Вы находитесь на странице: 1из 105

WHO/CDS/CSR/DRS/2001.

2
DISTR: GENERAL
ORIGINAL: ENGLISH

WHO GLOBAL STRATEGY FOR CONTAINMENT OF ANTIMICROBIAL RESISTANCE


WHO Global
Strategy for
Containment
of Antimicrobial
Resistance

Copies can be obtained from the CDS Information Resource Centre World Health Organization
World Health Organization, 1211 Geneva 27, Switzerland
WHO

fax: +41 22 791 42 85 • email: cdsdoc@who.int


WHO/CDS/CSR/DRS/2001.2
ORIGINAL: ENGLISH
DISTRIBUTION: GENERAL

WHO Global
Strategy for
Containment
of Antimicrobial
Resistance

World Health Organization


Acknowledgements
The World Health Organization (WHO) wishes to acknowledge with gratitude the significant support
from the United States Agency for International Development (USAID) and additional assistance for
this work from the United Kingdom Department for International Development and the Ministry of
Health, Labour and Welfare, Japan.
This strategy is the product of collaborative efforts across WHO, particularly in the clusters of Commu-
nicable Diseases, Health Technology and Pharmaceuticals, and Family and Community Health, with
significant input from the staff at WHO Regional Offices and from many partners working with WHO
worldwide. In particular, WHO would like to acknowledge the important contributions made to the
drafting of the strategy by Professor W Stamm, Professor ML Grayson, Professor L Nicolle and Dr M
Powell, and the generosity of their respective institutions—Infectious Diseases Department, Harborview
Medical Center, University of Washington, Seattle, USA; Infectious Diseases and Clinical Epidemiol-
ogy Department, Monash Medical Centre, Monash University, Melbourne, Australia; Department of
Internal Medicine, University of Manitoba, Winnipeg, Canada; Medicines Control Agency, London
UK—that enabled them to spend time at WHO.
WHO also wishes to thank all those who participated and contributed their expertise in the consulta-
tions (see Annex B) and those individuals and organizations that provided valuable comments on drafts
of this document.

© World Health Organization 2001


This document is not a formal publication of the World Health Organization (WHO), and all rights are reserved by the
Organization. The document may, however, be freely reviewed, abstracted, reproduced and translated, in part or in whole, but
not for sale or for use in conjunction with commercial purposes.
The views expressed in documents by named authors are solely the responsibility of those authors.
The designations employed and the presentation of the material in this document, including tables and maps, do not imply the
expression of any opinion whatsoever on the part of the secretariat of the World Health Organization concerning the legal status
of any country, territory, city or area or of its authorities, or concerning the delimitation of its frontiers or boundaries. Dotted
lines on maps represent approximate border lines for which there may not yet be full agreement.
The mention of specific companies or of certain manufacturers’ products does not imply that they are endorsed or recom-
mended by WHO in preference to others of a similar nature that are not mentioned. Errors and omissions excepted, the names
of proprietary products are distinguished by initial capital letters.
Designed by minimum graphics
Printed in Switzerland
Contents

Executive Summary 1

Summary of recommendations for intervention 3

Part A. Introduction and background 9


Introduction 11
Antimicrobial resistance is a global problem that needs urgent action 11
A global problem calls for a global response 12
Implementation of the WHO Global Strategy 13
Background 15
What is antimicrobial resistance? 15
Appropriate use of antimicrobials 15
Surveillance of antimicrobial resistance 15
The prevalence of resistance 16
Conclusion 16

Part B. Appropriate antimicrobial use and emerging resistance: issues and interventions 19
Chapter 1. Patients and the general community 21
Chapter 2. Prescribers and dispensers 25
Chapter 3. Hospitals 31
Chapter 4. Use of antimicrobials in food-producing animals 37
Chapter 5. National governments and health systems 41
Chapter 6. Drug and vaccine development 47
Chapter 7. Pharmaceutical promotion 51
Chapter 8. International aspects of containing antimicrobial resistance 55

Part C. Implementation of the WHO Global Strategy 61


Introduction 63
Prioritization and implementation 63
Implementation guidelines 66
Monitoring outcomes 66
Summary 67
Recommendations for intervention 68
Tables 71
Suggested model framework for implementation of core interventions 76

References 83

Annexes 93
Annex A. National Action Plans 95
Annex B. Participation in WHO Consultations 96

iii
EXECUTIVE SUMMARY
Executive Summary

■ Deaths from acute respiratory infections, ■ Resistance is only just beginning to be consid-
diarrhoeal diseases, measles, AIDS, malaria and ered as a societal issue and, in economic terms, as
tuberculosis account for more than 85% of the a negative externality in the health care context.
mortality from infection worldwide. Resistance to Individual decisions to use antimicrobials (taken
first-line drugs in most of the pathogens causing by the consumer alone or by the decision-making
these diseases ranges from zero to almost 100%. combination of health care worker and patient)
In some instances resistance to second- and third- often ignore the societal perspective and the per-
line agents is seriously compromising treatment spective of the health service.
outcome. Added to this is the significant global
■ The World Health Assembly (WHA) Resolu-
burden of resistant hospital-acquired infections,
tion of 1998 (1) urged Member States to develop
the emerging problems of antiviral resistance and
measures to encourage appropriate and cost-
the increasing problems of drug resistance in the
effective use of antimicrobials, to prohibit the dis-
neglected parasitic diseases of poor and mar-
pensing of antimicrobials without the prescription
ginalized populations.
of a qualified health care professional, to improve
■ Resistance is not a new phenomenon; it was practices to prevent the spread of infection and
recognized early as a scientific curiosity and then thereby the spread of resistant pathogens, to
as a threat to effective treatment outcome. How- strengthen legislation to prevent the manufacture,
ever, the development of new families of sale and distribution of counterfeit antimicrobials
antimicrobials throughout the 1950s and 1960s and the sale of antimicrobials on the informal
and of modifications of these molecules through market, and to reduce the use of antimicrobials in
the 1970s and 1980s allowed us to believe that we food-animal production. Countries were also en-
could always remain ahead of the pathogens. By couraged to develop sustainable systems to detect
the turn of the century this complacency had come resistant pathogens, to monitor volumes and pat-
to haunt us. The pipeline of new drugs is running terns of use of antimicrobials and the impact of
dry and the incentives to develop new anti- control measures.
microbials to address the global problems of drug
■ Since the WHA Resolution, many countries
resistance are weak.
have expressed growing concern about the prob-
■ Resistance costs money, livelihoods and lives lem of antimicrobial resistance and some have
and threatens to undermine the effectiveness of developed national action plans to address the
health delivery programmes. It has recently been problem. Despite the mass of literature on anti-
described as a threat to global stability and na- microbial resistance, there is depressingly little on
tional security. A few studies have suggested that the true costs of resistance and the effectiveness of
resistant clones can be replaced by susceptible ones; interventions. Given this lack of data in the face
in general, however, resistance is slow to reverse of a growing realization that actions need to be
or is irreversible. taken now to avert future disaster, the challenge is
■ Antimicrobial use is the key driver of resistance. what to do and how to do it.
Paradoxically this selective pressure comes from a ■ The WHO Global Strategy for Containment
combination of overuse in many parts of the world, of Antimicrobial Resistance addresses this chal-
particularly for minor infections, misuse due to lenge. It provides a framework of interventions to
lack of access to appropriate treatment and under- slow the emergence and reduce the spread of anti-
use due to lack of financial support to complete microbial-resistant microorganisms through:
treatment courses.

1
WHO GLOBAL STRATEGY FOR CONTAINMENT OF ANTIMICROBIAL RESISTANCE • WHO/CDS/CSR/DRS/2001.2
— reducing the disease burden and the spread provision of public goods such as information, in
of infection surveillance, analysis of cost-effectiveness and
— improving access to appropriate anti- cross-sectoral coordination.
microbials
■ Given the complex nature of antimicrobial re-
— improving use of antimicrobials
sistance, the strategy necessarily contains a large
— strengthening health systems and their sur-
number of recommendations for interventions.
veillance capabilities
Prioritization of the implementation of these in-
— enforcing regulations and legislation
terventions needs to be customized to national
— encouraging the development of appropri-
realities. To assist in this process an implementa-
ate new drugs and vaccines.
tion approach has been defined together with
■ The strategy highlights aspects of the contain- indicators for monitoring implementation and
ment of resistance and the need for further research outcomes.
directed towards filling the existing gaps in knowl-
■ Recognition that the problem of resistance
edge.
exists and the creation of effective national inter-
■ The strategy is people-centred, with interven- sectoral task forces are considered critical to the
tions directed towards the groups of people who success of implementation and monitoring of
are involved in the problem and need to be part interventions. International interdisciplinary co-
of the solution, i.e. prescribers and dispensers, operation will also be essential.
veterinarians, consumers, policy-makers in hos-
■ Improving antimicrobial use must be a key
pitals, public health and agriculture, professional
action in efforts to contain resistance. This requires
societies and the pharmaceutical industry.
improving access and changing behaviour; such
■ The strategy addresses antimicrobial resistance changes take time.
in general rather than through a disease-specific
■ Containment will require significant strength-
approach, but is particularly focused on resistance
ening of the health systems in many countries and
to antibacterial drugs.
the costs of implementation will not be negligi-
■ Much of the responsibility for implementation ble. However, such costs must be weighed against
of the strategy will fall on individual countries. future costs averted by the containment of wide-
Governments have a critical role to play in the spread antimicrobial resistance.

2
SUMMARY OF RECOMMENDATIONS FOR INTERVENTION
Summary of recommendations
for intervention

Patients and the general community &


prescribers and dispensers 1.3 Educate patients on simple measures that
The emergence of antimicrobial resistance is a may reduce transmission of infection in the
household and community, such as
complex problem driven by many interconnected
handwashing, food hygiene, etc.
factors, in particular the use and misuse of
antimicrobials. Antimicrobial use, in turn, is in- 1.4 Encourage appropriate and informed
health care seeking behaviour.
fluenced by an interplay of the knowledge, expec-
tations and interactions of prescribers and patients, 1.5 Educate patients on suitable alternatives to
antimicrobials for relief of symptoms and
economic incentives, characteristics of the health
discourage patient self-initiation of treat-
system(s) and the regulatory environment. In the
ment, except in specific circumstances.
light of this complexity, coordinated interventions
are needed that simultaneously target the behav-
iour of providers and patients and change impor- 2 PRESCRIBERS AND DISPENSERS
tant features of the environments in which they Education
interact. These interventions are most likely to be 2.1 Educate all groups of prescribers and dis-
successful if the following factors are understood pensers (including drug sellers) on the im-
within each health setting: portance of appropriate antimicrobial use
and containment of antimicrobial resist-
• which infectious diseases and resistance prob- ance.
lems are important 2.2 Educate all groups of prescribers on dis-
• which antimicrobials are used and by whom ease prevention (including immunization)
• what factors determine patterns of antimi- and infection control issues.
crobial use 2.3 Promote targeted undergraduate and
• what the relative costs and benefits are from postgraduate educational programmes on
changing use the accurate diagnosis and management
• what barriers exist to changing use. of common infections for all health care
workers, veterinarians, prescribers and dis-
Although the interventions directed towards
pensers.
providers and patients are presented separately (1
2.4 Encourage prescribers and dispensers to
and 2) for clarity, they will require implementa-
educate patients on antimicrobial use and
tion in an integrated fashion.
the importance of adherence to prescribed
treatments.

1 PATIENTS AND THE GENERAL 2.5 Educate all groups of prescribers and dis-
COMMUNITY pensers on factors that may strongly influ-
ence their prescribing habits, such as
Education economic incentives, promotional activi-
1.1 Educate patients and the general commu- ties and inducements by the pharmaceu-
nity on the appropriate use of antimicro- tical industry.
bials.
1.2 Educate patients on the importance of Management, guidelines and formularies
measures to prevent infection, such as im- 2.6 Improve antimicrobial use by supervision
munization, vector control, use of bednets, and support of clinical practices, especially
etc. diagnostic and treatment strategies.

3
WHO GLOBAL STRATEGY FOR CONTAINMENT OF ANTIMICROBIAL RESISTANCE • WHO/CDS/CSR/DRS/2001.2

2.7 Audit prescribing and dispensing practices Diagnostic laboratories


and utilize peer group or external stand- 3.5 Ensure access to microbiology laboratory
ard comparisons to provide feedback and services that match the level of the hospi-
endorsement of appropriate antimicrobial tal, e.g. secondary, tertiary.
prescribing. 3.6 Ensure performance and quality assurance
2.8 Encourage development and use of guide- of appropriate diagnostic tests, microbial
lines and treatment algorithms to foster identification, antimicrobial susceptibility
appropriate use of antimicrobials. tests of key pathogens, and timely and rel-
2.9 Empower formulary managers to limit evant reporting of results.
antimicrobial use to the prescription of an 3.7 Ensure that laboratory data are recorded,
appropriate range of selected anti- preferably on a database, and are used to
microbials. produce clinically- and epidemiologically-
useful surveillance reports of resistance
Regulation patterns among common pathogens and
2.10 Link professional registration requirements infections in a timely manner with feed-
for prescribers and dispensers to require- back to prescribers and to the infection
ments for training and continuing educa- control programme.
tion.
Interactions with the pharmaceutical
industry
Hospitals 3.8 Control and monitor pharmaceutical com-
pany promotional activities within the hos-
Although most antimicrobial use occurs in the pital environment and ensure that such
community, the intensity of use in hospitals is far activities have educational benefit.
higher; hospitals are therefore particularly impor-
tant in the containment of antimicrobial resist-
ance. In hospitals it is crucial to develop integrated Use of antimicrobials in food-producing
approaches to improving the use of antimicrobials, animals
reducing the incidence and spread of hospital-ac-
A growing body of evidence establishes a link
quired (nosocomial) infections, and linking thera-
between the use of antimicrobials in food-produc-
peutic and drug supply decision-making. This will
ing animals and the emergence of resistance among
require training of key individuals and the alloca-
common pathogens. Such resistance has an im-
tion of resources to effective surveillance, infec-
pact on animal health and on human health if
tion control and therapeutic support.
these pathogens enter the food chain. The factors
affecting such antimicrobial use, whether for thera-
3 HOSPITALS peutic, prophylactic or growth promotion pur-
poses, are complex and the required interventions
Management need coordinated implementation. The underly-
3.1 Establish infection control programmes,
ing principles of appropriate antimicrobial use and
based on current best practice, with the
containment of resistance are similar to those
responsibility for effective management of
applicable to humans. The WHO global princi-
antimicrobial resistance in hospitals and
ensure that all hospitals have access to
ples for the containment of antimicrobial resist-
such a programme. ance in animals intended for food (2) were adopted
at a WHO consultation in June 2000 in Geneva.
3.2 Establish effective hospital therapeutics
committees with the responsibility for They provide a framework of recommendations
overseeing antimicrobial use in hospitals. to reduce the overuse and misuse of antimicrobials
in food animals for the protection of human
3.3 Develop and regularly update guidelines
for antimicrobial treatment and prophy- health. Antimicrobials are widely used in a vari-
laxis, and hospital antimicrobial formular- ety of other settings outside human medicine, e.g.
ies. horticulture and aquaculture, but the risks to
3.4 Monitor antimicrobial usage, including the human health from such uses are less well under-
quantity and patterns of use, and feedback stood and they have not been included in this
results to prescribers. document.

4
SUMMARY OF RECOMMENDATIONS FOR INTERVENTION
4 USE OF ANTIMICROBIALS IN FOOD- 5 NATIONAL GOVERNMENTS AND
PRODUCING ANIMALS HEALTH SYSTEMS
This topic has been the subject of specific con-
Advocacy and intersectoral action
sultations which resulted in “WHO global prin-
5.1 Make the containment of antimicrobial
ciples for the containment of antimicrobial
resistance a national priority.
resistance in animals intended for food”*. A
complete description of all recommendations — Create a national intersectoral task
is contained in that document and only a sum- force (membership to include health
mary is reproduced here. care professionals, veterinarians,
agriculturalists, pharmaceutical manu-
Summary facturers, government, media repre-
4.1 Require obligatory prescriptions for all sentatives, consumers and other
antimicrobials used for disease control in interested parties) to raise awareness
food animals. about antimicrobial resistance, organ-
4.2 In the absence of a public health safety ize data collection and oversee local
evaluation, terminate or rapidly phase out task forces. For practical purposes such
the use of antimicrobials for growth pro- a task force may need to be a govern-
motion if they are also used for treatment ment task force which receives input
of humans. from multiple sectors.

4.3 Create national systems to monitor antimi- — Allocate resources to promote the
crobial usage in food animals. implementation of interventions to
contain resistance. These interventions
4.4 Introduce pre-licensing safety evaluation
should include the appropriate utiliza-
of antimicrobials with consideration of
tion of antimicrobial drugs, the control
potential resistance to human drugs.
and prevention of infection, and re-
4.5 Monitor resistance to identify emerging search activities.
health problems and take timely corrective
— Develop indicators to monitor and
actions to protect human health.
evaluate the impact of the antimicro-
4.6 Develop guidelines for veterinarians to re- bial resistance containment strategy.
duce overuse and misuse of antimicrobials
in food animals. Regulations
5.2 Establish an effective registration scheme
for dispensing outlets.
* http:// www.who.int/emc/diseases/zoo/
who_global_principles.html 5.3 Limit the availability of antimicrobials to
prescription-only status, except in special
circumstances when they may be dis-
pensed on the advice of a trained health
National governments and health systems
care professional.
Government health policies and the health care 5.4 Link prescription-only status to regulations
systems in which they are implemented play a cru- regarding the sale, supply, dispensing and
cial role in determining the efficacy of interven- allowable promotional activities of antimi-
tions to contain antimicrobial resistance. National crobial agents; institute mechanisms to
commitment to understand and address the prob- facilitate compliance by practitioners and
lem and the designation of authority and respon- systems to monitor compliance.
sibility are prerequisites. Effective action requires 5.5 Ensure that only antimicrobials meeting
the introduction and enforcement of appropriate international standards of quality, safety
regulations and allocation of appropriate resources and efficacy are granted marketing
for education and surveillance. Constructive in- authorization.
teractions with the pharmaceutical industry are 5.6 Introduce legal requirements for manufac-
critical, both for ensuring appropriate licensure, turers to collect and report data on anti-
promotion and marketing of existing microbial distribution (including import/
export).
antimicrobials and for encouraging the develop-
ment of new drugs and vaccines. For clarity, in- 5.7 Create economic incentives for the appro-
terventions relating to these interactions with the priate use of antimicrobials.

industry are shown in separate recommendation


groups (6 and 7).

5
WHO GLOBAL STRATEGY FOR CONTAINMENT OF ANTIMICROBIAL RESISTANCE • WHO/CDS/CSR/DRS/2001.2

Policies and guidelines 6 DRUG AND VACCINE DEVELOPMENT


5.8 Establish and maintain updated national 6.1 Encourage cooperation between industry,
Standard Treatment Guidelines (STGs) and government bodies and academic institu-
encourage their implementation. tions in the search for new drugs and
5.9 Establish an Essential Drugs List (EDL) con- vaccines.
sistent with the national STGs and ensure 6.2 Encourage drug development pro-
the accessibility and quality of these drugs. grammes which seek to optimize treat-
5.10 Enhance immunization coverage and other ment regimens with regard to safety,
disease preventive measures, thereby re- efficacy and the risk of selecting resistant
ducing the need for antimicrobials. organisms.
6.3 Provide incentives for industry to invest in
Education the research and development of new
5.11 Maximize and maintain the effectiveness antimicrobials.
of the EDL and STGs by conducting appro-
6.4 Consider establishing or utilizing fast-track
priate undergraduate and postgraduate
marketing authorization for safe new
education programmes of health care
agents.
professionals on the importance of appro-
priate antimicrobial use and containment 6.5 Consider using an orphan drug scheme
of antimicrobial resistance. where available and applicable.

5.12 Ensure that prescribers have access to ap- 6.6 Make available time-limited exclusivity for
proved prescribing literature on individual new formulations and/or indications for
drugs. use of antimicrobials.
6.7 Align intellectual property rights to pro-
vide suitable patent protection for new
Surveillance of resistance, antimicrobial antimicrobial agents and vaccines.
usage and disease burden
6.8 Seek innovative partnerships with the
5.13 Designate or develop reference microbiol-
pharmaceutical industry to improve access
ogy laboratory facilities to coordinate
to newer essential drugs.
effective epidemiologically sound surveil-
lance of antimicrobial resistance among
common pathogens in the community, 7 PHARMACEUTICAL PROMOTION
hospitals and other health care facilities. 7.1 Introduce requirements for pharmaceuti-
The standard of these laboratory facilities cal companies to comply with national or
should be at least at the level of recom- international codes of practice on promo-
mendation 3.6. tional activities.
5.14 Adapt and apply WHO model systems 7.2 Ensure that national or international codes
for antimicrobial resistance surveillance of practice cover direct-to-consumer
and ensure data flow to the national inter- advertising, including advertising on the
sectoral task force, to authorities responsi- Internet.
ble for the national STGs and drug policy, 7.3 Institute systems for monitoring compli-
and to prescribers. ance with legislation on promotional
5.15 Establish systems for monitoring antimi- activities.
crobial use in hospitals and the community, 7.4 Identify and eliminate economic incentives
and link these findings to resistance and that encourage inappropriate antimicro-
disease surveillance data. bial use.
5.16 Establish surveillance for key infectious 7.5 Make prescribers aware that promotion in
diseases and syndromes according to accordance with the datasheet may not
country priorities, and link this information necessarily constitute appropriate antimi-
to other surveillance data. crobial use.

6
SUMMARY OF RECOMMENDATIONS FOR INTERVENTION
8 INTERNATIONAL ASPECTS OF 8.5 Encourage the establishment of interna-
CONTAINING ANTIMICROBIAL tional inspection teams qualified to con-
RESISTANCE duct valid assessments of pharmaceutical
8.1 Encourage collaboration between govern- manufacturing plants.
ments, non-governmental organizations, 8.6 Support an international approach to the
professional societies and international control of counterfeit antimicrobials in line
agencies to recognize the importance of with the WHO guidelines**.
antimicrobial resistance, to present consist-
8.7 Encourage innovative approaches to
ent, simple and accurate messages regard-
incentives for the development of new
ing the importance of antimicrobial use,
pharmaceutical products and vaccines for
antimicrobial resistance and its contain-
neglected diseases.
ment, and to implement strategies to con-
tain resistance. 8.8 Establish an international database of
potential research funding agencies with
8.2 Consider the information derived from the
an interest in antimicrobial resistance.
surveillance of antimicrobial use and anti-
microbial resistance, including the contain- 8.9 Establish new, and reinforce existing, pro-
ment thereof, as global public goods for grammes for researchers to improve the
health to which all governments should design, preparation and conduct of re-
contribute. search to contain antimicrobial resistance.

8.3 Encourage governments, non-governmen-


tal organizations, professional societies and * Interagency Guidelines. Guidelines for Drug
international agencies to support the es- Donations, revised 1999. Geneva, World
tablishment of networks, with trained staff Health Organization, 1999. WHO/EDM/PAR/
and adequate infrastructures, which can 99.4.
undertake epidemiologically valid surveil- ** Counterfeit drugs. Guidelines for the develop-
lance of antimicrobial resistance and anti- ment of measures to combat counterfeit drugs.
microbial use to provide information for Geneva, World Health Organization, 1999.
the optimal containment of resistance. WHO/EDM/QSM/99.1.
8.4 Support drug donations in line with the UN
interagency guidelines*.

7
PART A

Introduction and
background
INTRODUCTION
Introduction

Antimicrobial resistance is a global expensive drugs to treat a fraction of the


problem that needs urgent action population needing treatment, or to increase
Deaths from acute respiratory infections, diar- health care expenditure.
rhoeal diseases, measles, AIDS, malaria and
• Ineffective therapy leads to increased costs
tuberculosis account for more than 85% of the
mortality from infection worldwide (3). Resist- associated with prolonged illness, more
ance to first-line drugs in the pathogens causing frequent hospital admissions and longer
these diseases ranges from zero to almost 100%. periods of hospitalization. In addition,
In some instances resistance to second- and third- resistant pathogens in the hospital environ-
line agents is seriously compromising treatment ment result in hospital-acquired infections
outcome. Added to these major killers is the which are expensive to control and extremely
significant global burden of hospital-acquired difficult to eradicate.
(nosocomial) infections usually caused by resist-
• The use of antimicrobials outside the field
ant pathogens, the emerging problems of antivi-
of human medicine also has an impact on
ral resistance and the increasing threats of drug
human health. Resistant microorganisms in
resistance in parasitic diseases such as African
food-producing animals may have major
trypanosomiasis and leishmaniasis.
financial implications for both farmers and
The massive increases in trade and human
consumers. Resistant animal pathogens in
mobility brought about by globalization have
some food products, especially meat, may
enabled the rapid spread of infectious agents, in-
cause infections in humans that are difficult
cluding those that are drug resistant. While richer
to treat. In addition, loss of public confi-
countries, to a large extent, are still able to rely on
dence in the safety of food affects the de-
the latest antimicrobials to treat resistant infec-
mand for products, with potentially serious
tions, access to these life-saving drugs is often lim-
economic effects on the farming sector.
ited or totally absent in many parts of the world.
Urgent global action is needed, as outlined below.
Risk management and national security
Costs of resistance Antimicrobial resistance threatens other health
care gains. For example, co-infection with HIV
The relentless emergence of antimicrobial resist-
and antimicrobial-resistant pathogens, e.g. tuber-
ance has an impact on the cost of health care
culosis, salmonellosis, other sexually transmitted
worldwide. Ineffective therapy due to antimicro-
infections, may result in rapid disease progression
bial resistance is associated with increased human
in the infected individual and has a potential
suffering, lost productivity and often death. De-
multiplier effect on the dissemination of resistant
spite a dearth of data on the costs of resistance
pathogens to the rest of the population—thereby
(4), there is growing consensus about the follow-
placing more demands on health care resources.
ing points.
The emergence of antimicrobial resistance is
• In many regions the prevalence of resistance regarded as a major future threat to the security
among common pathogens to readily avail- and political stability of some regions (5).
able cheap antimicrobials is so high that
these agents are now of limited clinical
Antimicrobial resistance is frequently irreversible
effectiveness. Increasingly, this results in dif-
ficult choices: to spend money on cheap Although a few studies (6,7) have suggested that
useless drugs, to use more effective but more resistant clones can be replaced by susceptible ones,

11
WHO GLOBAL STRATEGY FOR CONTAINMENT OF ANTIMICROBIAL RESISTANCE • WHO/CDS/CSR/DRS/2001.2
resistance is generally slow to reverse or is irre- • raise awareness of the problems posed by
versible. This suggests that interventions to stop antimicrobial resistance
the development of resistance should be imple-
• promote the sharing of information about
mented early, before resistance becomes a prob-
and understanding of resistance
lem. The earlier interventions are implemented,
the slower will be the development of resistance • provide strategic and technical guidance on
(4). However, this implies taking action before the interventions to contain resistance
prevalence of resistant infections climbs, based on • assist Member States to implement these
decisions made whilst the number of people suf- interventions
fering resistant infections is low. Antimicrobial
resistance is only just beginning to be considered • stimulate research to address the knowledge
as a societal issue and, in economic terms, as a gaps and improve understanding of antimi-
negative externality (8,9). Individual decisions to crobial resistance and to encourage research
use antimicrobials (taken by the consumer alone and development of new antimicrobial
or by the decision-making combination of pre- agents.
scriber and patient) often ignore the societal per-
spective and the perspective of the health service. Development of the WHO Global Strategy
Following the Resolution on Antimicrobial Re-
A dwindling supply of new antimicrobials sistance in 1998 (1), WHO has worked with many
The development of new antimicrobial agents partners to develop the WHO Global Strategy for
effective against resistant pathogens and of alter- Containment of Antimicrobial Resistance (referred
native approaches such as vaccines is crucial to re- to as the WHO Global Strategy hereafter). The
duce the future impact of resistance. However, new aim of this strategy is to provide, for all Member
agents are expensive and time-consuming to States, a framework of interventions to stimulate
develop. Interest in antimicrobial research and de- the prevention of infection, to slow the emergence
velopment among the research-based pharmaceu- of resistance and to reduce the spread of resistant
tical industry has declined as infectious diseases microorganisms, in order to reduce the impact of
in richer country populations appear to have been resistance on health and health care costs, while
conquered and as priorities have shifted to the improving access to existing agents and encour-
development of lifestyle drugs. Unless the current aging the development of new agents. The strat-
rate of emerging resistance is controlled and slowed egy has been formulated on the basis of expert
to preserve the life of existing drugs, this decline opinion, published evidence, commissioned re-
in new antimicrobial development, even if reversed views and the deliberations of international and
now, is likely to result in the absence of effective national bodies (see Annex B) on the key factors
therapies for some pathogens within the next ten contributing to antimicrobial resistance and the
years. interventions needed for its containment. Based
on these inputs, a series of recommendations is
proposed, directed towards the aims stated above.
A global problem calls for a global response Part B of this document provides a summary of
There can be no doubt that antimicrobial resist- the evidence on which the recommendations are
ance poses a global challenge. No single nation, based.
however effective it is at containing resistance It is important to recognize that much remains
within its boundaries, can protect itself from the to be learnt about the interplay between the fac-
importation of resistant pathogens through travel tors responsible for the emergence and spread of
and trade. The global nature of resistance calls for resistance and the optimization and cost-effective-
a global response, not only in the geographic sense, ness of appropriate interventions. However, the
i.e. across national boundaries, but also across the urgency of the situation requires that implemen-
whole range of sectors involved. Nobody is ex- tation of the WHO Global Strategy moves for-
empt from the problem, nor from playing a role ward on the evidence currently available.
in the solution.
The response of the World Health Organiza-
tion is to:

12
INTRODUCTION
Implementation of the tainment of antimicrobial resistance presented
WHO Global Strategy here, there is a practical need for prioritization and
The approach to implementation is crucial to its customization to the individual national setting.
efficacy and success. Much of the responsibility To assist in the implementation of the WHO Glo-
for implementing interventions will fall on indi- bal Strategy, an approach to defining a smaller core
vidual Member States. There are certain actions set of recommendations is presented (Part C).
that only governments can assure, including the Furthermore, since antimicrobial resistance is a
provision of public goods such as information, clearly a global issue, international interdiscipli-
surveillance and analysis of cost-effectiveness of nary cooperation is critical and the areas in which
interventions, and the cross-sectoral coordination this can be most effective are outlined (Part B,
critical for an effective response (10). Given the Chapter 8).
large number of recommendations for the con-

13
BACKGROUND
Background

What is antimicrobial resistance? Appropriate use of antimicrobials


Resistance to antimicrobials is a natural biologi- The WHO Global Strategy defines the appropri-
cal phenomenon. The introduction of every anti- ate use of antimicrobials as the cost-effective use of
microbial agent into clinical practice has been antimicrobials which maximizes clinical therapeu-
followed by the detection in the laboratory of tic effect while minimizing both drug-related toxic-
strains of microorganisms that are resistant, i.e. ity and the development of antimicrobial resistance.
able to multiply in the presence of drug concen- The general principles of appropriate antimi-
trations higher than the concentrations in humans crobial use (11) are the same as those for all other
receiving therapeutic doses. Such resistance may medicinal products. An additional dimension for
either be a characteristic associated with the en- antimicrobials is that therapy for the individual
tire species or emerge in strains of a normally sus- may affect the health of society as a result of the
ceptible species through mutation or gene transfer. selective pressure exerted by all use of antimicro-
Resistance genes encode various mechanisms bial agents. In addition, therapeutic failures due
which allow microorganisms to resist the inhibi- to drug-resistant pathogens or superinfections lead
tory effects of specific antimicrobials. These mech- to an increased potential for the spread of these
anisms offer resistance to other antimicrobials of organisms throughout hospitals and the commu-
the same class and sometimes to several different nity. Although these risks occur even when
antimicrobial classes. antimicrobials are used appropriately, inappropri-
All antimicrobial agents have the potential to ate use increases the overall selective pressure in
select drug-resistant subpopulations of microor- favour of drug-resistant microorganisms.
ganisms. With the widespread use of antimi- The choice of an appropriate antimicrobial
crobials, the prevalence of resistance to each new agent may be straightforward when the causative
drug has increased. The prevalence of resistance pathogen(s) is/are known or can be presumed with
varies between geographical regions and over time, some certainty from the patient’s clinical presen-
but sooner or later resistance emerges to every tation. However, in the absence of reliable micro-
antimicrobial. biological diagnosis or when several pathogens may
While much evidence supports the view that be responsible for the same clinical presentation,
the total consumption of antimicrobials is the criti- empiric treatment, often with broad-spectrum
cal factor in selecting resistance, the relationship antimicrobials, is common. Ideally, the choice of
between use and resistance is not a simple correla- antimicrobial should be guided by local or national
tion. In particular, the relative contribution of resistance surveillance data and treatment guide-
mode of use (dose, duration of therapy, route of lines. The reality is often far removed from this
administration, dosage interval) as opposed to to- ideal.
tal consumption is poorly understood. Paradoxi-
cally, underuse through lack of access, inadequate
Surveillance of antimicrobial resistance
dosing, poor adherence and sub-standard
antimicrobials may play as important a role as Surveillance of antimicrobial resistance is essen-
overuse. There is consensus, however, that the in- tial for providing information on the magnitude
appropriate use of antimicrobial agents does not and trends in resistance and for monitoring the
achieve the desired therapeutic outcomes and is effect of interventions. The actions taken on the
associated with the emergence of resistance. For basis of surveillance data will depend on the level
this reason, improving use is a priority if the emer- at which the data are being collected and analysed.
gence and spread of resistance is to be controlled. For example, local surveillance data should be used
to guide clinical management and to update treat-

15
WHO GLOBAL STRATEGY FOR CONTAINMENT OF ANTIMICROBIAL RESISTANCE • WHO/CDS/CSR/DRS/2001.2
ment guidelines, educate prescribers and guide Modern techniques have enabled the develop-
infection control policies. The frequency at which ment and application of molecular methods to
surveillance information is updated is also determine the presence of specific resistance genes
important given that the rise in prevalence of a in microbes. They are most widely used to detect
resistance phenotype may be rapid and the imple- genotypic resistance in viruses such as HIV and
mentation of policy changes is often slow. HBV and, in the future, may form the basis of
Nationally collected surveillance data may be systems to monitor antiviral resistance. However
used to inform policy decisions, update national these molecular methods rely on sophisticated
formularies or lists of essential drugs and stand- technology that is not available in many settings.
ard treatment guidelines and evaluate the cost-
effectiveness of interventions. Since resistance is a
Epidemiologically valid patient selection
global problem, international collation of resist-
ance data may also have a useful role (see Chapter Currently, epidemiological methods are not ap-
8). plied in most resistance surveillance studies. The
terms incidence and prevalence tend to be used in-
terchangeably and usually refer to the number of
National surveillance systems
resistant isolates among the total number of iso-
WHO and its partners have been successful in lates surveyed. In contrast, from a public health
supporting the surveillance of drug-resistant standpoint, one of the goals of surveillance is to
tuberculosis in many countries (12,13). Despite detect the incidence of resistant infections among
the many ongoing activities worldwide in moni- the total number of infections in a population (15).
toring resistance among other bacteria, few coun- Further bias arises since tests to detect resistance
tries have well-established national networks that are performed on a subset of patients presenting
regularly collect and report relevant data. In many for treatment who may be more likely to have
developing countries and countries whose econo- failed empiric therapy previously or to have other
mies are in transition, microbiology laboratory complications. Much greater epidemiologic rig-
facilities and information networks will require our and more active surveillance approaches are
considerable strengthening before reliable surveil- needed to better understand the impact of resist-
lance of resistance is a reality. ance. In this respect, surveillance of drug resist-
ance in tuberculosis is more advanced than that
Standardization of methods to detect resistance of other bacteria (12).
Surveillance of antimicrobial resistance is fun-
Current methods for monitoring antimicrobial
damental to understanding trends in resistance,
resistance can be classified as in vivo, in vitro and
to developing treatment guidelines accurately and
molecular methods. The extent to which each of
to assessing the effectiveness of interventions ap-
these is used depends on the pathogen/disease and
propriately. Without adequate surveillance, the
the facilities available. In vivo methods or thera-
majority of efforts to contain emerging antimi-
peutic efficacy tests are the gold standard for moni-
crobial resistance will be difficult.
toring resistance to antimalarial drugs (14) but are
not used routinely to monitor resistance in other
pathogens. However, linking clinical outcome of The prevalence of resistance
treatment with in vitro detection of resistance is The prevalence of resistance varies widely between
of critical importance to understand the predic- and within countries, and over time.
tive value of in vitro tests. Data on the prevalence of resistance in acute
In vitro methods are the techniques of choice respiratory infections, diarrhoeal diseases, malaria,
for monitoring resistance in the vast majority of tuberculosis and gonorrhoea can be found in re-
bacterial pathogens, including Mycobacterium tu- cent reviews (16,17,18,19,20,21).
berculosis. However, there is no single international
standard method. Different methods have gained
popularity in different parts of the world—at least Conclusion
ten different methods for antimicrobial suscepti- While it is difficult to quantify the total impact of
bility tests are used in Europe and more than twelve resistance on health, published data clearly indi-
worldwide. International quality assurance stand- cate that morbidity and mortality are increased
ards can help to overcome the potential difficul- by delays in administering effective treatment for
ties arising from the use of different methods. infections caused by resistant pathogens. The pro-

16
BACKGROUND
longed illness and hospitalization of patients with financial resources that could otherwise be used
resistant infections and the additional procedures for improving health and threatens the success of
and drugs that they may require carry financial global efforts to combat the major infectious dis-
implications. There may also be economic impli- eases of poverty. In this light, implementation of
cations for the patient in terms of lost productiv- the WHO Global Strategy can be considered
ity. Antimicrobial-resistant infections in appropriate risk management to protect current
food-producing animals may have major finan- health care initiatives and the availability of treat-
cial implications for both farmers and consumers. ment for future generations.
In addition, antimicrobial resistance diverts

17
PART B

Appropriate
antimicrobial use
and emerging
resistance:
issues and
interventions
CHAPTER 1. PATIENTS AND THE GENERAL COMMUNITY
CHAPTER 1

Patients and the general community

Recommendations for intervention Patients’ misperceptions


Education Many patients believe that most infections, regard-
1.1 Educate patients and the general community less of etiology, respond to antimicrobials and thus
on the appropriate use of antimicrobials. expect to receive a prescription from their physi-
cian for any perceived infection. In a study by
1.2 Educate patients on the importance of meas- Macfarlane et al., 85% of patients thought their
ures to prevent infection, such as immuniza- respiratory symptoms were caused by infection and
tion, vector control, use of bednets, etc. 87% believed that antimicrobials would help.
1.3 Educate patients on simple measures that may One-fifth of these patients specifically asked their
reduce transmission of infection in the house- physician to prescribe an antimicrobial (22).
hold and community, such as handwashing, Another study showed that patients’ expectations
food hygiene, etc. for a prescription were met 75% of the time by
prescribers (23). In a survey of 3610 patients con-
1.4 Encourage appropriate and informed health ducted by Branthwaite and Pechère (24), over 50%
care seeking behaviour. of interviewees believed that antimicrobials should
1.5 Educate patients on suitable alternatives to be prescribed for all respiratory tract infections
antimicrobials for relief of symptoms and dis- with the exception of the common cold. It was
courage patient self-initiation of treatment, noted that 81% of patients expected to see a defi-
except in specific circumstances. nite improvement in their respiratory symptoms
after three days and that 87% believed that feel-
ing better was a good reason for cessation of anti-
Introduction
microbial therapy. Most of these patients also
Patient-related factors are major drivers of inap- believed that any remaining antimicrobials could
propriate antimicrobial use and therefore contrib- be saved for use at a later time. Physicians’
ute to the increasing prevalence of antimicrobial perceptions of patient expectations are clearly also
resistance. In particular, the perception of patients crucial (see Chapter 2).
that most episodes of suspected infection require Many patients believe that new and expensive
antimicrobial therapy notably influences the medications are more efficacious than older agents;
prescribing practices of providers. The direct-to- this belief is shared by some prescribers and dis-
consumer marketing by the pharmaceutical indus- pensers and often results in the unnecessary use
try increasingly influences patient expectations and of the newer agents. In addition to causing un-
behaviour. necessary health care expenditure, this practice
Patient-related factors that are thought to con- encourages the selection of resistance to these
tribute to the problem of antimicrobial resistance newer agents as well as to older agents in their
include the following: class.
• patients’ misperceptions Patients commonly misunderstand the phar-
macological actions of antimicrobial agents.
• self-medication Experience suggests that many people do not know
• advertising and promotion the difference between antimicrobials and other
classes of drugs and thus will not understand the
• poor adherence to dosage regimens. issues of resistance uniquely related to antimi-
crobials. In the Philippines, isoniazid is viewed as
a “vitamin for the lungs” and mothers purchase
isoniazid syrup for children with “weak lungs” in

21
WHO GLOBAL STRATEGY FOR CONTAINMENT OF ANTIMICROBIAL RESISTANCE • WHO/CDS/CSR/DRS/2001.2
the absence of documented tuberculosis (25). the consumer’s relative lack of sophistication about
Patients also fail to recognize that many brand the evidence supporting the use of one treatment
names may actually be the same antimicrobial— over another” (35). These advertising methods are
resulting in the unnecessary overstocking of some apparently quite effective, since pharmacists are
agents. For example, specific patient demand frequently able to guess the feature advertisements
caused one pharmacy in South India to stock more of the previous day’s television programmes based
than 25 of the 100 or so brands of co-trimoxazole upon daily customer requests for specific medica-
available (26). tions (31). A survey of physicians in the USA dem-
A greater interaction between health providers onstrated that, on average, each had encountered
and consumers for health and drug (antimicro- seven patients within the previous six months who
bial)-related education has been proposed (27). had specifically requested prescription-only drugs
The WHO Action Programme on Essential Drugs as a result of direct-to-consumer advertising (36).
convened a consultation to address the need for Over 70% of the physicians reported that requests
public education in rational drug use (28) and has from patients as a result of direct-to-consumer
since produced a document “Rational Drug Use: advertisements had led them to prescribe a phar-
Consumer Education and Information” (29). This maceutical agent that they might not have other-
document discusses the practical issues and dilem- wise chosen.
mas related to the need for rational drug use edu- In a telephone survey of consumers regarding
cation, its priority and content, underlying direct advertising, 66% believed that advertise-
principles and target population. In a study car- ments for medications would provide useful
ried out in Peru, a multifaceted educational inter- information but 88% said that they would seek
vention directed at the community using media, out more information about a drug that they saw
face-to-face meetings and training on the use of advertised on television or in print before purchas-
medicines was successful in decreasing the inap- ing it. On the other hand, only one-third of inter-
propriate use of antidiarrhoeals and antimicrobials viewees agreed with the statement that most people
for simple diarrhoea (30). would know if they were being misled by the ad-
vertisements (37).
Recently, the United States Food and Drug
Self-medication
Administration proposed new guidelines that lift
Self-medication with antimicrobials is often cited previous restrictions on direct-to-consumer adver-
as a major factor contributing to drug resistance tising and allow pharmaceutical manufacturers
(31). In a Brazilian study, it was determined that greater freedom on advertised health claims. A
the three most common types of medication used two-year evaluation period was proposed to assess
by villagers were antimicrobials, analgesics and the impacts and implications of these guidelines
vitamins. The majority of antimicrobials were pre- (27).
scribed by a pharmacy attendant or were purchased Advertising and promotion can also be used to
by the patient without prescription (32) despite improve the appropriate use of antibiotics. Public
having prescription-only legal status. In addition education campaigns in India, which include the
to obvious uncertainty as to whether the patient use of mass media such as television, appear to
has an illness that will benefit from antimicrobial have effectively educated even illiterate popula-
treatment, self-medicated antimicrobials are often tions about antimicrobial resistance in some
inadequately dosed (33) or may not contain regions (Bhatia, personal communication).
adequate amounts of active drug, especially if they Interventions to address the effects of advertis-
are counterfeit drugs (34). This is especially ing and promotion are discussed in Chapter 7.
important in the treatment of diseases such as
tuberculosis.
Poor adherence to dosage regimens
In a 1988 literature search, over 4000 English
Advertising and promotion
language articles were available on the topic of
Direct-to-consumer advertising allows pharmaceu- patient adherence to dosing instructions, more
tical manufacturers to market medicines directly than 75% of which had been published within
to the public via television, radio, print media and the previous ten years (38). In the majority of stud-
the Internet. Where permitted, this practice has ies, it was reported that a lack of patient under-
“the potential to stimulate demand by playing on standing and provider communication led to most

22
CHAPTER 1. PATIENTS AND THE GENERAL COMMUNITY
instances of non-adherence (39,40). Patients who adverse effects of their medication(s) has been used
fail to complete therapy have a higher likelihood to increase adherence (49) (see also Chapter 5,
of relapse, development of resistance and need for Recommendations).
re-treatment; this applies especially to those Price is a powerful factor in determining how
patients requiring prolonged treatment, e.g. those consumers use antimicrobials—economic hard-
with tuberculosis or HIV infection. Previous ship can lead to early cessation of therapy. For
antimicrobial treatment and excessive duration of example, antimicrobials are purchased in single
treatment are considered two of the most impor- doses in many developing countries and are taken
tant factors in the selection of resistant microor- for only a fraction of the recommended effective
ganisms (41,42). duration, until the patient feels better. This prac-
Many methods have been used to ensure ad- tice has the potential for fostering the selection of
herence to antimicrobial therapy. These include resistant microorganisms and therefore has a
the use of fixed dose combinations to minimize higher likelihood of treatment failure (50,51). This
the number of tablets or capsules, special calen- is especially important for diseases such as tuber-
dars, blister packing, DOT (directly observed culosis and endocarditis (43,52). Government
therapy) for tuberculosis (12,13,43,44), other schemes which subsidize the cost of certain pre-
course-of-therapy packaging using symbols in ferred antimicrobials are one economic means of
labelling, and more simplified therapy (45,46). improving the appropriateness of antimicrobial
Directly observed therapy, short-course (DOTS) use. Where insurance systems exist, charging dif-
is the WHO strategy for TB control that has been ferential co-payments to patients, with lower pay-
shown to significantly decrease acquired resistance ments for the more desirable drugs, may encourage
in tuberculosis (47,48). Education of patients on appropriate use.
the name, dosage, description and common

23
CHAPTER 2. PRESCRIBERS AND DISPENSERS
CHAPTER 2

Prescribers and dispensers

Recommendations for intervention Regulation


Education 2.10 Link professional registration requirements
2.1 Educate all groups of prescribers and dispens- for prescribers and dispensers to require-
ers (including drug sellers) on the importance ments for training and continuing educa-
of appropriate antimicrobial use and contain- tion.
ment of antimicrobial resistance.
2.2 Educate all groups of prescribers on disease
Introduction
prevention (including immunization) and Prevention of infection should be the primary goal
infection control issues. to improve health and reduce the need for anti-
microbial therapy. Where appropriate, vaccine
2.3 Promote targeted undergraduate and post-
uptake should be improved to achieve this. Both
graduate educational programmes on the ac-
the emergence and maintenance of resistant
curate diagnosis and management of common
microorganisms are promoted by antimicrobial
infections for all health care workers,
use. Furthermore, once they are widespread, re-
veterinarians, prescribers and dispensers.
sistant strains are difficult to replace by their sus-
2.4 Encourage prescribers and dispensers to edu- ceptible counterparts. Early action to optimize
cate patients on antimicrobial use and the prescribing patterns and to reduce inappropriate
importance of adherence to prescribed treat- use is thus crucial. The difficulty is that multiple
ments. factors influence prescribers and dispensers in de-
2.5 Educate all groups of prescribers and dispens- ciding when to use antimicrobials. These factors
ers on factors that may strongly influence their appear to vary in importance depending on geo-
prescribing habits, such as economic incen- graphical region, social circumstances and the
tives, promotional activities and inducements prevailing health care system. Often, the most im-
by the pharmaceutical industry. portant factors are interlinked. Many traditional
approaches to improving antimicrobial use rely on
Management, guidelines and formularies providing correct information about drugs or dis-
eases, with the implicit assumption that prescrib-
2.6 Improve antimicrobial use by supervision and
ers and dispensers will incorporate the new
support of clinical practices, especially diag-
knowledge and make appropriate adjustments in
nostic and treatment strategies.
their practice. However, experience and reviews
2.7 Audit prescribing and dispensing practices of well-designed research studies (53,54,55) have
and utilize peer group or external standard shown that this is rarely the case. Effective inter-
comparisons to provide feedback and endorse- ventions to improve antimicrobial use must
ment of appropriate antimicrobial prescrib- address the underlying causes of current practice
ing. and barriers to change (56).
2.8 Encourage development and use of guidelines
and treatment algorithms to foster appropri- Lack of knowledge and training
ate use of antimicrobials.
Lack of knowledge about differential diagnoses,
2.9 Empower formulary managers to limit anti- infectious diseases and microbiology and about the
microbial use to the prescription of an appro- appropriate choice of antimicrobials for various
priate range of selected antimicrobials. infections all play a role in inappropriate prescrib-
ing practices (34). Even in developed countries,

25
WHO GLOBAL STRATEGY FOR CONTAINMENT OF ANTIMICROBIAL RESISTANCE • WHO/CDS/CSR/DRS/2001.2
the pharmacology of antimicrobial agents, their nating targeted educational messages to their peer
modes of action and spectrum of activity and group (73,74). Unfortunately, none of these stud-
issues relating to resistance receive limited cover- ies looked at resistance as an outcome or impact
age in medical school curricula, resulting in poorly indicator. Increasing problem-based pharmaco-
informed prescribers (57). It is not uncommon therapy training for medical and paramedical stu-
for drug company sales representatives and the dents can have a positive impact on long-term
commercially oriented publications they provide good prescribing habits. The use of a WHO
to be the main sources of information for prescrib- manual (75) designed to support problem-based
ers (58). learning for medical students has been demon-
Lack of knowledge is a major factor responsi- strated to have a positive impact on prescribing
ble for inappropriate antimicrobial use globally. skills of students in seven medical schools (76).
In one study in China, 63% of antimicrobials In countries with limited resources, the dispens-
selected to treat proven bacterial infections were ing of antimicrobials by unauthorized persons
found to be inappropriate (59). In a retrospective lacking appropriate knowledge is common. In a
study in Viet Nam, more than 70% of patients study of 40 randomly selected health facilities in
were prescribed inadequate dosages (60). Gumo- Ghana, only 8.3% of dispensers had received for-
doka et al. (61) reported that one in four patients mal training (77). Bruneton et al. (78) found that
in their medical districts received antimicrobials drug sellers in seven sub-Saharan African coun-
by injection and that approximately 70% of these tries frequently recommended antimicrobials not
injections were unnecessary. Studies in many present on the regions’ Essential Drugs List and
European countries and in the USA demonstrate rarely suggested that the patient should consult a
widespread unnecessary use of antimicrobials in physician.
patients with viral upper respiratory tract infec- Improving the drug education of non-physi-
tions (62). cian prescribers and dispensers is another recom-
Printed materials are the most common and mended step in improving drug use. A study in
least expensive educational interventions but in- Ghana showed that educational interventions
appropriate prescribing is rarely due to a lack of aimed at dispensers significantly improved drug
knowledge alone. Many studies have found that use by increasing the percentage of appropriately
the use of printed material only, without other labelled containers and by increasing patient
forms of supporting interventions, is ineffective knowledge of their medications (77). Interventions
in altering prescribing behaviours (63,64,65). targeted at local drug sellers in the Philippines sig-
Continuing education and in-service training nificantly improved their quality of practice (79).
programmes have traditionally involved lecture-
and seminar-style presentations oriented to the
Lack of access to information
presentation of factual information. A large body
of research has shown that these approaches are Even relatively well-trained prescribers often lack
not necessarily the most effective for improving the up-to-date information required to make
practice (66). Academic detailing, or unadvertise- appropriate prescribing decisions. This tends to
ments, and educational programmes directed to result in the excessive use of newer antimicrobial
physicians have been shown to decrease antimi- agents, often with a broader spectrum of action.
crobial use/misuse (67,68,69,70). One consist- Conversely, lack of surveillance data and updated
ently successful method has been educational treatment guidelines may lead to the inappropri-
outreach, which consists of brief, targeted, face- ate prescription of older antimicrobials which are
to-face educational visits to clinicians by specially either no longer effective due to the emergence of
trained staff (67,71,72). In developing countries resistance or should have been replaced by newer
where individual educational outreach visits may agents with improved cost-effectiveness or reduced
not be practical or cost-effective, interactive prob- toxicity.
lem-oriented educational sessions with small Use of clinical practice guidelines is a core
groups of physicians, paramedics, or pharmacy managerial strategy in every health system for
counter attendants have demonstrated similar suc- improving diagnosis and therapy. Despite the
cess, especially when sessions are repeated over time abundance of guidelines, research has shown that
or reinforced with improved clinical supervision they have little effect on clinical practice unless
(53). Another promising approach involves engag- they are actively disseminated (80). Factors that
ing local opinion leaders in the process of dissemi- increase the likelihood of guidelines being adopted

26
CHAPTER 2. PRESCRIBERS AND DISPENSERS
include local involvement of end-users in the proc- algorithms have been developed (55,85,86). These
ess of development, the presentation of key ele- diagnostic and treatment algorithms are based on
ments in a simple algorithm or protocol, and detailed research studies, generally in resource-
dissemination in a multi-component programme poor regions, in which the patients’ clinical pres-
that includes interactive education, monitoring of entations have been correlated with subsequent
adherence and reinforcement of positive changes. microbiological confirmation of disease. This
A combination of national prescribing guidelines syndromic approach is particularly useful in health
and educational campaigns on the appropriate use care settings where diagnostic capabilities are lim-
of antimicrobials targeted at prescribers has had ited, since it allows a rational approach to deter-
some success in reducing the prevalence of spe- mining the need for antimicrobial therapy and the
cific antimicrobial resistance (7). There has also most appropriate agents.
been some success in the use of educational cam-
paigns targeted at prescribers and patients to
Fear of bad clinical outcomes
recognize that not all infections require the use of
antimicrobials. Included in one such campaign was Prescribers may overuse antimicrobials because
a message to parents discouraging them from they fear that their patients may suffer poor out-
sending their sick children to day care in order to comes in the absence of such therapy. Prescribing
reduce the opportunities for transmission of in- just to be safe increases when there is diagnostic
fection (81). uncertainty, lack of prescriber knowledge regard-
ing optimal diagnostic approaches, lack of oppor-
tunity for patient follow-up, or fear of possible
Lack of diagnostic support
litigation (87,88).
Lack of access to or use of appropriate diagnostic
facilities and slow or inaccurate diagnostic results
encourage prescribers to cover the possibility that Perception of patient demands and
preferences
infection may be responsible for a patient’s illness,
even when this is not the case (58). In particular, Prescribers’ perceptions regarding patient expec-
the lack of accurate tests at point-of-care to achieve tations and demands substantially influence pre-
a rapid diagnosis is a significant problem for many scribing practices (22,23,58,87,89). Although
diseases and is an area in which future research these perceptions may be incorrect, they can lead
could be very beneficial. Empiric treatment of in- to a perpetual cycle whereby patients who repeat-
fections with a reasonably well-defined clinical edly receive unnecessary antimicrobials develop the
presentation is more likely to be appropriate than misconception that antimicrobials are frequently
that of infections with an undifferentiated pres- necessary for most ailments and therefore request
entation e.g. malaria presenting as fever alone. In them excessively (22,90). Prescribers and dispens-
this latter situation the differential diagnosis may ers may also respond to patient demand for par-
be wide and therefore empiric treatment protocols ticular formulations of antimicrobials, e.g. capsules
will necessarily need to be broad—leading to a rather than tablets. In some cultural settings,
higher likelihood of unnecessary antimicrobial antimicrobials given by injection are considered
therapy. A careful history and access to adequate more efficacious than oral formulations. This tends
diagnostic facilities allow the differential diagno- to be associated with the overprescribing of broad-
sis to be narrowed and therapy more targeted. A spectrum injectable agents when a narrow-
study of barefoot doctors in a district of Bangla- spectrum oral agent would be more appropriate
desh found that antimicrobials were prescribed for (61).
60% of all patients seen in areas without diagnos- In an effort to reduce re-consultation of
tic services—a higher rate than noted in other patients, Macfarlane et al. (23) utilized a patient
districts (82). Other studies have had similar find- information leaflet regarding coughs. Among
ings (83,84). In developed countries, empiric patients not prescribed antimicrobials, those given
antimicrobial therapy is sometimes considered to the educational leaflet appeared less likely to
be more cost-effective than awaiting laboratory re-consult; this result, however, was not statisti-
proof of infection before commencing treatment. cally significant. The use of delayed prescribing
For conditions such as acute respiratory infec- techniques has been proposed when physicians feel
tions, diarrhoea and malaria in children, and pressured by their patients into prescribing
sexually transmitted disease in adults, treatment antimicrobials (45,87). Some physicians say that

27
WHO GLOBAL STRATEGY FOR CONTAINMENT OF ANTIMICROBIAL RESISTANCE • WHO/CDS/CSR/DRS/2001.2
they promise a free return visit if the patient feels bonuses tied to practice pharmaceutical budgets.
that a re-consultation is necessary because they did While these strategies may reduce inappropriate
not receive antimicrobials (87). use of antimicrobials, they may also reduce
appropriate use. Nevertheless, a number of
Scandinavian studies have suggested that national
Economic incentives
antibiotic policies together with changes in reim-
Many health providers practise in environments bursement policy can be safe and effective
with financial incentives to prescribe or dispense (95,96,97).
greater numbers of drugs overall or of specific
types. Prescribers may fear the potential loss of
future patient custom and revenue if they do not
Peer pressure and social norms
respond to perceived demands for antimicrobials In focus group studies, prescribers expressed con-
(91). Furthermore, in some countries, prescribers cern that, if they did not prescribe antimicrobials,
profit from both prescribing and dispensing patients would seek other sources of care where
antimicrobials, such that it is in their financial they could obtain antimicrobials (91). In addition,
interest to prescribe antimicrobials even when they the physician offering the latest and often the most
are not clinically indicated. Additional profit is expensive and broad-spectrum antibiotic may be
sometimes gained by recommending newer more perceived to be the most informed and desirable
expensive antimicrobials in preference to older source of care.
cheaper agents. In countries where physicians are Understanding prescribing patterns is crucial
poorly paid, pharmaceutical companies have been to identifying areas for potential intervention to
known to pay commissions to prescribers who use improve use (58). Drug use patterns and prescrib-
their products (92). Other less direct incentives ing behaviour, including the influence of various
such as financial support for attendance at meet- social and patient pressures, can be described us-
ings, entertainment, or payment for enrolling ing the indicators and methods in the WHO
patients in marketing research studies may also manual “How to investigate drug use in health
influence prescribing practices. Even in health facilities” (98). After undertaking interventions to
systems where there is no overt incentive to improve drug use, these same indicators can be
prescribe, there is usually no incentive not to used to measure impact.
prescribe (8).
It is desirable to minimize financial conflicts
Factors associated with the prescriber’s
of interest in therapeutic decision-making by working environment
health providers, such as allowing physicians to
profit from dispensing drugs that they prescribe In busy clinical practices, health care providers may
or allowing pharmacists who sell drugs to prescribe not have time to explain to patients why they have
them as well. Coast et al. (9) and Smith and Coast chosen to prescribe or not prescribe antimicrobial
(93) explored economic perspectives of policies therapy (99). Some clinicians in this situation may
used to decrease antimicrobial resistance. They believe it is simply most time-effective to prescribe
discuss techniques such as regulation (controlling an antimicrobial. Lack of privacy in consultation
prescription practices by means of policies and facilities may also impact on prescribing behav-
guidelines or by enforcing a global limit to the iour since some conditions, such as urinary tract
prescription of antimicrobials), permits (allowing sepsis and sexually transmitted diseases, require
physicians to prescribe up to a certain quantity of diagnostic specimens and/or physical examination
antimicrobials per permit) and charges (levying that are difficult to undertake in public. The lack
taxes on antimicrobials). In their model, they sug- of opportunity for health care workers to follow
gest that the use of permits may offer a method up their patients to assess progress after treatment
for reducing antimicrobial resistance. and poor continuity of care in general negatively
Several countries have introduced health pro- influence communication and the development
vider reimbursement strategies that are designed of trust between the patient and health care pro-
to encourage physicians to limit overall use of vider. It is thus often easier for both prescriber
medicines and often to share in the resulting and patient if an antimicrobial agent is prescribed
financial savings. Examples of these strategies are on first contact.
capitation payments that include pharmaceutical
costs, general practice fundholding (94) and

28
CHAPTER 2. PRESCRIBERS AND DISPENSERS
Lack of appropriate legislation or Inadequate drug supply infrastructure
enforcement of legislation
In many parts of the world, the ability of prescrib-
Absence of appropriate legislation or its enforce- ers and dispensers to provide appropriate antimi-
ment may result in the proliferation of locations crobial therapy is limited by the lack of availability
where untrained or poorly trained persons dispense of the necessary drugs (100).
antimicrobials, leading to overuse and inappro-
priate use (see Chapter 5).

29
CHAPTER 3. HOSPITALS
CHAPTER 3

Hospitals

Recommendations for intervention Introduction


Management Hospitals are a critical component of the antimi-
3.1 Establish infection control programmes, based crobial resistance problem worldwide. The com-
on current best practice, with the responsi- bination of highly susceptible patients, intensive
bility for effective management of antimicro- and prolonged antimicrobial use, and cross-
bial resistance in hospitals and ensure that all infection has resulted in nosocomial infections
hospitals have access to such a programme. with highly resistant bacterial pathogens such as
multi-resistant Gram-negative rods, vancomycin-
3.2 Establish effective hospital therapeutics com- resistant enterococci (VRE) and methicillin-
mittees with the responsibility for overseeing resistant Staphylococcus aureus (MRSA) as well as
antimicrobial use in hospitals. resistant fungal infections. Some of these resist-
3.3 Develop and regularly update guidelines for ant strains have now spread outside the hospital
antimicrobial treatment and prophylaxis, and causing infections in the community. Hospitals
hospital antimicrobial formularies. are also the eventual site of treatment for many
patients with severe infections due to resistant
3.4 Monitor antimicrobial usage, including the pathogens acquired in the community, including
quantity and patterns of use, and feedback penicillin-resistant Streptococcus pneumoniae,
results to prescribers. multi-resistant salmonellae and multi-resistant
Mycobacterium tuberculosis. In the wake of the
Diagnostic laboratories
AIDS epidemic, the prevalence of such infections
3.5 Ensure access to microbiology laboratory can be expected to increase, both in the commu-
services that match the level of the hospital, nity and in hospitals. Hospitals can thus serve both
e.g. secondary, tertiary. as a point of origin of and as a reservoir for highly
3.6 Ensure performance and quality assurance of resistant pathogens which may later enter the com-
appropriate diagnostic tests, microbial iden- munity or chronic care facilities.
tification, antimicrobial susceptibility tests of
key pathogens, and timely and relevant Infection control
reporting of results.
Transmission of highly resistant bacteria from
3.7 Ensure that laboratory data are recorded, pref- patient to patient within the hospital environment
erably on a database, and are used to produce (nosocomial transmission) amplifies the problem
clinically- and epidemiologically-useful of antimicrobial resistance and may result in the
surveillance reports of resistance patterns infection of patients who are not receiving anti-
among common pathogens and infections in microbials. Transmission of antimicrobial-resist-
a timely manner with feedback to prescribers ant strains from hospital personnel to patients or
and to the infection control programme. vice versa may also occur. The key element in mini-
mizing such horizontal transmission of infection
Interactions with the pharmaceutical industry within hospitals is careful attention to infection
3.8 Control and monitor pharmaceutical control practices (101).
company promotional activities within the Failure to implement simple infection control
hospital environment and ensure that such practices such as handwashing and changing gloves
activities have educational benefit. before and after contact with patients is common
(102,103,104,105). In some cases, especially in
resource-poor regions, this may be due to the ab-

31
WHO GLOBAL STRATEGY FOR CONTAINMENT OF ANTIMICROBIAL RESISTANCE • WHO/CDS/CSR/DRS/2001.2
sence of suitable handwashing facilities. However, The key elements of an effective infection con-
inadequate handwashing is generally due to lack trol programme include:
of recognition of its importance in maintaining
— development and implementation of appro-
good infection control, understaffing or health care
priate barrier precautions (handwashing,
worker forgetfulness. Regardless of the reasons,
wearing of gloves and gowns) and isolation
poor infection control practices result in the
procedures
increased dissemination of resistant bacterial
strains in hospital and health care facilities. The — adequate sterilization and disinfection of
spread of resistance appears to be widening as supplies and equipment
patients move more rapidly from intensive care — the use of aseptic techniques for medical
wards to general wards and then to the commu- and nursing procedures
nity or between hospitals and nursing homes
— training of health care personnel in appro-
(51,106,107).
priate sterile techniques and infection con-
Infections can also be transmitted via non-
trol procedures
sterile injection and surgical equipment. In a study
of health facilities in Tanzania, 40% of suppos- — maintenance of appropriate disinfection
edly sterilized needles and syringes were bacteri- and sanitary control of the hospital envi-
ally contaminated (61). Poor decontamination or ronment, including air
failures in sterilization of equipment can have an
— active surveillance of infections and anti-
enormous impact on the spread of viral infections
microbial resistance, with data analysis and
such as HIV (108), hepatitis B and C. Re-use of
feedback to prescribers and other staff
single-use needles and syringes has played a major
role in the spread of viral hepatitis following im- — recognition and investigation of outbreaks
munization programmes in some countries and or clusters of infections.
among intravenous drug users (109,110,111,112). The programme should have a qualified chair-
Any practice that permits the spread of infection person and staff and adequate resources to accom-
permits the spread of resistant infections. plish these goals. The most effective infection
Infection control activities are best coordinated control team consists of a physician (preferably
by an active and effective infection control pro- trained in infectious diseases), a microbiologist,
gramme. The SENIC study, conducted by the infection control nurses, pharmacist(s) and
Centers for Disease Control (CDC) and which hospital management representatives, with the
included a large sample of hospitals in the USA, responsibility for the day-to-day management of
demonstrated that hospitals having infection con- resistance issues. Increased efficiency may be
trol programmes with both active surveillance and achieved by an overlap in the membership of the
control elements were effective in reducing rates infection control team and the hospital therapeu-
of nosocomial infection (113,114,115). In par- tics committee.
ticular, interventions such as education and moti- Appropriate facilities for optimal infection
vation programmes, improvement of equipment, control practices, including sufficient basins and
and performance feedback can increase adherence clean towels to regularly wash hands between
to improved handwashing (104). Barrier precau- patient contacts, may be difficult to achieve in
tions have been shown to be effective in reducing some countries. Nevertheless, such facilities are
infection transmission rates and thereby the spread vital if nosocomial transmission of infections is to
of resistance. Mayer et al. (116) showed that im- be controlled. Handwashing, isolation practices,
proved handwashing and the use of gloves and sufficient beds (and space between them), as well
gowns decreased infection rates. as clean ventilation are needed in hospitals to pre-
With respect to the control of antimicrobial vent the spread of bacteria, including resistant
resistance within the hospital setting, the major strains.
evidence of effectiveness stems from the manage-
ment of outbreaks or clusters of resistant infec-
tions. In these situations, a variety of techniques
Control of antimicrobial use in hospitals
including targeted cohorting of infected patients, Hospitals provide an important training ground
enhanced surveillance, isolation or rigorous for students to learn about prescribing practices.
barrier precautions, early discharge and alteration Unfortunately, antimicrobial prescribing in hos-
in antimicrobial usage have been effective. pitals is often irrational. In an analysis of prescrib-

32
CHAPTER 3. HOSPITALS
ing practices in ten studies from teaching hospi- bial considered necessary without any peer-review
tals worldwide, 41% to 91% of all antimicrobials process is generally inconsistent with optimizing
prescribed were considered inappropriate (117). antimicrobial use. All clinicians should be prepared
Patterns of prescribing become entrenched and, if to justify their antimicrobial usage patterns.
they are not consistent with appropriate antimi- The following activities represent some of the
crobial treatment guidelines, they may have an key roles of an effective therapeutics committee.
enormous effect on the emergence of resistant
• Development of written policies and guide-
pathogens and on the pharmacy budget of a hos-
lines for appropriate antimicrobial usage in
pital if the drugs are expensive. For many clini-
the hospital, based on local resistance sur-
cians, a common source of information regarding
veillance data. Policies should be developed
hospital antimicrobial use is the literature provided
locally, with broad input and consensus from
by pharmaceutical representatives. Such informa-
health care providers and microbiologists.
tion is less likely to be objective than national or
regional treatment guidelines (see Chapter 7). • Selection and provision of appropriate anti-
Antimicrobial prophylaxis for surgical proce- microbials in the pharmacy after considera-
dures is a common reason for excessive prescrib- tion of local clinical needs.
ing in many hospitals. Numerous studies have • Establishment of formal links with an
outlined those procedures in which patients infection control committee, preferably with
benefit from such prophylaxis and those in which some overlap in membership.
they do not (118,119,120,121,122,123,124), but
inappropriate prophylaxis is still widely used. A • Definition of an antimicrobial utilization
further problem is the continuation of anti- review programme, with audit and feedback
microbials, initially administered as prophylaxis, on a regular basis to providers, and promo-
well beyond the required 12 to 24 hour post- tion of active surveillance of the nature
surgical period without clear medical indication and amount of antimicrobial use in the
other than the opinion of the surgeon. Such pre- hospital.
scribing patterns result in high rates of antimicro- • Overseeing antimicrobial use through a
bial exposure among hospitalized patients, system of monitoring the quantity used and
potentially leading to high colonization rates of the indications for use.
resistant nosocomial pathogens and antibiotic-
With regard to the last point, it is important to
associated diarrhoea. For these reasons a variety
recognize that such seemingly basic data collec-
of approaches have been utilized to modify anti-
tion can be difficult to undertake accurately, even
microbial prescribing practices within the hospi-
in the best medical centres. Nevertheless, accu-
tal setting. These have the overall goal of reducing
rate antimicrobial usage information is crucial to
the total consumption of antimicrobials and of
rational decision-making and the interpretation
altering the type of usage in favour of regimens
of antimicrobial resistance data. In systems where
less likely to foster the emergence of resistant
prescribing data are collected routinely, utilization
strains.
review (or audit) combined with feedback of per-
formance data to prescribers has become a com-
Hospital therapeutics committees mon strategy to influence patterns of prescribing
practice. The success of audit and feedback
An active and effective hospital therapeutics com-
programmes is mixed (127). Audit and feedback
mittee is considered a key element for the control
programmes using manually collected samples of
of antimicrobial usage in hospitals, although there
prescribing data and simple performance indica-
are only limited published data to support this
tors have been successful at improving antibiotic
view and there are few data regarding the impact
prescribing in some developing country settings
of hospital therapeutics committees in develop-
(53). Although a decrease in resistance prevalence
ing countries. However, their beneficial role in the
can be achieved with the use of control
promotion of rational prescribing habits, moni-
programmes, once monitoring is relaxed, the
toring of drug usage and cost containment is well
prevalence of drug-resistant organisms may quickly
established in developed countries (125,126). For
increase again (128).
this reason, the establishment of such a commit-
tee is considered important. The premise that any
clinician should be allowed to use any antimicro-

33
WHO GLOBAL STRATEGY FOR CONTAINMENT OF ANTIMICROBIAL RESISTANCE • WHO/CDS/CSR/DRS/2001.2
Formularies regionally with wide input and consensus and
Hospital formularies, or lists of drugs routinely should utilize information from local surveillance
stocked by the hospital pharmacy for inpatient and data whenever possible. Programmes that utilize
outpatient use, guide the parallel processes of an- clinical practice guidelines supported by other in-
timicrobial selection, procurement and supply, and terventions such as education and peer review are
represent a means for decreasing inappropriate more effective than those without such support
antimicrobial prescribing and reducing expendi- (135). In an observational study of one hospital
tures. In conjunction with clinical guidelines, with a computerized prescribing guideline system
formularies encourage the proper use of preferred that encouraged appropriate use of antimicrobials,
drugs within each category of antimicrobial. trend analysis showed that resistance patterns in
Antimicrobial formularies should relate to local selected hospital-acquired infections stabilized over
or regional treatment guidelines and should ide- a seven-year period (136). Another study noted a
ally be based on relative efficacy, cost-effectiveness reduction in one type of resistance when controls
data and local patterns of resistance (129). This, on selected agents were applied, but an increase
however, is difficult in many hospitals. In a USA in resistance to other antimicrobials which were
survey in which a great majority of hospitals not controlled—the so-called “squeezing the bal-
had implemented formularies as a method of loon” effect (137). Nevertheless, treatment guide-
lines are particularly useful in resource-poor
decreasing antimicrobial costs, many noted that
countries where they can be used to streamline
expenditures actually increased—this was usually
treatment protocols and limit the range of
considered to be due to drug resistance (130).
antimicrobials stocked in pharmacies. However,
Although some authors have suggested that a
such treatment guidelines need to be developed
restrictive hospital formulary may actually con-
carefully and their implementation reviewed regu-
tribute to the selection of resistant bacteria by nar-
larly. Their appropriateness is dependent on
rowing and focusing selective pressures, there are
accurate and updated resistance surveillance and
few data to support this view (131). Thus, formu-
clinical outcome data.
laries are useful in avoiding the unnecessary keep-
ing in stock of a range of antimicrobials that
duplicate their spectrum and in reinforcing the Other techniques to control or modify
importance for clinicians to understand an appro- antimicrobial use in hospitals
priate range of antimicrobials well. However, the Several types of innovative tools to guide antimi-
specific effectiveness of a formulary in reducing crobial prescribing and dispensing have been
the emergence of antimicrobial resistance is un- tested; some have been shown to be effective in
clear. changing antimicrobial use in hospital settings.
Antimicrobial order forms have been used with
Cycling of antibiotics mixed success, showing improvement in antimi-
crobial prescribing in some hospitals but not in
The cycling of antimicrobials within a health care others (138,139,140). Automatic review of the use
institution has been suggested as a possible inter- of selected antimicrobials by a consultant or
vention to decrease drug resistance. This technique automatic cessation of antimicrobial administra-
alternates formulary antimicrobials between drug tion after a defined period may also reduce
classes every couple of months and theoretically unnecessary use (46). However, these control
reduces the selective pressure of one antimicro- measures are either labour-intensive or require
bial class (132). However, in a recent review of reasonably sophisticated computerized pharmacy
the topic, there was no evidence that cycling records—both of which are not generally avail-
reduced antimicrobial resistance (133). Cycling able.
may have only a temporary effect on resistance
patterns and ultimately may simply replace one
Integrated interventions
resistance problem with another (134).
Multi-disciplinary and integrated approaches to
reduce antimicrobial use in hospitals have been
Use of clinical practice or treatment guidelines proposed as a solution (105,141,142,143). Hos-
Clinical practice guidelines (80) can improve pital administrators, clinicians, infectious diseases
decision-making and therefore improve patient specialists, infection control practitioners,
care. They should be developed locally or microbiologists, clinical epidemiologists and

34
CHAPTER 3. HOSPITALS
hospital pharmacists all have a role—but coordi- rials and equipment, and internal quality control
nation of their activities is vital. Such activities and external quality assurance procedures, are
include the selection of formulary drugs, the essential. The laboratory should produce and dis-
development of formulary-based guidelines, moni- seminate meaningful local surveillance data both
toring and evaluating drug use, surveillance and with respect to the predominant pathogens/syn-
reporting of bacterial resistance patterns, detec- dromes and their antimicrobial resistance patterns.
tion and appropriate care of patients with resist- The laboratory should work closely with hospital
ant organisms, and promotion and monitoring of infection control personnel, with the hospital
basic infection control practices (143). Interactions therapeutics committee and with providers to
with the pharmaceutical industry must also be con- ensure that appropriate antimicrobials are tested
sidered, including appropriate control of the and reported in order to recognize outbreaks or
access of sales representatives to clinical staff and unusual infections and identify trends in antimi-
monitoring industry-sponsored educational pro- crobial resistance. Software tools such as
grammes for providers. Targeted antimicrobial WHONET are available to facilitate analysis and
control policies in combination with improved data sharing (147). Depending on resources, the
hygiene and education have reduced antimicro- laboratory should also provide specialized testing,
bial resistance in some settings (144,145). How- e.g. molecular typing of bacterial strains, to assist
ever, in one study, prescriber education combined epidemiological investigations.
with hospital antimicrobial control policies led to
decreased antimicrobial costs and improved pre-
Interactions between the hospital
scribing, but only limited change in resistance and the community
(146).
Following discharge from hospital, patients may
still be colonized or infected with resistant bacte-
The microbiology laboratory and ria acquired in hospital. In general, little action is
antimicrobial resistance necessary in such circumstances if the patient is
Delayed or incorrect laboratory diagnostic data healthy and discharged home. However, this is the
frequently result in prolonged empiric antimicro- likely mechanism through which highly resistant
bial therapy (see also Chapters 2 and 5). The hos- hospital-acquired pathogens eventually become
pital microbiology laboratory plays an important widespread in the community. Of greater concern
role in the recognition and surveillance of antimi- is the transfer of such patients to chronic care
crobial resistance, both within the hospital and in facilities where they have been shown to be the
the community. The laboratory must provide high source of strains that subsequently spread through-
quality diagnostic testing to correctly identify in- out the facility. Patients known to be colonized or
fection and accurate antimicrobial susceptibility infected with resistant pathogens upon discharge
testing to guide appropriate treatment. Appropri- to a care facility should generally be identified so
ately trained personnel, adequate supplies, mate- that appropriate precautions can be taken.

35
CHAPTER 4. USE OF ANTIMICROBIALS IN FOOD-PRODUCING ANIMALS
CHAPTER 4

Use of antimicrobials in
food-producing animals

This topic has been the subject of specific consul- — an increased risk for resistant pathogens to
tations which resulted in “WHO global princi- be transferred to humans by direct contact
ples for the containment of antimicrobial with animals or through the consumption
resistance in animals intended for food”*. A com- of contaminated food or water
plete description of all recommendations is con-
— the transfer of resistance genes from ani-
tained in that document and only a summary is
mal to human bacterial flora.
reproduced here.
Increasingly, data suggest that inappropriate
antimicrobial use poses an emerging public health
Recommendations for intervention
risk (148,149,150,151).
Summary Factors associated with the emergence of anti-
4.1 Require obligatory prescriptions for all microbial resistance in food-producing animals
antimicrobials used for disease control in food and the farming industry appear to be similar to
animals. those responsible for such resistance in humans.
Inadequate understanding about and training on
4.2 In the absence of a public health safety evalu- appropriate usage guidelines and the effects of in-
ation, terminate or rapidly phase out the use appropriate antimicrobial use on resistance are
of antimicrobials for growth promotion if they common among farmers, veterinary prescribers
are also used for treatment of humans. and dispensers.
4.3 Create national systems to monitor antimi- There are three modes of antimicrobial use in
crobial usage in food animals. animals—prophylaxis, treatment and growth
promotion. Overall, the largest quantities of anti-
4.4 Introduce pre-licensing safety evaluation of
microbials are used as regular supplements for
antimicrobials with consideration of poten-
prophylaxis or growth promotion in the feed of
tial resistance to human drugs.
animal herds and poultry flocks. This results in
4.5 Monitor resistance to identify emerging health the exposure of a large number of animals, irre-
problems and take timely corrective actions spective of their health, to frequently subthera-
to protect human health. peutic concentrations of antimicrobials (152).
Furthermore, a lack of diagnostic services and their
4.6 Develop guidelines for veterinarians to reduce
perceived high cost means that most therapeutic
overuse and misuse of antimicrobials in food
antimicrobial use in animals is empiric, rather than
animals.
being based on laboratory-proven disease. For
animals and birds that are farmed in large herds
Introduction or flocks, the identification of a few ill individuals
generally results in the entire herd or flock being
Antimicrobial use in food-producing animals may treated to avoid rapid dissemination and stock
affect human health by the presence of drug losses. Clearly this is a different situation to most
residues in foods and particularly by the selection human diseases where decisions are generally made
of resistant bacteria in animals. The consequences about the need for individual therapy, rather than
of such selection include: the empiric treatment of an entire population. In
addition to these issues, veterinarians in some
countries earn as much as 40% or more of their
income by the sale of drugs, so there is a disincen-
* http:// www.who.int/emc/diseases/zoo/who_global_
principles.html tive to limit antimicrobial use (153,154).

37
WHO GLOBAL STRATEGY FOR CONTAINMENT OF ANTIMICROBIAL RESISTANCE • WHO/CDS/CSR/DRS/2001.2
As in human medicine, inefficient and inad- use in animals contributes to an increased pool of
equately enforced regulatory mechanisms regard- vancomycin-resistant enterococci (VRE) (160,
ing antimicrobial supply contribute to excessive 161). However, the extent to which the microbial
and inappropriate drug use. Discrepancies between gene pool in animals contributes to the prevalence
regulatory requirements and prescribing/dispens- of VRE colonization and infection in humans is
ing realities for animal antimicrobial use are often less well defined. VRE cause serious infections,
worse than in human medicine (155). In addi- mainly in hospitalized immunocompromised
tion, antimicrobials that are used as growth pro- patients. Such infections are difficult to cure due
moters are generally not even considered as drugs to the limited number of effective treatment
and are either not licensed or licensed solely as options and are thus associated with increased
feed additives. Poor manufacturing quality assur- morbidity and mortality. There are also concerns
ance in some settings results in the supply of that the genes that cause resistance to vancomy-
sub-standard drugs. Marketing practices of cin may spread from enterococci to other bacte-
antimicrobials for therapeutic, prophylactic or ria, such as Staphylococcus aureus, for which
growth promoter purposes in animals by private vancomycin is one of the drugs of last resort.
industry influence the prescribing patterns and Studies in Denmark have shown that the ban
behaviour of veterinarians, feed producers and of avoparcin in animals has led to a reduction in
farmers. the prevalence of VRE in poultry and pigs
In North America and Europe it is estimated (162,163). Similarly, studies in Germany and the
that about 50% in tonnage of all antimicrobial Netherlands suggest that banning avoparcin has
production is used in food-producing animals and led to a reduction in the prevalence of VRE in
poultry (156). The increased intensity of meat healthy individuals in the community (164,165).
production under crowded industrialized condi- Sweden banned the use of growth promoters in
tions contributes to the increased use of anti- livestock and poultry in 1987 and focused on the
microbials since they are used in subtherapeutic implementation of disease prevention methods
doses as growth promoters, given as prophylaxis that did not involve antimicrobials and on the
for disease prevention and used therapeutically for prudent use of antimicrobials for therapeutic pur-
the treatment of infected animals. In addition, the poses. The subsequent national antimicrobial con-
impact of antimicrobial metabolites and non- sumption has reduced by approximately 50%
metabolized drug in animal sewage that is released (166,167). Furthermore, the prevalence of anti-
into the environment is not clear. microbial resistance in pathogenic bacteria isolated
from animals in Sweden has been maintained at a
low prevalence since 1985 (168).
Use of antimicrobials as growth promoters
Some antimicrobials, particularly those that Use of antimicrobials that affect food-
target Gram-positive bacteria, are associated with borne pathogens such as Salmonella and
an increase in the rate of animal growth when they Campylobacter spp.
are provided in subtherapeutic quantities in stock Non-typhoidal Salmonella spp. and Campylobacter
feed to food-producing animals. The mechanism jejuni are among the most commonly identified
of this effect is uncertain. However, these drugs causes of bacterial diarrhoea in humans. Such
also alter the gut flora of exposed animals such species are generally transmitted to humans via
that they frequently contain bacteria that are re- food or direct contact with animals (169). Data
sistant to the antimicrobial used. When such demonstrate that antimicrobial use in animals
antimicrobial growth promoters belong to a class selects for resistance among non-typhoidal Salmo-
similar to that of antimicrobials used in human nella spp., thus limiting the effective available treat-
medicine, these resistant animal bacteria are ment options (170,171,172). A recent example is
often also resistant, i.e. cross-resistant, to impor- a clone of Salmonella typhimurium DT104 that
tant human use antimicrobials (157). Five growth has become prevalent in many countries includ-
promoters (bacitracin, tylosin, spiramycin, ing the UK, Germany and the USA—it is resist-
virginiamycin and avoparcin [a similar agent to ant to commonly used agents including ampicillin,
vancomycin]) have recently been banned by the tetracycline, streptomycin, chloramphenicol and
European Union due to fears of such cross-resist- sulphonamides (171,173,174). Multi-drug resist-
ance (158,159). ance has likewise been noted in other Salmonella
Scientific data strongly suggest that avoparcin spp. (175).

38
CHAPTER 4. USE OF ANTIMICROBIALS IN FOOD-PRODUCING ANIMALS
Following the introduction of fluoroquinolones in the prevalence of fluoroquinolone-resistant
for use in food-producing animals, the emergence Campylobacter jejuni isolated in live poultry,
of Salmonella serotypes with reduced susceptibil- poultry meat and from infected humans (180,181,
ity to fluoroquinolones has been observed in 182). Prior to fluoroquinolone use in poultry, no
countries such as France, Germany, Ireland, the resistant strains were reported in individuals with-
Netherlands, the Russian Federation, Spain and out previous exposure to these agents (178,183).
the UK (176,177,178). Little has been docu- Because of their broad antibacterial spectrum,
mented about the impact of this resistance on fluoroquinolones are often used for empiric treat-
human health to date, but there is concern about ment of gastrointestinal infections in severely ill
the potential human health consequences. This or immunocompromised patients. Fluoro-
has been substantiated by a recent outbreak of quinolone resistance among Campylobacter spp.
quinolone-resistant S. typhimurium DT104 result- is associated with a higher rate of clinical treat-
ing in treatment failures in hospitalized patients ment failure than for susceptible strains when
in Denmark (179). fluoroquinolones are used for treatment of disease
The introduction of fluoroquinolone use in (184,185,186). A recent review by APUA (187)
poultry has been associated with a dramatic rise provides further material on this topic.

39
CHAPTER 5. NATIONAL GOVERNMENTS AND HEALTH SYSTEMS
CHAPTER 5

National governments and health systems

Recommendations for intervention 5.5 Ensure that only antimicrobials meeting in-
Advocacy and intersectoral action ternational standards of quality, safety and
efficacy are granted marketing authorization.
5.1 Make the containment of antimicrobial
resistance a national priority. 5.6 Introduce legal requirements for manufactur-
ers to collect and report data on antimicro-
— Create a national intersectoral task force bial distribution (including import/export).
(membership to include health care pro-
fessionals, veterinarians, agriculturalists, 5.7 Create economic incentives for the appropri-
pharmaceutical manufacturers, govern- ate use of antimicrobials.
ment, media representatives, consumers
Policies and guidelines
and other interested parties) to raise
awareness about antimicrobial resistance, 5.8 Establish and maintain updated national
organize data collection and oversee Standard Treatment Guidelines (STGs) and
local task forces. For practical purposes encourage their implementation.
such a task force may need to be a gov- 5.9 Establish an Essential Drugs List (EDL) con-
ernment task force which receives input sistent with the national STGs and ensure
from multiple sectors. the accessibility and quality of these drugs.
— Allocate resources to promote the imple- 5.10 Enhance immunization coverage and other
mentation of interventions to contain disease preventive measures, thereby reduc-
resistance. These interventions should ing the need for antimicrobials.
include the appropriate utilization of
antimicrobial drugs, the control and Education
prevention of infection, and research
5.11 Maximize and maintain the effectiveness of
activities.
the EDL and STGs by conducting appro-
— Develop indicators to monitor and evalu- priate undergraduate and postgraduate
ate the impact of the antimicrobial education programmes of health care pro-
resistance containment strategy. fessionals on the importance of appropriate
antimicrobial use and containment of anti-
Regulations microbial resistance.
5.2 Establish an effective registration scheme for 5.12 Ensure that prescribers have access to approved
dispensing outlets. prescribing literature on individual drugs.
5.3 Limit the availability of antimicrobials to
prescription-only status, except in special cir- Surveillance of resistance, antimicrobial usage
cumstances when they may be dispensed on and disease burden
the advice of a trained health care professional. 5.13 Designate or develop reference microbiol-
5.4 Link prescription-only status to regulations ogy laboratory facilities to coordinate effec-
regarding the sale, supply, dispensing and tive epidemiologically sound surveillance of
allowable promotional activities of antimicro- antimicrobial resistance among common
bial agents; institute mechanisms to facilitate pathogens in the community, hospitals and
compliance by practitioners and systems to other health care facilities. The standard of
monitor compliance. these laboratory facilities should be at least
at the level of recommendation 3.6.

41
WHO GLOBAL STRATEGY FOR CONTAINMENT OF ANTIMICROBIAL RESISTANCE • WHO/CDS/CSR/DRS/2001.2
5.14 Adapt and apply WHO model systems for Government legislation—drug licensing
antimicrobial resistance surveillance and Marketing authorization
ensure data flow to the national intersectoral
task force, to authorities responsible for the Many countries have legislation that requires all
national STGs and drug policy, and to pre- medicinal products to undergo licensure before
scribers. being placed on the market. Marketing authori-
zation usually follows a detailed assessment of data
5.15 Establish systems for monitoring antimicro- provided by the applicant, generally by a desig-
bial use in hospitals and the community, and nated government department and sometimes
link these findings to resistance and disease with input from an expert advisory group(s). Some
surveillance data. countries are willing to license new medicines
5.16 Establish surveillance for key infectious dis- based on their prior approval in other countries,
eases and syndromes according to country such as the USA or EU. Whatever the process,
priorities, and link this information to other the fundamental requirement is that the data
surveillance data. should support the quality, safety and efficacy of
the product (188,189). The use of antimicrobials
that do not meet appropriate standards in each of
Introduction these three areas has implications for human health
Placing antimicrobial resistance high on the na- and for antimicrobial resistance.
tional agenda should be a priority in tackling the
problem of resistance. National governments and Quality
health care systems can have considerable impact
on limiting the emergence and development of As with all medicinal products, control of the
antimicrobial resistance through the introduction quality of antimicrobial agents is vital for the de-
of legislation and policies concerning the devel- livery of accurate dosage units to patients—doses
that have been shown to be safe and effective in
opment, licensing, distribution and sale of anti-
clinical trials (188,189,190). Antimicrobial agents
microbial agents. Health and pharmaceutical
containing less than the stated dose may produce
regulations shape the way antimicrobials are used.
suboptimal levels of circulating drug, which may
Key regulatory frameworks include professional
result in both therapeutic failure and selection of
licensing, the ability to prescribe and dispense
drug-resistant strains. Similar problems may arise
medicines, drug registration, product quality, pric-
as a result of counterfeit products, which com-
ing and movement of drugs in the supply system.
monly contain little or none of the active substance
Although pharmaceutical regulations represent a
stated on the label and may even contain entirely
powerful tool, implementing them to influence
different active ingredients. The Counterfeit
patterns of antimicrobial use can be a two-edged
Intelligence Bureau estimated that, in 1991, 5%
sword, achieving both intended and unintended
of all the world’s trade was in counterfeit goods,
effects. For example, active enforcement of regu-
with this percentage likely to be higher for phar-
lations regarding the sale of antimicrobials with-
maceuticals since they are easily transportable
out prescription in pharmacies and drug shops may
(191). Excessive drug content may lead to con-
reduce unnecessary use while at the same time
centrations in the body associated with certain
limiting access to appropriate therapy, especially
adverse events. Unnecessarily high concentrations
among the poor. The unintended effects of pro-
may also lead to a marked disruption of the nor-
posed regulations should be carefully considered
mal flora and an increased risk of superinfections
before and monitored during their implementa-
such as fungal disease and C. difficile enterocolitis.
tion.
Government-initiated inspection of drug
National governments also have the responsi-
manufacturing plants for adherence to Good
bility for coordinating surveillance networks and
Manufacturing Practice (GMP) with certification
for directing educational efforts to improve
for defined time periods, adherence to the prod-
understanding about appropriate antimicrobial
uct specifications agreed upon at the time of
use.
licensure, and the elimination of unauthorized
medicines from the market are essential. Strict
controls limiting drug importation and exporta-
tion to those products and manufacturers that have
been inspected and approved can serve to reduce

42
CHAPTER 5. NATIONAL GOVERNMENTS AND HEALTH SYSTEMS
the risks posed by substandard and counterfeit Prescribing information
medicines. Countries that carry out spot checks Wherever there are formal procedures for drug
and drug analyses are able to make a major con- licensure, the content of the prescribing informa-
tribution to reducing the production of poor tion is subject to approval by the licensing authori-
quality and counterfeit products. ties. Requirements for international alignment on
the essential content of the prescribing informa-
Safety and efficacy tion and on the reporting of safety data have led
to the development of core datasheets by many
The scope and quality of data presented to sup-
pharmaceutical companies (194,195). These
port the safety and efficacy of new drugs are
describe the minimal prescribing information,
determined mainly by the requirements of the US
including contraindications, warnings and poten-
Food and Drug Administration (FDA), the Euro-
tial adverse reactions, which should be available
pean Commission, and the Japanese Ministry of
to users in all countries where the product is mar-
Health, Labour and Welfare (MHLW) (188,189,
keted. However, it may not be feasible for all
192). The individual regulations issued by these
companies, especially those that are large and mul-
bodies, together with the activities of the Interna-
tinational, to regularly audit compliance with the
tional Conference on Harmonisation (193), have
use of core datasheets in all regions or to require
greatly influenced the content and conduct of pre-
that national or regional offices fully adopt stated
clinical and clinical development programmes for
corporate standards. Also, in countries where there
pharmaceuticals. Dossiers meeting these inter-
are inadequate regulations to ensure the availabil-
national standards are generally acceptable world-
ity of prescribing information to prescribers and
wide, although there may be additional local
users, health care professionals may have little or
stipulations. In this way, all countries may benefit
no access to independently-assessed material re-
from high quality development programmes that
garding antimicrobial agents (see also Chapters 2
better identify the safety and efficacy of new drugs.
and 7).
In one sense, the emergence of resistance associ- Failure to specify precisely in the prescribing
ated with the use of a particular antimicrobial information the types of infections for which safety
could be viewed as an adverse event. However, and efficacy have been demonstrated in clinical
current regulatory and licensure bodies do not trials may serve to encourage antimicrobial use for
regard the emergence of resistance in this man- conditions that have not been studied. An exam-
ner. ple is the use of the term “lower respiratory infec-
Countries that do not have systems for the tions” instead of specifying the types of pneumonia
adequate assessment of safety and efficacy before or bronchitis that were studied. Thus, without
and after drug licensure face an increased risk of careful attention to detail and to translation, even
exposure to drugs of inferior efficacy and unac- the approved prescribing information may inad-
ceptable toxicity, as well as a potentially higher vertently encourage inappropriate antimicrobial
market penetration of counterfeit drugs. The use.
establishment of Assessment Report Sharing The product literature usually reflects the dos-
Schemes has facilitated assessment of the safety age regimens shown to be efficacious in clinical
and efficacy of antimicrobial agents by resource- trials for each indication. Identification of opti-
poor countries. Participating countries are able to mal antimicrobial treatment regimens for various
request detailed reports of pharmaceutical, pre- diseases is important to ensure that the drug is
clinical and clinical data that have been prepared given in an appropriate dose and for an appropri-
by drug regulatory authorities in other countries. ate duration to maximize the likelihood of cure,
The Product Evaluation Report (PER) network while minimizing the risk of toxicity. Low dose
and the arrangements made by the European regimens may be associated with less toxicity, but
Agency for the Evaluation of Medicinal Products may result in insufficient drug concentrations at
(EMEA) are examples of schemes which allow the site of infection to effect bacterial eradication
countries access to information to assist in mak- and may therefore encourage the development of
ing licensing decisions. In addition, regional as- resistance among target pathogens. In contrast,
sociations of regulatory bodies, e.g. AFDRAN in higher dose regimens may result in greater effects
Africa, have contributed to the application of simi- on the host’s normal flora increasing the likeli-
lar standards and requirements for drug approvals hood of superinfections, including those caused
in many countries. by highly resistant nosocomial pathogens. How-

43
WHO GLOBAL STRATEGY FOR CONTAINMENT OF ANTIMICROBIAL RESISTANCE • WHO/CDS/CSR/DRS/2001.2
ever, clinical trials to support antimicrobial drug Nairobi, it was noted that 64% of chemists sold
approval are almost always designed to show antimicrobials without physicians’ prescriptions
equivalence to a licensed comparative agent. and most sold incomplete treatment courses at the
Therefore there is a tendency to use perhaps request of the patient (207). In a study of a rural
unnecessarily high dose or long duration regimens village in Bangladesh, 95% of all medications con-
so as to avoid any risk of treatment failure (196, sumed were obtained from pharmacies with only
197,198). Companies are often reluctant to ex- 8% having been prescribed by graduate physicians;
plore a variety of dosage and treatment regimens one-third of these medications were antimicrobials
in clinical trials with a new drug because of the (208). Poor enforcement of prescription-only regu-
study costs involved and the risk of failing to meet lations is almost universally associated with inap-
the specified regulatory requirement (199). Dose propriate antimicrobial usage.
regimens in clinical trials are often chosen by com- Although the cost of antimicrobial agents with-
paring the pharmacokinetics of the drug in man out prescription is generally carried by the patient,
with the in vitro susceptibilities of the main target in some regions this may actually be less expen-
pathogens. Increasingly, the pharmacokinetic and sive than the combined costs of a time-consum-
pharmacodynamic characteristics of new antimi- ing visit to a distant and/or very busy health care
crobial agents are being used in pre-clinical facility and the physician’s consulting fee. There-
studies to better predict the optimal clinical dos- fore, depending on the structure and funding of
ing regimens in man (200,201,202). While this the national health care system, restricting anti-
approach does not replace clinical trials, the phar- microbial agents to prescription-only may
maceutical industry and regulatory authorities actually limit the access of many patients to these
have both recognized that this may also have drugs, even when they are really needed. On the
benefits in terms of reducing the risk of selecting other hand, requiring a prescription for access to
for drug-resistant organisms. antimicrobial agents provides an opportunity to
dissuade patients from unnecessary antimicrobial
therapy and hopefully results in a trained health
Government legislation—control of drug care worker selecting the drug and the treatment
supply, distribution and sales
regimen. This potential point of intervention
Some countries are unable to control the supply, should help reduce inappropriate antimicrobial
distribution and sale of medicines. In many re- usage, especially if accompanied by an education
gions there is minimal control over public access programme on the appropriate use of antimicro-
to antimicrobials and these can be purchased over bial agents (see Chapter 2).
the counter without prescription (34,203). There With or without implementation of prescrip-
are also marked international differences in the tion-only access to antimicrobial agents, legisla-
types of retail outlets that provide access to pre- tion that restricts the sale of antimicrobials to
scription-only and non-prescription drugs, as well registered outlets would allow local policing and
as whether these outlets require government reg- prevention of over-the-counter non-prescription
istration. Where there is adequate legislation sales. Ideally, such registered outlets should be
regarding the licensure of medicinal products, a staffed by personnel with at least a basic knowl-
legal classification system generally determines the edge of antimicrobials. Legislation that compels
mode of sale, supply and dispensing. In such coun- registered outlets to keep records of the sources of
tries, antimicrobial agents are almost uniformly drugs purchased and quantities sold would allow
prescription-only medicines (POM), with dispens- the auditing of antimicrobial sales and possibly of
ing restricted to registered outlets and by suitably usage data. Such surveillance may result in greater
qualified personnel (204). In reality, however, the restriction of the sales of counterfeit and substand-
degree of drug law enforcement and the penalties ard medicines. However, in regions where prescrib-
imposed for infringements vary enormously ers earn a considerable portion of their income
between countries. For example, all systemic either by directly dispensing antimicrobials or via
antibacterial agents are legally subject to prescrip- subsequent pharmacy sales, such legislation is
tion control in the EU, yet they can be purchased likely to be less effective. These circumstances
over the counter in pharmacies in several EU mem- provide a disincentive to appropriate antimicro-
ber states (205). Antimicrobials can be purchased bial prescribing and prescribers are more likely to
without prescription in many resource-poor coun- recommend antimicrobial use, particularly the
tries (59,129,206). In a study of chemist shops in more expensive agents, regardless of whether

44
CHAPTER 5. NATIONAL GOVERNMENTS AND HEALTH SYSTEMS
cheaper drugs may be just as appropriate (see also Surveillance of resistance and antimicrobial use
Chapter 2). Surveillance of both antimicrobial resistance and
antimicrobial use are fundamental to the effective
Government legislation—inspection implementation of any strategy for the contain-
and enforcement ment of antimicrobial resistance, as a means to
monitor the efficacy of various interventions.
The existence of appropriate legislation regarding
However, designing and implementing compre-
the manufacture, licensure, sale, supply and dis-
hensive surveillance systems that are practical, cost-
pensing of antimicrobial agents cannot improve
effective and interlink with the national healthcare
the quality and appropriate use of these drugs
system is a challenge. It is likely that in many
unless it is enforced. Individual countries may not
resource-poor countries, laboratory facilities and
have the financial or human resources needed to
information networks will require considerable
support policing activities by suitably qualified
strengthening before reliable surveillance of resist-
personnel. There may be reluctance on the part of
ance can be undertaken.
governments to take action because the introduc-
Epidemiologically sound surveillance of resist-
tion of restrictions could prove unpopular with
ance in key pathogens, using standardized micro-
patients, physicians and the pharmaceutical indus-
biological methods, may be developed on the basis
try. Increasing international recognition of inspec-
of an existing laboratory surveillance system for
tions of manufacturing plants by teams from other
antimicrobial resistance and routine diagnostic
countries has relieved the burden on some gov-
microbiology (see Part A, Background). To assist
ernments and facilitated the quality control of in this aim, WHO is developing “Surveillance
medicines and adherence to Good Manufactur- standards for antimicrobial resistance” which pro-
ing Practice (GMP), Good Laboratory Practice pose practical epidemiological methods for
(GLP) and Good Clinical Practice (GCP). The several infections and key pathogens (209). Where
possibility of expanding these international coop- possible, such surveillance should be integrated
erative efforts by using suitably qualified staff from with other national and hospital laboratory serv-
non-governmental organizations (NGOs) to aid ices to maximize efficiency and ensure surveillance
policing efforts in other areas of drug law compli- of clinically relevant isolates (see Chapter 3).
ance may be worthy of serious consideration by Measurement of antimicrobial usage could be
some countries (see also Chapter 8). approached through the registration of outlets that
dispense antimicrobials, requiring them to main-
Health care systems and drug policies tain accurate records of antimicrobial supply and
sales. Incomplete patient adherence to treatment
Health care systems
protocols means that antimicrobial dispensing data
The organization and funding of health care sys- will not necessarily be the same as antimicrobial
tems varies between countries, with a mixture of consumption, but it is likely to be the most
public- and privately-funded health care facilities accurate achievable surrogate available. Targeted
and diagnostic laboratories being common. The research to measure the correlation between the
structure and organization of these systems can quantity of antimicrobials dispensed and the quan-
be an important factor in determining the reli- tity consumed could be used to adjust national
ability and practicality of data collection regard- dispensing data, providing a more accurate assess-
ing antimicrobial use, surveillance of antimicrobial ment of antimicrobial consumption. Establishing
resistance and the impact of resistance on clinical surveillance systems of antimicrobial usage and
outcomes. In addition, the system may have a di- control of drug supply and dispensing outlets will
rect influence on undergraduate medical curricula, require a major commitment from national gov-
on the existence and maintenance of registration ernments in countries which do not currently have
systems for all health care professionals, and on effective prescription-only regulations for anti-
the attention paid to their continuing professional microbials. Implementation of an integrated sur-
education and accreditation. Whether or not veillance system for antimicrobial resistance and
antimicrobials are prescription-only, undergradu- antimicrobial usage will require national govern-
ate and postgraduate medical and pharmacist edu- ments to re-assess many regulatory aspects of their
cation concerning appropriate antimicrobial use health care system, including legislation related
is vital (see Chapter 2), as is the need for evidence- to drug licensure (including quality, safety and
based prescribing information. efficacy) and drug supply, distribution and sales.

45
WHO GLOBAL STRATEGY FOR CONTAINMENT OF ANTIMICROBIAL RESISTANCE • WHO/CDS/CSR/DRS/2001.2
Essential Drugs Lists and policies regions without such a programme. Thus, such
In 1977 the first WHO Model List of Essential programmes appear to improve access to essential
Drugs was developed to promote the availability drugs, especially when they are supported by edu-
of a selected number of drugs, including anti- cational programmes and follow-up (83,117).
microbials, and their rational use. The Model List
has been revised regularly and serves as a guide for Establishing national treatment guidelines
countries in determining their national drug poli-
Evidence-based national treatment guidelines en-
cies. At present over 120 countries have imple-
courage appropriate antimicrobial prescribing.
mented an Essential Drugs List. A retrospective
Using local laboratory and clinical surveillance
study of prescribing practices in Ethiopia found a
data on antimicrobial resistance, these guidelines
significant decrease in the prescribing of non-
can be appropriately modified for community and
essential drugs after the introduction of an Essen-
hospital use in various regions, but should be up-
tial Drugs List (210). Studies have demonstrated
dated regularly. The use of such guidelines is most
that in those areas in which an Essential Drugs
effective when combined with supportive inter-
Programme is in operation, significantly more
ventions such as educational training and super-
essential drugs are available, significantly fewer
vision programmes (83,211).
injections and antimicrobials are utilized, and drug
stocks last about three times longer than in

46
CHAPTER 6. DRUG AND VACCINE DEVELOPMENT
CHAPTER 6

Drug and vaccine development

Recommendations for intervention drug and vaccine research is now a crucial issue
6.1 Encourage cooperation between industry, for all nations given the emergence of antimicro-
government bodies and academic institutions bial resistance among human pathogens.
in the search for new drugs and vaccines.
6.2 Encourage drug development programmes New drug and vaccine development
which seek to optimize treatment regimens The fact that there are currently several novel
with regard to safety, efficacy and the risk of antimicrobial agents and vaccines in clinical trials
selecting resistant organisms. reflects the awareness of the industry of the prob-
6.3 Provide incentives for industry to invest in lems of antimicrobial resistance and the enormous
the research and development of new investment by some companies in anti-infective
antimicrobials. drug development. At the same time, however,
there are concerns within the industry that efforts
6.4 Consider establishing or utilizing fast-track to encourage the more appropriate use of anti-
marketing authorization for safe new agents. microbials may have a negative impact on sales.
6.5 Consider using an orphan drug scheme where This concern may potentially discourage compa-
available and applicable. nies from either initiating or maintaining invest-
ment in antimicrobial research and development.
6.6 Make available time-limited exclusivity for
An overall drop in the antimicrobial-generated
new formulations and/or indications for use
revenues of pharmaceutical companies may also
of antimicrobials.
influence the quantity of antimicrobial agents and
6.7 Align intellectual property rights to provide vaccines that they donate, or provide at reduced
suitable patent protection for new antimicro- cost, to some regions of the world.
bial agents and vaccines. Schemes that encourage investment in antimi-
6.8 Seek innovative partnerships with the phar- crobial and vaccine research must therefore
maceutical industry to improve access to recognize the need for companies to recoup their
newer essential drugs. development costs as well as make a profit from
post-licensing sales. A range of incentives to the
industry, including both push and pull mecha-
Introduction nisms, are currently under discussion (212). Some
The pharmaceutical industry is the predominant countries, such as Australia, have devised provi-
source of new antimicrobial agents and new dis- sions by which companies which conduct research
ease prevention modalities, including novel aimed at identifying new therapies and which
vaccines and immunomodulating therapies. It is perform some sections of the development pro-
vital that there are incentives for companies to gramme in the home country can benefit from
invest resources into research and development in tax reductions and incentive payments. This
these areas even though the development of other approach also attracts some companies to estab-
types of medicinal products may ultimately be lish research facilities in supportive countries,
more profitable. Encouraging research into which may have employment and other benefits.
vaccines and antimicrobial agents that will pre- Drug discovery may also be stimulated by co-
dominantly be used in low-resource countries operative research agreements between companies
poses particular challenges given the need for phar- and academic institutions. These agreements can
maceutical companies to make a profit. However, stimulate basic science research and the sharing of
the continuation and expansion of anti-infective knowledge which may speed up the identification

47
WHO GLOBAL STRATEGY FOR CONTAINMENT OF ANTIMICROBIAL RESISTANCE • WHO/CDS/CSR/DRS/2001.2
of promising compounds or vaccines. This hepatitis C and HIV could likewise have enor-
approach may potentially reduce overall costs by mous clinical impact.
reducing the duplication of research activities (see
Chapter 8). Public-private partnerships are increas- Licensure and patent protection
ingly being exploited for speeding up drug
discovery and development and addressing unmet To hasten the licensure of some new products, fast-
medical needs where the market opportunities are track evaluation of innovative medicines is offered
less attractive (213). by some licensing authorities (188,224), allowing
truly innovative products to reach the public
domain as early as possible. Such schemes benefit
Vaccines both the companies and the community—
Vaccines potentially provide a major means of lim- although careful post-licensure surveillance of
iting the clinical impact of emerging antimicro- adverse effects is vital. Some products may be
bial resistance. Pneumococcal and Haemophilus considered clinically useful but of limited com-
influenzae type b (Hib) vaccines have had a mercial value due to infrequent disease occur-
dramatic effect in reducing the incidence of clini- rence—for these, some countries provide special
cal disease in some age groups and regions (214, licensure under an orphan drug scheme which has
215,216,217). Recent studies of a nonavalent variable eligibility requirements.
pneumococcal conjugate vaccine demonstrated a The safeguarding of intellectual property rights
significant reduction in the carriage of penicillin- is a major concern to the pharmaceutical indus-
resistant and cotrimoxazole-resistant strains of try. There may be opportunities to encourage
S. pneumoniae in children nine months after research by furthering international agreements
vaccination, compared to controls (214). Thus, and cooperation on innovative new approaches to
by reducing the incidence of disease and carriage patents and time-limited exclusivity arrangements.
of resistant strains, pneumococcal vaccination may Time-limited exclusivities on new clinically use-
limit the impact of antimicrobial resistance. When ful formulations and/or additional indications for
use on some current agents might serve to stimu-
the information is available, knowledge of a
late the additional pharmaceutical and clinical
patient’s vaccination status for pneumococcal, Hib
studies which are needed to support licensure for
and other diseases may aid the differential diag-
these additional indications.
nosis if the patient presents with an acute illness
and thereby allow narrower-spectrum agents to be
employed for empiric therapy. Clinical development programmes
Vaccines have also proven effective for some Clinical development programmes are designed
diarrhoeal diseases and enteric fever. A number of to undertake trials which will support drug regis-
typhoid vaccine preparations are now available, tration. These programmes offer possibilities to
but their use has previously been limited mainly investigate not only the most effective treatment
to travellers to endemic areas. With the new oral regimens but also those which are least likely to
preparations, wider use may now be more feasible result in the emergence of antimicrobial resistance.
once they can be produced at reasonable cost. However, these pre-registration clinical trials rarely
Given recent outbreaks of ciprofloxacin-resistant assess the degree to which in vitro susceptibility
typhoid fever, vaccination has been suggested as data correlate with the in vivo clinical outcomes
an adjunct to sanitation measures in some regions of infected patients receiving treatment. Although
(218,219,220,221). Vaccines for other diarrhoeal these correlations are of vital clinical importance,
diseases such as shigellosis, cholera, E. coli ETEC such trials can be difficult to perform. In most
and rotavirus are also under trial (218). pre-registration clinical antimicrobial trials, the
As the medical treatment of HIV, hepatitis B number of treatment failures is generally too few
and C becomes more widespread, antiviral resist- to allow such assessments and, in any case, the
ance will become a major limiting factor. Child- primary goal of these studies is to assess equiva-
hood vaccination against hepatitis B, either lence of efficacy or drug toxicity, not the correla-
universal or targeted to high risk groups depend- tion between in vitro and in vivo outcomes. In
ing on the prevalence of hepatitis B in the popu- addition, a number of design features of licensure
lation, is a very cost-effective means of controlling studies make such correlations even more diffi-
the disease and avoiding the problems of resist- cult. Firstly, some protocols require that enrolled
ance (109,222,223). Effective vaccines against patients found to be infected with pathogens that

48
CHAPTER 6. DRUG AND VACCINE DEVELOPMENT
are resistant in vitro to one of the trial drugs should Microbiological and pharmacological issues
be withdrawn from the study. However, others A number of important microbiological and phar-
allow such patients to continue receiving trial macological features of antimicrobials appear to
therapy if they are doing well, despite in vitro influence their likelihood of selecting and promot-
resistance. Such design issues have an important ing resistant strains (51,226). Pharmacodynamic
impact on the ability to accurately analyse corre- and pharmacokinetic parameters can be used to
lation data. Furthermore, the site of infection may help identify the optimal dose and dosing inter-
influence the level of antimicrobial penetration vals for each antimicrobial (202). The parameters
and therefore the likely concentrations of active most appropriate in terms of encouraging the
drug available under routine dosing conditions, emergence of resistance have been investigated and
e.g. drug concentrations in cerebrospinal fluid are debated extensively (132,227,228). In addition,
generally lower than those achievable in serum. the use of antimicrobials in combinations has been
Therefore, in vitro definitions of resistance will suggested for some infections, since a reduced
depend on potentially achievable drug concentra- incidence of resistance has been noted with com-
tions in vivo, meaning that the MIC breakpoints bination therapy (229,230).
for individual pathogens may need to vary depend-
ing on the site of infection.
Since clinical trials with antimicrobials are Cost-effectiveness
almost exclusively designed to demonstrate equiva- Cost-effectiveness studies are increasingly becom-
lence to an approved comparative agent, this ing a major component of clinical development
means that results cannot be used as a basis for programmes. While these are not required for
recommending one treatment over another. Thus, licensure, they may be needed in some countries
these studies generally do not provide clear evi- for negotiations on drug supply contracts. While
dence on which to base guidelines for the best companies may have some cost-effectiveness data
choice of antimicrobial or the optimal mode of available, few release such data to the public. Many
management of a particular infection. In addition, published cost-effectiveness studies are at risk of
clinical drug trials have not been designed to bias in favour of the new agent, since few studies
determine the most appropriate duration of anti- of older agents, that are often no longer patent-
microbial therapy. Many scientists and clinicians protected, are undertaken due to insufficient
believe that shorter treatment courses for many research funding support. Furthermore, studies
infections may be as effective as longer courses focus neither on the cost of resistance nor on the
(225). Potential benefits of shorter course thera- clinical impact of resistance and there is a need
pies are to decrease disruption of the normal flora for new approaches to incorporate such evalua-
and the selective pressure of antimicrobials favour- tions into cost-effectiveness studies (8). Thus,
ing drug-resistant microorganisms. Shorter current clinical development programmes rarely
durations of therapy are also likely to encourage support decision-making regarding the cost-effec-
patient adherence (see Chapter 1). It should be tiveness or optimal dose of various antimicrobials.
noted that, at the present time, relatively few clini- However, such programmes may provide some
cal antimicrobial studies are conducted on paedi- unique opportunities to gain more useful infor-
atric populations and that this may be an area for mation in future if innovative modifications are
greater attention in the future. made to current trial designs.

49
CHAPTER 7. PHARMACEUTICAL PROMOTION
CHAPTER 7

Pharmaceutical promotion

Recommendations for intervention macists, dentists, nurses and the general commu-
7.1 Introduce requirements for pharmaceutical nity. The close relationships between drug pro-
companies to comply with national or inter- motion, prescribing habits and drug sales have
national codes of practice on promotional been demonstrated in several studies (34,231).
activities. Since drug promotion increases usage, it may be
assumed that it can contribute to the prevalence
7.2 Ensure that national or international codes of antimicrobial resistance, particularly if it results
of practice cover direct-to-consumer advertis- in increased inappropriate use of antimicrobial
ing, including advertising on the Internet. agents.
7.3 Institute systems for monitoring compliance
with legislation on promotional activities. Promotional literature and prescribing information
7.4 Identify and eliminate economic incentives A number of studies have shown that advertise-
that encourage inappropriate antimicrobial ments and other promotional literature distrib-
use. uted by companies at conferences and symposia
7.5 Make prescribers aware that promotion in are major influential sources of information for
accordance with the datasheet may not nec- health professionals (231,232). Indeed, the con-
essarily constitute appropriate antimicrobial tent of advertisements and literature provided by
use. companies may be the only readily accessible
sources of information on antimicrobial agents in
some countries. In the absence of legislation or its
Introduction enforcement for promotional materials to reflect
National governments have an important legisla- approved prescribing information, companies may
tive role in ensuring the appropriate manufacture, present to potential prescribers, suppliers and
licensure and sale of antimicrobials (see Chapter users a very selective and biased view of the effi-
5) and also an important responsibility in ensur- cacy and safety of a drug. It has been suggested
ing that these drugs are promoted in a fair and that physicians may not even be aware of these
accurate manner. Government controls on drug influences. Avorn et al. (232) found that most pre-
promotional activities and compliance of the phar- scribers believed that drug advertisements and
maceutical industry with both legislation and pharmaceutical representatives played a role of
agreed codes of practice are important factors if minimal importance in influencing prescribing
appropriate antimicrobial use is to be encouraged. patterns whereas academic sources of information
were very important yet the opposite appeared to
be true. This finding was supported by a study of
The power of promotional activities
prescribing habits of physicians in Peru (231). The
Promotional activites include drug advertisements study concluded that advertising materials distrib-
in the media and over the Internet, personal con- uted by pharmaceutical companies appeared to be
tacts during visits from company representatives, a key source of information for prescribers,
sponsored symposia and guest lectures or lecture despite claims by more than two-thirds of the phy-
tours funded by companies, and other induce- sicians surveyed that their primary source of drug
ments to prescribe a particular product or brand. information came from medical literature. A re-
The target audience for promotional activities view of the literature on the interactions between
depends on the product and the local regulatory physicians and the pharmaceutical industry con-
environment, but generally includes doctors, phar- cluded that there was strong evidence that these

51
WHO GLOBAL STRATEGY FOR CONTAINMENT OF ANTIMICROBIAL RESISTANCE • WHO/CDS/CSR/DRS/2001.2
interactions influence prescribing behaviour (233). Patients
Thus, pharmaceutical promotional material that Health care professionals in all countries, includ-
contains misinformation may ultimately encour- ing those subject to prescription control, often feel
age inappropriate antimicrobial use and the pressured by patients to prescribe antimicrobials
emergence of resistance.
for minor infections which do not need specific
Promotional materials must not only reflect
therapy (see Chapter 2). Direct-to-public adver-
approved prescribing information correctly but
tisements in countries with prescription-only
must also be accurate, comprehensive and up to
restrictions on antimicrobial agents may enhance
date. Particular difficulties may be encountered
the pressures on health care professionals to pre-
with older antimicrobials for which the initially
scribe when their clinical judgement suggests that
licensed indications may now be considered
specific therapy is unnecessary. In addition,
excessively broad or vague, e.g. upper respiratory
advertising on the Internet is gaining market pen-
infections, and may no longer reflect current think-
etration yet is difficult to control with legislation
ing on the optimal evidence-based management
due to poor enforceability. To counter this prob-
of certain infections. Although pharmaceutical
lem, education campaigns directed at health care
companies may apply to update sections of the
professionals and the general public are underway
prescribing information, they are unlikely to do
in some countries where antimicrobial agents are
so voluntarily if there is a risk of a negative impact
available by prescription only. The aim of such a
on sales. Licensing authorities may require that
campaign in the UK, ongoing in 2000, was to
the prescribing information be updated or modi-
inform all parties about those infections least likely
fied but are unlikely to do so without strong evi-
to require antimicrobial treatment—thereby
dence to support the changes, due to the possibility
reducing patient expectation of the need for an
of a legal challenge from companies. Furthermore,
antimicrobial agent. Data on the effects of such
the fact that many older agents are no longer pat-
efforts are awaited.
ent-protected means that license holders may not
The effects of direct-to-public promotion on
consider that the drug’s market value warrants such
total and specific antimicrobial usage are likely to
applications and effort.
be much greater in countries where these agents
are available without prescription. In these circum-
Prescribers stances, even promotion in accordance with the
Promotion of products to health professionals in- prescribing information is likely to result in un-
forms prescribers about the range of drugs avail- necessary antimicrobial use as purchasers are less
able and alerts them to the availability of new able to fully appreciate the information provided
agents. Inherently, pharmaceutical marketing re- and to weigh the possible risks and benefits.
sults in the highlighting of potential benefits and Inappropriate antimicrobial use as a result of over-
advantages of new agents over existing agents to the-counter availability may therefore be greatly
the extent allowable. Under these circumstances exacerbated by direct-to-public advertising.
it is often difficult for prescribers to identify the
most appropriate role of the new agents within Sales
the context of existing protocols. Promotional
materials often emphasize simple messages in pref- Pharmaceutical promotion directed towards health
erence to more complex ones, not infrequently care professionals who sell antimicrobials may re-
resulting in over-prescribing. sult in a conflict of interest. The desire to profit
It is also difficult to regulate the provision of from making the sale and/or to favour a particu-
inducements such as meals, event tickets, and lar company’s product in expectation of rewards
travel to conferences. These perks may serve as may override clinical judgement. In this manner,
rewards for using a company’s products and as the decision regarding the necessity for treatment
enticements to prescribe newly-introduced drugs and the choice of the most suitable agent are less
(234,235). This may also encourage prescribers likely to reflect appropriate clinical management.
to prescribe using brand names rather than Sales of antimicrobial drugs through outlets not
generic names, which may markedly increase staffed by health care professionals are likely to be
specific company sales in those countries which driven predominantly by profit margins with only
do not allow pharmacists to substitute between limited potential for control of antimicrobial
brands of the same active substance when dispens- usage.
ing prescriptions (see Chapter 2).

52
CHAPTER 7. PHARMACEUTICAL PROMOTION
Control of drug promotion appropriate promotional activities that have been
Legislation and enforcement drawn up by national and international associa-
tions of pharmaceutical companies (239,240,241).
Where licensing, supply and sales legislation are Unfortunately, these codes vary between countries
in place, the regulation of promotional activities and in the manner in which they are executed
is frequently linked to the legal classification of (235), such that there are many pharmaceutical
medicines (236,237,238). In such countries, e.g. companies that have not agreed to any such code
the European Union, it is common to restrict the of practice. When these companies market prod-
promotion of prescription-only products to health ucts in countries in which there is little or no
professionals, whereas over-the-counter medicines governmental control on promotional activities,
may be promoted to the general public. Certain there is no way of monitoring the situation and
countries, e.g. the USA, adopt a middle course by preventing misinformation to health care profes-
allowing direct promotion to the public while sionals and to the public.
enforcing restricted access to prescription-only Some pharmaceutical associations carry out
products. Promotional activities that are consid- inspections of the promotional activities of their
ered acceptable, and the regulations regarding members in order to monitor compliance. Com-
them, vary by country. Any legislation applicable panies may also complain to these associations
to promotional activities may be supplemented by about the activities of rivals when these seem to
voluntary codes that have been agreed nationally go beyond the agreed codes of practice. Several
between companies, or internationally between non-governmental organizations undertake audits
federations of pharmaceutical companies. and investigate complaints regarding some forms
Advertisements in peer-reviewed journals, of promotion (234). Whereas none of these
magazines and newspapers and broadcast via bodies has legal empowerment, they may exert
radio or television can be reviewed and made sub- considerable pressure to improve compliance with
ject to controls. However, the advent of advertis- voluntary codes of practice and internationally
ing on the Internet has provided a means by which accepted standards. Nevertheless, despite these
companies can circumvent regulations, reaching codes and monitoring activities, there is clearly a
wide audiences and global markets with unre- need for greater effort to ensure that health pro-
strained messages about their products. fessionals receive accurate information regarding
the efficacy and safety of antimicrobial agents
Codes of practice (117) and of the problems of antimicrobial resist-
ance.
In addition to legislative control mechanisms,
there are various codes of practice regarding

53
CHAPTER 8. INTERNATIONAL ASPECTS OF CONTAINING ANTIMICROBIAL RESISTANCE
CHAPTER 8

International aspects of containing


antimicrobial resistance

Recommendations for intervention 8.7 Encourage innovative approaches to incen-


8.1 Encourage collaboration between govern- tives for the development of new pharma-
ments, non-governmental organizations, pro- ceutical products and vaccines for neglected
fessional societies and international agencies diseases.
to recognize the importance of antimicrobial 8.8 Establish an international database of poten-
resistance, to present consistent, simple and tial research funding agencies with an inter-
accurate messages regarding the importance est in antimicrobial resistance.
of antimicrobial use, antimicrobial resistance
8.9 Establish new, and reinforce existing,
and its containment, and to implement strat-
programmes for researchers to improve the
egies to contain resistance.
design, preparation and conduct of research
8.2 Consider the information derived from the to contain antimicrobial resistance.
surveillance of antimicrobial use and antimi-
crobial resistance, including the containment
Background—the changing global
thereof, as global public goods for health to
context of public health
which all governments should contribute.
Multiple global factors are influencing the epide-
8.3 Encourage governments, non-governmental miology of infectious diseases, the contexts in
organizations, professional societies and which they need to be managed, and thus the
international agencies to support the estab- demands on health care systems. Increasing
lishment of networks, with trained staff and urbanization, with its associated overcrowding and
adequate infrastructures, which can undertake inadequate housing, poor sanitation and lack of
epidemiologically valid surveillance of anti- clean water supplies, has a major influence on the
microbial resistance and antimicrobial use to burden of infectious disease. Pollution and envi-
provide information for the optimal contain- ronmental change, including deforestation, chang-
ment of resistance. ing weather patterns and desertification, may also
8.4 Support drug donations in line with the UN affect the incidence and distribution of infectious
interagency guidelines*. diseases. Demographic changes resulting in a
growing proportion of elderly people and the ex-
8.5 Encourage the establishment of international
panding use of modern medical interventions are
inspection teams qualified to conduct valid
increasing the risks of acquiring infections,
assessments of pharmaceutical manufacturing
especially those caused by multi-resistant hospital
plants.
pathogens. The AIDS epidemic has greatly en-
8.6 Support an international approach to the con- larged the population of immunocompromised
trol of counterfeit antimicrobials in line with patients at risk of infections. Changing patterns
the WHO guidelines**. of lifestyle also have an effect, e.g. the increase in
cigarette smoking in many societies and the con-
sequent increase in associated respiratory diseases,
including pneumonia.
* Interagency guidelines. Guidelines for Drug Donations, revised An increased incidence of infections leads to
1999. Geneva, World Health Organization, 1999. WHO/ more antimicrobial use and consequently a greater
EDM/PAR/99.4.
selection pressure in favour of resistant microor-
**Counterfeit drugs. Guidelines for the development of measures ganisms. Furthermore, the increased food require-
to combat counterfeit drugs. Geneva, World Health Organi-
zation, 1999. WHO/EDM/QSM/99.1. ments of expanding populations may promote an

55
WHO GLOBAL STRATEGY FOR CONTAINMENT OF ANTIMICROBIAL RESISTANCE • WHO/CDS/CSR/DRS/2001.2
increased use of antimicrobial agents in agricul- tions can encourage both the health and educa-
ture, in turn contributing to the emergence of tion sectors of national governments to ensure that
antimicrobial resistance in zoonotic pathogens. sufficient education on infectious disease, antimi-
Increases in global trade and travel have increased crobial use and infection control is provided to all
the speed with which both infectious diseases and students in health care professions.
resistant microorganisms can spread between Experience of the successful implementation of
continents. interventions to contain resistance is a resource
that should not be wasted; sharing information
A call for international cooperative action between nations should be given high priority to
maximize the success of national strategies. These
Containing antimicrobial resistance must involve are areas in which organizations such as WHO
concerted international action. While the major- can and should play a leading role. A summary of
ity of the interventions recommended in earlier currently available national programmes and strat-
chapters of this document are directed at the egies to contain antimicrobial resistance is shown
national level, interventions also need to be un- in Annex A; some of these have been analysed in
dertaken at an international level. It is no longer more detail (187).
justifiable for countries to exempt themselves from
taking action to contain resistance, since inaction
will have both national and international conse- Legal issues associated with
quences. antimicrobial resistance
At the same time it is important to recognize Existing laws at international level require report-
the barriers to action and work to remove them. ing of a limited number of infectious diseases (242)
Antimicrobial resistance is a multi-faceted prob- but do not extend to any systematic reporting of
lem which calls for a multi-sectoral response, but antimicrobial resistance. In the revision of the In-
it is a challenge to get all the sectors on board when ternational Health Regulations (IHR) currently
the magnitude of the problem is unknown. There under evaluation, potential international threats
is a lack of coordination between different groups posed by resistant infections could be recognized.
and disciplines working in this field and even a Some countries have now made certain multi-
lack of knowledge that the different groups exist. resistant pathogens, e.g. MRSA, notifiable at the
Thus messages concerning antimicrobial use and national level. However, the global nature of the
resistance are often confusing and conflicting. antimicrobial resistance problem means that
Many countries lack the money, the skilled pro- national legal measures alone are insufficient. At
fessionals and sufficient laboratory capacity to the same time, the creation of new international
tackle even the definition of the size of the prob- duties would be undermined if not incorporated
lem of resistance. into national law (88).
Closer cooperation between national govern-
ments and agencies, professional societies,
Antimicrobial resistance as a
non-governmental organizations (NGOs) and in-
Global Public Good for Health
ternational agencies would raise the importance
of antimicrobial resistance and its threat to health The concept of Global Public Goods for Health
and development up the political agenda and pro- (GPGH) and their development to assist in the
vide additional resources for the implementation prevention and containment of communicable
of the containment strategy. The development of diseases is growing in importance (243,244). In
consistent messages is critical. International the context of the current review by the Commis-
organizations and NGOs can be particularly sion for Macroeconomics and Health, epidemi-
effective in raising the awareness of their mem- ologically sound surveillance of antimicrobial use,
bers and the public about the importance of anti- resistance and the overall burden of infectious dis-
microbial resistance and in lobbying governments eases is an important component of GPGH. These
about the issue so that antimicrobial resistance is are public goods which have quasi-universal health
seen by governments as being important. By benefits in terms of countries, populations and
including the containment of antimicrobial resist- generations, both current and future, or at least
ance in their aims and objectives, relevant NGOs meet the needs of current generations without
and professional societies can educate their mem- foreclosing development options for future gen-
bers. Furthermore, such international organiza- erations (243). Given the increasing ease of trans-

56
CHAPTER 8. INTERNATIONAL ASPECTS OF CONTAINING ANTIMICROBIAL RESISTANCE
mission of infectious diseases between populations International surveillance
and across national boundaries, and the impor- Surveillance of antimicrobial resistance and anti-
tance to future generations of the current devel- microbial use should be performed at local and
opment of resistance, antimicrobial resistance is national levels to guide clinical management and
clearly a global public “bad” for health, with the infection control, to monitor treatment guidelines
inverse, i.e. the containment of resistance, thus and to update lists of essential drugs. Surveillance
being a global public “good” for health. is also a critical tool to monitor the effectiveness
Given that there is no global government as of interventions to contain resistance. Interna-
final arbiter, the challenge is to determine how tional collaboration on surveillance may also be
the containment of antimicrobial resistance as a of value, to share information as an early warning
GPGH can be implemented so as to benefit the of new or unusual resistance events. At present
world’s people. The large number of participants, there are no formal mechanisms or international
e.g. governments, private sector, NGOs and citi- legal instruments that require reporting (see
zens, complicates coordination of effort, especially above); such resistance events are detected through
in an area such as this where there is significant research studies published in scientific journals.
technical uncertainty. The potential for free Furthermore, surveillance for rare events such as
riding (nations benefiting from the action of a new resistance phenotype has different require-
others without reciprocation) and prisoners’ ments in terms of populations to test and sample
dilemma (lack of communication resulting in a size, etc. from those required for routine surveil-
suboptimal decision for all parties compared to lance. Given the lack of standardization of meth-
the decision which could have occurred with im- ods and the worldwide lack of national surveillance
proved communication) are significant considera- systems generating epidemiologically valid data on
tions. Thus, identifying who will define the global antimicrobial resistance, the first priority should
political agenda, the priorities for resource alloca- be at the national level. International organiza-
tion and the enforcement of penalties if needed, tions and donors should contribute to the streng-
are important international issues if the contain- thening of laboratory capacity in developing
ment of antimicrobial resistance is to successfully countries such that diagnostic services and resist-
become a GPGH. ance surveillance can be provided effectively. The
There are also practical problems in seeking to development of international surveillance stand-
implement global initiatives under the banner of ards is needed—for example, WHO antimicro-
the GPGH concept. For example, there may be bial resistance surveillance standards (209), WHO
financial and technological barriers to accessing guidelines for the management of drug-resistant
information about containing antimicrobial tuberculosis (245) and WHO protocols for
resistance. Some countries may not be able to detection of antimalarial drug resistance (14).
collaborate on certain global initiatives, such as International agencies, professional societies
surveillance or adhering to certain treatment and the pharmaceutical industry could play a role
protocols, due to deficiencies in their health care in defining the mechanisms for the establishment
infrastructure—in this context, strengthened and maintenance of an international resistance
health systems may themselves become a GPGH. alert. In addition, assurance should be sought from
Nevertheless, GPGH aspects of containment the editorial boards of international scientific jour-
could be of great benefit. For instance, surveil- nals that public notification of an international
lance systems could include mechanisms for alert- alert does not jeopardise subsequent publication.
ing governments about the emergence of new International cooperation should also be sought
resistant strains. The maintenance of a global to extend the availability of External Quality
database regarding antimicrobial resistance could Assurance Schemes to resource-poor nations to
be valuable to individual nations, although the dif- assist in improving the quality of surveillance data
ferences worldwide in interpretation of laboratory from microbiology laboratories.
susceptibility tests currently pose a challenge to
this idea. The availability of a database on the dis- Antimicrobial quality and availability
tribution of antimicrobials could assist countries,
especially those with limited resources, to under- Drug donations
take such data collection independently. Data Generous drug donations by pharmaceutical com-
gathering is likely to be most effective if coordi- panies, either in the form of actual drug or by re-
nated, or at least facilitated, internationally. lease of patent, have had a dramatic effect on the

57
WHO GLOBAL STRATEGY FOR CONTAINMENT OF ANTIMICROBIAL RESISTANCE • WHO/CDS/CSR/DRS/2001.2
availability of treatment in complex emergencies tional GMP inspection teams, made up of em-
and in elimination and eradication programmes ployees of larger agencies who might contribute a
for certain disabling diseases in resource-poor limited number of hours per year to the team. Such
settings e.g. leprosy, onchocerciasis (river blind- teams might conduct inspections of a selection of
ness) and lymphatic filariasis. Such donations manufacturing sites at the invitation of, and on
should be strongly encouraged, but in certain situ- behalf of, licensing authorities in resource-poor
ations may need to be better coordinated to countries.
optimize the selection of drugs provided and their
distribution and accessibility to avoid duplication Assessment report sharing schemes
and wastage.
Donors may inadvertently promote inappro- Drug licensing authorities in many resource-poor
priate use of antimicrobials, thereby contributing countries are often willing to license new medi-
to the resistance problem, through supporting cines on the basis of their prior approval by other
donations that are inappropriate in terms of the regulatory agencies, e.g. the US FDA or the EU.
type and quantity of drug, or because a lack of In other countries, where a formal national review
local infrastructure and capacity prevents the of application dossiers is performed, the assess-
appropriate use of the donated drugs. Thus, ment of safety and efficacy of new medicinal prod-
donor agencies should ensure that the advice they ucts has sometimes been assisted by the existence
of certain assessment report sharing schemes. Ex-
give to governments in drawing up their national
pansion of such schemes might benefit regulatory
health plans takes into account antimicrobial re-
authorities thereby expediting the licensing of new
sistance issues. International action should be
drugs.
taken to ensure that all donations follow the
The WHO Certification Scheme is an inter-
interagency guidelines (246). Alternatively, finan-
national voluntary agreement, devised to enable
cial donations to countries to ensure the purchase
countries with limited regulatory capacity to
of the most effective antimicrobials for their needs
obtain partial assurance from exporting countries
together with strategies for distribution and use
concerning the safety, quality and efficacy of the
may be more appropriate. International pro-
products they plan to import. The scheme requires
grammes concerned with drug donations should
that the regulatory authorities of exporting coun-
have capacity-building, training and supervision
tries issue certificates when requested by import-
components, and should be evaluated using indi-
ing countries.
cators that are relevant at the community level,
i.e. at household and primary health care facility,
where most antimicrobial use takes place. Counterfeits
Antimicrobials are among the most frequently
International inspections of pharmaceutical counterfeited drugs (191). Such drugs have
manufacturing potentially major clinical consequences in terms
of treatment failure and prolonged, or even in-
National quality control of medicines and the creased, suffering. Concerted action to reduce the
monitoring of compliance with Good Manufac- distribution of counterfeit drugs is beyond the
turing Practice (GMP) are important to ensure scope of this document and will require the im-
that products meet the required standards. Some plementation of a separate coordinated package
countries, e.g. in the EU, already accept findings of interventions. National and international
of inspections that have been performed by authorities should collaborate to ensure the
appropriately qualified persons from another enforcement of relevant laws.
country. However, not all countries have the re-
sources to perform regular detailed inspections of
manufacturing plants; consequently, these do not International codes of good marketing practice
get inspected unless they are the target of an Adherence to national and international codes of
inspection by a team from another country into marketing practices (240) is critical to maintain-
which the product(s) is/are exported. In these in- ing and improving the quality and accuracy of drug
stances, there may be scope for more extensive promotion practices. The effective policing of
sharing of inspection reports between the authori- adherence to these codes of practice requires
ties of the originator country and the inspecting international commitment, cooperation and
country. It may also be possible to set up interna- supervision (see also Chapter 7).

58
CHAPTER 8. INTERNATIONAL ASPECTS OF CONTAINING ANTIMICROBIAL RESISTANCE
Research and development of new drugs resistance and its successful containment, and
and vaccines keeping this summary up to date, could aid this
Research and development of new drugs and process.
vaccines is expensive and time-consuming. The The various research-funding bodies have dif-
establishment of international research networks ferent priorities in terms of geographical and
and further international cooperation on the scientific emphasis, and process individual appli-
standardization of new drug registration require- cation protocols rather than using one generic for-
ments could assist pharmaceutical companies with mat. The creation of a single entry point through
drug development programmes and so facilitate which researchers could access information about
the availability of new drugs and vaccines. potential funding agencies, including specific con-
International collaboration to improve and tact details, their areas of interest and application
standardize clinical trial designs in order to requirements, could be extremely beneficial. This
optimize the clinical relevance of the data pro- may also assist greater coordination of effort
duced would be helpful. More trials are needed between the various grant-giving bodies and avoid
that aim not only to demonstrate equivalence of unnecessary duplication. WHO may be well
the new drug to the comparative agent but also to placed to provide such a service if grant-giving
assist in identifying regimens which optimize treat- bodies were prepared to collaborate.
ment while minimizing resistance emergence. The quality of research proposals is the key to
Such studies are required for products already on their likelihood of getting funding and producing
the market as well as for new antimicrobials. useful data. Thus, programmes that educate
There is currently a general lack of interest potential researchers on the preparation of high-
among companies in developing therapies for in- quality research proposals would serve to improve
fections that primarily affect resource-poor regions the overall quality of research and reduce wasted
of the world. Innovative incentives, both push and research time and money. Greater coordination
pull mechanisms, need to be carefully considered of international effort to provide such training,
in collaboration with the pharmaceutical indus- either via the Internet or by means of targeted
try, so as to facilitate research into drugs and workshops, could be most beneficial.
vaccines that would have dramatic health benefits
but would likely not be profitable to develop. International support for national
International agreements and cooperation on antimicrobial resistance containment
intellectual property rights, and new approaches
Much of the responsibility for implementing in-
to patents and to time-limited exclusivity arrange-
terventions will fall on national governments and
ments should also be considered, particularly as a
there are certain actions that only governments
means to stimulate additional pharmaceutical and
can assure, including the provision of public goods.
clinical studies to support the licensure of older
However, many countries will need significant
products for additional, previously unregistered,
financial and technical support to address the
indications.
problem of antimicrobial resistance within the
wider priorities of strengthened health systems and
Research to address knowledge gaps disease control and prevention programmes. By
Understanding all the issues associated with anti- directing bilateral support to antimicrobial con-
microbial resistance is probably impossible, but it tainment, international donors can play a major
is clear that there are a number of key knowledge role in the containment of antimicrobial resist-
gaps. A clear research agenda highlighting the most ance, not only for the benefit of individual coun-
important knowledge gaps needs to be defined to tries, but for the global good.
guide future research efforts. In this manner, new
data that are important to understanding and com-
bating resistance can be channelled back to im-
prove future containment initiatives. To avoid
potentially wasteful duplication of effort and
finances, international cooperation to develop a
common, shared research agenda should be en-
couraged. Defining a summary of major gaps in
the current knowledge regarding antimicrobial

59
PART C

Implementation
of the WHO
Global Strategy
IMPLEMENTATION OF THE WHO GLOBAL STRATEGY
Implementation of the WHO Global Strategy

Introduction to simply as interventions) set out in the WHO


To control the most prevalent infectious diseases, Global Strategy, there is a practical need to iden-
especially those that are related to poverty and for tify priorities. The identification of a core set of
which vaccines are not available, antimicrobials interventions to contain resistance could provide
need to be used more wisely, and in some cases, great assistance to governments and health care
more widely. Appropriate access to effective anti- workers charged with the responsibility of imple-
microbial agents is a major public health issue. menting national policy.
Although many patients, especially in sub-
Saharan Africa, continue to die as a result of Prioritization and implementation
inadequate access to antimicrobials, an emerging
STEP 1
problem globally is the widespread indiscriminate
use of antimicrobials, especially antibacterial The diseases requiring antimicrobial therapy can
agents. As a result, many antimicrobials have now be used as the basis for the first step in priori-
become less effective due to the emergence of re- tization. National priorities for the containment
sistance. Simply expanding access to antimicrobials of antimicrobial resistance can be guided by iden-
is thus not sufficient; priority must also be given tifying those diseases that are major problems in
to their appropriate use. the country. On the basis of the evidence used to
Antimicrobial resistance affects a very broad formulate the WHO Global Strategy, the factors
range of human diseases, including tuberculosis, most relevant for antimicrobial resistance in
malaria, AIDS and infections caused by other bac- selected diseases can be identified (see Tables 2–
terial, viral, fungal and parasitic pathogens 5). For each of these factors, those groups of in-
(12,13,14,43,247,248). Despite this wide range terventions that are likely to be most effective are
of pathogens, the factors responsible for the emer- indicated. In this manner, the process for select-
gence of resistance are very similar, with excessive ing the necessary interventions to limit emerging
and inappropriate drug usage being the key driv- antimicrobial resistance can be based on the
ers. Thus the broad management approach to con- diseases most prevalent in the country. In some
taining antimicrobial resistance is similar for each instances, the interventions may be the most chal-
of these pathogens and diseases, although there lenging to implement. Countries in which all
are some differences such as clinical presentation, major disease infections are common will need to
diagnostic difficulty, treatment strategies and address all groups of interventions.
resistance detection, which are summarized in
Table 1. Effective implementation of the WHO Bacterial infections (other than tuberculosis)
Global Strategy needs to recognize and be coher-
ent with these differences. The bacterial infections which contribute most to
The various factors identified to be responsi- human disease are also those in which emerging
ble for the emergence of antimicrobial resistance antimicrobial resistance is most evident. In this
have been discussed in the Part B—Issues and document they are grouped as four key diseases:
interventions, and recommendations for inter- — diarrhoea (Table 2)
ventions have been developed on the basis of
— respiratory tract infections and meningitis
these factors. However, the identification and
(Table 3)
prioritization of those factors especially relevant
in each national and regional context is more dif- — sexually transmitted infections (Table 4)
ficult. In addition, given the large number of rec-
— hospital-acquired infections (Table 5)
ommendations for intervention (hereafter referred

63
WHO GLOBAL STRATEGY FOR CONTAINMENT OF ANTIMICROBIAL RESISTANCE • WHO/CDS/CSR/DRS/2001.2
The problems related to resistance to the treat- are not properly treated under a DOTS-based pro-
ments-of-choice for these diseases are presented gramme. However, the control of existing MDR-
in detail in accompanying documents TB also has an extremely high priority. The Global
(19,20,21,101). Tables 2–5 summarize the impor- Project on Anti-Tuberculosis Drug Resistance
tant factors influencing the emergence and spread Surveillance (managed jointly by WHO and the
of resistance and set out the groups of interven- International Union against Tuberculosis and
tions which need to be implemented to make an Lung Disease) has identified high prevalence of
impact. MDR-TB in some countries of Eastern Europe,
Latin America, Africa, and Asia (12,245,250).
Tuberculosis MDR-TB does not respond as effectively as drug-
susceptible TB to short-course chemotherapy with
Tuberculosis is a leading cause of morbidity and first-line drugs (48). Therefore, WHO and its
mortality worldwide and resistance to antituber- partners have launched DOTS-Plus (43,247,251)
culous therapy has increased dramatically in to manage MDR-TB with second-line drugs.
recent years with evidence of substantive clinical DOTS-Plus includes the five components of
treatment failures and increased person-to-person DOTS together with other aspects regarding long-
transmission (12,13,43). The spread of HIV in- term (18–24 months) therapeutic regimens with
fection, with its associated immunosuppression, second-line drugs, and the use of drug suscepti-
has resulted in an enormous increase in TB cases, bility testing for diagnosis and therapeutic follow-
most frequently among resource-poor communi- up. Recommendations for therapeutic regimens
ties and in regions with weak health care systems. for the treatment of MDR-TB were compiled by
Inadequate treatment, including insufficient drugs a panel of experts convened by WHO (245,249).
(inadequate supply or mono-therapy), poor qual- Pilot projects with some of the recommended
ity drugs, and/or poor adherence to treatment treatment regimens are underway to assess the
regimens have been major factors in the emergence feasibility and cost-effectiveness of using second-
of multi-drug resistant TB (MDR-TB). line drugs under programme conditions. Surveil-
Although tuberculosis is a bacterial infection, lance for drug resistance at the pilot sites is a
it is considered different enough to warrant a dis- prerequisite. Data generated through this initia-
tinct focus. In addition, WHO has initiated ap- tive will be used to design evidence-based policy
proaches to the containment of anti-tuberculosis guidelines for the management of MDR-TB,
drug resistance. Faced with the global emergency which in turn will play a critical role in the con-
of tuberculosis, WHO adopted the DOTS (di- tainment of drug resistance in tuberculosis.
rectly observed treatment, short-course) interven- Thus, many of the interventions that will need
tion strategy for effective TB control (245,249). to be implemented to contain resistance in other
The principles of DOTS are the following: bacterial infections, such as political commitment,
— government commitment to a National improvement in national regulatory frameworks,
Tuberculosis Programme drug distribution and educational initiatives re-
garding antimicrobial resistance, are in line with,
— case detection through case-finding by
and will further support, current initiatives to con-
sputum smear microscopy examination of
tain drug-resistant tuberculosis. Intervention
TB suspects in general health facilities
priorities have been identified for tuberculosis
— standardized short-course chemotherapy to, (Table 6).
at least, all smear-positive TB cases under
directly observed therapy (DOT) under
proper case management conditions Malaria
— regular uninterrupted supply of all essen- The majority of deaths in malarious areas con-
tial anti-TB drugs tinue to be due to the lack of drug availability
(252). However, emerging resistance is also greatly
— monitoring system for programme super-
undermining the efficacy of antimalarial treatment
vision and evaluation.
regimens in many regions and is likely to pose a
The implementation of DOTS, presently in major problem worldwide in the future.
119 countries (12,43), prevents the generation of As summarized in Table 7, one of the key driv-
MDR-TB through the cure of drug-susceptible ers behind the emergence of antimalarial resist-
TB patients, who will evolve to MDR-TB if they ance is poor patient understanding about the

64
IMPLEMENTATION OF THE WHO GLOBAL STRATEGY
disease and its appropriate treatment, resulting in fections other than tuberculosis. The valuable
indiscriminate short-course therapy with antima- lessons that will be learned during this first phase
larial agents. In addition, inappropriate prescrib- should impact on the implementation approaches
ing/dispensing and ineffective drug distribution used for containment of resistance in tuberculo-
systems encourage such behaviour. The frequent sis, malaria and viral infections. However, due to
lack of appropriate diagnostic facilities makes the commonality of factors leading to antimicro-
the decision to treat difficult, since malaria so bial resistance in all diseases, many of the inter-
frequently presents in an undifferentiated man- ventions, instigated for containing resistance in
ner, as fever with, or without, headache. Thus, bacterial infections—such as political commit-
without the ability to confirm the diagnosis, the ment, regulatory framework, laboratory strength-
tendency is to treat every patient with a fever with ening, surveillance and education—will also
antimalarials if they reside in a malaria endemic contribute to resistance containment in other
region. Systems for surveillance of antimicrobial diseases at national level.
resistance are often weak and thus unable to in-
form about the need to change treatment guide- STEP 2
lines. Despite early promising data, it appears that
vaccines effective against malaria are still some
Defining a core set of interventions to contain
antibacterial resistance
years away (253). The priority for the contain-
ment of antimalarial resistance is thus to concen- While prioritization by disease group provides
trate on the implementation of intervention some direction for implementation, the identifi-
groups 1, 2, 5 and 6. This is in line with WHO cation of a core set for national implementation is
policy as expressed in a document in preparation required, within each group of interventions. This
by the WHO Regional Office for Africa (254). is particularly relevant to intervention groups 1,
2, 3, 5 and 7. Issues related to group 4 (use of
antimicrobials in food-producing animals) have
Viral infections recently been the subject of an extensive consulta-
With the increasing development and use of tive process at WHO and primarily involve inter-
effective antiretroviral agents, resistance is becom- ventions in the agricultural industry (2). Thus they
ing apparent. In vitro resistance to antiretroviral are not considered further here. Interventions
agents among HIV strains appears to correlate with relating to drug and vaccine development and
prior antiretroviral therapy and with clinical treat- international aspects of containing antimicrobial
ment failure (255,256,257,258,259). Highly resistance are extremely important, but since they
effective combination therapy is considered to be depend on supra-national factors, a number of
less associated with the emergence of resistance. which involve the (multi-national) research-based
However, this is a rapidly progressing area of pharmaceutical industry, their prioritization at
scientific research in which the factors that drive national level is less relevant.
resistance are less clearly defined than for bacte- Implementation of the WHO Global Strategy
rial infections and malaria. As the knowledge base at national level therefore requires prioritization
expands, a prioritization of interventions can be among interventions in groups 1, 2, 3, 5 and 7.
developed. At present it seems clear that improved The prioritization presented in Step 3 is based on
patient and prescriber education (Intervention available evidence (summarized in Part B); where
Groups 1 and 2), government regulations regard- evidence is lacking, it is based on the consensus of
ing licensure and surveillance of resistance (Inter- a suitably qualified group of experts convened by
vention Group 5) and issues of drug and vaccine WHO for this purpose.
development (Group 6) will all be important.
STEP 3
Conclusion of Step 1 Intra-group prioritization of interventions
Given the disease-specific aspects of containment Interventions within each group have been
of antimicrobial resistance associated with tuber- prioritized according to the relative merits of each
culosis, malaria and HIV infections and the pro- intervention and ranked according to sequence
grammes already in place, it is proposed that the and importance of implementation. This complex
first phase of implementation of the WHO task required consideration of multiple factors
Global Strategy should be directed to bacterial in- relating to each intervention including:

65
WHO GLOBAL STRATEGY FOR CONTAINMENT OF ANTIMICROBIAL RESISTANCE • WHO/CDS/CSR/DRS/2001.2
— overall importance of the intervention to process only provides a guide to implementation
improving the appropriate use of antimi- and is not a rigid set of rules. Differences in
crobials and containing antimicrobial national circumstances, health care systems and
resistance burden of the different infections may influence
the practicality with which some interventions can
— likely impact, allowing for the expected cost
be implemented and the local importance of one
of implementation
intervention over another in a manner that is not
— complexity of implementation considering accurately reflected in Table 8. However, Table 8
the capacity of various health care systems provides a working guide to the prioritization and
and political realities sequence of implementation of interventions in
— time required for implementation and the groups 1, 2, 3, 5 and 7.
expected lag period before outcomes could
be expected Implementation guidelines
— the accuracy with which most health care Effective implementation requires a number of key
systems could assess the efficacy of each features, including a clear action plan, delegation
intervention of authority and power to act, resources and sound
— the interrelationship between various inter- mechanisms to assess the effectiveness of interven-
ventions, including the need to undertake tions, allowing feedback of results to influence
future implementation strategies. Thus, interven-
some interventions in a logical sequence.
tions identified in the prioritization process as
being of fundamental and first priority (see Table
Inter-group prioritization of interventions 8) have been considered in greater detail, specifi-
Following the prioritization within each group, cally identifying the following aspects as impor-
interventions were ranked according to their over- tant for successful implementation:
all importance and timing (sequence) of imple- — the optimal approach to implementation
mentation without consideration of their group.
Although it was recognized that some priorities — who should initiate the intervention,
might vary depending on the health care system undertake and manage the intervention,
in which they are to be implemented, it was found and evaluate the intervention
that this consideration did not impact to any — what process and outcome indicators
significant extent on the priority given to the should be used for evaluation.
majority of very high priority interventions.
The proposed guidelines for implementation
The results of Step 3 are shown in Table 8. In-
are detailed in “Suggested Model Framework for
terventions are grouped according to those which
Implementation of Core Interventions”.
should be undertaken first, through to those
which, although important, are either dependent
on the implementation of the earlier interventions Monitoring outcomes
or are of lower priority. Within each priority i.e. Ability to monitor the process to ensure that in-
first, second, third, interventions are not ranked terventions are appropriately designed and targeted
but listed in numerical order only and should be and their impact on the use of antimicrobials and
considered of equal importance. For example, for the prevalence of resistance will be crucial to the
group 1, both interventions 1.2 and 1.3 are con- successful implementation of the WHO Global
sidered to be of similar priority for implementa- Strategy. Without accurate information about
tion, but both 1.2 and 1.3 are given higher priority antimicrobial usage and antimicrobial resistance
than either 1.1 (second priority) or 1.4 and 1.5 and their respective trends, the impact of inter-
(third priority). ventions will be difficult to interpret. Thus, an
Comparisons across the groups of interventions early priority in the implementation of the WHO
are more difficult but are important to achieve a Global Strategy for all countries should be the es-
logical and effective implementation. Within the tablishment of an appropriate framework to moni-
national reality, consideration of the sectors tor accurately antimicrobial use and antimicrobial
involved in implementation of the interventions resistance (Intervention Group 5).
should allow a plan of action to be elaborated.
It must be emphasized that this prioritization

66
IMPLEMENTATION OF THE WHO GLOBAL STRATEGY
Summary
This model implementation plan for the WHO
Global Strategy is a guide only. Differences in
national circumstances, health care systems and
prevalent diseases may influence the approaches
taken by governments to contain antimicrobial
resistance. However, this is a complex area in which
it is often difficult to see the wood for the trees.
The stepwise approach described above attempts
to highlight the interventions that are most im-
portant and to identify a logical sequence for
implemention. The manner in which the WHO
Global Strategy for Containment of Antimicro-
bial Resistance is implemented will depend largely
on the decisions and actions of each nation, but
the consequences are likely to be felt worldwide.

67
WHO GLOBAL STRATEGY FOR CONTAINMENT OF ANTIMICROBIAL RESISTANCE • WHO/CDS/CSR/DRS/2001.2
Recommendations for intervention 2.9 Empower formulary managers to limit antimi-
crobial use to the prescription of an appropri-
1. PATIENTS AND THE GENERAL COMMUNITY
ate range of selected antimicrobials.

Education Regulation
1.1 Educate patients and the general community on
2.10 Link professional registration requirements for
the appropriate use of antimicrobials.
prescribers and dispensers to requirements for
1.2 Educate patients on the importance of meas- training and continuing education.
ures to prevent infection, such as immunization,
vector control, use of bednets, etc.
3. HOSPITALS
1.3 Educate patients on simple measures that may
reduce transmission of infection in the house-
Management
hold and community, such as handwashing,
3.1 Establish infection control programmes, based
food hygiene, etc.
on current best practice, with the responsibility
1.4 Encourage appropriate and informed health for effective management of antimicrobial
care seeking behaviour. resistance in hospitals and ensure that all hos-
1.5 Educate patients on suitable alternatives to pitals have access to such a programme.
antimicrobials for relief of symptoms and 3.2 Establish effective hospital therapeutics com-
discourage patient self-initiation of treatment, mittees with the responsibility for overseeing
except in specific circumstances. antimicrobial use in hospitals.
3.3 Develop and regularly update guidelines for
2. PRESCRIBERS AND DISPENSERS antimicrobial treatment and prophylaxis, and
hospital antimicrobial formularies.
Education
3.4 Monitor antimicrobial usage, including the
2.1 Educate all groups of prescribers and dispens-
quantity and patterns of use, and feedback
ers (including drug sellers) on the importance
results to prescribers.
of appropriate antimicrobial use and contain-
ment of antimicrobial resistance.
Diagnostic laboratories
2.2 Educate all groups of prescribers on disease 3.5 Ensure access to microbiology laboratory
prevention (including immunization) and infec- services that match the level of the hospital, e.g.
tion control issues. secondary, tertiary.
2.3 Promote targeted undergraduate and post-
3.6 Ensure performance and quality assurance of
graduate educational programmes on the
appropriate diagnostic tests, microbial identifi-
accurate diagnosis and management of cation, antimicrobial susceptibility tests of key
common infections for all health care workers, pathogens, and timely and relevant reporting
veterinarians, prescribers and dispensers. of results.
2.4 Encourage prescribers and dispensers to educate
3.7 Ensure that laboratory data are recorded, pref-
patients on antimicrobial use and the importance
erably on a database, and are used to produce
of adherence to prescribed treatments.
clinically- and epidemiologically-useful surveil-
2.5 Educate all groups of prescribers and dispens- lance reports of resistance patterns among
ers on factors that may strongly influence their common pathogens and infections in a timely
prescribing habits, such as economic incentives, manner with feedback to prescribers and to the
promotional activities and inducements by the infection control programme.
pharmaceutical industry.
Interactions with the pharmaceutical industry
Management, guidelines and formularies 3.8 Control and monitor pharmaceutical company
2.6 Improve antimicrobial use by supervision and promotional activities within the hospital envi-
support of clinical practices, especially diagnos- ronment and ensure that such activities have
tic and treatment strategies. educational benefit.
2.7 Audit prescribing and dispensing practices and
utilize peer group or external standard compari- 4. USE OF ANTIMICROBIALS IN
sons to provide feedback and endorsement of FOOD-PRODUCING ANIMALS
appropriate antimicrobial prescribing. This topic has been the subject of specific consulta-
2.8 Encourage development and use of guidelines tions which resulted in “WHO global principles for the
and treatment algorithms to foster appropriate containment of antimicrobial resistance in animals
use of antimicrobials. intended for food”*. A complete description of all rec-

* http:// www.who.int/emc/diseases/zoo/who_global_
principles.html

68
IMPLEMENTATION OF THE WHO GLOBAL STRATEGY
ommendations is contained in that document and 5.4 Link prescription-only status to regulations re-
only a summary is reproduced here. garding the sale, supply, dispensing and allow-
able promotional activities of antimicrobial
Summary agents; institute mechanisms to facilitate
4.1 Require obligatory prescriptions for all anti- compliance by practitioners and systems to
microbials used for disease control in food monitor compliance.
animals. 5.5 Ensure that only antimicrobials meeting inter-
4.2 In the absence of a public health safety evalua- national standards of quality, safety and efficacy
tion, terminate or rapidly phase out the use of are granted marketing authorization.
antimicrobials for growth promotion if they are 5.6 Introduce legal requirements for manufacturers
also used for treatment of humans. to collect and report data on antimicrobial
4.3 Create national systems to monitor antimicro- distribution (including import/export).
bial usage in food animals. 5.7 Create economic incentives for appropriate use
4.4 Introduce pre-licensing safety evaluation of of antimicrobials.
antimicrobials with consideration of potential
resistance to human drugs. Policies and guidelines
4.5 Monitor resistance to identify emerging health 5.8 Establish and maintain updated national Stand-
problems and take timely corrective actions to ard Treatment Guidelines (STGs) and encourage
protect human health. their implementation.

4.6 Develop guidelines for veterinarians to reduce 5.9 Establish an Essential Drugs List (EDL) consist-
overuse and misuse of antimicrobials in food ent with national STGs and ensure the accessi-
animals. bility and quality of these drugs.
5.10 Enhance immunization coverage and other
5. NATIONAL GOVERNMENTS AND
disease preventive measures, thereby reducing
HEALTH SYSTEMS the need for antimicrobials.

Advocacy and intersectoral action Education


5.1 Make the containment of antimicrobial resist- 5.11 Maximize and maintain the effectiveness of the
ance a national priority. EDL and STGs by conducting appropriate
undergraduate and postgraduate education
— Create a national intersectoral task force
programmes of health care professionals on the
(membership to include health care profes-
importance of appropriate antimicrobial use
sionals, veterinarians, agriculturalists,
and containment of antimicrobial resistance.
pharmaceutical manufacturers, govern-
ment, media representatives, consumers 5.12 Ensure that prescribers have access to approved
prescribing literature on individual drugs.
and other interested parties) to raise aware-
ness about antimicrobial resistance, organ-
ize data collection and oversee local task Surveillance of resistance, antimicrobial usage
forces. For practical purposes such a task and disease burden
force may need to be a government task 5.13 Designate or develop reference microbiology
force which receives input from multiple laboratory facilities to coordinate effective
sectors. epidemiologically sound surveillance of antimi-
crobial resistance among common pathogens
— Allocate resources to promote the imple-
in the community, hospitals and other health
mentation of interventions to contain resist-
care facilities. The standard of these laboratory
ance. These interventions should include
facilities should be at least at the level of rec-
the appropriate utilization of antimicrobial
ommendation 3.6.
drugs, the control and prevention of infec-
tion, and research activities. 5.14 Adapt and apply WHO model systems for anti-
microbial resistance surveillance and ensure
— Develop indicators to monitor and evaluate
data flow to the national intersectoral task force,
the impact of the antimicrobial resistance
to authorities responsible for the national STGs
containment strategy.
and drug policy, and to prescribers.
Regulations 5.15 Establish systems for monitoring antimicrobial
5.2 Establish an effective registration scheme for use in hospitals and the community, and link
dispensing outlets. these findings to resistance and disease surveil-
lance data.
5.3 Limit the availability of antimicrobials to
prescription-only status, except in special 5.16 Establish surveillance for key infectious diseases
circumstances when they may be dispensed on and syndromes according to country priorities,
the advice of a trained health care professional. and link this information to other surveillance
data.

69
WHO GLOBAL STRATEGY FOR CONTAINMENT OF ANTIMICROBIAL RESISTANCE • WHO/CDS/CSR/DRS/2001.2
6. DRUG AND VACCINE DEVELOPMENT 8. INTERNATIONAL ASPECTS OF CONTAINING
ANTIMICROBIAL RESISTANCE
6.1 Encourage cooperation between industry,
government bodies and academic institutions 8.1 Encourage collaboration between govern-
in the search for new drugs and vaccines. ments, non-governmental organizations, pro-
fessional societies and international agencies to
6.2 Encourage drug development programmes
recognize the importance of antimicrobial
which seek to optimize treatment regimens
resistance, to present consistent, simple and
with regard to safety, efficacy and the risk of
accurate messages regarding the importance of
selecting for resistant organisms.
antimicrobial use, antimicrobial resistance and
6.3 Provide incentives for industry to invest in the its containment, and to implement strategies to
research and development of new antimi- contain resistance.
crobials.
8.2 Consider the information derived from the sur-
6.4 Consider establishing or utilizing fast-track veillance of antimicrobial use and antimicrobial
marketing authorization for safe new agents. resistance, including the containment thereof,
6.5 Consider using an orphan drug scheme where as global public goods for health to which all
available and applicable. governments should contribute.
6.6 Make available time-limited exclusivity for new 8.3 Encourage governments, non-governmental
formulations and/or indications for use of organizations, professional societies and inter-
antimicrobials. national agencies to support the establishment
6.7 Align intellectual property rights to provide suit- of networks, with trained staff and adequate
able patent protection for new antimicrobial infrastructures, which can undertake epidemi-
agents and vaccines. ologically valid surveillance of antimicrobial
resistance and antimicrobial use to provide
6.8 Seek innovative partnerships with the pharma-
information for the optimal containment of
ceutical industry to improve access to newer
resistance.
essential drugs.
8.4 Support drug donations in line with the UN
interagency guidelines*.
7 PHARMACEUTICAL PROMOTION
8.5 Encourage the establishment of international
7.1 Introduce requirements for pharmaceutical inspection teams qualified to conduct valid
companies to comply with national or inter- assessments of pharmaceutical manufacturing
national codes of practice on promotional plants.
activities.
8.6 Support an international approach to the
7.2 Ensure that national or internationally codes of control of counterfeit antimicrobials in line
practice cover direct-to-consumer advertising, with the WHO guidelines**.
including advertising the Internet.
8.7 Encourage innovative approaches to incentives
7.3 Institute systems for monitoring compliance for the development of new pharmaceutical
with legislation on promotional activities. products and vaccines for neglected diseases.
7.4 Identify and eliminate economic incentives that 8.8 Establish an international database of potential
encourage inappropriate antimicrobial use. research funding agencies with an interest in
7.5 Make prescribers aware that promotion in antimicrobial resistance.
accordance with the datasheet may not neces- 8.9 Establish new, and reinforce existing, pro-
sarily constitute appropriate antimicrobial use. grammes for researchers to improve the design,
preparation and conduct of research to contain
antimicrobial resistance.

* Interagency guidelines. Guidelines for Drug Donations, revised


1999. Geneva, World Health Organization, 1999. WHO/
EDM/PAR/99.4.
**Counterfeit drugs. Guidelines for the development of measures
to combat counterfeit drugs. Geneva, World Health Organi-
zation, 1999. WHO/EDM/QSM/99.1.

70
IMPLEMENTATION OF THE WHO GLOBAL STRATEGY
TABLE 1. COMPARISON OF DISEASE-RELATED RESISTANCE ISSUES

Issues Bacterial infections TB Malaria HIV

Appropriate use important Yes Yes Yes Yes

Inappropriate use contributes


to ↑ resistance Yes Yes Yes Yes

Need for new drug development Yes Yes Yes Yes

Detection of pathogen Reasonably easy Easy Easy Easy


& feasible

Detection of in vitro resistance Reasonably easy Feasible but Difficult, expensive Difficult, expensive
& feasible expensive rarely feasible limited availability

Treatment indication Generally pathogen- Pathogen-based Frequently Pathogen-based


based (± resistance) syndromic

Observed treatment No Yes–DOT No No

Antimicrobial treatment Single agent Multiple agents ≥1 agent Multiple agents


Short duration Long duration Short duration Lifelong

HIV interaction Some: Massive: Possibly —


Especially Personal &
nosocomial risk nosocomial risk

Potential impact of one Yes Little Some Yes


programme on another Some antibiotics Except: e.g. doxycyline, e.g. cotrimoxazole +
could affect malaria Rifampicin use on sulphadoxine- isoniazid prophylaxis
resistance. Staph. spp. pyrimethamine

71
WHO GLOBAL STRATEGY FOR CONTAINMENT OF ANTIMICROBIAL RESISTANCE • WHO/CDS/CSR/DRS/2001.2
TABLE 2. BACTERIAL INFECTIONS (OTHER THAN TUBERCULOSIS): DIARRHOEAL DISEASES

Pathogens Important factors

Human Human Misuse in animal Surveillance Vaccines


misuse in misuse in & agricultural of antibacterial potentially useful
the community hospitals industry resistance important future option

Campylobacter spp. +/– – +++ ++ –

Shigella spp. ++ – +/– ++ –

Salmonella spp:
S. typhi & S. paratyphi ++ – – +++ +
Non-typhoidal salmonellae –/+ – +++ +++ –

Vibrio cholerae + – – +++ +

Diarrhoeal disease overall +/++ ++/++ +++ –/+




High High High Moderate
priority priority priority priority
Intervention Intervention Intervention Intervention
Groups Groups Group Group
1, 2, 5 & 7 4&7 5 6

High priority interventions:


Group 1 Patients and the general community
Group 2 Prescribers and dispensers
Group 4 Use of antimicrobials in food-producing animals
Group 5 National governments and health systems
Group 7 Pharmaceutical promotion

TABLE 3. BACTERIAL INFECTIONS (OTHER THAN TUBERCULOSIS): RESPIRATORY TRACT INFECTIONS AND MENINGITIS

Pathogens Important factors

Human Human Misuse in animal Surveillance Vaccines


misuse in misuse in & agricultural of antibacterial potentially useful
the community hospitals industry resistance important future option

Streptococcus pneumoniae +++ + – +++ +++

Haemophilus influenzae ++ – – ++ +++

Neisseria meningitidis + – – + +

Respiratory disease overall +++ + ++/+++ +++



High Moderate High High


priority priority priority priority
Intervention Intervention Intervention Intervention
Groups Groups Group Group
1, 2, 5 & 7 3&7 5 6

High priority Interventions:


Group 1 Patients and the general community
Group 2 Prescribers and dispensers
Group 5 National governments and health systems
Group 6 Drug and vaccine development
Group 7 Pharmaceutical promotion

72
IMPLEMENTATION OF THE WHO GLOBAL STRATEGY
TABLE 4. BACTERIAL INFECTIONS (OTHER THAN TUBERCULOSIS): SEXUALLY TRANSMITTED INFECTIONS

Pathogens Important factors

Human Human Misuse in animal Surveillance Vaccines


misuse in misuse in & agricultural of antibacterial potentially useful
the community hospitals industry resistance important future option

Neisseria gonorrhoeae +++ – – +++ –

Haemophilus ducreyi +++ – – +++ –

Treponema pallidum – – – – –

Chlamydia trachomatis – – – – –

Sexually transmitted disease overall +++ +++



High High
priority priority
Intervention Intervention
Groups Group
1, 2, 5 & 7 5

High priority interventions:


Group 1 Patients and the general community
Group 2 Prescribers and dispensers
Group 5 National governments and health systems
Group 7 Pharmaceutical promotion

TABLE 5. BACTERIAL INFECTIONS (OTHER THAN TUBERCULOSIS) : HOSPITAL-ACQUIRED INFECTIONS

Pathogens Important factors

Human Human Misuse in animal Surveillance Vaccines


misuse in misuse in & agricultural of antibacterial potentially useful
the community hospitals industry resistance important future option

Gram-positive spp:
Staphyloccus aureus + +++ – +++ –
Streptococci – + – – –
Enterococci – +++ +/++ ++ –

Gram-negative spp:
Escherichia coli + ++ + ++ –
Enterobacter spp + +++ – +++ –
Klebsiella spp + +++ – +++ –
Pseudomonas aeruginosa – +++ – ++ –

Fungi – ++ – – –

Hospital-acquired infections overall + ++/+++ + +++



High High Moderate High


priority priority priority priority
Intervention Intervention Intervention Intervention
Groups Groups Group Group
1, 2, 5 & 7 3&7 4 5

High priority interventions:


Group 1 Patients and the general community
Group 2 Prescribers and dispensers
Group 3 Hospitals
Group 5 National governments and health systems
Group 7 Pharmaceutical promotion
73
WHO GLOBAL STRATEGY FOR CONTAINMENT OF ANTIMICROBIAL RESISTANCE • WHO/CDS/CSR/DRS/2001.2
TABLE 6. TUBERCULOSIS

Pathogens Important factors

Human Human Misuse in animal Surveillance Vaccines


misuse in misuse in & agricultural of antibacterial potentially useful
the community hospitals industry resistance important future option

Mycobacterium tuberculosis ++ – – +++ +/–

Tuberculosis overall ++ +++ +


High High Moderate
priority priority priority
Intervention Intervention Intervention
Groups Group Group
1, 2 & 5 5 6

High priority interventions:


Group 1 Patients and the general community
Group 2 Prescribers and dispensers
Group 5 National governments and health systems

TABLE 7. MALARIA

Pathogens Important factors

Human Human Misuse in animal Surveillance Vaccines


misuse in misuse in & agricultural of antibacterial potentially useful
the community hospitals industry resistance important future option

Plasmodium vivax / ovale / malariae + – – + –

Plasmodium falciparum +++ – – +++ +/–

Malaria overall ++ +++ +/–



High High Moderate
priority priority priority
Intervention Intervention Intervention
Groups Group Group
1, 2 & 5 5 6

High priority interventions:


Group 1 Patients and the general community
Group 2 Prescribers and dispensers
Group 5 National governments and health systems

74
IMPLEMENTATION OF THE WHO GLOBAL STRATEGY
TABLE 8. PRIORITIZATION OF INTERVENTIONS: CORE SET FOR NATIONAL IMPLEMENTATION
(EXCLUDING GROUPS 4 AND 6)

Intervention Group Priority of implementation

Fundamental First Second Third

1. Patients and the general community 1.2 1.1 1.4


1.3 1.5

2. Prescribers and dispensers 2.1 2.6 2.4


2.2 2.7 2.5
2.3 2.9 2.10

2.8

3. Hospitals 3.1 3.2


3.5 3.3
3.6 3.4

3.7
3.8

5. National governments and 5.1 5.3 5.2 5.6


health systems 5.13 5.5 5.4 5.7

5.8 5.12
5.9 5.14
5.11 5.15

5.16

7. Pharmaceutical promotion 7.1 7.4

7.2 7.5
7.3

75
WHO GLOBAL STRATEGY FOR CONTAINMENT OF ANTIMICROBIAL RESISTANCE • WHO/CDS/CSR/DRS/2001.2
Suggested model framework for implementation of core interventions (excluding group 4)

INTERVENTIONS—PRIORITY OF IMPLEMENTATION : FUNDAMENTAL


Intervention 5.1 Make the containment of antimicrobial resistance a national priority.
● Create a national intersectoral task force (membership to include
health care professionals, veterinarians, agriculturalists, pharmaceuti-
cal manufacturers, government, media representatives, consumers and
other interested parties) to raise awareness about antimicrobial re-
sistance, organize data collection and oversee local task forces. For
practical purposes such a task force may need to be a government
task force which receives input from multiple sectors.
● Allocate resources to promote the implementation of interventions
to contain resistance. These interventions should include the appro-
priate utilization of antimicrobial drugs, the control and prevention of
infection, and research activities.
● Develop indicators to monitor and evaluate the impact of the antimi-
crobial resistance containment strategy.

Implementation: ● Develop a National Strategy and make it a national priority

Who should initiate: ● Ministry of Health


● Other interested parties should contribute (e.g. Professional Societies)
● WHO to assist and contribute

Who should undertake and manage: ● National Intersectoral Task Force appointed by the Ministry of Health
● Sufficient resources should be allocated

Who should evaluate: ● WHO through the Regional Offices

Process Indicators: ● Appointment of the National Intersectoral Task Force


● Allocation of sufficient resources

Outcome Indicators: ● Has a National Strategy been developed?

Intervention 5.13 Designate or develop reference microbiology laboratory facilities to co-


ordinate effective epidemiologically sound surveillance of antimicrobial
resistance among common pathogens in the community, hospitals and
other health care facilities. The standard of these laboratory facilities
should be at least at the level of recommendation 3.6.

Implementation: ● Establishment by government mandate

Who should initiate: ● Ministry of Health


● Sufficient resources should be allocated

Who should undertake and manage: ● Reference laboratories—accountable to Government Health Depart-
ment

Who should evaluate: ● Internal and external, (e.g. international), quality assurance pro-
grammes and performance assessments
● National Intersectoral Task Force audit

Process Indicators: ● Evidence of overseeing national resistance surveillance


● Documentation of resistance data

Outcome Indicators: ● Regular communication of resistance data to National Intersectoral


Task Force and Government Health Department
● Commitment to teaching and training of laboratory staff including
technology transfer

76
IMPLEMENTATION OF THE WHO GLOBAL STRATEGY
INTERVENTIONS—INTERVENTION PRIORITY: FIRST
Intervention 1.2 Educate patients on the importance of measures to prevent infection,
such as immunization, vector control, use of bednets, etc.

Implementation: ● Develop a National Strategy and make it a national priority

Who should initiate: ● Ministry of Health


● Other interested parties should contribute (e.g. Professional Societies)
● WHO to assist and contribute

Who should undertake and manage: ● National Intersectoral Task Force (e.g. appointed by the Ministry of
Health)
● Sufficient resources should be allocated

Who should evaluate: ● WHO through the Regional Offices


● Ministry of Health

Process Indicators: ● Appointment of the National Intersectoral Task Force


● Allocation of sufficient resources

Outcome Indicators: ● Has a National Strategy been developed?


● Immunization rates

Intervention 1.3 Educate patients on simple measures that may reduce transmission of
infection in the household and community, such as handwashing, food
hygiene, etc.

Implementation: ● Develop a National Strategy and make it a national priority

Who should initiate: ● Ministry of Health


● Other interested parties should contribute (e.g. Professional Societies)
● WHO to assist and contribute

Who should undertake and manage: ● National Intersectoral Task Force (e.g. appointed by the Ministry of
Health)
● Sufficient resources should be allocated

Who should evaluate: ● WHO through the Regional Offices


● Ministry of Health

Process Indicators: ● Appointment of the National Intersectoral Task Force


● Allocation of sufficient resources

Outcome Indicators ● Has a National Strategy been developed?

Interventions 2.1 and 2.2 2.1 Educate all groups of prescribers and dispensers (including drug
sellers) on the importance of appropriate antimicrobial use and
containment of antimicrobial resistance.
2.2 Educate all groups of prescribers on disease prevention (including
immunization) and infection control issues.

Implementation: ● Develop a National Strategy and make it a national priority


● Identify interested organizations and opinion leaders, educators and
sources of appropriate information

Who should initiate: ● National Intersectoral Task Force

Who should undertake and manage: ● Organizations delegated by the National Intersectoral Task Force

77
WHO GLOBAL STRATEGY FOR CONTAINMENT OF ANTIMICROBIAL RESISTANCE • WHO/CDS/CSR/DRS/2001.2
INTERVENTIONS—INTERVENTION PRIORITY: FIRST (continued)
Who should evaluate: ● Ministry of Health
● National Intersectoral Task Force
● Professional organizations, universities, delegated organizations

Process Indicators: ● Opinion leaders identified, quantitative and qualitative assessments


of educational exposure

Outcome Indicators: ● Levels of knowledge, attitudes and beliefs about antibiotic use, aware-
ness of antimicrobial resistance and disease prevention issues in tar-
get populations

Intervention 2.3 Promote targeted undergraduate and postgraduate educational pro-


grammes on the accurate diagnosis and management of common in-
fections for all health care workers, veterinarians, prescribers and
dispensers.

Implementation: ● Develop a National Strategy and make it a national priority


● Identify interested organizations and opinion leaders, educators and
sources of appropriate information
● Create and/or strengthen in-service training, professional development
and continuing education for all health care workers appropriate to
local context and problems.

Who should initiate: ● National Intersectoral Task Force—delegating to suitable interested


organizations and opinion leaders

Who should undertake and manage: ● Organizations delegated by the National Intersectoral Task Force

Who should evaluate: ● National Intersectoral Task Force


● Professional organizations, universities and organizations delegated
by the National Intersectoral Task Force

Process Indicators: ● Opinion leaders identified


● Curriculum developed and implemented; quantitative and qualitative
assessments of educational exposure

Outcome Indicators: ● Levels of knowledge, attitudes and skills regarding management of


common infections and containment of antimicrobial resistance

Intervention 2.8 Encourage development and use of guidelines and treatment algorithms
to foster appropriate use of antimicrobials.

Implementation: ● National Intersectoral Task Force—delegating to suitable interested


organizations, opinion leaders and educators
● Use of evidence-based principles of effective guideline development,
including maximal participation of health care providers most involved
in managing the condition, involvement of end-users, systematic re-
view and appraisal of evidence, involvement of consumers

Who should initiate: ● National Intersectoral Task Force

Who should undertake and manage: ● Organizations delegated by the National Intersectoral Task Force

Who should evaluate: ● National Intersectoral Task Force


● Organizations delegated by the National Intersectoral Task Force

Process Indicators: ● Production of guidelines and dissemination plan

Outcome Indicators: ● Level of uptake and indicators of appropriate use of antimicrobials


among target health care providers

78
IMPLEMENTATION OF THE WHO GLOBAL STRATEGY
INTERVENTIONS—INTERVENTION PRIORITY: FIRST (continued)
Intervention 3.1 Establish Infection Control Programmes, based on current best practice,
with the responsibility for effective management of antimicrobial resist-
ance in hospitals and ensure that all hospitals have access to such a pro-
gramme.

Implementation: ● Establishment by government mandate


● Where possible the infection control programme should be part of
hospital (public and private) accreditation
● Sufficient resources should be allocated for implementation

Who should initiate: ● Hospital management delegating to an infection control committee

Who should undertake and manage: ● Infection Control Committee

Who should evaluate: ● National Intersectoral Task Force


● Ideally, external audit by a competent authority delegated by the Na-
tional Intersectoral Task Force; in the absence of external evaluation,
use benchmarking to other comparable institutions

Process Indicators: ● Infection control strategies, policies, guidelines documented


● Evidence of relevant data collection

Outcome Indicators: ● Data being used to reduce rates of hospital-acquired infection and
antimicrobial resistance below an agreed target

Intervention 3.5 Ensure access to microbiology laboratory services that match the level
of the hospital, e.g. secondary, tertiary.

Implementation: ● Hospital management, through government if appropriate


● Sufficient resources should be allocated for establishment and main-
tenance of laboratories

Who should initiate: ● Hospital management—in consultation with appropriately trained


staff and learned societies

Who should undertake and manage: ● Microbiologists, or medical/scientific staff adequately trained in micro-
biology

Who should evaluate: ● Benchmarking by Microbiology and Hospital management to other


laboratories servicing similar institutions about range of diagnostic
and susceptibility tests

Process Indicators: ● Implementation of Recommendations 3.6 and 3.7

Outcome Indicators: ● Implementation of Recommendations 3.6 and 3.7

Intervention 3.6 Ensure performance and quality assurance of appropriate diagnostic


tests, microbial identification, antimicrobial susceptibility tests of key
pathogens, and timely and relevant reporting of results.

Implementation: ● Microbiology laboratory

Who should initiate: ● Microbiology laboratory management

Who should undertake and manage: ● Microbiology laboratory management

Who should evaluate: ● An internal and external (national or international) quality assurance
programme
● National Laboratory accreditation schemes where they exist

Process Indicators: ● Evidence of participation in quality assurance activities

79
WHO GLOBAL STRATEGY FOR CONTAINMENT OF ANTIMICROBIAL RESISTANCE • WHO/CDS/CSR/DRS/2001.2
INTERVENTIONS—INTERVENTION PRIORITY: FIRST (continued)
Outcome Indicators: ● Performance level in quality assurance activities
● Continuing laboratory accreditation, where accreditation schemes
exist

Interventions 5.3 and 5.5 5.3 Limit the availability of antimicrobials to prescription-only status, ex-
cept in special circumstances when they may be dispensed on the
advice of a trained health care professional.
5.5 Ensure that only antimicrobials meeting international standards of
quality, safety and efficacy are granted marketing authorization.

Implementation: ● Ministry of Health establishing and delegating to a Government Drug


Regulation Authority

Who should initiate: ● Ministry of Health delegating to a Government Drug Regulation


Authority
● National Intersectoral Task Force

Who should undertake and manage: ● Government Drug Regulation Authority


● National Intersectoral Task Force

Who should evaluate: ● Ministry of Health via Government Drug Regulation Authority
● National Intersectoral Task Force

Process Indicators: ● Presence of appropriate legislation


● Categorization of drugs, GMP inspection in place, restriction of drugs
to registered outlets

Outcome Indicators: ● Results of Regulations enforcement—number of inspections, prosecu-


tions, etc.

Interventions 5.8 and 5.9 5.8 Establish and maintain updated national Standard Treatment Guide-
lines (STGs) and encourage their implementation.
5.9 Establish an Essential Drugs List (EDL) consistent with the national
STGs and ensure the accessibility and quality of these drugs.

Implementation: ● National Intersectoral Task Force—to establish a suitable Committee


consisting of interested organizations, opinion leaders and educators
● Government Drug Regulation Authority

Who should initiate: ● National Intersectoral Task Force—to establish a suitable Committee
consisting of interested organizations, opinion leaders and educators
● Government Drug Regulation Authority

Who should undertake and manage: ● Ministry of Health


● National Intersectoral Task Force

Who should evaluate: ● Ministry of Health


● National Intersectoral Task Force

Process Indicators: ● Production of national Standard Treatment Guidelines and EDL


● Plan for implementation and dissemination

Outcome Indicators ● Level of uptake, including indicators of appropriate use of


antimicrobials among target health care providers and use of EDLs

80
IMPLEMENTATION OF THE WHO GLOBAL STRATEGY
INTERVENTIONS—INTERVENTION PRIORITY: FIRST (continued)
Intervention 5.11 Maximize and maintain the effectiveness of the EDL and STGs by con-
ducting appropriate undergraduate and postgraduate education pro-
grammes of health care professionals on the importance of appropriate
antimicrobial use and containment of antimicrobial resistance.

Implementation: ● Ministry of Health


● National Intersectoral Task Force—delegating to universities and other
training institutions, including suitable interested organizations,
opinion leaders and educators

Who should initiate: ● Ministry of Health


● National Intersectoral Task Force

Who should undertake and manage: ● Training institutions and organizations delegated by the National
Intersectoral Task Force
● Professional bodies responsible for registration of health care profes-
sionals

Who should evaluate: ● National Intersectoral Task Force


● Training institutions and organizations delegated by the National
Intersectoral Task Force

Process Indicators: ● Curriculum developed and implemented; quantitative and qualitative


assessments of educational exposure
● Presence of specific registration requirements for health care profes-
sionals

Outcome Indicators: ● Levels of knowledge, attitudes and skills regarding appropriate anti-
microbial use and containment of antimicrobial resistance
● Assessment of Registration suitability based on continuing education
on antimicrobial use and containment of antimicrobial resistance

81
REFERENCES
References

1. World Health Organization. World Health Assem- 13. World Health Organization. Global tuberculosis
bly (fifty-first). Emerging and other communicable control: WHO Report 2000. Geneva, 2000. WHO/
diseases: antimicrobial resistance. WHA51.17, 1998, CDS/TB/2000.275.
agenda item 21.3.
14. World Health Organization. Assessment of therapeu-
2. World Health Organization. WHO global princi- tic efficacy of antimalarial drugs for uncomplicated
ples for the containment of antimicrobial resistance falciparum malaria in areas of intense transmission.
in animals intended for food. 2000. WHO/CDS/ Geneva, 1996. WHO/MAL/96.1077.
CSR/APH/2000.4. www.who.int/emc/diseases/
15. Williams R. Resistance as a worldwide problem.
zoo/who_global_principles.html
In: Lewis K et al., eds. Bacterial resistance to anti-
3. World Health Organization. WHO report on infec- microbials. Marcel Dekker Inc., 2001 (in press).
tious diseases: Removing obstacles to healthy develop-
16. Bloland P. Drug resistance in malaria. Geneva,
ment. Geneva, 1999. WHO/CDS/99.1.
World Health Organization, 2001. WHO/CDS/
4. Smith RD et al. Cost effectiveness analysis: interven- CSR/DRS/2001.4.
tions against anti-microbial resistance. Interim report
17. Espinal MA. Epidemiology of multidrug-resistant
to the Global Forum for Health Research. 2001 (in
tuberculosis in low and middle-income countries.
preparation).
In: Bastian I, Portaels F eds. Multidrug-resistant
5. Central Intelligence Agency. The global infectious tuberculosis. The Netherlands, Kluwer Academic
disease threat and its implications for the United Publishers, 2000.
States. 1999. www.odci.gov/cia/publications/nie/
18. Pablos-Méndez A et al. Global surveillance for
report/nie99-17d.html
antituberculosis-drug resistance, 1994–1997.
6. Rice LB et al. Outbreak of ceftazidime resistance World Health Organization-International Union
caused by extended-spectrum beta-lactamases at a against Tuberculosis and Lung Disease Working
Massachusetts chronic-care facility. Antimicrob Group on Anti-Tuberculosis Drug Resistance Sur-
Agents Chemother, 1990, 34:2193–2199. veillance. N Engl J Med, 1998, 338:1641–1649.
7. Seppälä H et al. The effect of changes in the con- 19. Sack DA et al. Antimicrobial resistance in shigellosis,
sumption of macrolide antibiotics of erythromy- cholera and campylobacteriosis. Geneva, World
cin resistance in group A streptococci in Finland. Health Organization, 2001. WHO/CDS/CSR/
N Engl J Med, 1997, 337:441–446. DRS/2001.8.
8. Coast J, Smith RD, Millar MR. Superbugs: should 20. Schrag S, Beall B, Dowell SF. Resistant pneumococ-
antimicrobial resistance be included as cost in cal infections: the burden of disease and challenges in
economic evaluation? Health Econ, 1996, 5:217– monitoring and controlling antimicrobial resistance.
226. Geneva, World Health Organization, 2001. WHO/
CDS/CSR/DRS/2001.6.
9. Coast J, Smith RD, Millar MR. An economic
perspective on policy to reduce antimicrobial re- 21. Tapsall, J. Antimicrobial resistance in Neisseria
sistance. Soc Sci Med, 1998, 46:29–38. gonorrhoeae. Geneva, World Health Organization,
2001. WHO/CDS/CSR/DRS/2001.3.
10. Ainsworth M, Teokul W. Breaking the silence: set-
ting realistic priorities for AIDS control in less- 22. Macfarlane J et al. Influence of patients’ expecta-
developed countries. Lancet, 2000, 356:55–60. tions on antibiotic management of acute lower
respiratory tract illness in general practice: ques-
11. Management Sciences for Health. Managing for
tionnaire study. BMJ, 1997, 315:1211–1214.
rational drug use. In: Quick JD et al., eds. Manag-
ing drug supply, 2nd ed. USA, Kumarian Press, 23. Macfarlane JT, Holmes WF, Macfarlane RM.
1997:422–429. Reducing reconsultations for acute lower respira-
tory tract illness with an information leaflet: a
12. World Health Organization. Anti-tuberculosis drug
randomized controlled study of patients in primary
resistance in the world. Report no. 2. Prevalence and
care. Br J Gen Pract, 1997, 47:719–722.
trends. The WHO/IUATLD global project on anti-
tuberculosis drug resistance surveillance. Geneva,
2000. WHO/CDS/TB/2000.278.

83
WHO GLOBAL STRATEGY FOR CONTAINMENT OF ANTIMICROBIAL RESISTANCE • WHO/CDS/CSR/DRS/2001.2
24. Branthwaite A, Pechère J-C. Pan-European survey 40. Buckalew LW, Sallis RE. Patient compliance and
of patients’ attitudes to antibiotics and antibiotic medication perception. J Clin Psychol, 1986, 42:49–
use. J Int Med Res, 1996, 24:229–238. 53.
25. Querubin MP, Tan ML. Old roles, new roles: 41. Bloom BR, Murray CJ. Tuberculosis: commentary
women, primary health care, and pharmaceuticals on a reemergent killer. Science, 1992, 257:1055–
in the Philippines. In: McDonnell K, ed. Adverse 1064.
effects: women and the pharmaceutical industry.
42. Harbath S et al. Prolonged antibiotic prophylaxis
Toronto, Women’s Education Press/Penang Inter-
after cardiovascular surgery and its effects on sur-
national Organization of Consumers’ Unions,
gical site infections and antimicrobial resistance.
1986.
Circulation, 2000, 101:2916–2921.
26. Nichter M, Vuckovic N. Agenda for an anthropol-
43. World Health Organization. Guidelines for estab-
ogy of pharmaceutical practice. Soc Sci Med, 1994,
lishing DOTS-Plus pilot projects for the management
39:1509–1525.
of multidrug-resistant tuberculosis (MDR-TB).
27. t’Hoen E. Direct-to-consumer advertising: for Geneva, 2000. WHO/CDS/TB/2000.279.
better profits or for better health? Am J Health Syst
44. Frieden TR et al. Tuberculosis in New York City—
Pharm, 1998, 55:594–597.
turning the tide. N Engl J Med, 1995, 333:229–
28. World Health Organization. Public education in 233.
rational drug use. Report of an informal consultation,
45. Cockburn J et al. Effects of intervention on antibi-
Geneva, 23–26 November 1993. Geneva, 1994.
otic compliance in patients in general practice. Med
WHO/DAP/94.1.
J Aust, 1987, 147:324–328.
29. World Health Organization. Rational drug use: con-
46. Avorn J, Solomon DH. Cultural and economic
sumer education and information. Geneva, 1996.
factors that (mis)shape antibiotic use: the
DAP/MAC/(8)96.6.
nonpharmacologic basis of therapeutics. Ann
30. Paredes P et al. Intervention trial to decrease unjus- Intern Med, 2000, 133:128–135.
tified use of pharmaceuticals drugs in the treatment
47. Weis SE et al. The effect of directly observed therapy
of childhood diarrhoea, Lima, Peru. Presented at
on the rates of drug resistance and relapse in tuber-
ICIUM Chang Mai 1997. http://www.who.int/
culosis. N Engl J Med, 1994, 330:1179–1184.
dap-icium/posters/3a2_text.html
48. Espinal MA et al. Standard short-course chemo-
31. Vuckovic N, Nichter M. Changing patterns of
therapy for drug-resistant tuberculosis: treatment
pharmaceutical practice in the United States. Soc
outcomes in 6 countries. JAMA, 2000, 283:2537–
Sci Med, 1997, 44:1285–1302.
2545.
32. Haak H. Pharmaceuticals in two Brazilian villages:
49. Sharpe TR, Mikeal RL. Patient compliance with
lay practices and perceptions. Soc Sci Med, 1988,
antibiotic regimens. Am J Hosp Pharm, 1974,
27:1415–1427.
31:479–484.
33. Guillemot D et al. Low dosage and long treatment
50. Couper, MR. Strategies for the rational use of
duration of beta-lactam: risk factors for carriage of
antimicrobials. Clin Infect Dis, 1997, 24 (Suppl
penicillin-resistant Streptococcus pneumoniae. JAMA,
l):S154–S156.
1998, 279:365–370.
51. Standing Medical Advisory Committee, Sub-Group
34. Kunin CM et al. Social, behavioral, and practical
on Antimicrobial Resistance. Main report: The path
factors affecting antibiotic use worldwide: report
of least resistance. London, UK Department of
of Task Force 4. Rev Infect Dis, 1987, 9(Suppl
Health, September 1998.
3):S270–S285.
52. Wilson WR et al. Antibiotic treatment of adults
35. Shapiro MF. Regulating pharmaceutical advertis-
with infective endocarditis due to streptococci,
ing: what will work? CMAJ, 1997, 156:359–361.
enterococci, staphylococci, and HACEK microor-
36. Lipsky MS, Taylor CA. The opinions and experi- ganisms. JAMA, 1995, 274:1706–1713.
ences of family physicians regarding direct-to-
53. Ross-Degnan D et al. Improving pharmaceutical use
consumer advertising. J Fam Pract, 1997, 45:495–
in primary care in developing countries: a critical re-
499.
view of experience and lack of experience. Washing-
37. Morris LA et al. The attitudes of consumers ton, DC, International Network for Rational Use
towards direct advertising of prescription drugs. of Drugs, 1997. (Presented at the International
Public Health Rep, 1986, 101:82–89. Conference on Improving Use of Medicines, April
1997).
38. Trostle JA. Medical compliance as an ideology. Soc
Sci Med, 1988, 27:1299–1308. 54. York University NHS Centre for Reviews and Dis-
semination. Effective health care: getting evidence
39. Sackett D, Snow JC. The magnitude of compli-
into practice. Bulletin on the Effectiveness of Health
ance and non-compliance. In: Haynes RB, Taylor
Service Interventions for Decision Makers, 1999, 5:1–
DW, Sackett D, eds. Compliance in Health Care.
16.
Baltimore, Johns Hopkins University Press, 1979.

84
REFERENCES
55. World Health Organization. Management of the 69. Mölstad S et al. Antibiotics prescription in primary
child with a serious infection or severe malnutrition: care: a 5-year follow-up of an educational pro-
guidelines for care at the first-referral level in devel- gramme. Fam Pract, 1994, 11:282–286.
oping countries. Geneva, 2000. WHO/FCH/CAH/
70. Gani L, Tangkilisan A, Pujilestari L. Improving ra-
00.1.
tional prescribing of physicians: an educational ap-
56. Management Sciences for Health, Drug Manage- proach for acute diarrhoea in children in Jakarta.
ment Project. Interventions and strategies to improve Presented at ICIUM Chang Mai 1997. http://
the use of antimicrobials in developing countries: a www.who.int/dap-icium/posters/2b2_text.html
review. Geneva, World Health Organization, 2001.
71. May FW et al. Outcomes of an educational-
WHO/CDS/CSR/DRS/2001.9.
outreach service for community medical practition-
57. Tomasz A. Multiple-antibiotic-resistant pathogenic ers: non-steroidal anti-inflammatory drugs. Med J
bacteria: a report on the Rockefeller University Aust, 1999, 170:471–474.
Workshop. N Engl J Med, 1994, 300:1247–1251.
72. Thomson O’Brien MA et al. Educational outreach
58. Bosu WK, Ofori-Adjei D. Survey of antibiotic pre- visits: effects on professional practice and health
scribing pattern in government health facilities of care outcomes. Cochrane Database of Syst Rev [com-
the Wassa west district of Ghana. East Afr Med J, puter file], 2000, (2):CD000409.
1997, 74:138–142.
73. Soumerai SB et al. Effect of local medical opinion
59. Hui L et al. Patterns and determinants of use of leaders on quality of care for acute myocardial inf-
antibiotics for acute respiratory tract infection in arction. A randomized controlled trial. JAMA,
children in China. Pediatr Infect Dis J, 1997, 1998, 279:1358–1363.
16:560–564.
74. Thomson O’Brien MA et al. Local opinion lead-
60. Chalker J, Phuong NK. Combating the growth of ers: effects on professional practice and health care
resistance to antibiotics: antibiotic dose as an indica- outcomes. Cochrane Database of Syst Rev [compu-
tor for rational drug use. Presented at ICIUM Chang ter file], 2000, (2):CD000125.
Mai 1997. http://www.who.int/dap-icium/posters/
75. de Vries TP et al. Guide to good prescribing.
2E1_ txtf.html
Geneva, World Health Organization, 1994. WHO/
61. Gumodoka B et al. Injection practices in Mwanza DAP/94.11.
region, Tanzania: prescriptions, patient demand and
76. de Vries TP et al. Impact of short course in phar-
sterility. Trop Med Int Health, 1996, 1:874–880.
macotherapy for undergraduate medical students:
62. Nyquist A-C et al. Antibiotic prescribing for chil- an international randomised controlled study.
dren with colds, upper respiratory tract infections, Lancet, 1995, 346:1454–1457.
and bronchitis. JAMA, 1998, 279:875–877.
77. Ameyaw MM, Ofori-Adjei D. The impact of three
63. Soumerai SB, McLaughlin T, Avorn J. Improving forms of educational interventions on dispensing prac-
drug prescribing in primary care: a critical analysis tices. Presented at ICIUM Chang Mai 1997. http:/
of the experimental literature. Milbank, 1989, /www.who.int/dap-icium/posters/2b1_txt1.html
67:268–317.
78. Bruneton C, Maritoux J, Fontaine D. Assessment
64. Mabadeje AFB, Taylor O, Abiose AK. Intervention in 7 African countries of the advice given in private
study to reduce prescription cost in the Lagos Univer- drugstores through local researchers role playing
sity Teaching Hospital. Presented at ICIUM Chang customers. Presented at ICIUM Chang Mai 1997.
Mai 1997. http://www.who.int/dap-icium/posters/ http:// www.who.int.int/dap-icium/posters/1b2_
2a3_txt.html fin.html
65. Freemantle N et al. Printed educational materials: 79. Sia IC, Valerio J. The effects of an intervention on
effects on professional practice and health care out- the selling behaviour of sarisari (variety) store keepers
comes. Cochrane Database of Syst Rev [computer in some villages in the Philippines. Presented at
file], 2000, (2):CD000172. ICIUM Chang Mai 1997. http://www.who.int/
dap-icium/posters/3c4_txtf.html
66. Davis D et al. Impact of formal continuing medi-
cal education: Do conferences, workshops, rounds, 80. Grimshaw JM, Russell IT. Effect of clinical guide-
and other traditional continuing education activi- lines on medical practice: a systematic review of
ties change physician behavior or health care out- rigorous evaluations. Lancet, 1993, 342:1317–
comes? JAMA, 1999, 282:867–874. 1322.
67. Avorn J, Soumerai SB. Improving drug-therapy 81. Kristinsson KG. Epidemiology of penicillin resist-
decisions through educational outreach. A ant pneumococci in Iceland. Microbial Drug Re-
randomized controlled trial of academically based sist, 1995, 1:121–125.
“detailing”. N Engl J Med, 1983, 308:1457–1463.
82. Mamun KZ. Prevalence and genetics of resistance to
68. Harvey KJ et al. Educational antibiotic advertis- commonly used antimicrobial agents in faecal
ing. Med J Aust, 1986, 145:28–32. enterbacteriaceae from children in Bangladesh [PhD
thesis]. University of Liverpool, 1991.

85
WHO GLOBAL STRATEGY FOR CONTAINMENT OF ANTIMICROBIAL RESISTANCE • WHO/CDS/CSR/DRS/2001.2
83. Hogerzeil HV et al. Impact of an essential drugs 100. World Health Organization. Progress of WHO
programme on the availability and rational use of Member States in developing national drug policies
drugs. Lancet, 1989, i:141–142. and in revising essential drug lists, September 1998,
WHO Action Programme on Essential Drugs. Ge-
84. Hogerzeil HV et al. Field tests for rational drug
neva, 1998. WHO/DAP/98.7.
use in twelve developing countries. Lancet, 1993,
342:1408–1410. 101. Nicolle L. Infection control programmes to contain
antimicrobial resistance. Geneva, World Health
85. World Health Organization. Management of patients
Organization, 2001. WHO/CDS/CSR/DRS/
with sexually transmitted diseases: Report of a WHO
2001.7.
Study Group, 1991. Geneva, 1991 (WHO Techni-
cal Report Series, No. 810). 102. Albert RK, Condie F. Hand-washing patterns in
medical intensive-care units. N Engl J Med, 1981,
86. World Health Organization. Integrated manage-
304:1465–1466.
ment of childhood illness: a WHO/UNICEF
initiative. WHO Bulletin, 1997, 75, Supplement 1. 103. Graham M. Frequency and duration of hand-
washing in an intensive care unit. Am J Infect
87. Butler CC et al. Understanding the culture of pre-
Control, 1990, 18:77–81.
scribing: qualitative study of general practitioners’
and patients’ perceptions of antibiotics for sore 104. Larson E, Kretzer EK. Compliance with hand-
throats. BMJ, 1998, 317:637–642. washing and barrier precautions. J Hosp Infect,
1995, 30(Suppl):88–106.
88. Fidler DP. Legal issues associated with antimicro-
bial drug resistance. Emerg Infect Dis, 1998, 4:169– 105. Goldmann DA, Huskins WC. Control of noso-
177. comial antimicrobial-resistant bacteria: a strate-
gic priority for hospitals worldwide. Clin Infect
89. Bauchner H. Parents’ impact on antibiotic use.
Dis, 1997, 24(Suppl 1):S139–S145.
APUA Newsletter, 1997, 15:1–3.
106. Riley LW et al. The significance of hospitals as
90. Little P et al. Open randomised trial of prescribing
reservoirs for endemic multiresistant Salmonella
strategies in managing sore throat. BMJ, 1997,
typhimurium causing infection in urban Brazil-
314:722–727.
ian children. J Infect Dis, 1984, 150:236–241.
91. Barden LS et al. Current attitudes regarding use of
107. Ayliffe GAJ. The progressive intercontinental
antimicrobial agents: results from physician’s and
spread of methicillin-resistant Staphylococcus
parents’ focus group discussions. Clin Pediatr, 1998,
aureus. Clin Infect Dis, 1997, 24(suppl 1):S74–
37:665–671.
S79.
92. Norrby SR. Antibiotic resistance: a self-inflicted
108. Pratt RD et al. Virologic characterization of pri-
problem. J Intern Med, 1996, 239:373–375 (edi-
mary human immunodeficiency virus type 1 in-
torial).
fection in a health care worker following
93. Smith RD, Coast J. Controlling antimicrobial needlestick injury. J Infect Dis, 1995, 172:851–
resistance: a proposed transferable permit market. 854.
Health Policy, 1998, 43:219–232.
109. Centers for Disease Control and Prevention. Im-
94. Rafferty T, Wilson-Davis K, McGavock H. How munization of health-care workers: recommen-
has fundholding in Northern Ireland affected pre- dations of the Advisory Committee on
scribing patterns? A longitudinal study. BMJ, 1997, Immunization Practices (ACIP) and the Hospi-
315:166–170. tal Infection Control Practices Advisory Commit-
tee (HICPAC). Morb Mortal Wkly Rep, 1997,
95. Friis H et al. The effect of reimbursement on the
46(RR-18):1–42.
use of antibiotics. Scand J Prim Health Care, 1993,
11:247–251. 110. Centers for Disease Control and Prevention. Rec-
ommendations for prevention and control of
96. Steffensen FH et al. Changes in reimbursement
hepatitis C virus (HCV) infection and HCV-re-
policy for antibiotics and prescribing patterns in
lated chronic disease. Morb Mortal Wkly Rep,
general practice. Clin Microbiol Infect, 1997, 3:653–
1998, 47(RR-19):1–39.
657.
111. Centers for Disease Control and Prevention.
97. Monnet DL, Sørensen TL. Interpreting the effec-
Leads from the MMWR. Acquired immunodefi-
tiveness of a national antibiotic policy and com-
ciency syndrome associated with intravenous-drug
paring antimicrobial use between countries. J Hosp
use—United States, 1988. JAMA, 1989,
Infect, 1999, 43:239–242 (letter).
261:2314–2316.
98. World Health Organization. How to investigate drug
112. Beltrami E et al. Risk and management of blood-
use in health facilities: selected drug use indicators.
borne infections in health care workers. Clin
Geneva, 1993. WHO/DAP/93.1.
Microbiol Rev, 2000, 13:385–407.
99. Hutchinson JM, Foley RN. Method of physician
113. Haley RW et al. The SENIC Project. Study on
remuneration and rates of antibiotic prescription.
the efficacy of nosocomial infection control
CMAJ, 1999, 160:1013–1017.

86
REFERENCES
(SENIC Project). Summary of study design. Am 130. Rifenburg RP et al. Benchmark analysis of strat-
J Epidemiol, 1980, 111:472–485. egies hospitals use to control antimicrobial ex-
penditures. Am J Health Syst Pharm, 1996,
114. SENIC finds that hospitals’ IC programs reduce
53:2054–2062.
infections. Hosp Infect Control, 1982, 9:149–154.
131. Schentag JJ. Understanding and managing mi-
115. Hughes JM. Nosocomial infection surveillance in
crobial resistance in institutional settings. Am J
the United States: historical perspective. Infect
Health Syst Pharm, 1995, 52(6 Suppl 2):S9–S14.
Control, 1987, 8:450–453.
132. Schentag JJ et al. Genesis of methicillin-resistant
116. Mayer JA et al. Increasing handwashing in an in-
Staphylococcus aureus (MRSA), how treatment of
tensive care unit. Infect Control, 1986, 7:259–262.
MRSA infections has selected for vancomycin-
117. Hogerzeil HV. Promoting rational prescribing: an resistant Enterococcus faecium, and the importance
international perspective. Br J Clin Pharmacol, of antibiotic management and infection control.
1995, 39:1–6. Clin Infect Dis, 1998, 26:1204–1214.
118. Woods RK, Dellinger EP. Current guidelines for 133. Kucers A, Street A. Rotation of antimicrobials:
antibiotic prophylaxis of surgical wounds. Am possibilities for success. WHO Drug Information,
Fam Physician, 1998, 57:2731–2740. 1999, 13(2):67–71.
119. Swedish-Norwegian Consensus Group. Antibiotic 134. Urban C et al. Effect of sulbactam on infections
prophylaxis in surgery: summary of a Swedish- caused by imipenen-resistant Acinetobacter
Norwegian Consensus Conference. Scand J calcoaceticus biotype anitratus. J Infect Dis, 1993,
Infect Dis, 1998, 30:547–557. 167:448–451.
120. Leaper DJ. Use of antibiotic prophylaxis in clean 135. Goldmann DA et al. Stategies to prevent and con-
non-implant wounds. J Antimicrob Chemother, trol the emergence and spread of antimicrobial-
1998, 41:501–504. resistant microorganisms in hospitals. A challenge
to hospital leadership. JAMA, 1996, 275:234–
121. McDonald M et al. Single- versus multiple-dose
240.
antimicrobial prophylaxis for major surgery: a
systematic review. Aust N Z J Surg, 1998, 68:388– 136. Pestotnik SL et al. Implementing antibiotic prac-
396. tice guidelines through computer-assisted deci-
sion support: clinical and financial outcomes. Ann
122. Song F, Glenny A-M. Antimicrobial prophylaxis
Intern Med, 1996, 124:884–890.
in colorectal surgery: a systematic review of
randomized controlled trials. Br J Surg, 1998, 137. Rahal JJ et al. Class restriction of cephalosporin
85:1232–1241. use to control total cephalosporin resistance in
nosocomial Klebsiella. JAMA, 1998, 280:1233–
123. Polk HC Jr, Christmas AB. Prophylactic antibi-
1237.
otics in surgery and surgical wound infections.
Am Surg, 2000, 66:105–111. 138. Avorn J et al. Reduction of incorrect antibiotic
dosing through a structured educational order
124. Smaill F, Hofmeyer GJ. Antibiotic prophylaxis for
form. Arch Intern Med, 1988, 148:1720–1724.
cesarean section. Cochrane Database of Syst Rev
[computer file], 2000, (2):CD000933. 139. Aswapokee N, Vaithayapichet S, Komoltri C. The
failure of a preprinted order form to alter physi-
125. Soumerai SB, Avorn J. Efficacy and cost-contain-
cians’ antimicrobial prescribing patterns. J Med
ment in hospital pharmacotherapy: state of the
Assoc Thai, 1992, 75:223–230.
art and future directions. Milbank Mem Fund Q
Health Soc, 1984, 62:447–474. 140. Gyssens IC et al. Implementation of an educa-
tional program and an antibiotic order form to
126. Weekes LM, Brooks C. Drugs and therapeutics
optimize quality of antimicrobial drug use in a
committees in Australia: expected and actual per-
department of internal medicine. Eur J Clin
formance. Br J Clin Pharmacol, 1996, 42:551–
Microbiol Infect Dis, 1997, 16:904–912.
557.
141. Hughes JM, Tenover FC. Approaches to limiting
127. Thomson O’Brien MA et al. Audit and feedback:
emergence of antimicrobial resistance in bacteria
effects on professional practice and health care
in human populations. Clin Infect Dis, 1997,
outcomes. Cochrane Database of Syst Rev [com-
24(Suppl 1):S131–S135.
puter file], 2000, (2):CD000259.
142. Acar JF, Goldstein FW. Consequences of increas-
128. US Congress Report. Office of Technology
ing resistance to antimicrobial agents. Clin Infect
Assessment. Impacts of antibiotic resistant bacte-
Dis, 1998, 27(Suppl 1):S125–S130.
ria. Washington, DC, US Government Printing
Office, 1995. OTA-H-629. 143. Struelens MJ. The epidemiology of antimicrobial
resistance in hospital acquired infections: prob-
129. Levy SB, Burke JP, Wallace CK. Epilogue. Rev
lems and possible solutions. BMJ, 1998, 317:652–
Infect Dis, 1987, 9(Suppl 3):S313–S316.
654.

87
WHO GLOBAL STRATEGY FOR CONTAINMENT OF ANTIMICROBIAL RESISTANCE • WHO/CDS/CSR/DRS/2001.2
144. Barber M et al. Reversal of antibiotic resistance from food-animals. FEMS Microbiol Letter, 1998,
in hospital staphylococcal infection. BMJ, 1960, 168:145–151.
1:11–17.
159. Welton LA et al. Antimicrobial resistance in
145. Giamarellou H, Antoniadou A. The effect of enterococci isolated from Turkey flocks fed
monitoring of antibiotic use on decreasing anti- virginiamycin. Antimicrob Agents Chemother,
biotic resistance in the hospital. In: Antibiotic re- 1998, 42:705–708.
sistance: origins, evolution, selection and spread.
160. van den Bogaard AE et al. High prevalence
Ciba Found Symp, Chichester, Wiley, 1997,
of colonization with vancomycin- and
207:76–92.
pristinamycin-resistant enterococci in healthy
146. Recco R et al. Antibiotic control in a municipal humans and pigs in The Netherlands: is the ad-
hospital. JAMA, 1979, 241:2283–2286. dition of antibiotics to animal feeds to blame?
J Antimicrob Chemother, 1997, 40:454–456.
147. World Health Organization. WHONET 5.
Microbiolog y laboratory database software. 161. Wegener HC et al. Use of antimicrobial growth
Geneva,1999. WHO/CDS/CSR/DRS/99.1. promoters in food animals and Enterococcus
faecium resistance to therapeutic antimicrobial
148. Levy SB, FitzGerald GG, Macone AB. Changes
drugs in Europe. J Emerg Infect Dis, 1999, 5:329–
in the intestinal flora of farm personnel after
335.
introduction of tetracycline-supplemented feed
on a farm. N Engl J Med, 1976, 295:583–588. 162. Bager F et al. Glycopeptide resistance in Entero-
coccus faecium from broilers and pigs following
149. Levy SB. Antibiotic use for growth promotion in
discontinued use of avoparcin. Microb Drug
animals: ecologic and public health consequences.
Resist, 1999, 5:53–56.
J Food Protection, 1987, 50:616–620.
163. Danish Integrated Resistance Monitoring and
150. Stöhr K. Impact of zoonotic salmonella on pub-
Research Programme. DANMAP 99—Consump-
lic health and economics. Southeast Asian J Trop
tion of antimicrobial agents and occurrence of anti-
Med Public Health, 1995, 26(Suppl. 2):7–13.
microbial resistance in bacteria from food animals,
151. Piddock J. Does the use of antimicrobial agents food and humans in Denmark. Statens Serum
in veterinary medicine and animal husbandry Institut, Danish Veterinary and Food Adminis-
select antibiotic-resistant bacteria that infect man tration, Danish Medicines Agency and Danish
and compromise antimicrobial chemotherapy? Veterinary Laboratory, July 2000.
J Antimicrob Chemother, 1996, 38:1–3.
164. Klare I et al. Decreased incidence of VanA-type
152. Dupont HL, Steele JH. Use of antimicrobial vancomycin-resistant enterococci isolated from
agents in animal feeds: implications for human poultry meat and from fecal samples of humans
health. Rev Infect Dis, 1987, 9:447–460. in the community after discontinuation of
avoparcin usage in animal husbandry. Microb
153. Advisory Committee on the Microbiological
Drug Resist, 1999, 5:45–52.
Safety of Food. Report on microbial antibiotic
resistance in relation to food safety. London, UK 165. van den Bogaard AE, Bruinsma N, Stobberingh
Department of Health, 1999. EE. The effect of banning avoparcin on VRE car-
riage in The Netherlands. J Antimicrob Chemother,
154. Advisory Committee on the Microbiological
2000, 46:146–147.
Safety of Food. Antibiotic resistance: Government
accepts the recommendations from the ACMSF. 166. Wierup M et al. Animal consumption of antibi-
Vet Rec, 2000, 146:478–479. otics and chemotherapeutic drugs in Sweden
during 1980, 1982 and 1984. Vet Res Commun,
155. World Health Organization. The medical impact
1987, 11:397–405.
of the use of antimicrobials in food animals: Report
of a WHO meeting, Berlin, Germany, 13–17 Octo- 167. Wierup M. Ten years without antibiotic growth
ber 1997. Geneva, 1997. WHO/EMC/ZOO/ promoters—results from Sweden with special ref-
97.4. erence to production results, alternative disease
preventive methods and the usage of antibacte-
156. European Federation of Animal Health. Survey
rial drugs. In: The medical impact of the use of
of antimicrobial usage in animal health in the Eu-
antimicrobials in food animals. Report and proceed-
ropean Union and Switzerland.1998 (unpub-
ings of a WHO meeting, Berlin, Germany 13–17
lished).
October 1997. Geneva, World Health Organiza-
157. Aarestrup FM et al. Surveillance of antimicrobial tion, 1997:229–235. WHO/EMC/ZOO/97.4.
resistance in bacteria isolated from food animals
168. Franklin A. Current status of antibiotic resistance
to antimicrobial growth promoters and related
in animal production in Sweden. In: The medical
therapeutic agents in Denmark. APMIS, 1998,
impact of the use of antimicrobials in food animals.
106:606–622.
Report and proceedings of a WHO meeting, Berlin,
158. Hammerum AM, Jensen LB, Aarestrup FM. Germany 13–17 October 1997. Geneva, World
Detection of the satA gene and transferability of Health Organization, 1997:223–227. WHO/
virginiamycin resistance in Enterococcus faecium EMC/ZOO/97.4.

88
REFERENCES
169. Ryan CA et al. Massive outbreak of antimicro- 183. Piddock LJV. Quinolone resistance and
bial-resistant salmonellosis traced to pasteurized Campylobacter. In: The medical impact of the use
milk. JAMA, 1987, 258:3269–3279. of antimicrobials in food animals. Report and
proceedings of a WHO meeting, Berlin, Germany
170. Holmberg SD et al. Drug-resistant Salmonella
13–17 October 1997. Geneva, World Health Or-
from animals fed antimicrobials. N Engl J Med,
ganization, 1997:191–199. WHO/EMC/ZOO/
1987, 311:617–622.
97.4.
171. Glynn MK et al. Emergence of multidrug-resist-
184. Bowler I, Day D. Emerging quinolone resistance
ant Salmonella enterica serotype typhimurium
in campylobacters. Lancet, 1992, 340:245
DT104 infections in the United States. N Engl J
(letter).
Med, 1998, 338:1333–1338.
185. Sánchez R et al. Evolution of susceptibilities of
172. Vasallo FJ et al. Failure of ciprofloxacin therapy
Campylobacter spp. to quinolones and macrolides.
for invasive nontyphoidal salmonellosis. Clin
Antimicrob Agents Chemother, 1994, 38:1879–
Infect Dis, 1998, 26:535–536.
1882.
173. Wall PG et al. A case control study of infection
186. Food and Drug Administration. Draft risk assess-
with an epidemic strain of multiresistant Salmo-
ment on the human health impact of fluoroquinolone
nella typhimurium DT104 in England and Wales.
resistant Campylobacter associated with the con-
Comm Dis Rep CDR Rev, 1994, 4:R130–R135.
sumption of chicken. 2000. http://www.fda.gov/
174. Ridley A, Threlfall EJ. Molecular epidemiology cvm/antimicrobial/ra/risk.html
of antibiotic resistance genes in multiresistant
187. Alliance for the Prudent Use of Antibiotics. Anti-
epidemic Salmonella typhimurium DT 104.
biotic resistance: synthesis of recommendations by
Microb Drug Resist, 1998, 4:113–118.
expert policy groups. Geneva, World Health
175. Ramos JM et al. Changes in susceptibility of Organization, 2001. WHO/CDS/CSR/DRS/
Salmonella enteritidis, Salmonella typhimurium, 2001.10.
and Salmonella virchow to six antimicrobial
188. Food and Drug Administration. US New Drug
agents in a Spanish hospital, 1980–1994. Eur J
Application—NDA. 21 CFR section 314.50.
Clin Microbiol Infect Dis, 1996, 15:85–88.
189. European Union. Guidelines on the safety, quality
176. Frost JA, Kelleher A, Rowe B. Increasing
and efficacy of medicinal products. The rules gov-
ciprofloxacin resistance in salmonellas in England
erning medicinal products in the European Union.
and Wales 1991–1994. J Antimicrob Chemother,
Vol. III, 1996.
1996, 37:85–91.
190. Bryant R. The pharmaceutical quality control hand-
177. Threlfall EJ, Ward LR, Rowe B. Increasing inci-
book. Aster Publishing Corporation, 1989.
dence of resistance to trimethoprim and cipro-
floxacin in epidemic Salmonella typhimurium 191. World Health Organization. Counterfeit drugs:
DT104 in England and Wales. Eurosurveillance, report of a joint WHO/IFPMA workshop 1–3 April
1997, 2:81–84. 1992. Geneva, 1992. WHO/DMP/CFD/92.
178. World Health Organization. Use of quinolones in 192. Hvidberg EF. Regulatory implications of good
food animals and potential impact on human health. clinical practice. Towards harmonisation. Drugs,
Report and proceedings of a WHO meeting, Geneva, 1993, 45:171–176.
Switzerland, 2–5 June 1998. Geneva, 1998.
193. de Crémiers F. ICH M4/ The common technical
WHO/EMC/ZDI/98.12.
document (CTD); comparison of clinical docu-
179. Molbak K et al. An outbreak of multidrug-resist- ments and summaries of assessment practices in
ant, quinolone-resistant Salmonella enterica the United States, Europe and Japan. Drug Inf J,
serotype typhimurium DT104. N Engl J Med, 1999, 33:601–614.
1999, 341:1420–1425.
194. Council for International Organizations of Medi-
180. Endtz HP et al. Quinolone resistance in campylo- cal Sciences. Report of the CIOMS Working Group
bacter isolated from man and poultry following III. Guidelines for preparing core clinical safety in-
the introduction of fluoroquinolones in veteri- formation on drugs. 1995.
nary medicine. J Antimicrob Chemother, 1991,
195. t’Hoen E. ISDB: dedicated to ensuring reliable
27:199–208.
drug information. Essential Drugs Monitor, 1997,
181. Tee W et al. Emergence of multidrug resistance 24:11.
in Campylobacter jejuni isolates from three patients
196. British Society of Antimicrobial Chemotherapy.
infected with human immunodeficiency virus.
The clinical evaluation of antibacterial drugs.
Clin Infect Dis, 1995, 21:634–638.
Report of a Working Party of the British Society
182. Smith KE et al. Quinolone-resistant Campylo- of Antimicrobial Chemotherapy. J Antimicrob
bacter jejuni infections in Minnesota, 1992–1998. Chemother, 1989, 23(Suppl B):1–42.
N Engl J Med, 1999, 340:1525–1532.

89
WHO GLOBAL STRATEGY FOR CONTAINMENT OF ANTIMICROBIAL RESISTANCE • WHO/CDS/CSR/DRS/2001.2
197. Beam TR Jr, Gilbert DN, Kunin CM. European 213. World Health Organization. MMV comes of age.
guidelines for anti-infective drug products. Clin TDR News, 1999, 60:6.
Infect Dis, 1993, 17:787–788.
214. Mbelle N et al. Immunogenicity and impact on
198. Jones B et al. Trials to assess equivalence: the im- nasopharyngeal carriage of a nonavalent pneumo-
portance of rigorous methods. BMJ, 1996, coccal conjugate vaccine. J Infect Dis, 1999,
313:36–39. 180:1171–1176.
199. DiMasi JA et al. Research and development costs 215. Mulholland K. Strategies for the control of pneu-
for new drugs by therapeutic category. A study of mococcal diseases. Vaccine, 1999, 17(Suppl
the US pharmaceutical industry. Pharmaco- 1):S79–S84.
economics, 1995, 7:152–169.
216. Mulholland K. Evaluation of vaccines to prevent
200. Craig WA. Pharmacokinetic / pharmacodynamic childhood pneumonia: lessons relevant to plan-
parameters: rationale for antibacterial dosing of ning tuberculosis vaccine trials. Clin Infect Dis,
mice and men. Clin Infect Dis, 1998, 26:1–12. 2000, 30(Suppl 3):S206–S209.
201. MacGowan A. Concentration controlled and con- 217. Mulholland K et al. A randomised trial of a
centration defined clinical trials: do they offer any Haemophilus influenzae type b conjugate vaccine
advantages for antimicrobial chemotherapy? in a developing country for the prevention of
J Antimicrob Chemother, 1996, 37:1–5. pneumonia—ethical considerations. Int J Tuberc
Lung Dis, 1999, 3:749–755.
202. Committee for Proprietary Medicinal Products.
Points to consider on pharmacokinetics and phar- 218. Ivanoff B, Neira M. Vaccination against diarrheal
macodynamics in the development of antibacterial diseases and typhoid fever. Current status and
medicinal products. July 2000. CPMP/EWP/ prospects. Ann Med Interne (Paris), 1998,
2655/99. 149:340–350.
203. Kennedy JG. Over-the-counter drugs: changing 219. Licciardone J. Emerging drug resistance and
the roles of doctors and pharmacists. BMJ, 1996, vaccination for typhoid fever. JAMA, 1998,
312:593–594. 279:579–580 (letter).
204. The Council of European Communities. Euro- 220. Zenilman JM. Emerging drug resistance and vac-
pean Council Directive concerning the classifi- cination for typhoid fever. JAMA, 1998, 279:580.
cation for the supply of medicinal products for
221. Tarr PE et al. Considerations regarding mass vac-
human use. Council Directive 92/26/EEC, 1992.
cination against typhoid fever as an adjunct to
205. Commission of the European Communities. sanitation and public health measures: potential
Opinion of the scientific steering committee on an- use in an epidemic in Tajikistan. Am J Trop Med
timicrobial resistance. European Commission DG Hyg, 1999, 61:163–170.
XXIV, 1999. www.europa.eu.int/comm/dg24/
222. Centers for Disease Control and Prevention.
health/sc/ssc/out50_en.html
Hepatitis B virus: a comprehensive strategy for
206. Hart CA, Kariuki S. Antimicrobial resistance in eliminating transmission in the United States
developing countries. BMJ, 1998, 317:647–650. through universal childhood vaccination. Recom-
mendations of the Immunization Practices Advi-
207. Indalo AA. Antibiotic sale behaviour in Nairobi:
sory Committee (ACIP). Morb Mortal Wkly Rep,
a contributing factor to antimicrobial drug resist-
1991, 40(RR-13):1–25.
ance. East Afr Med J, 1997, 74:171–173.
223. Chen WN, Oon CJ. Human hepatitis B virus
208. Hossain MM, Glass RI, Khan MR. Antibiotic use
mutants: significance of molecular changes. FEBS
in a rural community in Bangladesh. Int J
Lett, 1999, 453:237–242.
Epidemiol, 1982, 11:402–405.
224. Committee for Proprietary Medicinal Products.
209. World Health Organization. Surveillance stand-
Accelerated evaluation of products indicated for
ards for antimicrobial resistance. Geneva, 2001.
serious diseases (life-threatening or heavy disa-
CDS/CSR/DRS 2001.5 (in preparation).
bling diseases). CPMP, 1996, 495/96.
210. Lindtjorn B. Essential drugs list in a rural hospi-
225. Pichichero ME, Cohen R. Shortened course of
tal. Does it have any influence on drug prescrip-
antibiotic therapy for acute otitis media, sinusitis
tion? Trop Doct, 1987, 17:151–155.
and tonsillopharyngitis. Ped Infect Dis J, 1997,
211. Kafuko JM, Zirabamuzaale C, Bagenda D. 16:680–695.
Rational drug use in rural health units of Uganda:
226. Loulergue J et al. Changes in microbial ecology
effect of national standard treatment guidelines on
and use of cloxacillin. J Hosp Infect, 1994, 27:275–
rational drug use. Presented at ICIUM Chang Mai
283.
1997. http://www.who.int/dap-icium/posters/
2f3_text.html 227. Drusano GL. Infection in the intensive care unit:
β-lactamase-mediated resistance among enter-
212. Kettler H. Narrowing the gap between provision
bacteriaceae and optimal antimicrobial dosing.
and need for medicines in developing countries.
Clin Infect Dis, 1998, 27(suppl 1):S111–S116.
London, The Office of Health Economics, 2000.

90
REFERENCES
228. Thomas JK et al. Pharmacodynamic evaluation 244. Commission of the European Communities.
of factors associated with the development of bac- Communication from the Commission to the
terial resistance in acutely ill patients during Council and the European Parliament. Pro-
therapy. Antimicrob Agents Chemother, 1998, gramme for Action: accelerated action on HIV/
42:521–527. AIDS, malaria and tuberculosis in the context of
poverty reduction. COM(2001)96 final, 2001.
229. Milatovic D, Braveny I. Development of resist-
ance during antibiotic therapy. Eur J Clin 245. World Health Organization. Guidelines for the
Microbiol, 1987, 6:234–244. management of drug-resistant tuberculosis. Geneva,
1997. WHO/TB/96.210.
230. Hilf M et al. Antibiotic therapy for Pseudomonas
aeruginosa bacteremia: outcome correlations in a 246. World Health Organization. Interagency Guide-
prospective study of 200 patients. Am J Med, lines. Guidelines for Drug Donations, revised 1999.
1989, 87:540–546. Geneva, 1999. WHO/EDM/PAR/99.4.
231. Zarate CE, Llosa IL. Prescribing habits of Peru- 247. World Health Organization. Multidrug resistant
vian physicians and factors influencing them. Bull tuberculosis. Basis for the development of an
Pan Am Health Organ, 1995, 29:328–337. evidence-based case-management strategy for MDR-
TB within the WHO’s DOTS strategy. Proceedings
232. Avorn J, Chen M, Hartley R. Scientific versus
of 1998 meetings and protocol recommendations.
commercial sources of influence on the prescrib-
Geneva, 1999. WHO/TB/99.260.
ing behavior of physicians. Am J Med, 1982, 73:4–
8. 248. World Health Organization. WHO report on in-
fectious diseases 2000. Overcoming antimicrobial
233. Lexchin J. Interactions between physicians and
resistance. Geneva, 2000. WHO/CDS/2000.2.
the pharmaceutical industry: what does the lit-
erature say? CAMJ, 1993, 149:1401–1407. 249. World Health Organization. Treatment of tuber-
culosis: guidelines for national programmes, 2nd ed.
234. Mansfield P, Lexchin J. MaLAM: networking for
Geneva, 1997. WHO/TB/97.220.
scientific integrity in drug promotion. Essential
Drugs Monitor, 1997, 24:5. 250. World Health Organization. Anti-tuberculosis drug
resistance in the world. The WHO/IUATLD global
235. Lexchin J. Enforcement of codes governing phar-
project on anti-tuberculosis drug resistance surveil-
maceutical promotion: what happens when com-
lance. Geneva, 1997. WHO/TB/97.229.
panies breach advertising guidelines? CMAJ,
1997, 156:351–356. 251. Espinal MA et al. Rational ‘DOTS Plus’ for the
control of MDR-TB. Int J Tuberc Lung Dis, 1999,
236. The Council of European Communities. Euro-
3:561–563.
pean council directive on the advertising of
medicinal products for human use. Council 252. Kidane G, Morrow RH. Teaching mothers to
Directive 92/28/EEC, 1992. provide home treatment of malaria in Tigray,
Ethiopia: a randomised trial. Lancet, 2000,
237. Food and Drug Administration. Draft policy
356:550–555.
statement on industry-supported scientific and
educational activities (notice). Federal Register, 253. Nosten F et al. Randomised double-blind
1992, 57:56412–56414. placebo-controlled trial of SPf66 malaria vaccine
in children in northwestern Thailand. Lancet,
238. Food and Drug Administration. Advertising and
1996, 348:701–707.
promotion; guidances (notice). Federal Register,
1996, 61:52800–52801. 254. World Health Organization. Framework for de-
veloping, implementing and updating antimalarial
239. World Health Organization. Ethical criteria for
treatment policy in Africa. A guide for country
medicinal drug promotion. Geneva, 1988.
malaria control programmes. Harare, 2001 (in
240. International Federation of Pharmaceutical preparation).
Manufacturers Associations. IFPMA code of phar-
255. Hammer SM, Yeni P. Antiretroviral therapy:
maceutical marketing practices. 1994.
where are we? AIDS, 1998, 12(Suppl A):S181–
241. Association of the British Pharmaceutical Indus- S188.
try. ABPI code of practice for the pharmaceutical
256. Erickson JW, Gulnik SV, Markowitz M. Protease
industry. In: ABPI Compendium. Datapharm Pub-
inhibitors: resistance, cross-resistance, fitness and
lications Limited, 1998.
the choice of initial and salvage therapies. AIDS,
242. World Health Organization. International health 1999, 13(Suppl A):S189–S204.
regulations (1969), 3rd annotated ed. Geneva,
257. Condra JH et al. Drug resistance and predicted
1983.
virologic responses to human immunodeficiency
243. The United Nations Development Programme. virus type 1 protease inhibitor therapy. J Infect
Global public goods. International cooperation in Dis, 2000, 182:758–765.
the 21st century. Oxford, Oxford University Press,
1999.

91
WHO GLOBAL STRATEGY FOR CONTAINMENT OF ANTIMICROBIAL RESISTANCE • WHO/CDS/CSR/DRS/2001.2
258. Swanstrom R, Erona J. Human immunodefi- 260. World Health Organization. Containing antimi-
ciency virus type-1 protease inhibitors: therapeu- crobial resistance. Review of the literature and re-
tic successes and failures, suppression and port of a WHO workshop on the development of a
resistance. Pharmacol Ther, 2000, 86:145–170. global strategy for the containment of antimicrobial
resistance. Geneva, Switzerland, 4–5 February
259. Vella S, Palmisano L. Antiretroviral therapy: state
1999. Geneva, 1999. WHO/CDS/CSR/DRS/
of the HAART. Antiviral Res, 2000, 45:1–7.
99.2.

92
Annexes
ANNEX A
ANNEX A

National Action Plans

Canada:
http://www.hc-sc.gc.ca/hpb/lcdc/bid/nosocom/fact1.html

European Union:
http://www.earss.rivm.nl/

France:
http://www.invs.sante.fr/

Norway:
http://odin.dep.no/shd/norsk/publ/handlingsplaner/030005-990326/index-dok000-b-n-a.html

Sweden:
http://www.sos.se/FULLTEXT/0000-044/0000-044.htm

United Kingdom:
http://www.doh.gov.uk/publications/pointh.htm

USA (Centers for Disease Control and Prevention, Atlanta):


http://www.cdc.gov/drugresistance/actionplan/

95
WHO GLOBAL STRATEGY FOR CONTAINMENT OF ANTIMICROBIAL RESISTANCE • WHO/CDS/CSR/DRS/2001.2

ANNEX B

Participation in WHO Consultations

WHO Global Strategy for the Containment Dr Keith Klugman, The South African Institute for
of Antimicrobial Resistance Medical Research, PO Box 1038, Johannesburg
2000, South Africa
Workshop to develop the framework document (260)
Geneva, 4–5 February 1999 Dr Richard Laing, Associate Professor, Department of
International Health, Boston University School of
Public Health, 715 Albany St, Boston, MA 02118-
List of participants 2526, USA

Dr Tasleem Akhtar, Pakistan Medical Research Coun- Dr David Lee, Deputy Director, Drug Management
cil, Shahnaki-e-Jamurait Sector G5/2, Islamabad, Program, Management Sciences for Health,
Pakistan Arlington, USA

Dr Susan Bacheller, Office of Health and Nutrition, Dr Joel Lexchin, 121 Walmer Road, Toronto, Canada
USAID/G/PHN/HN/HPSR, Washington, USA Dr Donald E Low, Microbiologist-in-Chief, Mount
Dr Richard Bax, Director and Vice-President, Anti- Sinai Hospital, The Toronto Hospital, Toronto,
infective Therapeutic Unit, Clinical Research and Canada
Development, SmithKline Beecham Pharmaceuti- Dr Peter Mansfield, Director, MaLAM, Australia
cals, Harlow, Essex, UK
Dr Shaheen Mehtar, Western Cape, South Africa
Dr Tom Bergan, President, International Society of
Chemotherapy, Institute of Medical Microbiology, Dr Le Van Phung, Central Biomedical Laboratory,
Rikshospitalet (National Hospital), Oslo, Norway Hanoi Medical School, Hanoi, Vietnam

Dr Nancy Blum, United States Pharmacopeia, Dr Mair Powell, Medicines Control Agency, Market
Rockville, USA Towers, Room 1534, 1 Nine Elms Lane, London,
UK
Dr Otto Cars, Department of Infectious Diseases,
Uppsala University Hospital, Uppsala, Sweden Dr Gro Ramster Wesenberg, Norwegian Medicine
Control Authority, Sven Oftedsalsvei 6, Oslo 0950,
Dr Keryn Christiansen, Clinical Microbiologist, De- Norway
partment of Microbiology & Infectious Diseases,
Royal Perth Hospital, Western Australia Dr Dennis Ross-Degnan, DACP, Drug Policy Research
Group, Department of Ambulatory Care and Pre-
Dr Andres de Francisco, International Health Special- vention, Harvard Medical School, Boston, USA
ist, Global Forum for Health Research, c/o World
Health Organization, 1211 Geneva 27, Switzerland Dr Budiono Santoso, Department of Clinical Pharma-
cology, Faculty of Medicine, Gadjah Mada Uni-
Dr David Fidler, Indiana University School of Law, 211 versity Sekip, Yogyakarta, Indonesia
South Indiana Avenue, Bloomington IN 47405-
1001, USA Dr Anthony Savelli, Director, Rational Pharmaceuti-
cal Management, Management Sciences for Health,
Professor Widjoseno Gardjito, Department of Surgery, Arlington, USA
Dr Soetomo Hospital, Jalan Professor Dr Moestopo
6–8, Surabaya 60286, Indonesia Dr Ben Schwartz, National Center for Infectious Dis-
eases, Centers for Disease Control and Prevention,
Dr Judy Gilley, (British Medical Association), Corn- Atlanta, USA
wall House Surgery, Cornwall Road, London N3
1LD, UK Dr Wing Hong Seto, Department of Microbiology,
Queen Mary Hospital, Hong Kong
Dr Neal Halsey, Director of Division of Disease Con-
trol, Johns Hopkins University, Baltimore, USA Dr Walter Stamm, Head, Division of Allergy and In-
fectious Diseases, University of Washington, Seattle,
Professor Pentti Huovinen, Antimicrobial Research USA
Laboratories, National Public Health Institute,
Turku, Finland Professor Mark Steinhoff, Department of International
Health, School of Hygiene and Public Health,
Johns Hopkins University, Baltimore, USA

96
ANNEX B
Dr J Todd Weber, National Center for Infectious Dis- Dr Marcelo F Galas, Profesional Servicio Anti-
eases, Centers for Disease Control and Prevention, microbianos, Instituto Nacional de Enferme-dades
Atlanta, USA Infecciosas—ANLIS—”Dr. Carlos G. Malbran”,
Buenos Aires, Argentina
Dr H Wegener, Danish Zoonosis Centre, National
Veterinary Laboratory, Copenhagen, Denmark Dr Manuel Guzmán-Blanco, President of the Commit-
tee on Antibiotics of the Sociedad Panamericana
Professor M Wierup, Swedish Animal Health Service,
de Infectología, (Pan American Society of Infec-
Johanneshov, Sweden
tious Diseases), Unidad de Microbiología y Enf.
Infecciosas, Hospital Vargas, Centro Médico de
Caracas, Caracas, Venezuela
Representatives from USAID
Professor King Holmes, University of Washington,
Dr Susan Bacheller Harborview Medical Center, Seattle, USA
Dr Anthony Boni Dr Abdulrahman Hassan Ishag, Hospitals Administra-
Dr Caryn Miller tion, Department of Curative Medicine, Ministry
of Health, Riyadh, Kingdom of Saudi Arabia
Professor KK Kafle, Institute of Medicine, TU Teach-
WHO Global Strategy for the ing Hospital, Kathmandu, Nepal
Containment of Antimicrobial Resistance Dr Adeeba Kamarulzaman, Associate Professor, Head,
Prioritization and Implementation Workshop Infectious Diseases Unit, Department of Medicine,
Geneva, 12–14 September 2000 University Malaya, Kuala Lumpur, Malaysia
Dr Göran Kronvall, Clinical Microbiology—MTC,
List of participants Karolinska Hospital, Stockholm, Sweden

Dr Samuel Azatyan, Head of the Department of Dr David Lee, Deputy Director, Drug Management
Pharmacovigilance and Rational Use of Drugs, Program, Management Services for Health,
Armenian Drug and Medical Technology Agency Arlington, USA
(ADMTA), Yerevan, Armenia Dra Alina Llop, Directora del Laboratorio Nacional de
Dr Luis Bavestrello, Infectious Diseases Specialist and Referencia de Microbiología, Sub-Directora
Clinical Pharmacologist, Jefe, Unidad de Instituto Medicina Tropical “Pedro Kouri”, La
infectología, Hospital dr. Gustavo Fricke, Viña del Habana, Cuba
Mar, Chile Mrs Precious Matsoso, Department of Health, Preto-
Dr Mike Bennish, Director, Africa Centre for Health ria, South Africa
and Population Studies, Mtubatuba, South Africa Dr Thomas O’Brien, Microbiology Laboratory,
Dr Richard E Besser, Respiratory Diseases Branch (C- Brigham and Women’s Hospital, Boston, USA
23), Centers for Disease Control and Prevention, Dr David Ofori Adjei, Director, Nogouchi Memorial
Atlanta, USA Institute for Medical Research, University of
Dr Christopher C Butler, Senior Lecturer, Department Ghana, Legon, Accra, Ghana
of General Practice, University of Wales College of Dr Philip Onyebujo, Department of Health, Pretoria,
Medicine, Llanedeyrn Health Centre, Cardiff, UK South Africa
Dr John Chalker, Management Services for Health, Associate Professor Neil Paget, Royal Australasian Col-
Arlington, USA lege of Physicians, Sydney, Australia
Professor Ranjit Roy Chaudhury, National Institute of Dr Ricardo Pérez-Cuevas, Investigador Asociado,
Immunology, Shahid Jeet Sing Marg, New Delhi, Unidad de Investigacion Epidemiologica y en
India Servicios de Salud CMN Siglo XXI, Instituto
Dr Narong Chayakula, Secretary General, Food and Mexicano del Seguro Social, Mexico, Mexico
Drug Administration, Ministry of Public Health, Dr Mair Powell, Medical Assessor, Licensing Division,
Muang, Nonthaburi, Thailand Department of Health, Medicines Control Agency,
Professor Thomas Cherian, Christian Medical College, London, UK
Vellore, India Dr Dennis Ross-Degnan, Associate Professor, Drug
Mrs Parichard Chirachanakul, Food and Drug Admin- Policy Research Group, Department of Ambula-
istration, Ministry of Public Health, Muang, tory Care and Prevention, Harvard Medical School,
Nonthaburi, Thailand Boston, USA

Dr Scott Fridkin, Medical Epidemiologist, Hospital Professor Sidorenko Sergei, Department of Microbiol-
Infections Program (E-55), Centers for Disease ogy, Russia Medical Academy of Postgraduate
Control and Prevention, Atlanta, USA Studies, National Research Centre of Antibiotics,
Moscow, Russia

97
WHO GLOBAL STRATEGY FOR CONTAINMENT OF ANTIMICROBIAL RESISTANCE • WHO/CDS/CSR/DRS/2001.2
Dr Richard Smith, Senior Lecturer, Health Economics European Society for Clinical Microbiology and Infectious Dis-
Group, School of Health Policy and Practice, Uni- ease (ESCMID)
versity of East Anglia, Norwich, UK Peter Schoch, ESCMID Basel, Switzerland
Soeparmanto, Dr Sri Astuti S, Kepala Badan Litbang Global Forum for Health Research
Kesehatan, Head, National Institute of Health Re-
search and Development, Jakarta, Indonesia Andres De Francisco, Senior Public Health Specialist,
c/o World Health Organization, Geneva, Switzer-
Dr Christian Trigoso, Head of the Bacteriology De- land
partment, Instituto de Laboratorio de Salud, La Paz,
Bolivia International Association of Medical Laboratory Technolo-
gists (IAMLT)
Dr Peet Tüll, Medical Director, Division of Commu-
Martha A Hjálmarsdóttir, President, Reykjavík, Iceland
nicable Diseases Control, The National Board of
Health and Welfare, Stockholm, Sweden International Committee of the Red Cross
Associate Professor John Turnidge, Women’s and Chil- Ann Aerts, Head of Health Services, Geneva, Switzer-
dren’s Hospital, North Adelaide, Australia land
Dr Kris Weerasuriya, Professor of Pharmacology and International Council of Nurses
Secretary of the Drug Evaluation Sub-Committee Tesfamicael Ghebrehiwet, ICN Consultant, Nursing
(DESC), Ministry of Health, Department of & Health Policy, Geneva, Switzerland
Pharmacology, Faculty of Medicine, University of
Colombo, Colombo, Sri Lanka International Council of Women
Pnina Herzog Ph. C.M.R. Pharm.S., President, Jeru-
salem , Israel
Representatives from WHO Regional Offices
International Federation of Infection Control (IFIC)
Dr Massimo Ciotti, Communicable Diseases, WHO Anna Hambraeus, Division for Hospital Control, Uni-
Regional Office for Europe, Copenhagen versity Hospital, Uppsala, Sweden
Dr Sudarshan Kumari, Regional Advisor, Blood Safety International Federation of Pharmaceutical Manufacturers’
and Clinical Technology, WHO Regional Office Association (IFPMA)
for South East Asia, New Delhi, India
Peter Hohl, Pharma Research Preclinical Infectious
Diseases, F. Hoffmann—La Roche Ltd, Basel, Swit-
zerland
WHO Meeting on International Aspects of
the Containment of Antimicrobial Patricia Hogan, Senior Manager, Pfizer Inc., New York,
Resistance USA

Geneva, 11–12 January 2001 Tony White, Anti-Infectives Strategic Product Devel-
opment, Smithkline Beecham Pharmaceuticals,
Harlow, Essex ,UK
List of participants
International Pharmaceutical Federation (FIP)
Alliance for the Prudent Use of Antibiotics (APUA)
Diane Gal, FIP Project Coordinator, Den Haag, The
Kathleen T Young, Executive Director, Boston, USA Netherlands
American International Health Alliance International Society of Chemotherapy
Thomas O’Brien, Head, Department of Microbiology, Jean-Claude Pechère, Secrétaire général, Université de
Brigham and Women’s Hospital, Boston, USA Génétique et Microbiologie, Université de Geneva
James P Smith, Executive Director, Washington, USA CHU, Geneva 4, Switzerland

Centers for Disease Control and Prevention (CDC) International Society for Infectious Diseases (ISID)
David Bell, Assistant to the Director for Antimicrobial Keryn Christiansen, Co-Chair, ISID Antibiotic Task
Resistance, National Center for Infectious Diseases, Force, Department Microbiology and Infectious
Atlanta, USA Diseases, Royal Perth Hospital, Perth, Australia

Conféderation Mondiale de L’Industrie de la Santé Animale Permanent Mission of Norway to the United Nations Office
(COMISA) and other International Organizations at Geneva
Anthony J Mudd, Vice President/Secretary General, O Christiansen, Counsellor, Geneva, Switzerland
Representative Body of the Worldwide Animal
The Wellcome Trust
Health Industry, Brussels, Belgium
Robert E Howells, Director of Science Programmes,
European Commission—Luxembourg London, UK
Hartmut Buchow, Euroforum Building, Luxembourg Richard Lane, Head of International Programmes, Lon-
don, UK

98
ANNEX B
UNICEF WHO Temporary Advisors
Abdel W El Abassi, UNICEF, New York, USA M Lindsay Grayson, Austin and Repatriation Medical
USAID Rational Pharmaceutical Management Project Centre, Melbourne, Australia
John Chalker, Arlington, USA Stuart B Levy, President APUA, Boston, USA

UK Department of Health Jean-Claude Pechère, also representing the International


Society of Chemotherapy
Jane Leese, Senior Medical Officer, Skipton House,
London, UK Mair Powell, Medicines Control Agency, London, UK
US Department of Health and Human Services /National In- Richard Smith, School of Health Policy and Practice,
stitute of Allergy and Infectious Diseases University of East Anglia, Norwich, UK
Marissa A Miller, Antimicrobial Resistance Program
Officer, Bethesda, Maryland, USA

World Self-Medication Industry (WSMI) Representatives from WHO


Jerome A Reinstein, Director-General, London, UK David Heymann, Executive Director, Communicable
Diseases
World Trade Organization
Guénaël Rodier, Director CSR
João Magalhães, Counsellor, Agriculture and Com-
modities Division, Centre William Rappard, Ge- Hans Troedsson, Director CAH
neva, Switzerland

World Veterinary Association


Herbert P Schneider, Vice-President, AGRIVET Con-
sultants, Windhoek, Namibia

99

Вам также может понравиться