonization was after preliminary contacts be-
shaped in the UK where a Committee on the Safety tween officials from Japan, EU and US discussed
of Drugs (CSD) was started in 1963 followed by during the International Conference of Drug Reg-
a voluntary adverse drug reaction reporting system ulatory Authorities (ICDRA – organized by WHO
(Yellow Card Scheme) in 1964. In the United States, every second year) in Paris in 1989. The prelimi-
The Drug Amendments Act of 1962 was passed by nary informal discussions had revealed a need for the
Congress requiring the FDA to approve all new drug harmonization of requirements relating to the new
applications (NDA) and, for the first time, demanded innovative drugs and the green light given in Paris
that a new drug should be proven to be effective led to the establishment in 1990 of the International
and safe. Of equal importance, the FDA was also Conference on Harmonization of Technical Require-
given the authority to require compliance with cur- ments for the Registration of Pharmaceuticals for
rent Good Manufacturing Practices (GMP), to of- Human Use (ICH), a collaborative initiative between
ficially register drug establishments and implement the EU, Japan and the United States with observers
other requirements. The EEC Directive 65/65/EEC
from WHO, EFTA and Canada. ICH harmoniza-
on the approximation of provisions laid down by
tion focuses primarily on technical requirements for
law, regulation and administrative action relating to
new, innovative medicines. However, countries with
medicinal products was also induced by the thalido-
limited resources are mostly generic markets and
mide disaster.
may have difficulties of implementing numerous so-
It took almost ten years for the European Com-
munity (EC), since Council Directive 65/65/EEC phisticated ICH standards. Pharmaceutical regula-
was introduced, to further develop harmonization tory harmonization facilitates the availability of safe,
in the Community. In 1975 two Council Direc- effective and good quality pharmaceuticals. World
tives were introduced, the first on approximation Health Organization (WHO)2 supports harmoniza-
of the laws of Member States relating to analyt- tion on national, regional, inter-regional and inter-
ical, pharmacotoxicological and clinical standards national levels. International consensus on quality,
and protocols in respect of the testing of proprietary safety and efficacy standards can accelerate the in-
medicinal products (75/318/EEC), and the second troduction of new medicines and increase availabil-
on the approximation of provisions laid down by ity of generic medicines through fair competition,
law, regulation and administrative action relating to thereby lowering prices.
medicinal products (75/319/EEC). The latter estab-
lished an ‘old’ Committee on Proprietary Medicinal
Products (CPMP) as an advisory committee to the II. WHY REGULATING DRUGS?
EC and introduced the multistate procedure known
now as the mutual recognition procedure. Directive Drugs are not ordinary consumers’ products. In most
87/22/EEC introduced the concentration procedure instances, consumers are not in a position to make
which is now known as the centralized procedure. decisions about when to use drugs, which drugs to
These directives, and following council regulation, use, how to use them and to weigh potential bene-
were the landmarks for starting harmonization in- fits against risks as no medicine is completely safe.
side the European Union with the final longstand- Professional advise from either prescribers or dis-
ing aim of creating a ‘common market’ for medi- pensers are needed in making these decisions. How-
cines. The Council Regulation EEC/2309/93 estab- ever, even healthcare professionals (medical doc-
lished the European Medicines Evaluation Agency tors, pharmacists) nowadays are not in capacity to
(EMEA) in 1993 and re-established the CPMP as a
‘new’ CPMP to formulate the opinion of the Agency
2 WHO is the directing and coordinating technical agency for
on questions relating to the submission of applica-
health within the United Nations system. It is responsible for pro-
tions and granting marketing authorizations in ac-
viding leadership on global health matters, shaping the health re-
cordance with the centralized procedure. The details search agenda, setting norms and standards, articulating evidence-
of European marketing authorization procedure are based policy options, providing technical support to countries and
described in detail in other publications. monitoring and assessing health trends.
Drug Regulation: History, Present and Future 67

take informed decisions about all aspects of medi- Table 1. Principal medicines regulatory functions
cines without special training and access to nec-
• Licensing of the manufacture, import, export, distrib-
essary information. The production of medicines, ution, promotion and advertising of medicines
their distribution and dispensing also requires spe- • Assessing the safety, efficacy and quality of medi-
cial knowledge and expertise. Among medical disci- cines, and issuing marketing authorization for individ-
plines clinical pharmacology could be considered as ual products
a discipline that covers most comprehensively clini- • Inspecting and surveillance of manufacturers, im-
cal aspects of medicines safety and efficacy. Among porters, wholesalers and dispensers of medicines
medical specialists clinical pharmacologists have the • Controlling and monitoring the quality of medicines
most comprehensive training to understand all the on the market
complexities of the clinical use of medicines. Due • Controlling promotion and advertising of medicines
• Monitoring safety of marketed medicines including
to sophisticated scientific issues related to medicines
collecting and analysing adverse reaction reports
just any medical training may not be enough to take • Providing independent information on medicines to
fair judgments about their safety and efficacy. Also professionals and the public
only basic training in pharmacy may not enable to
take proper judgments about medicines quality.
Source: WHO Policy Perspectives on Medicines no 7, 2003.
The use of ineffective, poor quality, harmful
medicines can result in therapeutic failure, exac-
erbation of disease, resistance to medicines and and skills, and operates within a legal framework.
sometimes death. It also undermines confidence in Regulatory functions involve interactions with vari-
health systems, health professionals, pharmaceuti- ous stakeholders (e.g. manufacturers, traders,
cal manufacturers and distributors. Money spent on consumers, health professionals, researchers and
ineffective, unsafe and poor quality medicines is governments) whose economic, social and political
wasted – whether by patients/consumers or insur- motives may differ, making implementation of reg-
ance schemes/governments. Governments have the ulation both politically and technically challenging.
responsibility to protect their citizens in the areas Medicines regulation has administrative part but far
where the citizens themselves are not able to do more important is the scientific basis for it. All medi-
so. Thus, Governments need to establish strong na- cines must meet three criteria: be of good quality,
tional regulatory authorities (NRAs), to ensure that safe and effective. The judgments about medicines
quality, safety and efficacy should be based on solid
the manufacture, trade and use of medicines are
science. There are several general and specific fac-
regulated effectively. In broad terms the mission of
tors contributing to effective regulation by NRAs.
NRAs is to protect and promote public health. Medi-
General factors include political will and commit-
cines regulation demands the application of sound
ment to regulation, adequate availability of medi-
scientific (including but not limited to medical, phar-
cines that are accessible (to avoid smuggling and il-
maceutical, biological and chemical) knowledge and
legal use), strong public support for drug regulation,
specific technical skills, and operates within a legal effective cooperation between the NRA and other
framework. The basic elements of effective drug reg- government institutions including those dealing with
ulation have been laid down in several WHO docu- law enforcement (e.g. customs and police), and suf-
ments. ficient qualified and experienced pharmaceutical,
medical and other professionals. Political environ-
ment favouring independent science based decision-
III. WHAT IS MEDICINES REGULATION? making and control of import/export and distribution
(including e-commerce) of medicines is essential.
Medicines regulation incorporates several mutually The specific factors for NRA include clear mission
reinforcing activities all aimed at promoting and statement, adequate medicines legislation and regu-
protecting public health. These activities vary from lation, appropriate organizational structure and facil-
country to country in scope and implementation, but ities, clearly defined NRA roles and responsibilities,
generally include the functions listed in Table 1. adequate and sustainable financial resources, includ-
What makes medicines regulation effective? ing resources to retain and develop staff and appro-
Medicines regulation demands the application of priate tools, such as standards, guidelines and proce-
sound medical, scientific and technical knowledge dures. International collaboration with other NRAs
68 Drug Benefits and Risks

Table 2. Minimum regulatory functions for a national regulatory authority (NRA)

As an absolute minimum NRAs should


• Ensure that all medicines manufacturing, importation, exportation, wholesale and distribution establishments are li-
censed. Activities and premises must comply with Good Manufacturing Practices (GMP) and Good Distribution Prac-
tice requirements
• Before medicines are marketed, assess their safety, efficacy and quality
• Monitor the quality and safety of medicines on the market to prevent harmful, substandard and counterfeit medicines
from reaching the public
• Regularly inspect and control the informal market, including e-commerce, to prevent illegal trade of medicines
• Monitor advertising and promotion of medicines, and provide independent information on their rational use to the
public and professionals
• Participate in sub-regional and regional regulatory networks and international meetings of drug regulatory authorities
to discuss issues of mutual interest and concern, facilitate timely exchange of information and promote collaboration
• Monitor and evaluate performance to assess if perceived regulatory objectives have been met, to identify weaknesses
and take corrective action

Source: WHO Policy Perspectives on Medicines no 7, 2003.

(for example, in the EU national regulators are re- regulations and regulations in many other countries
quired to collaborate in line with respective Com- do not allow direct to patient advertising of prescrip-
munity regulations) and internal collaboration with tion only medicines (in US it is allowed and has
all stakeholders, transparency (making transparent increased sales of several medicines dramatically).
how and based on which information decisions are These guidelines remain also useful today and pro-
made) and accountability combined with good man- vide ethical criteria for different promotional activi-
agement and effective internal quality system con- ties and cover, among others, advertisements to pre-
tribute to the success of a regulatory authority. Min- scribers and to the general public, the availability of
imum functions that a NRA should be able to carry free samples of prescription drugs for prescribers or
out are laid down in Table 2. of non-prescription drugs to the general public, med-
Excessive promotion of pharmaceuticals has been ical symposia and other scientific meetings, activi-
associated in many countries with serious problems ties of medical representatives, packaging and label-
of irrational drug use. Unethical medicines promo- ing and the information for patients in the package
tion activities often convey misleading information inserts.
about drugs to the different target audiences. Misin- There are few in depth comparative studies of
formation can be in the form of an expansion of in- regulatory systems in different countries globally.
dications or an exaggeration of efficacy but can also The study by Ratanwijitrasin and Wondemageg-
present itself as downplaying the seriousness or the nehu (2002) revealed that in spite of similarities
incidence of adverse reactions. Such misleading in- there are still substantial differences existing in how
formation will create a wrong perception of the effi- regulatory systems in different countries carry out
minimum functions required for effective medicines
cacy and safety of medicinals among prescribers and
regulation. A huge variety in national regulatory
consumers and it will lead to a significant increased
capacity does exist and not all national regulators
demand for drugs. In many countries, relevant pro-
can effectively implement even minimum regulatory
visions regarding such control measures have been
oversight of pharmaceutical market in their jurisdic-
stipulated in legislation. For example, only product
tion. Substandard and counterfeit medicines are still
information approved during the registration process
common in many parts of the world.
can be included in the package inserts, leaflets or
promotional materials. Regulatory or legal provi-
sions with respect to drugs usually appreciate the IV. DRUG REGISTRATION
right of patients or consumers on proper informa-
tion about the drugs they take. WHO has developed Registration of drugs, also known as product licens-
guidelines on Ethical Criteria for Medicinal Drug ing or marketing authorization, is an essential ele-
Promotion. These guidelines in line with European ment of drug regulation. All drugs that are marketed,
Drug Regulation: History, Present and Future 69

distributed and used in the country should be regis- most countries compulsory. As important as assess-
tered by the national competent regulatory authority. ment of quality, safety and efficacy is ensuring ap-
Only the inspection of manufacturing plants and lab- propriateness, accuracy and availability of approved
oratory quality control analysis certainly does not by regulators product information. When marketing
guarantee product quality and safety. Drug regula- authorization is granted for medicines a set of clin-
tion should therefore include the scientific evalua- ical information including indications are approved.
tion of products before registration, to ensure that The use of medicines for indications that have not
all marketed pharmaceutical products meet the cri- been approved by a regulator is called ‘off-label’
teria of safety, efficacy and quality. Although these use. This means that the safety and efficacy of medi-
criteria are applicable to all medicines including bi- cines for these indications has not been assessed and
ological products (including vaccines, blood prod- approved by a regulator. One of the most common
ucts, monoclonal antibodies, cell and tissue thera- off-label use areas is pediatric medicine.
pies) and herbal medicines (also other traditional In the next section we are concentrating on giv-
and complementary medicines) there are substan- ing general overview of registration requirements for
tial differences in the regulatory requirements for two major groups of medicines: innovative (origina-
some groups of medicines. There should also be tor) and multisource (generic) medicines.
clear distinctions between medicines which can be
dispensed without prescription (over the counter or IV.a. Innovative Medicines
OTC medicines) and those for which a prescription
is needed. Usually new medicines are introduced as Innovative medicines (originator products) are new
prescription only medicines and only after obtain- medicines that have not been used in humans ear-
ing knowledge and experience about their safe use lier and contain new active ingredients (usually ex-
they may be considered being used as OTC for self- pressed using INN system). Nowadays these medi-
medication. This is valid only in case patients are cines are usually first approved by regulators in
expected to be able for adequate self-diagnosis as well resourced countries using regulatory require-
well. WHO has issued Guidelines for the Regula- ments harmonized in the framework of International
tory Assessment of Medicinal Products for Use in Conference on Harmonization of Technical Require-
Self-Medication. In regulatory practice active phar- ments for the Registration of Pharmaceuticals for
maceutical ingredients used in medicines are ex- Human Use (ICH – see also web site: www.ich.org).
pressed using International Nonproprietary Names The terms of reference for ICH include to maintain
(INNs). INNs are assigned upon request to a mole- a forum for constructive dialogue between regula-
cular entity responsible for the pharmacological ac- tory authorities and the pharmaceutical industry on
tion by WHO. The INN system as it exists today the real and perceived differences in the technical
was initiated in 1950 by a World Health Assem- requirements in the EU, USA and Japan in order to
bly resolution WHA3.11 and began operating in ensure a more timely introduction of new medicinal
1953. Chemical names and entire formulas are of- products, and their availability to patients, to monitor
ten difficult to remember and may be incomprehen- and update harmonized technical requirements lead-
sible for a non specialist (for example, perhaps few ing to a greater mutual acceptance of research and
medical doctors know that 4 -hydroxyacetanilide or development data and to contribute to the protection
N -(4-hydroxyphenyl) acetamide is paracetamol). of public health from international perspective.
The cumulative list of INN now stands at some 7500 The ICH technical Topics are divided into four
plus names designated since that time, and this num- major categories and specific ICH Topic Codes
ber is growing every year by some 120–150 new (such as Q1, E6, S1 and M4) are assigned accord-
INN (INNs are proposed also for biological medi- ing to these categories. Q means ‘Quality’ Topics
cines such as monoclonal antibodies and gene ther- i.e., those relating to chemical and pharmaceutical
apy products). INNs are also widely used in scien- Quality Assurance (examples: Q1 Stability Test-
tific literature and in teaching basic and clinical phar- ing, Q3 Impurity Testing). S means ‘Safety’ Top-
macology. The lists of International Nonproprietary ics, i.e., those relating to in vitro and in vivo pre-
Names are published in regular manner. Use of INNs clinical studies (examples: S1 Carcinogenicity Test-
in product labeling and information is nowadays in ing, S2 Genotoxicity Testing). E means ‘Efficacy’
70 Drug Benefits and Risks

Topics, i.e., those relating to clinical studies in hu- Safety section (M4S) and the Efficacy section (M4E)
man subject (examples: E4 Dose Response Stud- of the harmonized application. Module 1 contains
ies, Carcinogenicity Testing, E6 Good Clinical Prac- ICH region specific administrative data and prescrib-
tices; Clinical Safety Data Management is also clas- ing information and is not part of CTD. Module 2
sified as an ‘Efficacy’ Topic – E2). M designates contains CTD summaries, Module 3 is dedicated to
‘Multidisciplinary’ Topics, i.e., cross-cutting Top- quality, Module 4 for non-clinical study reports and
ics which do not fit uniquely into one of the above Module 5 on clinical study reports. The structure
categories (examples here are M1 Medical Termi- of Common Technical Document (CTD) is given in
nology – MedDRA, M2 Electronic Standards for the Fig. 1. The content for CTD has to be compiled
Transmission of Regulatory Information – ESTRI, taking into consideration technical requirements in
M3 Timing of Pre-clinical Studies in Relation to more than 56 ICH guidelines for Quality, Safety and
Clinical Trials, M4 The Common Technical Doc- Efficacy plus 5 multidisciplinary (M) topics. Reg-
ument – CTD and M5 Data Elements and Stan- istration of new medicines by less resourced regu-
dards for Drug Dictionaries). ICH guidelines are not latory agencies is often based on first approval ei-
mandatory for anybody per se but the strength of ther by US FDA or EMEA from EU. Indirectly ICH
ICH process lies in the commitment for implemen- guidelines used by these regulatory agencies have
tation by the ICH ‘regions’ (EU, USA and Japan) major impact on approval of new medicines beyond
using appropriate national/regional tools. For exam- ICH regions. Many ICH guidelines, especially those
ple, in the EU all ICH guidelines are submitted to the concerning preclinical and clinical research, are of
Committee for Human Medicinal Products (CHMP) interest to the research community and can serve
associated to European Medicines Agency (EMEA, also as educational tools.
see web site: http://www.emea.europa.eu/) for en- Clinical pharmacologists should be familiar with
available ICH guidelines concerning medicines ef-
dorsement once they have reached certain matu-
ficacy and safety. Those involved in clinical re-
rity phase ICH process. The CHMP, in consultation
search have to know in depth Good Clinical Prac-
with the European Commission decides on the dura-
tice (GCP – ICH E6) guidelines as well the guide-
tion for consultation with interested parties (up to 6
lines concerning the research ethics. WHO has its
months). The European Medicines Agency (EMEA)
own GCP guidelines which do not contradict ICH
publishes and distributes the Step 2 guidelines for
guideline but which in addition describe the role
comments. At Step 4 the guidelines are endorsed by
of regulatory authorities. In addition, WHO has de-
the CHMP and a time frame for implementation is veloped a tool for implementation of GCP which
established (usually 6 months). The guidelines are provides practical advice on the principles of GCP
subsequently published by the European Commis- and has an interactive CD which incorporates many
sion in the Rules Governing Medicinal Products in texts related to GCP and research ethics. In research
the European Union (http://ec.europa.eu/enterprise/ ethics the fundamental principle that “no one shall
pharmaceuticals/eudralex/index.htm). Step 2 and be subjected without his free consent to medical
Step 4 guidelines are also available from the EMEA or scientific experimentation” has found further in-
site on the Internet (http://www.emea.europa.eu). terpretation in a set of principles laid down in the
As more than 95% of new medicines are worked World Medical Association (WMA) Declaration of
out in the ICH “regions” the technical requirements Helsinki (first edition 1964, current version from
for the safety, efficacy and quality of new medi- 2004 under revision). In case of research ethics and
cines is determined at large by ICH technical guide- medicines safety the work of the Council for Interna-
lines. The application format for registration (mar- tional Organizations of Medical Sciences (CIOMS)
keting authorization) of new medicines in ICH and should be referred to. CIOMS was founded un-
associated countries (such as Canada, Switzerland der the auspices of the World Health Organization
and Australia) has to follow The Common Technical (WHO) and the United Nations Educational, Scien-
Document (CTD) which provides harmonized struc- tific and Cultural and Organization (UNESCO) in
ture and format for new product applications. This 1949. In the late 1970s, CIOMS set out, in cooper-
Common Technical Document is divided into four ation with WHO, to prepare guidelines “to indicate
separate sections and 5 modules (see Fig. 1). The how the ethical principles that should guide the con-
four sections address the application organization duct of biomedical research involving human sub-
(M4: Organization), the Quality section (M4Q), the jects, as set forth in the Declaration of Helsinki,
Drug Regulation: History, Present and Future 71

Fig. 1. Diagrammatic representation of the organization of the ICH Common Technical Document (CTD).

could be effectively applied, particularly in develop- Most important of them are International Report-
ing countries”. In 1991, CIOMS published the Inter- ing of Adverse Drug Reactions, which has been ba-
national Guidelines for Ethical Review of Epidemi- sis for ICH guideline E2A (pre-approval reporting)
ological Studies; and, in 1993, International Ethical and ICH E2B (electronic case submission of indi-
Guidelines for Biomedical Research Involving Hu- vidual case safety reports – ICSRs). CIOMS Inter-
man Subjects. This guideline was updated and pub- national Reporting of Periodic Drug-Safety Update
lished in 2002 and is designed to be of use, particu- Summaries has been basis for ICH E2C (periodic
larly to low-resource countries, in defining the ethics safety update report – PSUR). The latest CIOMS
of biomedical research, applying ethical standards working group resulted in publishing The Develop-
in local circumstances, and establishing or redefin- ment Safety Update Report (DSUR): Harmonizing
ing adequate mechanisms for ethical review of re- the Format and Content for Periodic Safety Report-
search involving human subjects. In addition, WHO ing During Clinical Trials. CIOMS has also been
has created several guidance documents how to es- involved in discussing issues related to pharmaco-
tablish and run Ethics Committees dealing with clin- genetics with regulators, industries and academia
ical research. Several CIOMS guidelines have also which resulted in publishing Pharmacogenetics: To-
influenced regulatory approach to medicines safety. wards Improving Treatment with Medicines.
72 Drug Benefits and Risks

IV.b. Multisource (Generic) Medicines in different contexts. In the pharmaceutical sense,


the pharmacopoeia is an official (legally binding)
Multisource (generic) medicines are formulated
publication containing recommended quality speci-
when patent and other exclusivity rights expire.
fications for the analysis and determinations of drug
These medicines have an important role to play in
substances, specific dosage forms, excipients and
public health as they are well known to medical com-
finished drug products. A quality specification is
munity and usually more affordable due to compe-
composed of a set of appropriate tests which will
tition. The key for generic medicines is their ther- confirm the identity and adequate purity of the prod-
apeutic interchangeability with originator products. uct, ascertain the strength (or amount) of the ac-
To ensure the therapeutic interchangeability generic tive substance and, when possible, certain its per-
products must be pharmaceutically interchangeable formance characteristics. General requirements are
(contain the same amount of active ingredient and also given in the pharmacopoeia on important sub-
have the same dosage form) and bioequivalent to the jects related to drug quality, such as microbiological
originator product. Bioequivalence is usually estab- purity, dissolution testing and stability.
lished using comparative in vivo pharmacokinetic The underlying principles of a pharmacopoeia
studies with originator products. The detailed de- are that pharmaceutical substances and products in-
scription how it is carried out is described in respec- tended for human use should be manufactured in
tive WHO document and national regulatory guide- sites that are adequately equipped, dispose of ap-
lines. Well resourced regulatory authorities require propriate professional and technical knowledge and
that a multisource (generic) medicine must meet cer- that are operated by qualified staff. General rules
tain regulatory criteria. These are presented in Ta- of appropriate pharmaceutical manufacture are con-
ble 3. tained in the Good Manufacturing Practices (GMP)
WHO has developed comprehensive set of guide- requirements recommended by WHO and/or those
lines for generic drug registration which are useful laid down by the competent national (or regional,
for drug authorities in developing countries: Market- such as European Commission) regulatory authority.
ing Authorization of Pharmaceutical Products with In regulatory terms GMP could belong to ABC of
Special Reference to Multisource (Generic) Prod- regulatory requirements for medicines and compli-
ucts – A Manual for Drug Regulatory Authorities ance with it is vital for products quality. GMP is ap-
(first edition 1999, updated version to be published plicable for both innovator and generic products. It is
in 2008). applicable for manufacture of active pharmaceutical
In the context of generic medicines it is appro- ingredients and finished dosage forms. Even manu-
priate to ask what is a “pharmacopoeia” (word is facture of investigational drugs should follow GMP.
derived from Greek pharmako-poios “drug-maker”) Without GMP consistency of manufacture clinical
and how it fits in nowadays regulatory systems? performance of medicines cannot be assured.
The answer to this question may seem obvious, but There is a practical distinction between phar-
the term “pharmacopoeia” is used in a varied way macopoeial standards and manufacturers’ release
specifications, although both comprise sets of tests
Table 3. Regulatory requirements for multisource to which a given product should conform. Release
(generic) medicines specifications are applied at the time of manufacture
of a pharmaceutical product to confirm its appropri-
A generic medicines must: ate quality but they also need to have a predictive
(1) contain the same active ingredients as the innovator value, to support the notion that the manufacturer is
drug responsible for the product during its entire shelf-
(2) be identical in strength, dosage form, and route of life. In many cases pharmacopoeial monographs are
administration based on the specifications developed by the manu-
(3) have the same use indications
facturers of innovator (originator) products.
(4) be bioequivalent (as a marker for therapeutic inter-
In order to launch innovator products pharma-
changeability)
(5) meet the same batch requirements for identity,
copoeial specifications are not necessary as the man-
strength, purity and quality ufacturers quality specifications have to pass rigor
(6) be manufactured under the same strict standards of scientific assessment by the competent regulatory
GMP required for innovator products authorities in conjunction with pre-clinical and clin-
ical safety and efficacy data. It is important to notice
Drug Regulation: History, Present and Future 73

that the focus in regulatory environment has been WHO hosts The International Pharmacopoeia.
shifting from finished dosage form quality control This pharmacopoeia is based on specifications val-
to the control of the whole complex of processes and idated internationally, through an independent inter-
procedures involved in the manufacture of both ac- national scientific process.
tive pharmaceutical ingredients (APIs) and finished Unlike national (such as British Pharmacopoeia,
dosage forms. The objective of a nowadays regula- Indian Pharmacopoeia or US Pharmacopoeia) and
tory approval is to ensure that the manufacturer has regional (such as European Pharmacopoeia) phar-
built quality into the product from A to Z. macopoeias, The International Pharmacopoeia has,
In case of multisource (generic) medicines (which a priori, no determined legal status, but WHO Mem-
are formulated after the patents and other exclu- ber States are free to adopt it and to incorporate it
sivity rights expire) pharmacopoeial monographs into national legislation, either in part or in whole.
are more important as they enable manufacturers The first edition was published in two volumes (1951
not to elaborate their own specifications but rather and 1955). The latest fourth edition of The Interna-
develop the products to meet the requirements of tional Pharmacopoeia was published in 2006 and an
pharmacopoeial standards (both for APIs and fin- update is to be published in 2008.
ished dosage forms). It should be noted that not all Most importantly, a new series of monographs
pharmacopoeias present monographs (quality stan- has been added for antiretrovirals. These mono-
dards) for finished dosage forms. Pharmacopoeial graphs have been developed as part of the WHO
standards have also certain limitations. For example,
strategy to make quality antiretroviral medicines
testing using pharmacopoeial methods is not neces-
more widely available to HIV-positive patients. Such
sarily identifying all possible dangerous impurities.
specifications support the joint United Nations –
Pharmacopoeial methods are usually designed to
WHO Prequalification project, managed by WHO
catch the impurities that are likely to occur dur-
(web site: http://mednet3.who.int/prequal/). Interna-
ing the route of synthesis that has been utilized by
tional Chemical Reference Substances (ICRS) are
the originator. In case of different route of synthe-
primary chemical reference standards used in con-
sis or accidental contamination with other chem-
icals it may not necessarily pick up the impuri- junction with International Pharmacopoeia mono-
ties even if they pose danger to the health. This is graphs. They are supplied primarily for use in phys-
why nowadays well resourced regulatory authorities ical and chemical tests and assays described in the
never base their marketing authorizations of mul- specifications for quality control of drugs published
tisource (generic) products only on quality control in The International Pharmacopoeia or proposed in
testing based on pharmacopoeial monographs. In draft monographs.
fact, the pre-marketing quality control testing has di- WHO gives advice on the establishment and man-
minished constantly and more accent is put on mar- agement of national quality control laboratories,
ket surveillance after the product is put on the mar- prepares guidelines on their functioning, publishes
ket. guidance and gives advice on Good Manufacturing
Pharmacopoeial monographs help to verify the Practices (GMP) and other regulatory issues, fol-
quality and in case of multisource (generic) medi- lowing the underlying principle that quality must be
cines they may indicate also on pharmaceutical in- built into a product from the very beginning of the
terchangeability with the originator product. How- manufacturing process. The whole area of work is
ever, pharmacopoeial monographs even for finished overseen by the WHO Expert Committee on Specifi-
dosage forms may have limitations in proving thera- cations for Pharmaceutical Preparations. The WHO
peutic interchangeability which is very important for Expert Committee on Specifications for Pharmaceu-
clinical use of medicines (Box 1). tical Preparations is the highest level advisory body

Box 1. Pharmacopoeial standards

Pharmacopoeial standards should be used in the framework of all regulatory measures such as Good Manufacturing
Practice (GMP) inspection of active pharmaceutical ingredient and finished dosage form manufacturing, scientific as-
sessment of all quality specifications, interchangeability data and labeling information provided by the manufacturer.
The most of their value is in post-marketing surveillance of the quality of multisource (generic) medicine.
74 Drug Benefits and Risks

to WHO’s Director-General and its Member States activity involves assessment of regulatory activi-
in the area of quality assurance. The advice and rec- ties on country level and various technical train-
ommendations provided by this Expert Committee ing courses (such as GMP and GCP, how to assess
are intended to help national and regional authorities generic medicines, bioequivalence, safety monitor-
(in particular drug regulatory authorities), procure- ing and pharmacovigilance, quality assurance and
ment agencies, as well as major international bodies quality control) and customized technical assistance
and institutions to combat problems of substandard (in cooperation with numerous WHO collaborating
and counterfeit medicines. centers and other partners) to the countries. Third,
The importance and role of WHO in the field of in selected areas of essential products, ensuring the
quality assurance of medicines, especially for those quality, safety and efficacy of limited high public
countries that have no or little means to develop their health value essential medicines (such as antiretro-
own quality control specifications, persists. WHO voirals to treat HIV/AIDS, or medicines to treat
has numerous activities to support member states malaria) and vaccines (used in national vaccina-
such as creating necessary nomenclatures, guide- tion programs) through “prequalification”. De facto
lines and guidance (WHO GMP being a good exam- prequalification, although primarily meant for UN
ple) but also delivering training courses and work- procurement and international donors, is a regula-
shops on various topics of regulatory sciences ded- tory activity mimicking medicines registration (mar-
icated to assessment of safety, efficacy and quality keting authorization) in its all elements to ensure
of medicines in order to build national capacity to that products prequalified meet all international stan-
regulate medicines. dards for quality, safety and efficacy. Prequalifica-
tion program has also a very strong capacity build-
ing element built into it. Fourth, WHO plays a very
important role in facilitating exchange of regulatory
V. ROLE OF WHO IN DRUG REGULATION
information for which it has developed a number of
tools. Since 1980 WHO convenes every second year
WHO is the directing and coordinating authority for
International Conference of Drug Regulatory Au-
health within the United Nations system (see more thorities (ICDRA) and publishes their proceedings.
on web site: http://www.who.int/en/). It is responsi- These conferences provide drug regulatory authori-
ble for providing leadership on global health matters, ties of WHO Member States with a forum to meet
shaping the health research agenda, setting norms and discuss ways to strengthen collaboration. The
and standards, articulating evidence-based policy ICDRAs have been instrumental in guiding through
options, providing technical support to countries and its recommendations regulatory authorities, WHO
monitoring and assessing health trends. In the 21st and interested stakeholders and in determining pri-
century, health is a shared responsibility, involving orities for action in national and international reg-
equitable access to essential care and collective de- ulation of medicines, vaccines, biomedicines and
fence against transnational health threats. herbals.
WHO’s role in drug regulation is fourfold. First, WHO manages also a system for regular ex-
issuing necessary norms and standards (see exam- change of information between Member States on
ples above) through its Expert Committees (such as the safety and efficacy of pharmaceutical products,
WHO Expert Committee on Specifications for Phar- using a network of designated national drug infor-
maceutical Preparations and WHO Expert Com- mation officers. WHO ensures the prompt transmis-
mittee on Biological Standardization) and Expert sion to national health authorities of new information
Committee like bodies (such as International Non- on serious adverse effects of pharmaceutical prod-
proprietary Names Expert Group and International ucts and it also responds to individual requests
Working Group for Drug Statistics Methodology – for information. These goals are achieved by the
issuing Anatomical, Therapeutic and Chemical or regular publication of regulatory information in
ATC codes and Daily Defined Doses or DDDs for the WHO Pharmaceuticals Newsletter (http://www.
drug utilization research). Second, supporting reg- who.int/medicines/publications/newsletter/en/index.
ulatory capacity building leading to implementa- html) and by the dissemination of one-page Drug
tion of drug regulation on national level and its Alerts on an ad hoc basis. Relevant restrictive regula-
harmonization on regional and Global level. This tory decisions are ultimately compiled in the United
Drug Regulation: History, Present and Future 75

Nations Consolidated List of Products Whose Con- and new products will create new regulatory chal-
sumption and/or Sale Have Been Banned, With- lenges. For example, how will increasing public at-
drawn, Severely Restricted or not Approved by Gov- tention and expectations on medicines safety shape
ernments. WHO publishes updates to this list: Phar- the regulations? How using new technologies such
maceuticals: Restrictions in use and availability. as nanotechnologies change the medicines regula-
WHO publishes also quarterly WHO Drug Infor- tion? Issues relating to the understanding of how
mation (http://www.who.int/druginformation/) jour- the nanoparticles are presented to organs, cells and
nal which provides an overview of topics of cur- organelles are of the highest importance when try-
rent relevance relating to drug development, safety ing to understand the different mechanisms for in-
and regulation. Latest lists of proposed and recom- tracellular trafficking and use their full therapeu-
mended International Nonproprietary Names (INN) tic potential. Those aspects cannot be established
for Pharmaceutical Substances are also published in without improving appropriate basic knowledge of
cell and molecular biology at the intracellular level.
this journal.
However, at the same time important quality prob-
WHO cooperates very actively with national reg-
lems can rise. In order to assure quality physical and
ulatory authorities of all of its Member States. It tries
chemical properties of nanopharmaceuticals, includ-
to facilitate spreading best practices and experience.
ing residual solvents, processing variables, impuri-
Through its observer role in the international Confer- ties and excipients, should all be well known. There
ence of Harmonization (ICH) WHO is liaising be- will be a need for well-established standard tools to
tween ICH and non-ICH countries trying to ensure be used in the characterization of nanopharmaceu-
that information exchange between highly industri- ticals, including availability of validated assays to
alized and less resourced countries is taking place. detect and quantify nanoparticles in tissues, medici-
nal products and processing equipment. Toxicologi-
cal aspects of nanomedicines have been highlighted
VI. FUTURE OF MEDICINES REGULATION with focus on long-term toxicity. Carbon nanotubes,
quantum dots and other nonbiodegradable and po-
Medicines regulation has been developing together tentially harmful materials should be given closer
with the sciences involved in developing new drugs. attention weather associated with medicines or di-
Also developments in health delivery systems have agnostics. A special set of standards must be grad-
plaid role as those involved in health service delivery ually established in the global regulatory environ-
are interested in safe and effective treatments which ment. In fact, some elements already do exist. In Eu-
would be cost effective and affordable. Both costs rope Directive 2004/27/EEC on medicines addresses
of research and development and regulatory assess- directly the need for the study of environmental im-
ment of products is increasing. There is likely no al- pact of medicines which will have major impact for
ternative for more harmonization (international, re- new nanomaterails to be used in medicines. To ex-
amine and predict environment impact is a new task
gional and sub-regional) of regulatory requirements
for regulators.
and work sharing (together with information shar-
Using genetic information to create safe and ef-
ing) between different national regulatory authori-
fective medicines offers potential for more individ-
ties. The cost of full regulatory assessment of a new
ualized therapies and patient benefits but will also
drug is increasingly becoming not affordable (both have an impact on the use of healthcare resources.
in terms of financial and human resources) for less Pharmacogenetics has been viewed as something
resourced smaller regulatory agencies. What are the for the future, but real clinical examples now exist.
new areas of development beyond better harmoniza- Some pharmacogenetic tests, such as the thiopurine
tion, information exchange and gradual building of methyltransferase (TPMT) test that aims to predict
trust in each others decisions leading to recognition the risk of severe neutropenia for the purine drugs
instead of duplication? azathioprine and 6-mercaptopurine, have already
Although even quality issues are still a prob- relatively low unit costs (approximately 50$ US).
lem (poor quality of starting materials including However, even low unit cost tests may have a signifi-
active pharmaceutical ingredients, quality problems cant cost impact if they have a high volume of uptake
with finished dosage forms, spreading of counter- in a healthcare system. There may be added value as-
feit medicines) it is likely that new technologies sociated with introducing a pharmacogenetic test to
76 Drug Benefits and Risks

guide a prescribing decision, in terms of improved Ceci A, Felisi M, Catapano M, Baiardi P, Cipollina L, Rav-
health-related quality of life resulting from fewer se- era S et al. Medicines for children licensed by the Euro-
vere side effects and improved treatment response pean Agency for the Evaluation of Medicinal Products.
in the patient population taking the medicine. Phar- Eur J Clin Pharmaco 2002;58:495-500.
macogenetic tests broadly fall into one of two cate- CIOMS. International reporting of adverse drug reactions
(Report of CIOMS Working Group I). Geneva: Counsil
gories, those provided through clinical laboratories,
for International Organizations of Medical Sciences;
such as the TPMT test, and those for which a prod-
1990.
uct license has been granted in a similar way to new CIOMS. International reporting of periodic drug-safety
medicines, such as Third Wave Technologies’ (WI, update summaries (Report of CIOMS Working Group
USA) Invader® UGT1A1 Molecular Assay, which II). Geneva: Counsil for International Organizations of
was approved by the US FDA in 2005. The last op- Medical Sciences; 1993.
tion means that regulators are directly involved. Reg- CIOMS. International ethical guidelines for biomedical
ulators are starting to regulate pharmacogenetics and research involving human subjects. Geneva: Counsil
some guidance already exists in Canada, EU and US. for International Organizations of Medical Sci-
Recently also ICH started to deal with pharmacoge- ences; 2002 [cited 2008 Jan 13]. Available from: URL:
nomics and pharmacogenetics. The E15 guideline http://www.cioms.ch/frame_guidelines_nov_2002.htm
Definitions for Genomic Biomarkers, Pharmacoge- CIOMS. Pharmacogenetics: towards improving treat-
ment with medicines (Report of the CIOMS Working
nomics, Pharmacogenetics, Genomic Data and Sam-
Group). Geneva: Counsil for International Organiza-
ple Coding Categories has been finalized.
tions of Medical Sciences; 2005.
Another area of challenges includes biological CIOMS. The development safety update report (DSUR):
medicines including ‘generic’ biological medicines. harmonizing the format and content for periodic safety
New product groups are emerging and even with reporting during clinical trials (Report of CIOMS
known product groups there are challenges ahead, Working Group VII). Geneva: Counsil for International
especially from the point of view of safety. Other Organizations of Medical Sciences; 2006.
important areas for drug regulators remain pharmo- Declaration of Helsinki. World Medical Association; 2004
covigilance, pediatric medicines, orphan medicines [cited 2008 Feb 25]. Available from: URL:http://
and medicines for diseases outside ICH regions. www.wma.net/e/policy/b3.htm
There are few financial incentives to create medi- Fargher EA, Tricker K, Newman B, Elliott R, Roberts SA,
cines for tropical and neglected diseases but recently Shaffer JL, Payne K. Current use of pharmacogenetic
testing: a national survey of thiopurine methyltrans-
due to public private partnerships for drug develop-
ferase (TPMT) testing prior to azathioprine prescrip-
ment and creation of specific regulatory pathways tion. J Clin Pharm Ther 2007;32:187-95.
such EU Article 58 procedure that enables European Gaspar R. Regulatory issues surrounding nanomedicines:
Medicines Agency to assess these products and pro- setting the scene for the next generation of nanophar-
vide scientific advise for WHO has improved the sit- maceuticals. Nanomed 2007;2(2):143-7.
uation. There are even calls for ‘complete rethink’ Gazarian M, Kelly M, McPhee JR, Graudins LV, Ward RL,
of the regulatory systems in order to prepare for the Campbell TJ. Off-label use of medicines: consensus
next 20–30 year. recommendations for evaluating appropriateness. Med
The present short overview of medicines regula- J Aust 2006;185:544-8.
tion is clearly not comprehensive but rather an at- Grabinski JL. Pharmacogenomics of anticancer agents:
tempt to give idea about the complexities of this im- implications for clinical pharmacy practice. J Pharm
Pract 2007;20(3):246-51.
portant area of work that has many direct links with
Griffin JP, Shah RR. History of drug regulation in the
clinical pharmacology. Clinical pharmacologists as
UK. In: O’Grady J, Griffin JP, editors. The regulation
medical specialists equipped with unique knowledge of medical products. London: Blackwell BMJ Books;
about medicines have a role and responsibility to de- 2003. p. 3-12.
velop and contribute to medicines regulation. Hari P, Jain Y, Kabra SK. Fatal encephalopathy and renal
failure caused by diethylene glycaol poisoning. J Trop
Pediatr 2006;52(6):442-4.
BIBLIOGRAPHY Irs A, De Hoog TJ, Rägo L. Development of marketing
authorization procedures for pharmaceuticals. In: Free-
Bankowski, Z. editor. International guidelines for ethical mantle N, Hill S, editors. Evaluating pharmaceuticals
review of epidemiological studies. Geneva: Counsil for for health policy and reimbursement. London: Black-
International Organizations of Medical Sciences; 1991. well BMJ Books; 2004. p. 3-24.
Drug Regulation: History, Present and Future 77

Maynard A, Aitken RJ, Butz T, Colvin V, Donaldson K, WHO. Marketing authorization of pharmaceutical prod-
Oberdörster G et al. Safe handling of nanotechnology. ucts with special reference to multisource (generic)
Nature 2006; 444(7117):267-9. products. A manual for a drug regulatory au-
O’Brien KL, Selanikio JD, Hecdivert C, Placide MF, thority. Geneva: World Health Organization; 1999.
Louis M, Barr DB et al. Epidemic of pediatric deaths (WHO/DMP/RGS/98.5).
from acute renal failure caused by diethylene glycol WHO. Guidelines for the regulatory assessment of medic-
poisoning. Acute Renal Failure Investigation Team. inal products for use in self-medication. Geneva: World
JAMA 1998;279(15):1175-80.
Health Organization; 2000. (WHO/EDM/QSM/00.1).
Permanand G, Mossialos E, McKee M. Regulating medi-
WHO. Operational guidelines for ethics committees that
cines in Europe: the European Medicines Agency, mar-
keting authorisation, transparency and pharmacovigi- review biomedical research. Geneva: WHO; 2000
lance. Clin Med 2006;6(1):87-90. [cited 2008 Jan 13]. Available from: URL:http://www.
Ratanwijitrasin S, Wondemagegnehu E. Effective drug who.int/tdr/publications/publications/ethics.htm
regulation. A multicountry study. Geneva: World WHO. Surveying and evaluating ethical review practices.
Health Organization; 2002. A complementary guideline to the operational guide-
Rägo L. ICH and global cooperation in the new millen- lines for ethics committees that review biomedical
nium: WHO perspective. In: Cone M, editor. Proceed- research. Geneva: World Health Organization; 2002
ings of the fifth international conference on harmonisa- [cited 2008 Jan 13]. Available from: URL:http://www.
tion, San Diego, 2000. London: PJB Publications Ltd; who.int/tdr/publications/publications/ethics.htm
2001. p. 299-304. WHO. Introduction to drug utilization research, Geneva:
Rägo L. Global disequilibrium of quality. In: Prince R, ed- World Health Organization; 2003 [cited 2008 Feb 25].
itor. Pharmaceutical quality. River Grove (IL): Davies Available from: URL:www.who.int/medicines/areas/
Health Care International Publishing; 2004. p. 3-21. quality_safety/safety_efficacy/utilization/en/
The important world of prequalification [editorial]. Lancet WHO. Effective medicines regulation: ensuring safety, ef-
2004;346:1830.
ficacy and quality. Geneva: World Health Organiza-
The International Pharmacopoeia, 4th ed. Vol 1: General
tion; 2003 [cited 2008 Jan 7]. (WHO policy perspec-
notices; monographs for pharmaceutical substances
tives on medicines; no 7). Available from: URL: http://
(A–O). Vol 2: Monographs for pharmaceutical sub-
whqlibdoc.who.int/hq/2003/WHO_EDM_2003.2.pdf
stances (P–Z); monographs for dosage forms and ra-
diopharmaceutical preparations; methods for analysis; WHO. Handbook for good clinical research prac-
reagents. Geneva: World Health Organization; 2006. tice (GCP): guidance for implementation. Geneva:
Trouiller P, Olliaro P, Torreele E, Orbiniski J, Laing R, World Health Organization; 2005 [cited 2008
Ford N. Drug development for neglected diseases: a de- Jan 13]. Available from: URL:www.who.int/prequal/
ficient market and a public health policy failure. Lancet info_general/documents/GCP/gcp1.pdf
2002;359:2188-94. WHO. Multisource (generic) pharmaceutical products:
WHO. Ethical criteria for medicinal drug promotion. guidelines on registration requirements to establish
Geneva: World Health Organization; 1988. interchangeability. In: WHO Expert Committee on
WHO. Guiding principles for small national drug regula- Specifications for Pharmaceutical Preparations. Forti-
tory authorities. In: WHO Expert Committee on Spec- eth report. Geneva: WHO; 2006 [cited 2008 Jan 19].
ifications of Pharmaceutical Products. Geneva: WHO; p. 347-390. (WHO Technical report series; no 937,
1990. p.64-79. (Technical report series; no 790). annex 7). Available from: URL:http://www.who.int/
WHO. A legislative scheme for regulating medicinal prod- medicines/publications/pharmprep/en/index.html
ucts for adaptation by small national drug regula- WHO. International nonproprietary names (INN) for
tory authorities with limited manpower and other re-
pharmaceutical substances [CD-ROM]. Geneva:
sources. Geneva: World Health Organization; 1993.
World Health Organization; 2007. Available from:
(WHO/PHARM/93.244; annex 3).
URL:http://bookorders.who.int/bookorders/anglais/
WHO. Guidelines for good clinical practice (GCP) for
qsearch1.jsp?sesslan=1
trials on pharmaceutical products. Geneva: World
Health Organization; 1995 [cited 2008 Jan 13]. WHO. Quality assurance of pharmaceuticals. A com-
(WHO Technical report series; no. 850, annex pendium of guidelines and related materials, vol 2, 2nd
3). Available from: URL: http://www.who.int/ update ed. Good manufacturing practices and inspec-
medicines/library/par/ggcp/ GGCP.shtml tion. Geneva: World Health Organization; 2007.

You might also like