Vous êtes sur la page 1sur 218

ORGANISATION MONDIALE DE LA SANT ANIMALE WORLD ORGANISATION FOR ANIMAL HEALTH ORGANIZACIN MUNDIAL DE SANIDAD ANIMAL

R E V U E
SCIENTIFIQUE ET TECHNIQUE SCIENTIFIC AND TECHNICAL

R E V I E W R E V I S TA
CIENTFICA Y TCNICA Animal vaccination Part 2: scientic, economic, regulatory and socio-ethical aspects Vaccination animale Partie 2 : aspects scientiques, conomiques, rglementaires et socio-thiques Vacunacin animal Parte 2: aspectos cientcos, econmicos, reglamentarios y socio-ticos
Co-ordinated by Coordonn par Coordinado por

P.-P. Pastoret, M. Lombard & A.A. Schudel

VOL. 26 (2)
AUGUST AOT AGOSTO 2007
12, rue de Prony 75017 Paris France Tl. : 33 (0)1 44 15 18 88 E-mail : oie@oie.int Fax : 33 (0)1 42 67 09 87

All OIE publications are protected by international copyright law. Extracts may be copied, reproduced, translated, adapted or published in journals, documents, books, electronic media and any other medium destined for the public, for information, educational or commercial purposes, provided prior written permission has been granted by the OIE. The designations and denominations employed and the presentation of the material in this Review do not imply the expression of any opinion whatsoever on the part of the OIE concerning the legal status of any country, territory, city or area or of its authorities, or concerning the delimitation of its frontiers and boundaries. The views expressed in signed articles are solely the responsibility of the authors. The mention of specic companies or products of manufacturers, whether or not these have been patented, does not imply that these have been endorsed or recommended by the OIE in preference to others of a similar nature that are not mentioned.

Toutes les publications de lOIE sont protges par un copyright international. La copie, la reproduction, la traduction, ladaptation ou la publication dextraits, dans des journaux, des documents, des ouvrages ou des supports lectroniques et tous autres supports destins au public, des ns dinformation, didactiques ou commerciales, requirent lobtention pralable dune autorisation crite de lOIE. Les dsignations et dnominations utilises et la prsentation des donnes gurant dans la Revue ne retent aucune prise de position de lOIE quant au statut lgal de quelque pays, territoire, ville ou zone que ce soit, leurs autorits, aux dlimitations de leur territoire ou au trac de leurs frontires. Les auteurs sont seuls responsables des opinions exprimes dans les articles signs. La mention de socits spciques ou de produits enregistrs par un fabriquant, quils soient ou non protgs par une marque, ne signie pas que ceux-ci sont recommands ou soutenus par lOIE par rapport dautres similaires qui ne seraient pas mentionns.

Todas las publicaciones de la OIE estn protegidas por un copyright internacional. Extractos pueden copiarse, reproducirse, adaptarse o publicarse en publicaciones peridicas, documentos, libros o medios electrnicos, y en cualquier otro medio destinado al pblico, con intencin informativa, didctica o comercial, siempre y cuando se obtenga previamente una autorizacin escrita por parte de la OIE. Las designaciones y nombres utilizados y la presentacin de los datos que guran en la Revista no constituyen de ningn modo el reejo de cualquier opinin por parte de la OIE sobre el estatuto legal de los pases, territorios, ciudades o zonas ni de sus autoridades, fronteras o limitaciones territoriales. La responsabilidad de las opiniones profesadas en los artculos rmados incumbe exclusivamente a sus autores. La mencin de empresas particulares o de productos manufacturados, sean o no patentados, no implica de ningn modo que stos se benecien del apoyo o de la recomendacin de la OIE, en comparacin con otros similares que no hayan sido mencionados.

Ofce international des pizooties, 2007 ISSN 0253-1933 ISBN 978-92-9044-687-3 Vol. (1) ISBN 978-92-9044-688-0 Vol. (2) ISBN 978-92-9044-689-7

Le papier choisi pour limpression de cet ouvrage, tant recycl 50 % et exempt 100 % de chlore et dacide, ne peut pas nuire lenvironnement This book is printed on 50% recycled, 100% chlorine and acid-free environmentally friendly paper El papel escogido para la impresin de este libro est reciclado al 50% y no contiene cloro ni cidos, por lo que no puede causar perjuicio al medio ambiente

Conception maquette / Graphic design / Diseo de la maqueta: J. Prieur Tous les chemins Conception couverture/ Cover design / Diseo de cubierta: P. Blandin, OIE Images de la couverture / Images of the cover / Imgenes de la cubierta: A. Thiermann, P.-P. Pastoret

Rev. sci. tech. Off. int. Epiz., 26 (2), 2007

Contents Sommaire Contenido


Animal vaccination Part 2: scientic, economic, regulatory and socio-ethical aspects Vaccination animale Partie 2 : aspects scientiques, conomiques, rglementaires et socio-thiques Vacunacin animal Parte 2: aspectos cientcos, econmicos, reglamentarios y socio-ticos
Preface . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 307 Prface . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 308 Prlogo . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 310

The scientic and economic basis of vaccination Les fondements scientiques et conomiques de la vaccination Las bases cientcas y econmicas de la vacunacin
A. McLeod & J. Rushton Economics of animal vaccination
.....................................................

conomie de la vaccination animale (rsum) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Aspectos econmicos de la vacunacin animal (resumen)


...................................

313 323 324

K.A. Schat & E. Baranowski Animal vaccination and the evolution of viral pathogens . . . . . . . . . . . . . . . . . . . . . . . . 327 La vaccination des animaux et lvolution des virus (rsum) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 334 Vacunacin de animales y evolucin de los patgenos virales (resumen) . . . . . . . . . . . . . . . . . . . . 335 K. Grein, O. Papadopoulos & M. Tollis Safe use of vaccines and vaccine compliance with food safety requirements . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 339
Utilisation sans risques de la vaccination et conformit des vaccins avec les exigences de la scurit sanitaire des aliments (rsum)
..............

348

Utilizacin segura de vacunas y observancia de las normas sobre higiene de los alimentos en el uso de vacunas (resumen) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 349

P. Vannier, I. Capua, M.F. Le Potier, D.K.J. Mackay, B. Muylkens, S. Parida, D.J. Paton & E. Thiry Marker vaccines and the impact of their use on diagnosis and prophylactic measures . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 351
Les vaccins marqueurs et les consquences de leur utilisation sur le diagnostic et la prophylaxie (rsum) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 363 Vacunas marcadoras y sus consecuencias sobre el diagnstico y medidas de prolaxis (resumen) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 364

300

Rev. sci. tech. Off. int. Epiz., 26 (2)

Regulatory aspects Aspects rglementaires Aspectos reglamentarios


S. Edwards OIE standards for vaccines and future trends
...................................... .................. ..................

373 377 377

Normes de lOIE relatives aux vaccins et tendances pour lavenir (rsum) Normas de la OIE sobre vacunas y tendencias de cara al futuro (resumen)

P.G.H. Jones, G. Cowan, M. Gravendyck, T. Nagata, S. Robinson & M. Waits Regulatory requirements for vaccine authorisation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 379 Exigences rglementaires lies lagrment des vaccins (rsum) . . . . . . . . . . . . . . . . . . . . . . . . . . 392
Requisitos de las reglamentaciones relativas a la autorizacin de comercializacin de vacunas (resumen) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 392

C. Saegerman, M. Hubaux, B. Urbain, L. Lengel & D. Berkvens Regulatory issues surrounding the temporary authorisation of animal vaccination in emergency situations: the example of bluetongue in Europe . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 395
Questions rglementaires lies lautorisation temporaire de vacciner les animaux en situation durgence : lexemple de la vre catarrhale du mouton en Europe (rsum)
...

406

Cuestiones de reglamentacin en torno a la autorizacin temporal de vacunacin de animales en situaciones de emergencia: el ejemplo de la lengua azul en Europa (resumen) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 407

M. Holmes & R.E. Hill International harmonisation of regulatory requirements . . . . . . . . . . . . . . . . . . . . . . . . . 415 Harmonisation internationale des dispositions rglementaires (rsum) . . . . . . . . . . . . . . . . . . . . . 419 Armonizacin internacional de los requisitos reglamentarios (resumen) . . . . . . . . . . . . . . . . . . . . . 420

D.K.J. Mackay Authorisation within the European Union of vaccines against antigenically variable viruses responsible for major epizootic diseases . . . . . . . . . . . . . . . . . . . . . . . 421
Lagrment au sein de lUnion europenne des vaccins dirigs contre des virus pizootiques majeurs prsentant une variabilit antignique (rsum) . . . . . . . . . . . . . . . . . . . . . . . 426 Autorizacin de vacunas contra los virus con variaciones antignicas responsables de las principales enfermedades epizoticas en la Unin Europea (resumen) . . . . . . . . . . . . . . . . 426

L.A. Elsken, M.Y. Carr, T.S. Frana, D.A. Brake, T. Garland, K. Smith & P.L. Foley Regulations for vaccines against emerging infections and agrobioterrorism in the United States of America . . . . . . . . . . . . . . . . . . . . . . . . . . . 429
Rglements applicables aux vaccins contre les maladies infectieuses mergentes et lagrobioterrorisme aux tats-Unis dAmrique (rsum)
...................

440

Reglamentacin sobre vacunas contra infecciones emergentes y bioterrorismo en los Estados Unidos de Amrica (resumen) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 441

Rev. sci. tech. Off. int. Epiz., 26 (2)

301

Socio-ethical aspects Aspects socio-thiques Aspectos socio-ticos


J.-Ch. Audonnet, J. Lechenet & B. Verschuere Lexprimentation animale dans la dcouverte et la production de vaccins usage vtrinaire . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 443
Animal experimentation in the discovery and production of veterinary vaccines (summary) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 448 La experimentacin con animales en relacin con el descubrimiento y la produccin de vacunas de uso veterinario (resumen) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 448

J.M. Scudamore Consumer attitudes to vaccination of food-producing animals . . . . . . . . . . . . . . . . . 451


Comportement des consommateurs lgard de la vaccination des animaux destins lalimentation humaine (rsum) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 457 Actitud del consumidor frente a la vacunacin de animales destinados al consumo humano (resumen) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 458

C.M. Hardy & A.L. Braid Vaccines for immunological control of fertility in animals . . . . . . . . . . . . . . . . . . . . . . . 461 Les vaccins et le contrle immunologique de la fcondit chez les animaux (rsum) . . . . . . . 466 Vacunas para el control inmunolgico de la fertilidad en animales (resumen) . . . . . . . . . . . . . . . 466

D. OBrien & S. Zanker Animal vaccination and the veterinary pharmaceutical industry . . . . . . . . . . . . . . . 471 La vaccination animale et lindustrie pharmaceutique vtrinaire (rsum) . . . . . . . . . . . . . . . . . . 476 Vacunacin animal e industria farmacutica veterinaria (resumen) . . . . . . . . . . . . . . . . . . . . . . . . . . 477

L. Miguens Opinin del sector ganadero sobre el rol de las vacunas en el control y erradicacin de las enfermedades del ganado en Argentina

....

479

Le point de vue des leveurs argentins sur le rle de la vaccination dans le contrle et lradication des maladies du btail (rsum) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 483

The opinion of the production sector on the role of vaccines in the control and eradication of livestock diseases in Argentina

.............

485

P.-P. Pastoret, A.A. Schudel & M. Lombard Conclusions Future trends in veterinary vaccinology . . . . . . . . . . . . . . . . . . . . . . . 489-494 Conclusions Tendances futures de la vaccinologie vtrinaire . . . . . . . . . . 495-501 Conclusiones Tendencias futuras del estudio de vacunas veterinarias . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 503-509

302

Rev. sci. tech. Off. int. Epiz., 26 (2)

Appendices Annexes Anexos & M.C. Horzinek E. Thiry


Vaccination guidelines: a bridge between ofcial requirements and the daily use of vaccines . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 511
Les lignes directrices de la vaccination : un pont entre les besoins ofciels et lutilisation quotidienne des vaccins (rsum) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 516 Las directrices de vacunacin como nexo entre los requisitos ociales y el uso cotidiano de las vacunas (resumen) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 516

A.A. Schudel & M. Lombard Recommendations of the OIE International Conference on the Control of Infectious Animal Diseases by Vaccination, Buenos Aires, Argentina, 13 to 16 April 2004 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 519

A.A. Schudel Vaccines and OIE listed diseases

.....................................................

523

Rev. sci. tech. Off. int. Epiz., 26 (2)

303

Animal vaccination. Part 1: development, production and use of vaccines. Vol. 26 (1) Vaccination animale. Partie 1 : dveloppement, production et utilisation des vaccins. Vol. 26 (1) Vacunacin animal. Parte 1: desarrollo, produccin y utilizacin de vacunas. Vol. 26 (1)
Preface . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 11 Prface . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 12 Prlogo . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 14

P.-P. Pastoret, M. Lombard, A.A. Schudel, J. Plana-Durn & A. Wennberg Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 17 Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 21 Introduccin . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 25

M. Lombard, P.-P. Pastoret & A.-M. Moulin A brief history of vaccines and vaccination . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 29 Une brve histoire des vaccins et de la vaccination (rsum) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 45 Una breve historia de las vacunas y la vacunacin (resumen) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 46

Development and production of vaccines Dveloppement et production des vaccins Desarrollo y produccin de vacunas
C.G. Gay, R. Zuerner, J.P. Bannantine, H.S. Lillehoj, J.J. Zhu, R. Green & P.-P. Pastoret Genomics and vaccine development . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 49 La gnomique et la mise au point de vaccins (rsum) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 61 Genmica y desarrollo de vacunas (resumen) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 62

J.A. Mumford Vaccines and viral antigenic diversity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 69 Les vaccins et la variabilit antignique des virus (rsum) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 85 Vacunas y variabilidad antignica de los virus (resumen) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 86

M. Gottschalk & S. Laurent-Lewandowski Les vaccins face la diversit antignique des bactries . . . . . . . . . . . . . . . . . . . . . . . 91
Vaccine development: strategies for coping with the antigenic diversity of bacteria (summary) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Las vacunas ante la diversidad antignica de las bacterias (resumen)
.......................

99 99

304

Rev. sci. tech. Off. int. Epiz., 26 (2)

J. Vercruysse, T.P.M. Schetters, D.P. Knox, P. Willadsen & E. Claerebout Control of parasitic disease using vaccines: an answer to drug resistance? . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 105
Prophylaxie des maladies parasitaires au moyen de la vaccination : une rponse la rsistance aux mdicaments ? (rsum) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 111 El control de enfermedades parasitarias por las vacunas como posible solucin al problema de la farmacorresistencia (resumen)
....................

112

M. Lombard & A.-E. Fssel Antigen and vaccine banks: technical requirements and the role of the European antigen bank in emergency foot and mouth disease vaccination . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 117
Banques dantigne et de vaccins : prescriptions techniques, et rle de la banque dantigne de lUnion europenne dans la vaccination durgence contre la vre aphteuse (rsum) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 130 Bancos de antgenos y vacunas: requisitos tcnicos y papel del banco europeo de antgenos en vacunaciones de emergencia contra la ebre aftosa (resumen)
.............................................................

131

J.I. Todd Good manufacturing practice for immunological veterinary medicinal products . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 135
Bonnes pratiques de fabrication pour les mdicaments vtrinaires immunologiques (rsum) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 143 Buenas prcticas de fabricacin de productos inmunolgicos veterinarios (resumen) . . . . . . . . 144

G. Mutwiri, V. Gerdts, M. Lopez & L.A. Babiuk Innate immunity and new adjuvants . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 147 Limmunit inne et les nouveaux adjuvants (rsum) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 153 Inmunidad innata y nuevos adyuvantes (resumen) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 153

Why use vaccines? Pourquoi utiliser des vaccins ? Por qu utilizar vacunas?
D.B. Morton Vaccines and animal welfare
.......................................................... ............................................

Les vaccins et le bien-tre des animaux (rsum) Vacunas y bienestar animal (resumen)

.......................................................

157 161 162

D. Ltticken, R.P.A.M. Segers & N. Visser Veterinary vaccines for public health and prevention of viral and bacterial zoonotic diseases

.........................

165

Les vaccins vtrinaires en sant publique et la prvention des zoonoses virales et bactriennes (rsum) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 173 Vacunas veterinarias para la salud pblica y prevencin de enfermedades zoonticas virales y bacterianas (resumen)
................................

173

Rev. sci. tech. Off. int. Epiz., 26 (2)

305

J. Lubroth, M.M. Rweyemamu, G. Viljoen, A. Diallo, B. Dungu & W. Amanfu Veterinary vaccines and their use in developing countries . . . . . . . . . . . . . . . . . . . . . . 179 Les vaccins vtrinaires et leur utilisation dans les pays en dveloppement (rsum) . . . . . . . 196 Las vacunas veterinarias y su utilizacin en los pases en desarrollo (resumen) . . . . . . . . . . . . . 197

N. Marano, C. Rupprecht & R. Regnery Vaccines for emerging infections . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 203 Les vaccins contre les maladies infectieuses mergentes (rsum) . . . . . . . . . . . . . . . . . . . . . . . . . . 210 Vacunas contra infecciones emergentes (resumen) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 211

I. Capua Vaccination for notiable avian inuenza in poultry

.............................. ....

La vaccination des volailles contre linuenza aviaire dclaration obligatoire (rsum)

217 224

Vacunacin de aves de corral contra la inuenza aviar de noticacin obligatoria (resumen) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 225

G. Plumb, L. Babiuk, J. Mazet, S. Olsen, P.-P. Pastoret, C. Rupprecht & D. Slate Vaccination in conservation medicine . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
La vaccination et la mdecine environnementale (rsum) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . La vacunacin en medicina de la conservacin (resumen) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .

229 236 237

T. Wisniewski, J.A. Chabalgoity & F. Goni Is vaccination against transmissible spongiform encephalopathy feasible? . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 243
La vaccination contre lencphalopathie spongiforme transmissible est-elle une option raliste ? (rsum) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 247 Es factible la vacunacin contra la encefalopata espongiforme transmisible? (resumen)
..

248

P.L. Roeder & W.P. Taylor Mass vaccination and herd immunity: cattle and buffalo . . . . . . . . . . . . . . . . . . . . . . . . 253 La vaccination de masse et limmunit de troupeau : bovins et bufes (rsum) . . . . . . . . . . . . . 260 Vacunacin masiva e inmunizacin de los rebaos de vacunos y bfalos (resumen) . . . . . . . . . 261

S. Marangon & L. Busani The use of vaccination in poultry production . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 265 La vaccination dans les levages de volailles (rsum) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 272 Vacunacin en establecimientos avcolas (resumen) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 273

Rev. sci. tech. Off. int. Epiz., 2007, 26 (2), 313-326

Economics of animal vaccination


A. McLeod & J. Rushton
Livestock Information, Sector Analysis and Policy Branch, Animal Production and Health Division, Food and Agriculture Organization (FAO) The views presented in this paper are the authors own and do not constitute an official statement from FAO.

Summary This paper describes the steps that might be used in assessing the economic justification for using vaccination to control animal disease, and the way that vaccination is financed and administered. It describes decisions that have been taken with respect to preserving international trade, and issues related to protection of livelihoods. Regardless of the motivation for vaccination, its costs can usually be shared between the public and private sectors. Cost-effective vaccination requires methods of delivery to be adapted to livestock production systems. The paper concludes by suggesting questions around the use of vaccination that would merit further economic analysis. Keywords Animal disease Economics Livelihood Vaccination

Introduction
The paper addresses two subject areas, namely: the economic justification for using, or not using, vaccination in disease control; and the way that it is financed and administered. Interested parties may include the following: governments deciding whether to permit or forbid the use of vaccination, or make it compulsory farmers deciding whether or not to pay for vaccine for their herds and flocks, and whether to comply with requirements to present their animals for vaccination those who produce and supply vaccine those concerned with the welfare of farmers and their animals. Most readers will be familiar with the basic principles both of vaccination and of the economic analysis of animal health. The former are covered in this issue of the World Organisation for Animal Health (OIE) Scientific and Technical Review. Aspects of economic analysis and modelling of animal health have been dealt with in varying degrees of complexity by many authors (18, 30, 33, 37, 42,

45, 48). General thinking on animal health and livestock economics, including new institutional economics, has also been variously described by many authors (1, 12, 16, 26, 28, 40, 48, 49). A summary of economic methods for animal health was made recently by Rich et al. (32). Rather than repeating what has been well described, this paper concentrates on the practicality of applying economic analysis to assessment of vaccination, by identifying a series of key questions and reviewing the way in which decisions have actually been made in different situations. A stepwise process is used to lay out the most important issues, although in reality, decisions are seldom as linear or clear cut as they are in conceptual models. The process is as follows: a) The first step is to assess whether vaccination is technically viable; that is, whether a vaccine exists and can be applied safely and effectively. b) Assuming that vaccination is technically possible, two economic questions may underpin the decision to apply it, allow it or forbid it, they are: whether the use of vaccination will adversely affect international trade in livestock products

314

Rev. sci. tech. Off. int. Epiz., 26 (2)

whether the use of vaccination could protect livelihoods, particularly those of vulnerable people, or otherwise reduce distress. It is not impossible that conflicting answers will arise, and in this case, it is necessary to weigh objectives and make a choice, or to look for an acceptable compromise. c) The next factors to be considered are the institutional and operational conditions that affect the assumptions underlying economic analysis. Here, two closely related points must be examined: the funding source for vaccination, which may be public or private or a mixture of both regardless of the source of funding, the delivery of vaccination should be cost effective. This means that the delivery process needs to be adapted to the production systems in which vaccination is applied, and the potential recurrent costs of vaccination should be examined carefully before a programme is started. The remainder of the paper examines each of the above points in turn, and then concludes by identifying areas where animal health planners would benefit from further support or new economic analyses to assist in decisions about the use of vaccination in animal disease control.

to be slaughtered in the immediate outbreak control action. For this strategy to work, necessary conditions would include the existence of a safe and effective vaccine, and rapid onset of protection at individual level. Vaccination has been used in stamping out of foot and mouth disease (FMD) outbreaks, for example in the Netherlands, when insufficient personnel and equipment were available for culling. At the time of writing, ring vaccination was being reviewed by several countries for the control of highly pathogenic avian influenza (HPAI) since vaccines have been developed that provide good protection to poultry. The main technical concerns were that vaccinated birds must not increase the risk to humans, since they greatly reduce clinical disease but do not completely eliminate virus shed, and that birds could be adequately protected against spread in the poultry population when full protection was only conferred after two applications of vaccine. There is always a concern when using vaccination in an emergency situation that a sufficient amount of vaccine can be provided at short notice, since vaccine has a finite shelf life and manufacturers cannot afford to maintain large stocks against uncertain future orders. Vaccination has been an important component in eradication programmes, including those involving progressive zoning, such as those for rinderpest in Africa and FMD in Latin America. The technical challenge in using vaccination for eradication is to obtain on a consistent basis sufficiently high levels of herd immunity to prevent virus spread (the protection level needed depends on the reproductive rate of the virus. It can be as high as 80% or considerably lower, depending on the ease of transmission and the opportunities for animals to come into contact), often under difficult field conditions. Rinderpest devastated African cattle herds in the 19th Century and had become very widespread in less virulent forms in the 20th Century. An effective vaccine was produced several years ago that confers lifelong immunity, and later, a thermostable form was developed that could be delivered without a cold chain. It was therefore possible, with annual vaccination programmes, to maintain a level of immunity in the herd sufficient to drive back disease. In many countries disease has reduced to a point where vaccination could be stopped and the few remaining outbreaks controlled with surveillance and stamping out. Vaccination against FMD, although it has been used successfully in control programmes, is less straightforward because the vaccines available against FMD offer only a short span of protection, and bi- or tri-valent vaccines are needed to protect against the many and changing virus strains. When vaccination is used as a preventive measure to keep incidence below certain levels, a vaccine is needed that can be applied effectively (by farmers as well as veterinarians). It is applied in this way to deal with classical swine fever

Technical viability of vaccination


Vaccination may be considered for the following purposes: to stamp out an outbreak in a country, zone (a geographically continuous area separated from infected areas by a surrounding buffer zone) or compartment (a component of a management system under a common biosecurity regime, e.g. the biosecure production, slaughter and processing units of an industrial value chain, with animals and feed moved between units in biosecure transportation) that has previously been free of disease or free of virus to eradicate disease (when it is used in the early stages of a programme to reduce virus load) to establish and then protect a disease-free zone to achieve a low level of incidence of clinical disease in areas where it is endemic. In the first case, that of stamping out a new outbreak, vaccination together with reduced culling may be considered as an alternative to widespread culling. Vaccinating in a ring around an outbreak and culling in a smaller inner ring reduces the number of animals that need

Rev. sci. tech. Off. int. Epiz., 26 (2)

315

(CSF) in parts of Asia and brucellosis in many countries. It is also used against Newcastle disease (ND) and rabies, where there may be reservoirs of infection in wild animals. ND occurs all over the world and sweeps through smallholder poultry flocks from time to time, causing high mortality. Well-tested vaccines exist that can be used as a preventive measure in less than optimal field conditions, and applied even without injection, and they offer the means for individual farmers to protect their flocks. For some important diseases, such as African swine fever (ASF) and bovine spongiform encephalopathy (BSE) vaccines do not exist and the only possibilities for their control are good hygiene management to improve biosecurity of herds or stamping out measures (culling and disinfection together with movement control) to remove outbreaks. Even where good vaccines do exist, vaccination programmes must always be supported by surveillance and backed up by other measures. When the basic technical requirements for using vaccination have been met, it can be examined for economic viability. Two important considerations facing decision makers are the use of vaccination in countries engaged in international trade, and the possible benefits of vaccination in protecting livelihoods, particularly those of vulnerable people. Each of these will now be examined in turn.

reach 47% (23, based on Food and Agriculture Organization [FAO] data). Developing and emerging economies, whose resource base allows them to be competitive, have increasing importance in production, in particular the big three, Brazil, China and India (39). In many countries with strong livestock production growth, there continue to be problems with animal disease control. There has been a shift towards processed products among the industrial producers, in large part because of increased concerns about animal health and food safety regulations. The ability to export livestock and their products depends on equivalence of animal health and food safety conditions, particularly with regard to animal diseases that have been defined as notifiable by the OIE (50). Currently, countries where notifiable diseases are present have limited potential to export unprocessed livestock products or live animals to those that can prove they do not have the same diseases, although it is possible to export processed products such as heat-treated or de-boned meat. For exporting countries wishing to access premium markets, freedom from notifiable diseases forms a major preoccupation for their Veterinary Services and the commercial livestock sector. Increasingly it is being recognised and defined in international guidelines that disease freedom may apply not only to a country but also to a zone or a compartment. While the concept of a zone is quite well accepted, that of the compartment is less so. Thailand has been exploring the possibility of defining disease-free and vaccination-free compartments for export of poultry (22) which might allow targeted vaccination in smallholder flocks outside the compartments. It has been difficult to arrive at a technically and financially viable design that is acceptable to importers. The concept of safe commodity trading also allows for some flexibility. Essentially, it argues that a certified processed product such as de-boned beef from a hazard analysis critical control point (HACCP)-accredited slaughterhouse should be exportable even if vaccination is being applied in other parts of the livestock sector. This has always been possible to some extent, since processed products are exported from countries that vaccinate against certain diseases. It is of great interest in the Horn of Africa, where ruminant meat from extensive systems is the main exportable product. Gradient systems (those that allow different conditions for different parts of the livestock sector) have many attractions. There are, however, concerns that flexible standards in animal health and food safety might actually mean double standards, with premium markets getting better care and attention while others are offered a second-class product. One way to prevent that happening is to pay attention to providing suitable services to all types of production system. Importing countries free of disease and not vaccinating may decline to take animal products from countries, or

Vaccination and international trade


Figure 1 reminds us of the well recognised trend towards increasing international trade in livestock commodities, particularly from developing countries. In 1990 developing countries accounted for 30% of international trade while by the end of 2006 the figure was expected to

14,000 Million tonnes 12,000 10,000 8,000 6,000 4,000 2,000 0 1990 1992 1994 1996 1998 Year Developed countries Developing countries 2000 2002 2004 2006

Fig. 1 Growth in world meat exports Source: Morgan, 2006 (23)

316

Rev. sci. tech. Off. int. Epiz., 26 (2)

zones, that vaccinate, and will try to control rare outbreaks by culling, disinfection and movement control rather than vaccination. A country trying to achieve official recognition of disease freedom can only do so after a progressive control programme and months or years free of any outbreak (the OIE regulations define precise periods for some diseases, for others it is a matter of negotiation between trading partners). Disease freedom when vaccination is applied, even if it can reliably be demonstrated that there is no clinical disease, is never considered equivalent to freedom without vaccination by importing countries that are free without vaccination (e.g. FMD vaccination can be used for a short time to control an outbreak in a previously free area, but it must quickly be withdrawn and the area monitored for some time before trade can recommence), even if the OIE Terrestrial Animal Health Code may not differentiate the requirement. The benefits of achieving and maintaining a premium export market can be considerable. Uruguay spent between US$7 million and US$9 million a year on FMD vaccination prior to 1997. Once it was declared free of FMD, it gained access to a beef market in the United States of America (USA) worth around US$20 million a year above the value of previous domestic sales (17). When a country trades in livestock products on the basis of freedom from certain diseases without vaccination, the critical economic question will be: in the event of a disease outbreak, should vaccination be used to control it? When Botswana experienced a contagious bovine pleuropneumonia (CBPP) outbreak in the 1990s, the decision was made to use slaughter and compensation rather than vaccination for control, in order to re-establish its export market to the European Union (EU) more quickly, although a programme based on vaccination cost only 78% of one based on culling and compensation (19). Thailand was one of the first countries to report HPAI when a series of outbreaks began in Asia in 2003. By March 2004, the cost of compensation alone had been US$46.5 million, paid to 407,338 farmers. With disease in 51 provinces and a silently infected duck population in the wetland areas, it would have been a reasonable decision to move to widespread vaccination. However, Thailands poultry meat export industry was valued at US$597,634,000 in 2003 (it held fifth place in the global export market, falling to 17th in 2004 [6]), so the decision was made to try to eradicate disease without vaccination. A house-to-house x-ray survey was implemented to seek out disease, and all subsequent cases were rapidly investigated, reported and stamped out. These efforts greatly reduced disease but have not yet eliminated it, and recovery of the export trade was achieved largely by expanding processed products. Countries in trading blocs try to create harmonious rules about animal health, so that countries within the bloc can trade with each other under reduced transactions costs.

The countries of the EU try to remain free of notifiable animal diseases without vaccination, and when a member of the bloc experiences an outbreak there is peer pressure to control it by stamping out measures. In the United Kingdom (UK), an outbreak of FMD in 2001 was controlled without the use of vaccination, although at considerable cost in terms of lost animals. There were no major markets threatened at that time, as beef markets were closed due to BSE, the predominant lamb markets were for low value product to Spain and Italy and pork markets were for cuts that did not sell in the UK, but there was a strong political will to eradicate the disease. This was eventually achieved, but not without the loss of considerable numbers of animals, losses in other rural industries, psychological effects on farmers and concerns among the general public about slaughter of healthy animals (36). Risk modelling carried out after the outbreak suggests that vaccination with reduced culling might reduce the numbers of animals culled on disease grounds by 15% to 50%, although culling on welfare grounds might increase slightly (welfare culling may be permitted when animals are kept beyond their normal selling age and farmers cannot afford to feed them) (34). In 2001, the European Commission (EC) and OIE guidelines on the use of vaccination were very inflexible. Since 2001, the EC has revised its directives on FMD control to provide greater flexibility in the use of vaccination. At the same time, the OIE has agreed a reduction in the use of the holding period before freedom from disease can be declared. The development of differentiating infected from vaccinated animals (DIVA) strategies making it possible to distinguish in serological tests between antibodies from disease and those from vaccination offers further potential for limited use of vaccination by exporting countries. However, there has not yet been a reexamination of the UK data to assess whether vaccination would be economically and technically viable under current regulatory conditions. For countries engaged in international trade of live animals or unprocessed products, therefore, there will be hesitation in permitting vaccination to be used in disease control. In the event of an outbreak, a stamping out process that relies on culling and movement control rather than vaccination will promote a faster return to trade, provided that it is successful.

Protecting livelihoods and human well-being


Animal vaccination may have a role to play in protecting the livelihoods of producers and traders of livestock, and individuals employed in the livestock sector, particularly small-scale operators whose livelihoods are vulnerable

Rev. sci. tech. Off. int. Epiz., 26 (2)

317

because they have limited safety nets. Vaccination is likely to be beneficial to livelihoods in the following situations: when it can be used to control disease effectively with minimal depopulation, particularly in situations where adequate compensation for culled animals is not available when it prevents the disruption of national markets or allows markets to be restored quickly, thereby protecting the livelihoods of those who sell into the markets, and the providers of services, and those employed in the livestock sector as an insurance against disease losses by private farmers or an investment in protection of an industry when it minimises the disruption of other sectors linked to livestock, such as leisure and tourism. When stamping out measures are used to deal with an emerging or re-emerging disease, the effect on the national economy of the culling of even quite large numbers of animals may be minimal. Unfortunately, the effect on the livelihoods of those immediately affected may be severe. Depopulation of animals other than occasionally and on a very small scale can be badly damaging to livelihoods of smallholders and small-scale traders who rely on regular or instantly accessible cash flow from their livestock. Where the animals are owned or managed by women, as is often the case with smaller species, income from them tends to be used directly for buying food, and loss of this income has an adverse effect on household food security (7). In small herds and flocks, a certain level of risk is accepted and occasional disease outbreaks are taken as part of normal operation. However, in a widespread outbreak or culling operation where depopulation spans several villages, the safety-net herds and flocks kept with relatives may also be destroyed, leaving no easy means to restock. Even where effective compensation schemes are in place, they seldom cover the cost of lost production time and lost cash flow. In many countries, compensation is limited or faces considerable administrative challenges, which results in payments being inadequate or very delayed. Distress suffered by farmers over destruction of apparently healthy animals is an additional although unquantified impact of widespread culling. Many farm families in Britain suffered from psychological shock after the 2001 FMD outbreak in Britain as did many of the culling teams. More recently, scenes of distraught children hiding birds from culling teams, and farmers refusing to let cullers enter their villages, have been a regular feature of HPAI control. If vaccination reduces culling or the incidence of clinical disease, the positive impact on animal welfare can be seen as a benefit in itself, aside from the psychological impact on people who care for livestock. In the CSF outbreak in the Netherlands in 1997 and 1998 and the FMD outbreak

in the UK in 2001, the large number of animals slaughtered as well as the high costs of control raised the question that much earlier use of vaccination in epidemic control may be appropriate (4). Animal welfare has an increasing importance in the livestock standards of Member Countries of the Organisation for Economic Co-operation and Development (OECD) and in premium markets for animal products. At what point, then, should a government decide to bring vaccination into the control process for epidemic disease? An unchecked outbreak of a rapidly spreading disease may kill large numbers of animals. Halting it by means of a rapid and effective culling scheme will cause short-term distress but very little impact on livelihoods in the long term, particularly if it is possible to provide some form of compensation. When it becomes evident that culling will need to be widespread, or that outbreaks are spreading beyond control, a rapid decision to implement ring vaccination may save the livelihoods of many smallholders. In the areas of China with median poultry density, using ring vaccination in a 5 km zone with limited culling to stamp out an HPAI outbreak, instead of culling in a 3 km zone, has the potential to save the destruction of over 50,000 poultry in just one outbreak (38). Vietnam and Hong Kong are using targeted vaccination for control of HPAI, which has reduced both the number of new cases and the scale of culling. Market shocks from disease outbreaks affect small and large producers, but in different ways, and have varying impacts according to the disease. Market shocks can be caused by consumer worries leading to loss of demand, by very severe depopulation, or by closing of markets on animal health grounds. Zoonotic diseases (i.e. those that can affect both animals and people and can be passed between them) that cause human death have the greatest effect on demand, as consumers lose confidence and switch from eating a product thought to be dangerous to others considered to be safer. Concern over BSE still closes markets from certain exporters (8). HPAI has caused shortterm consumption and price drops for poultry in many affected countries, and in some countries not yet infected. In some cases the prices of substitute proteins have risen. Other diseases such as brucellosis, tuberculosis and cysticercosis are an accepted daily risk in many countries and do not create market shocks. Large exporters have safety nets to deal with immediate production losses, but are very concerned about the maintenance of their markets and for reasons already discussed may prefer not to have vaccination applied. Small-scale operators have limited capacity to deal with loss of animals and the immediate outbreak costs. Large or small producers selling into domestic markets may be worried about quarantine measures, which can require

318

Rev. sci. tech. Off. int. Epiz., 26 (2)

animals to be kept beyond their normal sale time. Prices may fall if animals grow beyond a normal weight range, particularly if many are released onto the market at the same time when quarantine is lifted, and meanwhile the costs of keeping and feeding the animals are higher than normal. Can the use of vaccination prevent any of these market disruptions? Using vaccination to control zoonotic disease outbreaks is unlikely to mitigate market shocks caused by consumer fear, unless certification can be provided and consumers are convinced that products from vaccinated animals are safe to eat. For a purely animal disease, where an outbreak occurs in an exporting country or zone, there may be benefits to both export and local markets from using control methods based on culling rather than vaccination, provided that the outbreak is halted quickly. In Botswana, the export market for beef to the EU from a disease-free zone is an important generator of revenue, and the domestic market is supplied mostly by local producers outside the free zone, where vaccination can be used. The previously mentioned CBPP outbreak resulted in a ban on exports of beef to the EU, which proved damaging to domestic producers as well as exporters, because animals bred for export had to be sold on the domestic market, severely depressing prices (46). Analysis of the Zimbabwe beef sector in 2003 suggested that a similar effect would occur there if an outbreak of FMD closed export markets (27). The decision was made in Botswana to use culling rather than vaccination because it was believed that this would result in the fastest recovery of the export market, and this would be beneficial to all producers. Unfortunately, the outbreak took longer than anticipated to control, and resulted in the destruction of many animals outside the export zone (24). Where the domestic market is the primary market and the export market is small or non-existent, and particularly when disease is endemic, there should be a considerable interest in using vaccination. Domestic markets do not exclude vaccinated animals provided that they are safe for humans to consume and the vaccine has been properly applied. Vietnam has recently examined its options for the control of CSF Of all the measures that can be employed, . quarantine (which does not permit pigs to be moved outside of their communes and therefore limits access to markets) is the most inequitable, because most of the cost is borne by producers while traders have a good chance of maintaining their market margins even though their cash flow may be disrupted (20). If several small outbreaks occur simultaneously (not uncommon), it is more equitable and less costly to apply ring vaccination rather than quarantine. For short-cycle species, if the use of vaccination makes it possible to keep domestic markets open, then it may prove to be very livelihoods-friendly.

When using vaccination against a zoonotic disease, human safety and health must be taken into account and this makes the analysis more complicated. Economic analysis for human health generally uses a cost utility approach, where the expected rise in the number of quality adjusted life years (QUALYs) or disability adjusted life years (DALYs) in the population at risk is compared to the cost of achieving this result. By contrast, economic analysis of livestock production systems is more likely to estimate the monetary value of both benefits and costs. Few assessments of zoonotic disease control have been carried out that give equal attention to the human and animal side, but two examples are Roth et al. (35) on the control of brucellosis in Mongolia and Coleman et al. (5) on the poverty impacts of zoonotic diseases. Current planning for avian and human influenza control has looked at the costs of each side of the programme but it has been extremely difficult to make meaningful estimates of total benefits (3), since the risk of a human pandemic, while real, is almost impossible to quantify. Vaccination of wildlife against rabies as practised in Western Europe has the effect of reducing the costs involved in paying compensation to cattle owners as well as the threat to companion animals and people. In France, vaccination of red foxes was found to be more effective and cheaper than their depopulation as a way of controlling rabies (2). Vaccination can also be seen as private or public insurance to protect livestock. One example from a public perspective would be the widespread use of FMD vaccination in the EU until 1991, when policy shifted to maintaining disease freedom without vaccination. Vaccination is often used by producers as an insurance against loss of animals or loss of productivity, with the relatively small investment in vaccination far outweighing the potential reduction in productivity from disease. Large commercial producers, particularly those in value chains supplying supermarkets, work to tight delivery schedules and cannot afford delays caused by disease. Even for small producers under less stringent production requirements, investment in vaccination can be financially attractive. For example, in Vietnam it can be economically viable for smallholder pig farmers to pay for CSF vaccination, particularly for their breeding sows. The cost of annual vaccination breaks even when incidence is around 2% (20). The livestock sector is never independent of other sectors, and in some countries models exist that make it possible to estimate the full impact of livestock disease. In Botswana, for example, the cattle sector has positive impacts outside of agriculture which can be affected by livestock disease (46). In both Thailand (29) and Malaysia, HPAI outbreaks caused a depression in tourism. The 2001 FMD outbreak in Britain had severe negative effects on rural economies because the ban on walking across farmland discouraged local tourism and FMD and its control generally discouraged international tourism. Approximately

Rev. sci. tech. Off. int. Epiz., 26 (2)

319

3.1 million of the total cost is estimated to have been borne by the public sector and the agri-food sector, with losses of 4.5 billion to 5.3 billion to other sectors, mainly tourism and leisure (4). Vaccination will be most helpful in situations where it reduces the spread of virus, so that the most stringent control measures can be limited to a small area, or reduces the outbreak time, allowing movement controls to be lifted sooner. Where an important tourist industry is affected by a zoonotic disease, human health concerns will take precedence.

In Vietnam, where CSF is endemic, its control by vaccination is treated almost as a private good although it has externalities. Government campaigns are run but not with sufficient regularity to provide high levels of herd immunity, and vaccine is available for purchase from the government by private veterinarians for their clients. In some places it can be bought by farmers or animal health workers from local licensed feed and drug shops (15, 20). Many preventive vaccination programmes have a cost sharing element, even when vaccination is delivered by government campaigns. This reduces the strain on limited government animal health budgets, although governmentrun cost recovery schemes only transfer costs from one part of the economy to another without generating any production, while adding administrative charges (13). Payment at point of delivery is a popular way to get cost sharing in government vaccination programmes but not without problems. In the late 1990s there was a cost recovery scheme for CBPP in Uganda. People distrusted the vaccine and were unwilling to use it, and an increasing scale of charges over a short time compounded the problem. Some private practitioners were unwilling to recommend the use of CBPP vaccine because it adversely affected customer relations (31). Ring fenced taxation, reciprocal (joint funding) schemes and payment at point of delivery, have all been used in the livestock sector. Reciprocal arrangements are used in animal health funds in Australia, to promote risk sharing. Generally, it seems that animal owners are prepared to pay for vaccination when they perceive a benefit from it. Leonard (16) cites an example where externalities, rather than the private/public nature of the good, would lead Ugandan herders to choose vaccination against prevalent epidemic disease (with high private and public benefits) but not to opt for quarantine (with negative private, but high public benefits). In the Gambia farmers are willing to pay for vaccination against peste des petits ruminants and ND when there is an outbreak but less so as a routine preventive measure (personal communication from members of the Veterinary Services). In other situations where the risk is perceived differently by different stakeholders, large commercial producers have opted to cover the costs or even deliver vaccination to smallholders close to them, and thereby reduce the risk to their own herds. It may help to rationalise cost sharing if the direct cost of vaccination is split into its different elements and each considered separately. Vaccination direct costs can be split into sunk or investment costs (including vaccine development and vaccine delivery infrastructure) and variable or recurrent costs (including vaccine delivery and vaccine). The investment costs can be made by either the public or private sector, or a combination of the two. Public investments are justified for diseases where there are

Financing vaccination
Vaccination is financed from a variety of sources, including national governments, international bodies and private organisations; cost sharing is quite common. The policy for funding is usually derived from a mixture of economic concepts and practicality. Conceptually it may be determined on the basis of public and private goods, where a private good is one that is funded entirely or mostly by private individuals (in this case farmers). The conventional rationale behind this decision is that the good in question has high excludability (it is easy to prevent people from using a service if they have not paid for it) and high rivalry (use of it by one person limits use by others) (11). If it has low excludability and low rivalry, it is a public good. Alternatively, the externalities generated by an action may be considered (16). Externalities occur where the actions of one person have effects on others who are not involved in the original transaction, but the person causing the problem is not required to pay compensation for damages. Where high externalities occur and can be traced to a source, it may be possible to apply a polluter pays principle, but in the case of infectious animal diseases this is difficult. It is easy to understand why vaccination against, say, mastitis, or theileriosis might be considered a private good. The presence of mastitis in one herd or flock creates no significant externalities. It is possible to make vaccine available only to those who pay for it, and in circumstances where supply is limited, use by one person might limit use by others. Control of a transboundary disease such as CBPP or CSF , seems to be a different case since there are clear externalities, and it is hard to apply a polluter pays principle. Controlling it in one herd reduces the risk to neighbouring herds that may not have contributed to the control effort. Nevertheless, Twinamasiko (47) made a convincing argument that control of CBPP could be treated as either a private or a public good in different parts of Uganda depending on the prevailing epidemiological situation. Externalities in areas where the disease is widespread, and where the preference among herders is to treat clinical disease with antibiotic, would be different from those in areas where it seldom occurs.

320

Rev. sci. tech. Off. int. Epiz., 26 (2)

strong externalities and important social and poverty reduction issues. Public investments could also reduce the costs of vaccine and its delivery. Payment for variable costs of vaccination depends on the objectives of the disease control (control or eradication), the disease itself (particularly the externalities it generates) and the systems in which the disease is found (extensive/intensive; communal or private land ownership). McInerneys (18) theoretical work paid much attention to the variable cost issues at farm-level, but ignored the investment costs. His framework has been applied in various studies, for example in farm-level disease control measures (40), and has been extended to include fixed costs such as vaccine development and vaccine delivery infrastructure (9, 44). However, making the best use of investment costs in vaccination requires that campaigns be short and sharp in order to eradicate diseases as quickly as possible (36). Unfortunately, many vaccination campaigns are long processes, where herd immunity is developed slowly, and as the impact of disease is reduced, there is less private incentive to vaccinate. Long campaigns favour returns on the original investment cost but are not a useful option to society. It may even be better to delay vaccination until vaccine delivery infrastructure is ready and farmer interest is at a maximum so that herd immunity is built up very quickly.

which it needs to be applied, the number of doses needed to confer protection and the possibility of side effects. Factors specific to the vaccine interact with factors for the species and production system. Even where there is potential for economies of scale, the animal health system may not have the capability to deliver it, and there is a need for investment in veterinary services in many parts of the world (9). The cost of delivering vaccination to an animal under the Pan African Rinderpest Campaign ranged from 0.27 to 1.71 ECU, with vaccine making up 5% to 33% of the cost (41). For a cattle or sheep ranching system in Australia or Latin America, the largest element of vaccination cost is that of collecting animals together for vaccination. As well as the direct costs of assembling animals, there are indirect costs when stressed animals lose weight or even abort fetuses. There are considerable economies in using a vaccine that need only be applied once a year, at a time when animals are being mustered for other activities, and can be given easily to semi-wild animals passing through a crush. For a pastoralist system in Africa, the timing of vaccination may be critical. Rinderpest is the only significant livestock disease to have been almost eradicated. Without the widespread use of strategic vaccination, it is doubtful that eradication could even have been contemplated. When a thermostable vaccine was developed, it became possible to combine traditional, widespread campaigns using cold chains with unconventional and participatory approaches in remote areas, but institutional factors still hampered the design of vaccine delivery. A benefitcost analysis was used (25) to examine the reasons why pastoralists in northern Kenya did not present animals for vaccination, when an effective vaccine was available that created no adverse side effects. The answer was simple: at the time that the study was carried out, rinderpest in northern Kenya was occurring spasmodically, and sometimes in a form that did not cause high mortality in cattle. Because of the constraints of veterinary budgets, vaccination campaigns were held at times when animals were widely scattered. Bringing animals to a central vaccination point required a trek across country where cattle raiding was common. The chance of losing an animal to raiding when bringing it for vaccination was considerably higher than the risk from rinderpest in an unprotected animal. Another consideration in pastoralist systems may be a process that allows the animal owners or herders to administer the vaccine, even if this is done under the supervision of a veterinarian and in a mass campaign, since the indirect costs of lost production will certainly be lower if animals are not stressed. To further reduce indirect costs, a system that allows animals that normally graze together to be vaccinated in cohorts will minimise the chances of being infected with other diseases.

Cost-effectiveness and the delivery process


If vaccination is to be used as part of a disease control strategy, it needs to be delivered in ways that are costeffective (i.e. that minimise the recurrent costs per animal protected), paying equal attention to the effectiveness and cost sides of the equation. Effectiveness is increased if livestock owners want vaccination and it is provided in a form and manner that is convenient to the people who manage the animals and at the same time preserves the quality of the vaccine. Recurrent costs include direct costs (those of getting the vaccine into the animal) as well as indirect costs (lost productivity as a result of vaccination). Direct costs will be reduced if there is a streamlined delivery chain for getting the vaccine to the people who use it, while indirect costs are reduced if the vaccine does not cause adverse reactions and the vaccination process minimises disruption and stress. Because some of the direct costs are lumpy (they cannot be broken down into very small units), such as transport and human resources, there may be some economies of scale attached to mass campaigns, but it is unwise to assume that this will always be true. The cost per protected animal will be determined by a number of factors. Some factors are specific to the vaccine itself: the cost of a dose, the length of time over which it is protective, the way in

Rev. sci. tech. Off. int. Epiz., 26 (2)

321

Smallholder dairy systems require a different approach. Animals are housed in ways that make them easily accessible, in smallholdings that are often close together. Moving them to central vaccination points will result in indirect costs from lost milk production. For private good vaccines, such as the infection-and-treatment method of immunisation against theileriosis, immunisation at home by a private animal health practitioner or the owner is financially optimal. Even where vaccination against infectious diseases is provided through public campaigns, it may be preferable to have the animals vaccinated at home through contracts to animal health workers or private veterinarians and bear the additional direct costs of delivery. Monogastric livestock, with short breeding cycles, offer considerable challenges in designing vaccine delivery. To achieve 80% protection against CSF in the smallholder pig population of Vietnam requires three campaigns a year by the Veterinary Services (20). As an alternative, making vaccine available at local level, through registered providers, gives pig owners the opportunity to vaccinate at the most appropriate time in the production cycle. An established feed and drug shop that invests in vaccine sales can break even in the second year of sales. Successful operation is dependent on farmer confidence and understanding of the benefits, assured supply of quality vaccine and regular inspection to ensure that the cold chain as far as the supplier is maintained. Two provinces that adopted this approach found that vaccine uptake increased, even though owners were asked to pay the full cost of vaccination when it was privately delivered and only part of the cost when it was provided by government campaigns. To effectively immunise backyard chickens can require four to six campaigns a year, which is clearly impractical for Veterinary Services, even in densely populated Asian systems, where the cost of delivering a dose of vaccine can be less than US$0.06 (J. Hinrichs, personal communication), compared to US$0.38 in rural Africa (10). Recognising this, providers of ND vaccination have experimented with vaccines that can be given in feed and water by the poultry owners. Even for commercial systems with confined birds, handling individual birds to deliver vaccine by needle is time consuming and causes production losses, so other means are preferred. Where production cycles make it impractical for government Veterinary Services to administer vaccine, the role of vaccine delivery shifts to the private players in the animal health system, leaving the government with the responsibility for quality control and providing impartial information. Smallholder cattle, as well as sheep, goats, pigs and chickens, are often owned or managed by women, yet few

vaccine delivery processes are designed with womens requirements explicitly taken into account. These might include: small numbers of doses in a bottle to reduce cash payments; delivery to a place near the homestead; training women to recognise whether the provider has an effective quality management system; information on vaccine use in something other than written form, since in some countries illiteracy is highest in rural women; information about vaccination provided to children in schools, since children are often sources of information for their mothers and grandmothers. The Intermediate Technology Development Group in Kenya and FARM-Africa in Ethiopia deliberately trained female community animal health workers and paraveterinarians to work in production systems where women normally care for animals. It has been suggested that research into animal health technology should be measured against a deliverability checklist of accessibility, acceptability, affordability and sustainability in order to make a link from the development of the technology to the expected economic outcome of its application (21). The same factors might provide a check against the assumptions for effective delivery of vaccine. On the supply side, financial sustainability of the delivery is related to the performance of the value chain for vaccination. Value chain approaches have been used extensively in manufacturing and horticulture, and to a lesser extent in the supply of livestock products, but very little in the analysis of livestock service delivery. In addition to cost-effectiveness in protecting individual animals, which is critical for both short, sharp campaigns and longer-term programmes, the economics of vaccination are affected by the length of time over which it needs to be applied. Countries that use vaccination as part of an eradication programme, or to maintain an acceptable incidence level of an endemic disease, may be faced with considerable recurrent costs over a long period. Vietnam, intending to commercialise its pig sector and expand the pork export market, began exploring the possibility of a disease-free zone in 2002. Between ten and fifty outbreaks of FMD were being reported a year to the OIE, while CSF was considered to be endemic, with an incidence rate below 5% annually. After analysing the production and movement patterns of ruminant livestock and pigs, two neighbouring provinces on the north-east coast were identified, that might be progressively declared free, first of FMD and then of CSF over a ten-year period. , Vaccination was expected to play an important part in removing CSF from the area. Figure 2 shows the estimated costs of the scheme year by year, with a gradual shift in recurrent costs from vaccination (which was withdrawn into the buffer zone) towards changes in the management

322

Rev. sci. tech. Off. int. Epiz., 26 (2)

3.00 2.50 2.00 Cost $ million 1.50 1.00 0.50 0.00 1 2 3 4 5 6 7 Project year 8 9 10

Conclusions
Decision makers faced with emerging and recurring diseases need good decision support tools to decide when it makes economic sense to deploy vaccination; these should include not only epidemiological and economic models, although these are important, but information about consumer and producer behaviour that underpins the assumptions made by modellers. Ideally, they would have access to tools applicable for several species, adaptable for different types of major disease, to assess the probability of risk under a range of situations, as well as up to date production, price and trade data, and agreed priorities for different stakeholders against which to measure predictions. These conditions are rarely in place in advance of a crisis, and may not be met even when a longer planning horizon is available. Animal health planners, like planners in other sectors, are forced to make satisficing decisions that make the best use of the available data in a limited time span. Most of the published work on the economics of animal health is based on problems that have already passed, although they carry lessons for the future. The examples described in this paper, together with comparable analyses for other animal health problems, suggest certain imperatives for social scientists charged with supporting the development of animal health systems. a) It is never too early to make contingency plans. Even for emerging diseases with no vaccines currently available, it is possible to develop risk assessments and computer models and carry out sensitivity analysis for different prices and protection levels. There is a growing need to balance the needs of different stakeholders as livestock markets become more globalised, and this means that questions about the use of vaccination need to be asked early in the planning process. Public opinion on animal welfare, and concerns about loss of livelihoods, have thrown up questions about the automatic use of widespread culling to control outbreaks. However, vaccination programmes may require prior investment in the animal health system. b) We are not beginning from zero. There have been more than twenty-five years of work in combining epidemiological and economic models to analyse animal health programmes, including vaccination, in cattle, pig and poultry systems. While models always need to be adapted to make them situation-specific, a lot is now known about developing quantitative tools. However, there are only rare examples of the same tools being reused or further developed. It is time to examine carefully the role that models play in the animal health planning process and whether they might be better used. Some initiatives have been taken in this direction (14, 42).

Change production system Co-ordination Routine movement management

Outbreak combat direct costs Searching for disease Routine vaccination

Fig. 2 Costs of establishing a classical swine fever and foot and mouth disease free zone in two provinces of Vietnam Source: Taylor et al., 2003 (43)

of pigs (chiefly, removal of swill feeding). This pattern would also represent a shift from public to individual private costs, since the costs of compulsory vaccination would be borne by the government (even if revenues were partly raised from an earmarked tax on the livestock sector), while management costs would be borne by individual farmers. Hong Kong used an enormous and rapid culling operation to control an outbreak of HPAI in 1997, culling all commercially reared chickens. In 2001 all poultry in markets were culled. Since 2002, however, the strategy has changed to widespread vaccination and limited culling. Vaccination is now compulsory for all poultry, given that the risk of virus incursion will continue into the foreseeable future, but it is not being used in isolation, as strict biosecurity and surveillance measures have been introduced at markets and retail points. The number of poultry farms is gradually being reduced through ex gratia payments to farmers who surrender their farm licences (38). The examples in this paper suggest that decisions around the use of vaccination are seldom clear-cut and benefit from a wide range of information about the economic and institutional conditions of the livestock sector. The final section examines constraints in applying economic assessment to the planning of animal vaccination and highlights areas that would benefit from further work.

Rev. sci. tech. Off. int. Epiz., 26 (2)

323

c) Animal health economics has placed a lot of emphasis on quantitative modelling. There is a need now to place equal emphasis on the assumptions underlying models, which result from social and political conditions and institutional arrangements. Animal health economists can also borrow from business analysts, using tools such as value chain analysis to revisit the design of animal health systems and the delivery of vaccination and other inputs. Work has been done in all of these areas analysing these conditions, but we have yet to develop planning teams that are truly multi-skilled or contain people with T-shaped skills (i.e. a strong grounding in one disciplinary area, and sufficient training or experience in others to be comfortable with problems that cut across several disciplines). d) Some specific areas would benefit from more attention. One would be a review of the economics of vaccination under the latest regulations, including the use of DIVA. There would also be value in developing more country and production system specific models to decide when to switch to vaccination, and applying them with a range of cultural and institutional assumptions, since outbreaks of notifiable diseases are prone to taking longer than anticipated to control, and having a more costly impact

than expected. Given the rise in emerging zoonotics, and the increasing interest in food safety in most countries, these areas may require more attention in the future. Finally, there is room for a more detailed review of ways to make vaccination more cost-effective. These may include regional co-operation in vaccination task forces, and increasing use of animal health workers and private individuals in vaccine delivery, with a variety of organisational structures. This is an interesting time for animal health economists. With a body of knowledge on which to build, technology to assist with analysis, and increasing understanding of the importance of many different aspects of social science in planning and policy making, there are excellent opportunities to fine tune the planning of animal health, including the use of vaccination, to make it user-friendly and cost-effective under many circumstances.

conomie de la vaccination animale


A. McLeod & J. Rushton
Rsum Les auteurs proposent une mthode permettant dvaluer pas pas le bienfond conomique de la vaccination en tant quoutil de lutte contre les maladies animales et de vrifier le financement et la gestion des campagnes de vaccination. Ils dcrivent les mesures qui ont t prises pour prserver le commerce international et abordent dautres questions lies la protection des moyens dexistence. Quel que soit le motif lorigine de la vaccination, les secteurs public et priv devraient en partager les cots. Pour que la vaccination soit rentable, il convient de prvoir des modes d'administration adapts aux systmes de production animale. Les auteurs concluent en soulevant un certain nombre de questions conomiques lies la vaccination, qui mriteraient un examen plus approfondi. Mots-cls conomie Maladie animale Moyen dexistence Vaccination.

324

Rev. sci. tech. Off. int. Epiz., 26 (2)

Aspectos econmicos de la vacunacin animal


A. McLeod & J. Rushton
Resumen Tras explicar los pasos que cabra seguir para valorar la conveniencia, desde el punto de vista econmico, de utilizar las vacunas para controlar una enfermedad animal, los autores describen el modo en que se financia y gestiona la vacunacin. Despus exponen las decisiones que se han adoptado con respecto a la proteccin del comercio internacional y a temas relacionados con la salvaguarda de los medios de subsistencia. Con independencia de los motivos que subyazcan a la vacunacin, en general cabe repartir sus costos entre el sector pblico y el privado. Para una campaa rentable se necesitan mtodos para administrar la vacuna adaptados a los sistemas de produccin ganadera. Los autores concluyen proponiendo una serie de temas ligados al uso de la vacunacin que mereceran un anlisis econmico ms detenido. Palabras clave Aspectos econmicos Enfermedad animal Medio de subsistencia Vacunacin.

References
1. Ahuja V. (2004). The economic rationale of public and private sector roles in the provision of animal health services. In Veterinary institutions in the developing world: current status and future needs (C. de Haan, ed.). Rev. sci. tech. Off. int. Epiz., 23 (1), 33-45. 2. Aubert M.F (1999). Costs and benefits of rabies control .A. in wildlife in France. In The economics of animal disease control (B.D. Perry, ed.). Rev. sci. tech. Off. int. Epiz., 18 (2), 533-543. 3. Brambhatt M. (2005). Avian and human pandemic influenza: economic and social impacts. Address given at WHO Headquarters, Geneva, 7-9 November. Available at: http://web.worldbank.org/WBSITE/EXTERNAL/NEWS/0,, contentMDK:20715087~pagePK:34370~piPK:42770~the SitePK:4607,00.html (accessed on 30 May 2007). 4. Burrel A. (2002). Animal disease epidemics: implications for production, policy and trade. Outlook Agric., 31 (3), 151160. 5. Coleman P.G. (2002). Zoonotic diseases and their impact on the poor. In Investing in animal health research to alleviate poverty (B.D. Perry, T.F. Randolph, J.J. McDermott, K.R. Sones & P.K. Thornton, eds). Appendix 9, 1-21. International Livestock Research Institute, Nairobi, Kenya. 6. Food and Agriculture Organization (2006). Key statistics of food and agriculture external trade. Available at: http://www.fao.org/es/ess/toptrade/trade.asp?dir=exp&disp= countrybycomm&resource=1058&ryear=2003 (accessed on 30 May 2007).

7. Food and Agriculture Organization (FAO) (2006). Committee on World Food Security, Thirty-second Session, Rome, 30 October-4 November. Assessment of The World Food Security Situation. Available at: ftp://ftp.fao.org/docrep/ fao/meeting/011/j8096e.pdf (accessed on 30 May 2007).

8. Food and Agriculture Organization (FAO) (2006). June 2006 Meat Market Assessment and Meat Statistics. FAO, Rome.

9. Harrison S.R., Tisdell C. & Ramsey G. (1999). Economic issues in animal health programs. In Understanding animal health in South-East Asia. Advances in the collection, management and use of animal health information. (Australian Centre for International Agricultural Research [ACIAR] Monograph 58, P. Sharma & C. Baldock, eds). ACIAR, Canberra, Australia, 57-72.

Rev. sci. tech. Off. int. Epiz., 26 (2)

325

10. Hinrichs J., Sims L. & McLeod A. (2006). Some direct costs of control for avian influenza. Proc. 11th International Symposium on Veterinary Epidemiology and Economics, 6-11 August, Cairns, Australia. ISVEE, 11, 811. 11. Holden S., Ashley S. & Bazeley P. (1996). Improving the delivery of animal health services in developing countries: a literature review. Livestock in Development, Crewkerne, UK. 12. James A.D. & Carles A. (1996). Measuring the productivity of grazing and foraging livestock. Agricultural Syst., 52 (2), 271-291. 13. James A.D. & Upton M. (1995). Cost recovery for veterinary services. Int. Farm Manag. J., 1, 125-133. 14. Kitching R.P., Thrusfield M.V. & Taylor N.M. (2006). Use and abuse of mathematical models: an illustration from the 2001 foot and mouth disease epidemic in the United Kingdom. In Biological disasters of animal origin: the role and preparedness of veterinary and public health services (M. Hugh-Jones, ed.). Rev. sci. tech. Off. int. Epiz., 25 (1), 293-311. 15. Lan L.T.K. (2000). Epidemiology and economics of classical swine fever at smallholder level in Vietnam. MSc thesis, the University of Reading. 16. Leonard D.K. (2000). Africas changing market for health and veterinary services. The new institutional issues. MacMillan Press, London, 1-39. 17. Leslie J., Barozzi J. & Otte M.J. (1997). The economic implications of a change in FMD policy: a case study in Uruguay. In Proc. 8th International Symposium on Epidemiology and Economics, Paris 8-11 July. pidmiol. Sant anim., 31/32. 18. McInerney J.P. (1996). Old economics for new problems. Livestock disease. Presidential address. J. agric. Econ., 47, 295-214. 19. McLeod A. & Leslie J. (2001). Socio-economic impacts of freedom from livestock disease and export promotion in developing countries. Livestock Policy Discussion Paper No. 3. Food and Agriculture Organization, Rome, June. Available at: http://www.fao.org/ag/againfo/resources/en/ publications/ sector_discuss/PP_Nr3_Final.pdf (accessed on 6 June 2007). 20. McLeod A., Taylor N., Thuy N.T. & Lan L.T.K. (2003). Control of classical swine fever in the Red River Delta of Vietnam. A stakeholder analysis and assessment of potential benefits, costs and risks of improved disease control in three provinces, Phase 3 Report, June 2003. 21. McLeod A. & Wilsmore A. (2002). The delivery of animal health services to the poor: a review. In Investing in animal health research to alleviate poverty (B.D. Perry, T.F Randolph, J.J. McDermott, K.R. Sones & P.K. Thornton, . eds). Appendix 11, 1-24. International Livestock Research Institute, Nairobi, Kenya.

22. Marchot P.J. (2005). Initiative of compartmentalisation in the poultry sector for recovering a partial avian influenza disease free status. Paper for a DLD/EU Workshop on Poultry Farming Compartmentalisation Project in Thailand, August 15, Siam City Hotel, Bangkok, Thailand. 23. Morgan N. (2006). Meating the market: outlook and issues. Presentation made to the International Poultry Council at the seminar on global trends in meat production and the impact of animal diseases, VIV Europe, 16-18 May, Utrecht. 24. Mullins G., Fidzani B. & Koanyane M. (1999). At the end of the day: the socio-economic impacts of eradicating contagious bovine pleuropneumonia from Botswana. In Tropical diseases: control and prevention in the context of the new world order. Proc. 5th biennial conference of the Society for Tropical Veterinary Medicine, Key West, 12-16 June. New York Academy of Science. 25. Ngotho R., McLeod A., Wamwayi H. & Curry J. (1999). Economic effects of rinderpest in pastoralist communities in West Pokot and Turkana. Paper presented to the KARI/DFID NARP II Project End of Project Conference, Kenya Agricultural Research Institute Headquarters, Nairobi, Kenya, 23-26 March 1999. 26. Otte M.J., Nugent R. & McLeod A. (2004). Transboundary animal diseases: assessment of socio-economic impacts and institutional responses. Livestock Policy Discussion Paper No. 9. Food and Agriculture Organization, February. Available at: http://www.fao.org/ag/againfo/resources/en/ publications/sector_discuss/PP_Nr9_Final.pdf (accessed on 6 June 2007). 27. Perry B.D., Randolph T.F Ashley S., Chimedza R., Forman ., T., Morrison J., Poulton C., Sibanda L., Stevens C., Tebele N. & Yngstrom I. (2003). The impact and poverty reduction implications of foot and mouth disease control in southern Africa, with special reference to Zimbabwe. International Livestock Research Institute (ILRI), Nairobi, Kenya. 28. Pica-Ciamarra U. (2005). Livestock policies for poverty alleviation: theory and practical evidence from Africa, Asia and Latin America. Pro-poor Livestock Policy Initiative (PPLPI) Working Paper No. 27. Food and Agriculture Organization, Rome. Available at: http://www.fao.org/ag/ againfo/projects/en/pplpi/docarc/wp27.pdf (accessed on 6 June 2007). 29. Poapongsakorn N. (2004). Dynamics of South East Asian livestock markets and their sanitary and technical standards. Paper prepared for FAO expert consultation on Dynamics of sanitary and technical requirements in domestic livestock markets: assisting the poor to cope, 22-24 June 2004. 30. Putt S.N.H., Shaw A.P.M., Woods A.J., Tyler L. & James A.D. (1988). Veterinary epidemiology and economics in Africa. A manual for use in the design and appraisal of livestock health policy. ILCA Manual No. 3, International Livestock Centre for Africa (Now International Livestock Research Institute), Addis Ababa, Ethiopia.

326

Rev. sci. tech. Off. int. Epiz., 26 (2)

31. RDP Livestock Services Limited (2000). Final evaluation, Pan African Rinderpest Campaign Project Phase II, Uganda, Project 6 Acp Rpr 536/7 Acp Ug 048, Final Report. 32. Rich K.M., Miller G.Y. & Winter-Nelson A. (2005). A review of economic tools for the assessment of animal disease outbreaks. Rev. sci. tech. Off. int. Epiz., 24 (3), 833-845. 33. Rich K.M., Winter-Nelson A. & Miller G.Y. (2005). Enhancing economic models for the analysis of animal disease. Rev. sci. tech. Off. int. Epiz., 24 (3), 847-856. 34. Risk Solutions (2005). Cost benefit analysis of foot and mouth disease control. A report for the Department for Environment Food and Rural Affairs (DEFRA), May 2005. Available at: http://www.defra.gov.uk/animalh/diseases/fmd/ pdf/costben.pdf (accessed on 4 June 2007). 35. Roth F Zinstag J., Orkhon D., Chimed-Ochir G., Hutton G., ., Cosivi O., Carrin G. & Otte J. (2003). Human health benefits from livestock vaccination for brucellosis: case study. Bull. WHO, 81, 867-876. 36. Rushton J. (2003). Modelling the economic impact of FMD from farm to national level impact. In Proc. Foot and mouth disease international symposium organised by the European Directorate for the Quality of Medicines (EDQM), Strasbourg (France), 17-18 March 2003, 89-96. 37. Rushton J., Thornton P. & Otte M.J. (1999). Methods of economic impact assessment. In The economics of animal disease control (B.D. Perry, ed.). Rev. sci. tech. Off. int. Epiz., 18 (2), 315-342. 38. Sims L. (2006). Assessment of long-term costs for control of highly pathogenic avian influenza in Indonesia, Thailand, Vietnam, Cambodia, Laos and Hong Kong PDR China. Draft FAO working paper. 39. Steinfeld H. & Chilonda P. (2006). Old players, new players. In Livestock Report 2006 (A. McLeod, J. Crook, N. Forlano & C. Ciarlantini, eds). Food and Agriculture Organization, Rome. 40. Stott A.W. (2005). Costs and benefits of preventing animal diseases: a review focusing on endemic diseases. Report to Scottish Executive Environment and Rural Affairs Department (SEERAD) Advisory Activity 211. Available at: http://www.scotland.gov.uk/library5/environment/cbpad00.asp (accessed in October 2005). 41. Tambi E.N., Maina O.W., Mukhebi A.W. & Randolph T.E. (1999). Economic impact assessment of rinderpest control in Africa. In The economics of animal disease control (B.D. Perry, ed.). Rev. sci. tech. Off. Int. Epiz., 18 (2), 458-477.

42. Taylor N.M. & James A.D. (2003). Comparison of two cases where epidemiological modelling was used to support decisions regarding foot-and-mouth disease control in UK. In Proc. 10th Symposium of the International Society of Veterinary Epidemiology and Economics, 17-21 November, Santiago, Chile. 43. Taylor N.M, McLeod A., Thuy N.T., Stone M., Binh V.T., Lan L.T.K., Dung D.H. & Barwinek F (2003). Examining . the options for a livestock disease-free zone in the Red River Delta of Vietnam. Strengthening of Veterinary Services in Vietnam (SVSV). ALA/96/20. Supported by the European Commission. 44. Tisdell C.A. (1995). Assessing the approach to cost-benefit analysis of controlling livestock diseases of McInerney and others. Research papers and reports in animal health economics, No. 3, Department of Economics, University of Queensland, Brisbane. 45. Tisdell C.A. (1996). Economics of investing in the health of livestock: new insights? Research papers and reports in animal health economics, No. 29, Department of Economics, University of Queensland, Brisbane. 46. Townsend R., Sigwele H. & MacDonald S. (1998). The effects of livestock diseases in southern Africa: a case study of the costs and control of cattle lung disease in Botswana. Paper prepared for the 1998 Conference of Economic and Social Research Council Development Economics Study Group, the University of Reading, July 1998. 47. Twinamasiko E.K. (2002). Development of an appropriate programme for the control of contagious bovine pleuropneumonia in Uganda. PhD Thesis R8842, University of Reading, UK. 48. Upton M. (1989). Livestock productivity assessment and herd growth models. Agric. Syst, 29, 149-164. 49. Upton M. (1993). Livestock productivity assessment and modelling. Agric. Syst., 43, 459-472. 50. World Organisation for Animal Health (OIE) (2006). Chapter 2.1.1. Criteria for listing diseases. Terrestrial Animal Health Code, 15th Ed. OIE, Paris. Available at: http://www.oie.int/eng/normes/mcode/en_chapitre_2.1.1.ht m#chapitre_2.1.1. (accessed on 6 June 2007).

Rev. sci. tech. Off. int. Epiz., 2007, 26 (2), 327-338

Animal vaccination and the evolution of viral pathogens


K.A. Schat (1) & E. Baranowski (2)
(1) Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, United States of America (2) UMR INRA-ENVT 1225, Ecole Nationale Vtrinaire de Toulouse, 31076 Toulouse, France

Summary Despite reducing disease, vaccination rarely protects against infection and many pathogens persist within vaccinated animal populations. Circulation of viral pathogens within vaccinated populations may favour the development of vaccine resistance with implications for the evolution of virus pathogenicity and the emergence of variant viruses. The high rate of mutations during replication of ribonucleic acid (RNA) viruses is conducive to the development of escape mutants. In vaccinated cattle, unusual mutations have been found in the major antigenic site of foot and mouth disease virus, which is also involved in receptor recognition. Likewise, atypical changes have been detected in the immunodominant region of bovine respiratory syncytial virus. Large deoxyribonucleic acid (DNA) viruses are able to recombine, generating new genotypes, as shown by the potential of glycoprotein E-negative vaccine strains of bovine herpesvirus-1 to recombine with wild-type strains. Mareks disease virus is often quoted as an example of vaccine-induced change in pathogenicity. The reasons for this increase in virulence have not been elucidated and possible explanations are discussed. Keywords Evolution Mareks disease Pathogenicity Pathotype Receptor Tropism Vaccine Virulence Virus.

Introduction
The production of animal proteins for human consumption depends on the reduction or elimination of diseases, which can cause losses directly through morbidity and mortality, and indirectly through increased condemnations at processing plants, decreased growth rates and/or increased susceptibility to other pathogens. The tendency towards increases in size of production units, often associated with the presence of multi-age groups on a farm, complicates the control of pathogens. The developments in the poultry industry provide an excellent

example of the increase in size of production units, with some farms having over one million layers producing eggs for consumption. Although the introduction of many pathogens can be reduced by applying strict biosecurity measures, including the use of filtered-air positive pressure housing, the associated costs often make this approach impractical. The development of veterinary vaccinology has been essential in providing cost-effective approaches to prevent and control infectious diseases in animals. Besides improving the animal health sector, animal vaccination has also substantially enhanced public health by preventing the occurrence of several zoonotic diseases, and reducing the use of veterinary drugs and hence drug residues in the

328

Rev. sci. tech. Off. int. Epiz., 26 (2)

food chain (38). Recent advances in vaccinology have defined new directions for vaccine development strategies, and new generation vaccines are rapidly gaining scientific acceptance (22, 49, 56). Nevertheless, the rapid development of new veterinary vaccines, and their widespread use to protect large populations of animals against a growing number of infectious diseases, have raised questions about the potential consequences of mass vaccination strategies for the evolution of pathogens, in particular rapidly evolving viruses. Because of high population numbers, rapid replication cycles and elevated mutation rates, viral pathogens exhibit an important capacity for variation and adaptation. This is particularly true for ribonucleic acid (RNA) viruses, which lack proofreading capability and replicate at maximum viable mutation rates. Mutation rates during RNA replication have been estimated to be in the range of 10-3 to 10-5 errors per nucleotide copied (Fig. 1). As a consequence of such limited replication fidelity, individual RNA genomes have only a fleeting existence, and RNA viruses evolve as heterogeneous populations of variant genomes collectively termed viral quasispecies (12, 13, 15). Viral genomes within quasispecies are subjected to a continuous process of variation and competition. Genome

subpopulations best adapted to replicate in a given environment will dominate the population, while unfit mutants are kept at low levels. Unfit mutant populations in one environment may nevertheless be fit in a different environment, and modulation of frequencies of genome subpopulations is the key to adaptability of RNA viruses. The biological and medical relevance of the quasispecies dynamic of RNA viruses has been extensively documented in several recent publications (12, 13, 15). Although several small deoxyribonucleic acid (DNA) viruses can reach evolution rates similar to those of RNA viruses, elevated mutation rates would be incompatible with the maintenance of the genetic information contained in large DNA viruses with complex genomes over 100 kilobases (kb) in size. Large DNA viruses are thus less prone to error than RNA viruses (Fig. 1). Acquisition of new genetic information proceeds mainly by gene duplication, lateral gene transfer by recombination between related viral genomes, and host gene capture (47). Besides important differences in their genetic organisation, viruses can also produce a wide range of clinical manifestations upon replication in their hosts. Viral infections may be inapparent or they may cause acute or chronic diseases either directly, as a result of viral replication in infected tissues, or indirectly by triggering immunopathological responses (33). Many viral and host functions, as well as environmental factors, are presumed to influence the outcome of an infection. It is therefore often difficult to determine the nature of virushost interactions responsible for these disparate effects. A number of recent studies involving several important animal pathogens, such as bovine respiratory syncytial virus (BRSV), foot and mouth disease virus (FMDV), bovine herpesvirus-1 (BoHV-1) and Mareks disease virus (MDV), have provided evidence that evolution of viral pathogens within vaccinated populations has not only important implications for the development of vaccine resistance, but may also promote the emergence of variant viruses with altered pathogenicity or host tropism. In this review we will discuss some of the factors that may drive the development of pathogens with modified pathogenicity in vaccinated populations. The development of vaccine resistance to MDV will be examined in detail because MDV vaccines provide the best example of changes in field strains in association with vaccination. Mareks disease (MD) vaccines are probably the most intensively used vaccines against a DNA virus in any species. In the United States of America (USA) alone approximately 10 billion chickens are vaccinated against MD every year and at least an equal number of chickens are vaccinated elsewhere in the world. Moreover, the emergence of MDV strains with increased pathogenicity is often quoted as an example of vaccine-driven evolution of a DNA pathogen (18, 19, 39).

Genome size 1 kb 10 kb RNA viruses DNA viruses Bacteria and yeast 100 kb 1 Mb 10 Mb

Mutation frequencies 10-10 10-9 10-8 10-7 10-6 10-5 10-4 RNA genomes 10-3

DNA genomes ( >100 kb)

Fig. 1 Genome sizes and mutation frequencies for DNA and RNA microorganisms The tolerance of DNA and RNA microorganisms to accept mutations decreases with genome size. RNA viruses are confined to one log variation in genome size and display average mutation frequencies of 10-4 errors per nucleotide copied (14). DNA viral genomes span more than 2.5 logs in size, overlapping the smallest bacterial genomes such as Mycoplasma genitalium (580 kb). The evolution rate for DNA viruses with small genomes varies from 10-4 (parvovirus) to 10-8 (papovavirus) substitutions per nucleotide per year (14), while the rate is estimated at 3.5 10-8 for large DNA viruses such as human herpesvirus-1 (152 kb) (42)

Rev. sci. tech. Off. int. Epiz., 26 (2)

329

Imperfect vaccines
Although vaccination allows control of the clinical manifestations of a disease, vaccine-induced immunity generally does not protect against viral infections, and limited virus replication and shedding can still be observed in vaccinated animals. Optimal protection of each individual within large populations is also generally not achievable by mass vaccination strategies, because the level of protection conferred by vaccination can be influenced by multiple factors, the most important being: damage to vaccine improper administration immaturity of the host immune system inhibition by maternal antibodies immunosuppressed state of the host enhanced susceptibility of the host insufficient time between vaccination and exposure antigenic differences between circulating viruses and vaccine strains development of vaccine resistance. Thus, in the absence of sterilising immunity, or as a consequence of sub-optimal protection, many pathogens still persist within vaccinated populations. The selective pressure that vaccine-induced immunity may exert on evolving pathogens is largely unknown. A recent analysis of the genetic diversity of BRSV strains, using a large collection of field isolates, revealed a continuous evolution of BRSV, especially in countries where vaccination is widely used. Remarkably, unusual mutations mapping within the conserved central hydrophobic part of the G attachment protein of BRSV were observed in some recent French isolates (55). The biological significance of these mutations in the immunodominant region of BRSV G protein is not known, but they may contribute to the lack of cross-protection between vaccine and field isolates (55). Another example of interaction between vaccine-induced immunity and the genetic diversity of RNA viral populations replicating in the animal host is provided by a large-scale challenge experiment with FMDV in peptidevaccinated cattle (52). Synthetic peptides conferred only partial protection, and some immunised animals developed lesions upon challenge with virulent virus. FMDV mutants escaping neutralisation in peptidevaccinated animals were characterised by unusual single amino acid substitutions affecting both virus antigenic structure and receptor-binding specificity (Fig. 2) (4, 6, 52, 53). The risk of recombination between attenuated or vectored viral vaccines and their wild-type counterparts in coD R NG N I Cattle A S A R G D L A HL G P P

BHK cells 136 Y T A S A R G D L A H L T T T 150 G R GV V R P E S L P

Guinea-pig A S A R G D L A HL P

Fig. 2 Exploration of new antigenic/receptor binding structures by foot and mouth disease virus The overlap between antibody and receptor-binding sites at the surface G-H loop of capsid protein VP1 of foot and mouth disease virus (FMDV) favours the co-evolution between antigenicity and cell tropism in this important animal pathogen. The emergence of unusual FMDV variants displaying both altered antigenicity and modified host cell tropism has been reported in different biological situations including the selection of monoclonal antibody resistant mutants in baby hamster kidney (BHK) cells (31, 41), challenge experiments in peptide-immunised cattle (52, 53), and virus adaptation to the guinea pig (5, 37). The amino acid sequence of the capsid protein VP1 G-H loop region corresponding to antigenic site A of FMDV C-S8c1 is indicated in black boxes. The Arg-Gly-Asp (RGD) integrin-binding motif and flanking amino acid residues promoting FMDV serotype C binding to BHK cells are designated by white-framed boxes (32). Single amino acid replacements found in antigenic site A of FMDV variants are listed for each position

infected animals is generally considered to be a major safety concern for the development and use of live vaccines. The consequences of co-infections were recently assessed in a series of studies involving the alphaherpesvirus BoHV-1. These studies revealed that BoHV-1 exhibits a high potential for genetic diversification in co-infections of the animal host and that recombination is an important mechanism in alphaherpesvirus evolution (46). Several European countries have initiated control programmes based on the use of marker vaccines in which the gE gene of BoHV-1 has been deleted. These marker vaccines, combined with serological detection of gEspecific antibodies, allow differentiation between naturally infected and vaccinated animals. Remarkably, virulent BoHV-1 recombinants carrying the vaccine gE-negative phenotype can be generated in vitro by co-infection of attenuated gE-negative with virulent gE-positive BoHV-1 strains (35, 36). As live attenuated marker vaccines can be administered intranasally, at the natural portal of entry of BoHV-1, co-infections between marker vaccine strains and wild-type BoHV-1 can be encountered under natural conditions. This suggests the potential for emergence of recombinant BoHV-1 with the virulence of the wild-type strain and the phenotype of the vaccine strain (54). Recent epidemiological evidence supports the hypothesis that oncogenic strains of MDV evolve towards pathotypes

330

Rev. sci. tech. Off. int. Epiz., 26 (2)

with increased virulence, and this evolution is probably driven, at least in part, by MD vaccination (59, 60, 61). However, there are many factors involved in optimal MD vaccination, which will be discussed in detail in the section Mareks disease: a case study on evolution of virulence. Although it is often difficult to predict the consequences at the population level of mechanisms evidenced at the animal level, these different studies suggest that circulation of pathogens within vaccinated populations may have a number of implications for the development of vaccine resistance, the evolution of virus pathogenicity, and even for the emergence of mutant viruses with altered tissue or host tropism.

upon FMDV acquisition of RGD-independent mechanisms of cell recognition. The relaxation of the constraints imposed by integrin recognition allows FMDV to explore new antigenic structures at the G-H loop of VP1, and variant viruses with highly unusual substitutions, such as Arg-Gly-Gly (RGG) or even Gly-Gly-Gly (GGG) sequences instead of the RGD, can be selected (31, 41). This provides FMDV with a new repertoire of antigenic variants and an improved capacity to escape neutralisation (Fig. 2). Absence of integrin recognition by FMDV harbouring altered RGD motifs has confirmed that changes in FMDV antigenic structures can be linked to modifications in receptor usage, and suggests that viruses which use the same surface site for receptor recognition and antibody binding harbour the potential for co-evolution of antigenicity and receptor usage (5, 6, 53). The concept of a receptor binding site accessible to antibodies may also have important implications for adaptation of FMDV in the field. The genomic changes that can endow FMDV with the capacity to use alternative mechanisms of cell recognition are minimal (5, 6), and antigenic variants with altered receptor-binding specificities are likely to be present in the mutant spectrum of FMDV replicating in the animal host. A recent study analysing the genetic changes selected during adaptation of FMDV to guinea pigs documented the progressive dominance of an unusual amino acid replacement affecting both the antigenic structure of the G-H loop of VP1 and its interaction with the integrin molecules expressed in various cell lines commonly used to propagate FMDV (Fig. 2) (4, 37). Remarkably, the antigenic alteration found in the guinea pig-adapted virus was also identified in several FMDV mutants escaping neutralisation by antiFMDV antibodies in peptide-vaccinated cattle (Fig. 2) (52, 53). These results with FMDV mutants generated in vivo illustrate the high potential for RNA virus adaptation in the face of an immune response.

Antigenic variation and shifts in receptor usage


Despite early evidence that antigenic changes in influenza virus haemagglutinin could be linked to modifications in sialic acid recognition (50), it is commonly assumed that antibody-binding sites and receptor recognition motifs on virus particles are physically separated. The main argument is that amino acid residues involved in receptor recognition should be invariant, as variation would be lethal, while antigenic sites are inherently variable to allow the virus to cope with the host antibody response. In contrast to this conventional wisdom, structural studies have shown that there is some overlap between these two regions in a number of viral systems, including FMDV (5, 6, 57). In FMDV, amino acids which are critical for the recognition of integrin receptor molecules, in particular the highly conserved Arg-Gly-Asp (RGD) motif located at the protruding G-H loop of capsid protein VP1, are also involved in the interaction with multiple neutralising antibodies (14, 17). Although integrins are probably the major class of receptor molecules used by FMDV in the animal host, virus evolution in cell culture can render RGD-dependent interactions dispensable for infectivity and develop alternative pathways to recognise and enter cells (17, 24). Because the RGD motif located at the G-H loop of capsid protein VP1 is also a key part of several epitopes recognised by neutralising antibodies, the capacity of FMDV to develop and use RGD-independent mechanisms of cell recognition has considerable implications for the evolution of virus antigenicity. When FMDV entry into cells is restricted to RGD-dependent interactions, the amino acid residues which are critically involved in integrin recognition remain invariant while mutations in flanking residues allow the virus to escape immune surveillance. This situation changes dramatically

Mareks disease: a case study in evolution of virulence


Virulence, the capacity of a pathogen to cause disease, is a phenotypic trait which is subjected to variation and natural selection. MDV provides an excellent example of variation in its potential to cause different disease syndromes and natural selection towards increased virulence. Since the first description of polyneuritis in four roosters by Jzsef Marek in 1907 (29), MDV has shown a propensity to cause a number of different disease syndromes including lymphoproliferative syndromes, commonly referred to as MD, lymphodegenerative disease, central nervous system disease syndromes, and atherosclerosis (65). Selection for

Rev. sci. tech. Off. int. Epiz., 26 (2)

331

increased pathogenicity was already noticed before vaccines were introduced in the early 1970s. The changes in the broiler industry in the 1950s had two important consequences. First of all, the housing system changed, with a drastic increase in the density of chickens per square metre. The birds were also subjected to intensive genetic selection to improve production parameters. During the same period a dramatic increase in MD incidence occurred (8) and the term acute MD was introduced. It is impossible to prove if this change in pathogenicity was caused by a change in the virus, a change in the genetics of the host, a change in the environment of the host or a combination of these factors. The incidence of MD continued to increase in broilers, with condemnations reaching approximately 3% in Delmarva (a peninsula in the USA encompassing Delaware, Maryland and Virginia) and Georgia, until the herpesvirus of turkeys (HVT) vaccine became available around 1970. Since the introduction of MD vaccination in the USA, there have been two periods with increased MD condemnations, which were first noted in Delmarva. The first one, in the early 1980s, led to the introduction of the bivalent HVT+SB-1 vaccine. The second occurred in the mid 1990s and led to the introduction in the USA of the CVI988 vaccine (40), also known as Rispens. The increase in condemnations in the early 1980s was significantly lower than the level of condemnations before the introduction of vaccines, and the second spike was again lower than the first one. Notwithstanding these spikes in condemnations, MDV vaccines are still highly effective in preventing the disease (9, 62). In 2002, MD condemnations were in general less than 0.001% in the USA, although condemnations may be higher in some regions, e.g. Delmarva, which had a condemnation rate of 0.011% in 2002 (34). Worldwide there are few problems reported at this time (20, 34). Spikes in condemnations are sometimes seen in various parts of the world, but these may be caused by many factors and do not necessarily indicate the presence of more virulent strains.

broilers have ranged from approximately $0.10 to actual losses of approximately $0.01 per pound (0.45 kg) (J. Smith, personal communication). As a consequence the pressure to reduce costs is high. The down-time between production cycles is approximately 14 days in the USA (J. Smith, personal communication) and litter is frequently re-used for a number of cycles (34), thus providing an immediate challenge to newly placed birds before adequate acquired immune responses have developed. MD vaccines are some of the more expensive used in broilers, with average prices in the USA ranging from $2.25/1,000 doses for HVT to $10.75/1,000 doses for HVT/CVI988. To reduce costs these MD vaccines are routinely used at half or one quarter of the recommended dose in broilers, although these vaccines are used at the recommended levels in layers and broiler breeders. Although there are differences between the USA and other major poultry production areas of the world, the general trend to reduce costs leads to improper vaccination techniques and inadequate biosecurity, thus increasing the risk of selection for strains with increased pathogenicity.

Nomenclature of Mareks disease virus strains


Mareks disease virus is generally referred to as serotype 1 MDV (MDV-1), and all MDV-1 strains have oncogenic potential unless the strains are attenuated in cell culture (65). Serotype 2 MDV (MDV-2) strains are non-oncogenic chicken herpesviruses, while serotype 3 consists of HVT. These three serotypes belong to the recently established Mardivirus genus within the Alphaherpesvirinae subfamily of the Herpesviridae (http://www.ncbi.nlm.nih.gov /ICTVdb/Ictv/index.htm [accessed on 20 September 2006]). Witter et al. (58, 63) have developed a system to determine the pathotype of MDV-1 strains isolated from vaccination breakdowns. Briefly, genetically defined chickens (line 15 7), positive for maternal antibodies for the three serotypes and vaccinated with HVT or HVT+SB1 (a MDV-2 strain), are challenged with virus strains with known pathogenicity and with the new strains. Depending on the development of MDV in the groups vaccinated with HVT or the bivalent vaccine, new isolates are classified as virulent (v), very virulent (vv) or very virulent plus (vv+) MDV. European strains are sometimes classified as hypervirulent, which is probably comparable to the vv+ classification in the USA.

Economic considerations
Before discussing some of the biological factors of MDVhost interactions it is important to address manmade factors influencing these interactions. The poultry industry provides the best example to illustrate some of the problems preventing optimal vaccine-induced protection in production animals. Primary chicken lines are under continuous selection for improved production characteristics, in which selection for disease resistance constitutes only one of the many components. The consequence is that the host is continually changing, which may impact virushost interactions. In the USA profits per broiler are minimal and highly variable from year to year. Over the last five to six years profits for

Pathogenesis of Mareks disease


The pathogenesis of MD has been extensively reviewed and further references can be found in these reviews (2, 10). Infection occurs by inhalation of cell-free virus and is probably transferred to B lymphocytes by phagocytic cells. Until recently, macrophages were generally considered to be resistant to MDV infection and replication, but Barrow

332

Rev. sci. tech. Off. int. Epiz., 26 (2)

et al. (7) provided support for the role of macrophages by demonstrating the presence of MDV transcripts in the cytoplasm and nucleus of splenic macrophages after infection with a hypervirulent strain of MDV. However, virus particles could not be demonstrated in the nucleus and it is not known if the presence of transcripts represents an abortive or productive infection. The first lytic replication phase mainly occurs in B cells, although some T cells can also be involved. The lytic phase can normally be demonstrated as early as three to four days post infection (PI). Subsequently, activated T cells expressing major histocompatibility complex (MHC) class II and CD4 become infected with MDV, while resting T cells appear to be refractory to infection. Around seven days PI, latent infections are established in the activated T cells. Depending on factors such as genetic resistance of the host, virulence of the MDV strain, vaccination status and presence of immunosuppressive viruses, tumours may develop mostly in CD4+, MHC class II+ T cells. Infectious virus is produced in the feather follicle epithelium (FFE), and shedding of MDV starts around 14 days PI, which is before the onset of MD mortality, although experimentally infected birds may die within this period with the early mortality syndrome (65). Quantitative polymerase chain reaction (qPCR) assays have been developed to measure MDV genome copies in the FFE, and initial data suggest that peak shedding occurs between two and five weeks PI (1, 3, 23), although viral DNA has been detected as early as seven days PI (3).

RISPENS BIVAL HVT Relative virulence vv (vv+)

1940

1960 Years

1980

2000

m: mild v: virulent vv: very virulent vv+: very virulent plus (hypervirulent) HVT: herpesvirus of turkeys vaccine BIVAL: bivalent HVT+SB-1 vaccine RISPENS: CVI988 vaccine

Fig. 3 Evolution of Mareks disease virus isolates. Stepwise evolution of virulence of Mareks disease virus isolates: past history and future predictions. There seems to be a relationship between introduction of new vaccines and the development of more virulent pathotypes
From (59), with permission of Taylor and Francis Ltd (http://www.tandf.co.uk/journals)

Mareks disease vaccines, applications and protective mechanisms


Since the introduction of HVT in the USA in the early 1970s, new vaccines or new combinations have been introduced twice in the USA. In 1983 the bivalent combination HVT+SB-1 became available, and CVI988 was licensed in 1996 to protect against field viruses with increased pathogenicity (Fig. 3). However, CVI988, which is currently considered the gold standard for MD vaccines (9), has been used in the Netherlands since 1971 and afterwards in other European countries (see 34 for details). Thus, since the introduction of MDV-2 strains (SB-1 and 301B) no new MDV vaccines have been developed that are better than CVI988. Witter and Kreager (64) found that new vaccine strains could be obtained that were comparable to, but not better than, CVI988. They actually questioned if the efficacy of MD vaccines is limited by some, unspecified, type of biological threshold. Vaccine-induced immunity protects against viral replication, resulting in a significant reduction of the lytic replication phase of the challenge virus (43). However, vaccination does not prevent the establishment of infection in the lymphoid organs nor, and more importantly, in the

FFE. Interestingly, qPCR studies suggest that vaccination with HVT does not reduce shedding of MDV-1 at 56 days PI (23). Additional studies are needed to determine if different pathotypes differ quantitatively in virus shedding and if this is modified by vaccination.

Are virulence- and transmission-related traits Iinked to pathogen fitness in Mareks disease virus?
Although it has been clearly shown that more virulent MDV pathotypes have evolved since the introduction of vaccines, the molecular basis for the evolution of MDV has not been elucidated (45). Early virus replication in nonvaccinated birds is prolonged for more virulent pathotypes compared with less pathogenic strains (66) and is higher in nonvaccinated than in vaccinated birds independent of the pathotype (43), suggesting that the evolution of MDV is related to the early virushost interaction, perhaps by interference with immune responses. However, genes important for viral replication, e.g. viral interleukin (vIL)-8, pp38 (11, 21, 26) and several glycoproteins, are highly conserved across pathotypes (25, 48), which is of interest because these genes, with the

Rev. sci. tech. Off. int. Epiz., 26 (2)

333

exception of vIL-8, are recognised by cell-mediated and/or humoral immune responses (30, 44). Mutations in the Meq gene correlating with virulence were described by Shamblin et al. (48). These authors argued that Meq is not a likely target for vaccine-induced selection because the Meq gene is not encoded by HVT or SB-1 and Meq is mostly expressed during latency and in transformed cells. Moreover, deletion mutants for Meq can cause cytolytic infections comparable to infection with wild-type virus, but do not cause tumours (28). Gandon and colleagues (18) have suggested that vaccines reducing the growth rate of a pathogen, e.g. MD vaccines, would lead to evolution of the pathogen towards increased virulence, while vaccines blocking infection would not induce such effects and may even lead to decreased virulence. They also suggest that virulence and transmission are related traits intimately linked to pathogen fitness (39), although this hypothesis is not generally accepted (16). If this hypothesis is correct, it would suggest that the more virulent MDV pathotypes should be more efficient in virus shedding from the FFE than less virulent pathotypes. As was mentioned before, there is a lack of quantitative data on MDV shedding from the FFE. Baigent et al. (3) quantitated the number of CVI988 genome copies in FFE from different commercial chicken breeds and found differences between the breeds, but comparative studies with other pathotypes have not yet been reported. However, there may not be an intimate link between virulence and transmission in MD, based on studies using deletion mutants. Very virulent MDV or vv+MDV mutants lacking the pp38 gene (21), vIL-8 gene (11) or the first intron of vIL-8 (26) show a significant reduction in lytic virus replication and a subsequent reduction in tumour incidence, probably as a consequence of reduced virus load. However, these viruses do produce cell-free virus in the FFE (11, 21). It will be important to conduct quantitative studies to determine if the amount of virus in the FFE is directly related to replication of mutant viruses in the lymphoid organs and if there are quantitative differences between wild-type and mutant viruses in the amount of virus produced in the FFE.

to be used by the industry any new vaccine must be costeffective, reducing MD contamination at a cost comparable to current vaccine prices. Future strategies for protection against MD may have to depend on a combination of vaccination, increased genetic selection, and perhaps transgenic approaches. All current commercial and experimental vaccines are based on the reduction of virus replication rather than prevention of infection. It is not known if vaccines preventing MDV infection can be developed. To be successful these vaccines need to block entry of virus into phagocytic cells, which may be complicated by the nature of cell-free virus produced in the FFE. Most, if not all, cell-free virus particles are encased in keratin, and vaccines designed to block viruscellular receptor interactions may therefore not work. Mechanisms to block virus replication after entry into susceptible cells (using macrophages, B cells, activated T cells, epithelial cells) may be possible using RNA interference (RNAi) approaches. Transgenic approaches, which may not be acceptable to consumers, will be needed to express the RNAi sequences in all potential target cells.

Conclusion
In a global economy with open markets and frequent exchanges of animals and livestock products, the control of infectious diseases has become a major concern for the farming industry. Vaccination, when available, is probably the most effective method of protecting animal populations against economically relevant pathogens, and mass vaccination strategies have largely contributed to the control of infectious diseases in endemic areas. Despite reducing the clinical manifestations of the disease, vaccination rarely protects against viral infection, and many pathogens still persist within vaccinated populations. Epidemiological studies have documented a rapid and continuous diversification of different viral pathogens in large populations of animals protected by vaccination, suggesting that the evolution of these pathogens was probably driven, at least in part, by vaccination. The biological implications of vaccine-driven evolution of viral pathogens remain largely unknown, and recent evidence suggests that the development of vaccine resistance may also be associated with modifications in pathogenicity or host tropism, as illustrated by the emergence of MDV pathotypes of enhanced virulence and the selection of FMDV variants with modified receptorbinding specificities. Recombination between attenuated marker vaccines and field strains of BoHV-1, with the potential emergence of virulent BoHV-1 expressing the phenotypic traits of the marker vaccines, provides another

The future of Mareks disease protection


Based on past experience it is expected that more virulent pathotypes than vv+ strains of MDV will emerge. Based on the work by Witter and Kreager (64) it is less certain that improved vaccines will be available in the near future. Recombinant fowlpox virus (rFPV)-based vaccines expressing MD glycoproteins have shown some promise under experimental conditions (27). Based on experiences with rFPV vaccines for avian influenza (51) it is not clear if these vaccines will give sufficient protection if chicks are positive for FPV maternal antibodies. In addition, in order

334

Rev. sci. tech. Off. int. Epiz., 26 (2)

remarkable example of virus adaptability upon evolution in vaccinated populations. This has implications for control and eradication strategies based on serological differentiation between vaccinated and infected animals. The evolution of viral pathogens circulating in vaccinatedpopulations may represent a new challenge for the animal health sector. There is a need to better understand the dynamics of viral pathogens replicating in the field, in particular those evolving in animal populations protected by vaccination.

Acknowledgements
We wish to express our gratitude to E. Domingo and F Sobrino for their continuous support. This manuscript . was written with support from the French Ministry of Research (ACI Microbiologie) and the Institut National de la Recherche Agronomique (Trans-Zoonose).

La vaccination des animaux et lvolution des virus


K.A. Schat & E. Baranowski
Rsum Si la vaccination parvient rduire lincidence des maladies, il est rare quelle confre une protection contre le processus infectieux, de sorte que nombre dagents pathognes continuent circuler au sein des populations danimaux vaccins. Cette circulation virale risque de favoriser lapparition dune rsistance aux vaccins, avec des consquences sur lvolution de la pathognicit des virus et lmergence de virus variants. Les taux levs de mutation accompagnant la multiplication des virus acide ribonuclique (ARN) favorisent lapparition de mutants dchappement. Chez des bovins vaccins, des mutations inhabituelles ont t observes au niveau du site antignique majeur du virus de la fivre aphteuse qui est galement impliqu dans la reconnaissance de rcepteurs. De mme, des modifications atypiques ont t dtectes dans la rgion immunodominante du virus respiratoire syncytial bovin. Les grands virus acide dsoxyribonuclique (ADN) ont la capacit de se recombiner et de crer de nouveaux gnotypes, comme cest le cas des souches vaccinales de lherpsvirus bovin de type 1 porteuses dune dltion dans le gne codant pour la glycoprotine gE, qui prsentent un risque de recombinaison avec les souches sauvages. Le virus de la maladie de Marek est souvent cit comme exemple de la modification du pouvoir pathogne induite par la vaccination. Les causes exactes de cette virulence accrue restent lucider ; les auteurs examinent quelques explications possibles de ce phnomne. Mots-cls volution Maladie de Marek Pathognicit Pathotype Rcepteur Tropisme Vaccin Virulence Virus.

Rev. sci. tech. Off. int. Epiz., 26 (2)

335

Vacunacin de animales y evolucin de los patgenos virales


K.A. Schat & E. Baranowski
Resumen Pese a que reduce la incidencia de la enfermedad, la vacunacin pocas veces protege contra la infeccin y muchos agentes patgenos siguen presentes en poblaciones animales vacunadas. La circulacin de agentes patgenos virales en esas poblaciones puede favorecer el desarrollo de resistencia a las vacunas, con implicaciones para la evolucin de los virus y la emergencia de variantes virales. La elevada tasa de mutaciones durante la replicacin de los virus cido ribonucleico (ARN) favorece la emergencia de mutantes de escape. En ganados vacunados se han observado mutaciones inusuales en el sitio antignico principal del virus de la fiebre aftosa, implicado tambin en el reconocimiento de receptores. Del mismo modo, se han detectado cambios atpicos en la regin inmunodominante del virus respiratorio sincitial bovino. Los grandes virus cido desoxiribonucleico (ADN) pueden recombinarse, dando lugar a nuevos genotipos, como lo muestra el potencial de las cepas vacunales del herpesvirus bovino de tipo 1 con delecin en el gen de la glicoprotena E para recombinarse con cepas salvajes. El virus de la enfermedad de Marek suele citarse como un ejemplo de cambio de patogenicidad provocado por la vacunacin. An no se han elucidado los motivos por los que aumenta la virulencia y los autores contemplan las posibles explicaciones. Palabras clave Evolucin Enfermedad de Marek Patogenicidad Patotipo Receptor Tropismo Vacuna Virulencia Virus.

References
1. Abdul-Careem M.F Hunter B.D., Nagy E., Read L.R., ., Sanei B., Spencer J.L. & Sharif S. (2006). Development of a real-time PCR assay using SYBR Green chemistry for monitoring Mareks disease virus genome load in feather tips. J. virol. Meth., 133, 34-40. 2. Baigent S. & Davison F (2004). Mareks disease virus: . biology and life cycle. In Mareks disease, an evolving problem (F Davison & V. Nair, eds). Academic Press, Oxford, 62-77. . 3. Baigent S.J., Smith L.P., Nair V.K. & Currie R.J. (2006). Vaccinal control of Mareks disease: current challenges, and future strategies to maximize protection. Vet. Immunol. Immunopathol., 112, 78-86. 4. Baranowski E., Molina N., Nunez J.I., Sobrino F & Saiz M. . (2003). Recovery of infectious foot-and-mouth disease virus from suckling mice after direct inoculation with in vitrotranscribed RNA. J. Virol., 77, 11290-11295. 5. Baranowski E., Ruiz-Jarabo C.M. & Domingo E. (2001). Evolution of cell recognition by viruses. Science, 292, 11021105. 6. Baranowski E., Ruiz-Jarabo C.M., Pariente N., Verdaguer N. & Domingo E. (2003). Evolution of cell recognition by viruses: a source of biological novelty with medical implications. Adv. Virus Res., 62, 19-111.

336

Rev. sci. tech. Off. int. Epiz., 26 (2)

7. Barrow A.D., Burgess S.C., Baigent S.J., Howes K. & Nair V.K. (2003). Infection of macrophages by a lymphotropic herpesvirus: a new tropism for Mareks disease virus. J. gen. Virol., 84, 2635-2645. 8. Benton W.J. & Cover M.S. (1957). The increased incidence of visceral lymphomatosis in broiler and replacement birds. Avian Dis., 1, 320-327. 9. Bublot M. & Sharma J. (2004). Vaccination against Mareks disease. In Mareks disease, an evolving problem (F. Davison & V. Nair, eds). Academic Press, Oxford, 168-185. 10. Calnek B.W. (2001). Pathogenesis of Mareks disease virus infection. Curr. Top. Microbiol. Immunol., 255, 25-55. 11. Cui X., Lee L.F Reed W.M., Kung H.J. & Reddy S.M. (2004). ., Mareks disease virus-encoded vIL-8 gene is involved in early cytolytic infection but dispensable for establishment of latency. J. Virol., 78, 4753-4760. 12. Domingo E., Biebricher C., Eigen M. & Holland J. (eds) (2001). Quasispecies and RNA virus evolution: principles and consequences. Landes Bioscience, Austin, Texas. 13. Domingo E., Martin V., Perales C., Grande-Perez A., Garcia-Arriaza J. & Arias A. (2006). Viruses as quasispecies: biological implications. Curr. Top. Microbiol. Immunol., 299, 51-82. 14. Domingo E., Verdaguer N., Ochoa W.F Ruiz-Jarabo C.M., ., Sevilla N., Baranowski E., Mateu M.G. & Fita I. (1999). Biochemical and structural studies with neutralizing antibodies raised against foot-and-mouth disease virus. Virus Res., 62, 169-175. 15. Domingo E., Webster R.G. & Holland J.J. (eds) (1999). Origin and evolution of viruses. Academic Press, San Diego, 1-499. 16. Ebert D. & Bull J.J. (2003). Challenging the trade-off model for the evolution of virulence: is virulence management feasible? Trends Microbiol., 11, 15-20. 17. Fry E.E., Stuart D.I. & Rowlands D.J. (2005). The structure of foot-and-mouth disease virus. Curr. Top. Microbiol. Immunol., 288, 71-101. 18. Gandon S., Mackinnon M.J., Nee S. & Read A.F (2001). . Imperfect vaccines and the evolution of pathogen virulence. Nature, 414, 751-756. 19. Gandon S., Mackinnon M., Nee S. & Read A. (2003). Imperfect vaccination: some epidemiological and evolutionary consequences. Proc. roy. Soc. Lond., B, biol. Sci., 270, 1129-1136. 20. Gimeno I.M. (2004). Future strategies for controlling Mareks disease. In Mareks disease, an evolving problem (F Davison & V. Nair, eds). Academic Press, Oxford, 186. 199.

21. Gimeno I.M., Witter R.L., Hunt H.D., Reddy S.M., Lee L.F & . Silva R.F (2005). The pp38 gene of Mareks disease virus . (MDV) is necessary for cytolytic infection of B cells and maintenance of the transformed state but not for cytolytic infection of the feather follicle epithelium and horizontal spread of MDV. J. Virol., 79, 4545-4549. 22. Henderson L.M. (2005). Overview of marker vaccine and differential diagnostic test technology. Biologicals, 33, 203-209. 23. Islam A., Cheetham B.F., Mahony T.J., Young P.L. & Walkden-Brown S.W. (2006). Absolute quantitation of Mareks disease virus and herpesvirus of turkeys in chicken lymphocyte, feather tip and dust samples using real-time PCR. J. virol. Meth., 132, 127-134. 24. Jackson T., King A.M., Stuart D.I. & Fry E. (2003). Structure and receptor binding. Virus Res., 91, 33-46. 25. Jarosinski K.W., OConnell P.H. & Schat K.A. (2003). Impact of deletions within the Bam HI-L fragment of attenuated Mareks disease virus on vIL-8 expression and the newly identified transcript of open reading frame LORF4. Virus Genes, 26, 255-269. 26. Jarosinski K.W. & Schat K.A. (2007). Multiple alternative splicing to exons II and III of viral interleukin 8 (vIL-8) in the Mareks disease virus genome: the importance of vIL-8 exon I. Virus Genes, 34, 9-22. 27. Lee L.F Witter R.L., Reddy S.M., Wu P., Yanagida N. & ., Yoshida S. (2003). Protection and synergism by recombinant fowl pox vaccines expressing multiple genes from Mareks disease virus. Avian Dis., 47, 549-558. 28. Lupiani B., Lee L.F Cui X., Gimeno I., Anderson A., ., Silva R.F Witter R.L., Kung H.J. &. Reddy S.M. (2004). ., Mareks disease virus-encoded Meq gene is involved in transformation of lymphocytes but is dispensable for replication. Proc. natl Acad. Sci. USA, 101, 11815-11820. 29. Marek J. (1907). Multiple Nervenentzuendung (Polyneuritis) bei Huehnern. Dtsch. tierrztl. Wochenschr., 15, 417-421. 30. Markowski-Grimsrud C.J. & Schat K.A. (2002). Cytotoxic T lymphocyte responses to Mareks disease herpesvirusencoded glycoproteins. Vet. Immunol. Immunopathol., 90, 133-144. 31. Martinez M.A., Verdaguer N., Mateu M.G. & Domingo E. (1997). Evolution subverting essentiality: dispensability of the cell attachment Arg-Gly-Asp motif in multiply passaged foot-and-mouth disease virus. Proc. natl Acad. Sci. USA, 94, 6798-6802. 32. Mateu M.G., Valero M.L., Andreu D. & Domingo E. (1996). Systematic replacement of amino acid residues within an Arg-Gly-Asp-containing loop of foot-and-mouth disease virus and effect on cell recognition. J. biol. Chem., 271, 12814-12819.

Rev. sci. tech. Off. int. Epiz., 26 (2)

337

33. Mims C., Nash A. & Stephen J. (eds) (2001). Mims pathogenesis of infectious disease. Academic Press, San Diego. 34. Morrow C. & Fehler F (2004). Mareks disease: a worldwide . problem. In Mareks disease, an evolving problem (F Davison . & V. Nair, eds). Academic Press, Oxford, 49-61. 35. Muylkens B., Meurens F Schynts F de Fays K., Pourchet A., ., ., Thiry J., Vanderplasschen A., Antoine N. & Thiry E. (2006). Biological characterization of bovine herpesvirus 1 recombinants possessing the vaccine glycoprotein E negative phenotype. Vet. Microbiol., 113, 283-291. 36. Muylkens B., Meurens F Schynts F Farnir F Pourchet A., ., ., ., Bardiau M., Gogev S., Thiry J., Cuisenaire A., Vanderplasschen A. & Thiry E. (2006). Intraspecific bovine herpesvirus 1 recombinants carrying glycoprotein E deletion as a vaccine marker are virulent in cattle. J. gen. Virol., 87, 2149-2154. 37. Nunez J.I., Baranowski E., Molina N., Ruiz-Jarabo C.M., Sanchez C., Domingo E. & Sobrino F (2001). A single . amino acid substitution in nonstructural protein 3A can mediate adaptation of foot-and-mouth disease virus to the guinea pig. J. Virol., 75, 3977-3983. 38. Pastoret P.-P. & Jones P. (2004). Veterinary vaccines for animal and public health. Dev. Biol., 119, 15-29. 39. Read A.F Gandon S., Nee S. & Mackinnon M. (2004). The ., evolution of pathogen virulence in response to animal and public health interventions. In Evolutionary aspects of infectious diseases (K. Dronamraj, ed.). Cambridge University Press, Cambridge, 265-292. 40. Rispens B.H., van Vloten H.J., Mastenbroek N., Maas H.J.L. & Schat K.A. (1972). Control of Mareks disease in the Netherlands. I. Isolation of an avirulent Mareks disease virus (strain CVI988) and its use in laboratory vaccination trials. Avian Dis., 16, 108-125. 41. Ruiz-Jarabo C.M., Sevilla N., Davila M., Gomez-Mariano G., Baranowski E. & Domingo E. (1999). Antigenic properties and population stability of a foot-and-mouth disease virus with an altered Arg-Gly-Asp receptor-recognition motif. J. gen. Virol., 80, 1899-1909. 42. Sakaoka H., Kurita K., Iida Y., Takada S., Umene K., Kim Y.T., Ren C.S. & Nahmias A.J. (1994). Quantitative analysis of genomic polymorphism of herpes simplex virus type 1 strains from six countries: studies of molecular evolution and molecular epidemiology of the virus. J. gen. Virol., 75, 513-527. 43. Schat K.A., Calnek B.W. & Fabricant J. (1982). Characterisation of two highly oncogenic strains of Mareks disease virus. Avian Pathol., 11, 593-605. 44. Schat K.A. & Markowski-Grimsrud C.J. (2001). Immune responses to Mareks disease virus infection. Curr. Top. Microbiol. Immunol., 255, 91-120.

45. Schat K.A. & Nair V. (2008). Mareks disease. In Diseases of poultry, 12th Ed. (Y.M. Saif et al., eds). Academic Press, Ames (in press). 46. Schynts F Meurens F Detry B., Vanderplasschen A. & ., ., Thiry E. (2003). Rise and survival of bovine herpesvirus 1 recombinants after primary infection and reactivation from latency. J. Virol., 77, 12535-12542. 47. Shackelton L.A. & Holmes E.C. (2004). The evolution of large DNA viruses: combining genomic information of viruses and their hosts. Trends Microbiol., 12, 458-465. 48. Shamblin C.E., Greene N., Arumugaswami V., Dienglewicz R.L. & Parcells M.S. (2004). Comparative analysis of Mareks disease virus (MDV) glycoprotein-, lytic antigen pp38- and transformation antigen Meq-encoding genes: association of meq mutations with MDVs of high virulence. Vet. Microbiol., 102, 147-167. 49. Shams H. (2005). Recent developments in veterinary vaccinology. Vet. J., 170, 289-299. 50. Skehel J.J. & Wiley D.C. (2000). Receptor binding and membrane fusion in virus entry: the influenza hemagglutinin. Annu. Rev. Biochem., 69, 531-569. 51. Swayne D.E., Beck J.R. & Kinney N. (2000). Failure of a recombinant fowl poxvirus vaccine containing an avian influenza hemagglutinin gene to provide consistent protection against influenza in chickens preimmunized with a fowl pox vaccine. Avian Dis., 44, 132-137. 52. Taboga O., Tami C., Carrillo E., Nunez J.I., Rodrguez A., Saiz J.C., Blanco E., Valero M.L., Roig X., Camarero J.A., Andreu D., Mateu M.G., Giralt E., Domingo E., Sobrino F & . Palma E.L. (1997). A large-scale evaluation of peptide vaccines against foot-and-mouth disease: lack of solid protection in cattle and isolation of escape mutants. J. Virol., 71, 2606-2614. 53. Tami C., Taboga O., Berinstein A., Nunez J.I., Palma E.L., Domingo E., Sobrino F & Carrillo E. (2003). Evidence of . the coevolution of antigenicity and host cell tropism of footand-mouth disease virus in vivo. J. Virol., 77, 1219-1226. 54. Thiry E., Muylkens B., Meurens F Gogev S., Thiry J., ., Vanderplasschen A. & Schynts F (2006). Recombination in . the alphaherpesvirus bovine herpesvirus 1. Vet. Microbiol., 113, 171-177. 55. Valarcher J.F Schelcher F & Bourhy H. (2000). Evolution ., . of bovine respiratory syncytial virus. J. Virol., 74, 1071410728. 56. Van Oirschot J.T. (2001). Present and future of veterinary viral vaccinology: a review. Vet. Q., 23, 100-108. 57. Verdaguer N., Mateu M.G., Andreu D., Giralt E., Domingo E. & Fita I. (1995). Structure of the major antigenic loop of foot-and-mouth disease virus complexed with a neutralizing antibody: direct involvement of the Arg-Gly-Asp motif in the interaction. EMBO J., 14, 1690-1696.

338

Rev. sci. tech. Off. int. Epiz., 26 (2)

58. Witter R.L. (1997). Increased virulence of Mareks disease virus field isolates. Avian Dis., 41, 149-163. 59. Witter R.L. (1998). The changing landscape of Mareks disease. Avian Pathol., 27, S46-S53. 60. Witter R.L. (1998). Control strategies for Mareks disease: a perspective for the future. Poult. Sci., 77, 1197-1203. 61. Witter R.L. (2001). Mareks disease vaccines past, present and future (Chicken vs virus a battle of the centuries). In Current progress on Mareks disease research (K.A. Schat, R.W. Morgan, M.S. Parcells & J.L. Spencer, eds). American Association of Avian Pathologists, Kennett Square, Pennsylvania, 1-9. 62. Witter R.L. (2001). Protective efficacy of Mareks disease vaccines. Curr. Top. Microbiol. Immunol., 255, 57-90. 63. Witter R.L., Calnek B.W., Buscaglia C., Gimeno I.M. & Schat K.A. (2005). Classification of Mareks disease viruses according to pathotype: philosophy and methodology. Avian Pathol., 34, 75-90.

64. Witter R.L. & Kreager K.S. (2004). Serotype 1 viruses modified by backpassage or insertional mutagenesis: approaching the threshold of vaccine efficacy in Mareks disease. Avian Dis., 48, 768-782. 65. Witter R.L. & Schat K.A. (2003). Mareks disease. In Diseases of poultry, 11th Ed. (Y.M. Saif, H.J. Barnes, J.R. Glisson, A.M. Fadly, L.R. McDougald & D.E. Swayne, eds). Iowa State University Press, Ames, 407-464. 66. Yunis R., Jarosinski K.W. & Schat K.A. (2004). Association between rate of viral genome replication and virulence of Mareks disease herpesvirus strains. Virology, 328, 142-150.

Rev. sci. tech. Off. int. Epiz., 2007, 26 (2), 339-350

Safe use of vaccines and vaccine compliance with food safety requirements
K. Grein (1), O. Papadopoulos (2) & M. Tollis (3)
(1) European Medicines Agency, 7 Westferry Circus, Canary Wharf, London, E14 4HB, United Kingdom (2) Faculty of Veterinary Medicine, Aristotle University, 54124, Thessaloniki, Greece (3) Department of Food Safety and Veterinary Public Health, Istituto Superiore di Sanit, Viale Regina Elena 299, 0014, Rome, Italy

Summary Advanced technologies and regulatory regimes have contributed to the availability of veterinary vaccines that have high quality and favourable safety profiles in terms of potential risks posed to the target animals, the persons who come into contact with the vaccine, the consumers of food derived from vaccinated animals and the environment. The authorisation process requires that a range of safety studies are provided to evaluate the products. The design and production of vaccines, and their safe use, are primarily assessed by using data gathered from extensive pre-marketing studies performed on target animals and specific quality tests. The current post-marketing safeguards include good manufacturing practices, batch safety testing, inspections and pharmacovigilance. In addition to hazard identification, a full benefit/risk evaluation needs to be undertaken. The outcome of that evaluation will determine options for risk management and affect regulatory decisions on the safety of the vaccine; options might, for example, include special warnings on package inserts and labels. Keywords Animal Assessment Benefit Consumer Environment Health Pharmacovigilance Risk Safety User Vaccine.

Introduction
Historically, vaccination of animals has been a strategic component of policies, designed to prevent and control infectious diseases. In this respect, vaccination has had evident benefits for animal welfare and health. As well as the quality, safety and efficacy of the vaccine in relation to the target animal, the impact on public health and the environment must also be considered. By improving animal health, veterinary vaccines can reduce the circulation of animal pathogens and waste and may result in a reduction in the use of disinfectants and antimicrobials.

As a matter of principle, veterinary vaccines must be safe not only for the target animal species but also for the vaccine users, consumers of foodstuffs of animal origin and the environment. The risks associated with the use of vaccines are primarily related to the properties of the vaccine, care in handling and administration, and recipient host factors. Side effects may be local or systemic; they may occur at, or shortly after vaccination, or be delayed. In order to establish that a vaccine is safe, a risk assessment, followed by a benefit/risk analysis, must be undertaken for each vaccine before it can be authorised. Criteria and requirements for risk assessment have been designed to ensure that vaccine producers gather a package

340

Rev. sci. tech. Off. int. Epiz., 26 (2)

of safety data on the product through studies of both target animals and non-target animals, as well as quality data. Extensive testing is required as part of the vaccine development in order to obtain marketing authorisation. After development, testing is also routinely performed as a part of the batch release procedure on every batch of vaccine. New technologies have improved the safety and the efficacy of vaccines, thus ensuring satisfactory levels of immunity and reducing, or even eliminating, harmful and/or unexpected side effects in vaccinated animals. In some cases, a risk assessment may indicate that a vaccine poses a potential risk to the target animals, public health or the environment; if so, the benefit/risk analysis, weighing the benefits of the vaccine against its risks, will determine whether the vaccine can still be used. If the vaccine offers considerable advantages in preventing illness, but presents potential risks to the target animals or human beings or the environment, the benefits would need to outweigh the risks. If the vaccine is authorised for release despite the risks, its use should be accompanied by adequate risk management measures that would minimise or eliminate the risk. Marketing authorisation depends upon the risk assessment and benefit/risk analysis, which are based on the intended use and claimed benefits of a vaccine. The use and claim will be included in the summary of product characteristics (SPC), the package insert and the labelling, which should provide instructions for use, storage and waste disposal, as well as any appropriate safety warnings. These documents must make clear to the user that off-label use (use not specified by the manufacturers) can be extremely dangerous; such off-label use could include administering the vaccine to non-target animal species or by a route different from the recommended ones. Users should also be aware that the use of contaminated vaccines can cause the spread of extraneous or exotic microorganisms in specific agent-free farms or geographical areas (10). In recent years, and particularly in developed countries, public debate over the benefits of vaccination has increasingly been fuelled by fears that some risks have been underestimated. The suspected link between vaccination of cats and the development of sarcomas at the injection site of vaccines is an example (2, 12). Some cat owners are often so worried that they prefer to take the risk of not vaccinating their animals, instead of having them exposed to the potential risk of sarcoma development following vaccination. Another case for concern is the use of multiple simultaneous vaccinations for different diseases, the validity and benefits of which are being questioned by several research groups. There are currently investigations into the possibility that such multiple vaccinations could lead to harmful side reactions such as interference with and

overloading of the immune system of animals, particularly companion animals. Similarly, early life immunisation programmes are increasingly criticised due to the unpredictable outcome of vaccinating very young animals and the fear of inducing tolerance or autoimmunity mechanisms (11, 15). Vaccination of animals is sometimes undertaken to mitigate the effects of poor management systems; this can result in serious risks of disease and should be avoided. In general, there is no need to establish a maximum residue limit (MRL) for the active principles contained in specific vaccines, and a withdrawal period is not necessary after vaccination in most cases, because in general vaccination does not result in harmful residues of the active principles or immunological responses distinguishable from those that arise naturally (4). However, with certain live vaccines with zoonotic agents, withdrawal periods may be necessary. Furthermore, excipients and adjuvants as primary components of vaccines need to be considered when evaluating the safety of vaccines (16). The present paper will give an overview of the regulatory requirements for vaccines. It will also address the main outcomes of the risk/safety assessment of the use of vaccines in target animal species, and in relation to vaccine users, consumers of food derived from vaccinated animals, and the environment.

Regulatory requirements
Most countries have a range of legislation to ensure at least minimum standards for the quality, safety and efficacy of veterinary medicinal products. As a general rule, before a veterinary medicinal product can be sold or used, it must be authorised by the responsible authority of the country where it will be used. This applies for pharmaceutical veterinary medicines as well as vaccines. In order to obtain a marketing authorisation (or registration or licence, as appropriate) the company that intends to bring the product on the market must submit an application to the authority concerned, accompanied by a comprehensive package of data on the quality, safety and efficacy of the vaccine. The application should also address any precautionary measures to be taken when storing the veterinary medicinal product, administering it to animals and disposing of waste, together with an indication of potential risks that the product might pose to human and animal health and to the environment. Following the initial assessment of the application, additional questions usually arise (list of questions) that the applicant (or sponsor) of the veterinary medicinal product will have to answer. Once all questions have been satisfactorily addressed and it is established that there are no risks that would prevent the

Rev. sci. tech. Off. int. Epiz., 26 (2)

341

licensing of the vaccines, a marketing authorisation will be issued, imposing specific conditions of use, storage and waste disposal. Standards of manufacturing safety, including the design and production of vaccines, will often be regulated under different legislation and separate regulating authorities (e.g. workplace safety and environmental safety at the manufacturing plant are not usually covered by legislation governing veterinary medicines). Harmonisation of rules and testing among individual countries and regions is carried out by international organisations concerned with animal health on a worldwide scale, such as the World Organisation for Animal Health (OIE) and the World Trade Organization (WTO). There is an ongoing programme to harmonise data requirements for veterinary medicinal products (including vaccines) in a number of areas, e.g. quality monitoring and target animal safety studies, within the International Cooperation on Harmonisation of Technical Requirements for Registration of Veterinary Medicinal Products (VICH). To give an example, in the European Union (EU) and the countries of the European Economic Area, the requirements for obtaining a marketing authorisation are laid down in Directive 2001/82/EC, and subsequent amendments (7). Appendix I to this Directive describes in detail the data about the quality, safety and efficacy of the product that must be provided with an application for a marketing authorisation. Directive 2001/82/EC, as amended, also requires the applicant to provide in addition to the data on quality, safety and efficacy information on the test methods and any precautionary measures to be taken when storing the veterinary medicinal product, administering it to animals and disposing of waste, together with an indication of potential risks that the product might pose to human and animal health and to the environment. Further guidance is given in specific guidelines issued by the Committee for Medicinal Products for Veterinary Use (CVMP) of the European Medicines Agency (EMEA) (6) and the European Commission (3). For vaccines which are the subject of a European Pharmacopoeia monograph, specific requirements may be included in the relevant monograph (9). All data are assessed by the responsible authority, and a risk assessment and a benefit/risk analysis are carried out before a decision on the marketing authorisation is taken. Different procedures exist in the EU to obtain a marketing authorisation: in the centralised procedure, which is optional for new chemical entities and innovative products, and mandatory for products derived by biotechnological processes, the application is assessed by the CVMP. This procedure leads to a marketing authorisation binding in all EU Member States. In the mutual recognition procedure and the decentralised procedure the Member States, in which the product is

intended to be marketed, carry out the assessment aiming for consistent marketing authorisations. National marketing authorisations issued by individual Member States exist for veterinary medicinal products, which were on the market in the EU before 1995, and can be issued today, if a product is intended for one single EU Member State only. Depending on the process of the marketing authorisation chosen, the procedure is defined by Regulation (EC) No. 726/2004 (8) or described in Directive 2001/82/EC (7).

Principles of risk assessment/risk analysis


In order to ensure the safety of vaccines a risk assessment is necessary. The aim of the risk assessment is to identify hazards, to estimate the likelihood that the hazards will lead to actual harm, and to take decisions about appropriate measures for prevention and control. Risk assessment is a science-based process involving the following stages: hazard identification hazard characterisation exposure assessment risk characterisation. In the first steps the potential hazards associated with the vaccine are comprehensively identified and characterised. In parallell, the exposure scenarios for the different protection goals for the vaccine under consideration are identified and described. A risk characterisation then estimates the probability of the identified hazards occurring. If any risks are not as low as reasonably practicable, the process of risk assessment should be repeated to ascertain whether additional management techniques and risk mitigation measures could reduce the level of risk. Although, wherever possible, the risk assessment should be based on quantifiable outcomes, it is recognised that many of the judgments must necessarily be qualitative only. The risk can often be judged on a rating scale ranging from negligible through low and medium to severe or unacceptable. Once all risks have been assessed and if possible reduced the residual risks are weighed against the benefits of the vaccine, in order to decide whether to grant a marketing authorisation, and to consider possible risk mitigation measures, if appropriate. The approved conditions of use of a veterinary medicinal product should be described in a clear and unambiguous manner. The identified risks and risk mitigation measures must be communicated to the users of the vaccines.

342

Rev. sci. tech. Off. int. Epiz., 26 (2)

Safety of the target animals


Problem statement
Errors in the manufacturing process or incorrect use of vaccines are the two main causes of potential risks for target animal species. Unsafe live or poorly inactivated vaccines may harm the animal to which they are administered, and the causative agent may also spread and harm other animals or have serious ecological effects. Apart from the threat of potentially serious consequences related to contamination of vaccines by extraneous agents, which in extreme cases could lead to disease outbreaks and death of animals (10, 17), it is the likelihood of local toxicity and long-lasting side effects that generates the greatest concern for animal health and welfare (13). Adverse events can vary from carcass damage, as in the case of particularly aggressive adjuvants, to death of animals.

a once in a lifetime probability), using a dose corresponding to the quantity of the product to be recommended and of the maximum titre or potency. One dose, repeated doses and overdose tests are normally required for each of the target species, testing animals of the most sensitive class, age and sex identified on the label, and using all recommended routes and methods of administration. Essential parameters to be evaluated for the safety of a vaccine are local and systemic reactions to vaccination, including application site reactions and their resolution, and clinical observation of the animals. Other objective criteria, such as rectal temperature and performance measurements, are also recorded. Examination of the reproductive performance of breeding animals as well as of immunological functions must be considered when data suggest that the initial material from which the product is derived may be a risk factor. For live vaccines specific additional tests are required, e.g. testing the potential that different levels of pathogenicity may be retained in the case of specific animal species, or of a certain age or class of animal, or specific routes of administration (induction of clinical signs or lesions of disease, or persistency/latency of the microorganism in the body of a vaccinated animal). Special requirements for live vaccines include the definition of the biological characteristics of the vaccine strain (e.g. animal tissue tropism) and the exclusion of any potential for the vaccine strains to revert to virulence. The nature and severity of adverse reactions following the administration of a veterinary vaccine can be dependent either on the antigen that is used (e.g. crude bacterial antigen compared to well-purified viral antigens) or on the susceptibility of the different animal species. The depth, length and width of local reaction as well as any histological change in the tissue structure have to be measured. Local reactions can vary from granulomas of limited or larger extent, to abscesses; from inflammation to necrosis and fibrosis of tissues. In recent years, some excipients (e.g. as part of stabilisers) or adjuvants (particularly oil/emulsion adjuvants) and vaccine administration (in terms of both route and timing) have frequently been implicated as the cause of adverse reactions, both at injection sites and systemic (including fever, arthritis, anorexia, soreness and lethargy). Moreover, recent reports have raised concerns among professionals in relation to vaccination and delayed adverse events, particularly in companion animals and specifically in relation to vaccine-associated fibrosarcomas in cats and immune-mediated disease in dogs. Although their role is still debated, adjuvants have been associated with a local tissue irritation which in very extreme circumstances may result in metaplasia of fibrocytes and tumour formation (14).

Scope of the assessment


The purpose of the evaluation during the development phase is to determine whether the vaccine is safe if used at the proposed dose level. The evaluation is limited to the health and welfare of the target animals. Safety testing in target animal species must show the potential risks that may occur under the proposed conditions of use of vaccines. Laboratory safety studies have been designed to meet necessary basic requirements before initiating field trials. For the final assessment, safety data produced under controlled experimental conditions will be supported by results obtained during field trials. These trials are carried out with a statistically significant number of animals and designed to reflect, as accurately as possible, the practical conditions of use of the products being tested.

Risk assessment
Hazard identification and characterisation
The specific information that needs to be provided depends upon various factors, including: the type of vaccine (inactivated or live, bacterial or viral) the nature of the adjuvants that will be used the use history of similar products dose claims proposed usage regimen animal species, class and breed. As a general rule, the worst case or most severe potential harm should be assessed, even if it is highly unlikely (e.g.

Rev. sci. tech. Off. int. Epiz., 26 (2)

343

Most of the common systemic reactions such as pain, fever and anorexia, but also vomiting, reduction in milk yield, anaemia, arthritis, epilepsy, thyroid disease, liver failure, diabetes and allergies have been associated with the use of vaccines in animals. Residual endotoxins and pyrogenic effects of antigens and adjuvants (oil, saponin) may be responsible for adverse events which can more commonly be expected as a consequence of the use of vaccines. Anaphylactic and delayed allergic reactions can also occur but are more insidious and in most cases are not controllable. The risk of allergic reactions has been reported to increase after repeated injection of vaccines (13). Other risks may be posed by the immunosuppressive effects of modified live viruses, such as bovine herpesvirus 1 (BHV-1), bovine viral diarrhoea virus (BVDV), and the SG33 strain of myxomatosis virus used in specific vaccines. Parvovirus and canine distemper vaccines have been reported to be responsible for many diseases of the immune system in dogs (1, 15). Among the most deleterious and suspected effects on the immune system are tolerance and autoimmunity, overloading of the immune system by multiple and simultaneous administration of vaccines, interaction with other vaccines or reciprocal interference with the immune response against antigens.

terms of the harm they might cause. Two broad categories with qualitatively different risk profiles are commonly recognised. Inactivated and subunit vaccines, as well as conventionally designed and produced live vaccines, are generally considered to be low-risk products in respect to target animal safety. Live genetically modified organisms (GMO) or vectored vaccines are usually considered to have high-risk profiles. In order to balance the greater risks of such GMO or vectored vaccines, higher levels of efficacy and benefits are normally required from them than from conventional vaccines. Nevertheless, even low-risk products may entail higher risks for the target animals, e.g. if injected intramuscularly.

Risk management and risk communication


Improved manufacturing processes (e.g. targeted use of only protective epitopes from specific proteins, or reduced use of sensitising agents such as residues of bovine serum, cell debris, egg proteins, preservatives) and new generation adjuvants are key issues for the prevention of side effects in target animal species. In particular, adjuvants may be tailored for specific needs of the antigen or species and to enhance the type (e.g. humoral, mucosal, cellmediated), timing and magnitude of protective immunoresponse. The use of routes and methods of administration known to cause less adverse reactions should be preferred. Vaccination schemes should be developed taking into account which vaccines are more appropriate for animals of different ages and categories, and provide long-lasting immunity following a single immunisation, thus reducing the risks of potential immunosuppression or immunomediated diseases. Recommendations for the appropriate use of core vaccines in companion animals should be based on the severity of disease caused by the agent, the risk of the agent being transmitted to susceptible animals, and the potential for a particular infection to be zoonotic. Antibody titre testing before vaccination could be useful to determine if a vaccination is really needed. Non-core vaccines should be used when a known or likely risk of exposure is anticipated or when an individual animals lifestyle represents a significant risk of infection. Warnings about possible side effects should realistically reflect the data obtained in safety studies. The outcome of the risk assessment must be summarised within the characteristics of each product, and the label should include warnings about potential over-dosage, contra-indications, and undesirable side effects for each target species and category of animal (e.g. pregnant or lactating animals). If the product is incompatible with other vaccines or medicines that might be administered, that must also be reported, as must any special precautions for use. In the case of any identified risk, appropriate warnings must be incorporated in the package insert and/or label.

Exposure assessment
In principle, adverse events may potentially occur at any exposure level to vaccines, whether administered once in a lifetime or repeatedly, and with products containing minimum or larger combinations of antigens. In particular, risks may increase when vaccines are administered to pregnant or lactating animals, or are used for early life immunisation of animals, such as in ovo or day-old vaccination of birds. Concern has been expressed about this; some practitioners argue that the youngest recommended age for vaccinating various species should be increased, because early vaccination may interfere with persistent maternally derived antibodies, and may therefore affect both the safety and efficacy of vaccination. It is necessary to ensure a wide spectrum of protection against major infectious diseases early in the life of animals; consequently, in most cases animals have to be vaccinated repeatedly in a relatively short time period, which increases the risks of adverse reactions. In order to avoid unnecessary vaccination of animals, data generated by studies on the duration of immunity should be used when preparing vaccination schemes.

Risk characterisation
Once hazards have been identified as likely, the risks associated with those hazards should be characterised in

344

Rev. sci. tech. Off. int. Epiz., 26 (2)

User safety
Problem statement
Vaccines should be safe not only for the target species, but also for the user, i.e. the person administering the vaccine or any other person assisting or involved in the administration of the vaccine or likely to come into contact with it. The user must be informed of any specific hazard through the label and the package insert. This warning must identify any risk, and give advice on ways to prevent it occurring, as well as the measures to be taken in the case of exposure. The hazard could occur during normal use or through an accidental exposure during or following the administration. It is evident that user safety is a critical factor for vaccines containing well-known live zoonotic agents or certain adjuvants. However, for the majority of vaccines the situation is not so clear. Risk assessments need to be performed, considering the specific properties of vaccines and their intended use on a case-by-case basis.

Risk assessment
Hazard identification and characterisation
Risk assessment is primarily concerned with examining the effects of the active ingredients, but also, if necessary, of excipients and, especially, adjuvants. For live vaccines the human pathogenicity of the vaccine strain is the main concern. For certain vaccine strains, pathogenicity has been documented by study of human cases caused by the same strain (e.g. Brucella melitensis Rev.1). The probable pathogenicity of a strain could also be assessed from published information for related modified strains or field strains (e.g. rabies) or from pharmacovigilance data. Agents that are rarely pathogenic for humans may be of special interest in the case of immunocompromised individuals or pregnant women (e.g. toxoplasma). For inactivated, but also for some live vaccines, effects of adjuvants, such as local or systemic reactions as a result of accidental injection, are of most concern (e.g. oil adjuvants).

Exposure assessment
Factors to consider include: the method of preparation of a vaccine (e.g. recovery from liquid nitrogen, rehydration, dilution of ingredients, loading in application apparatus or system) the route of administration of the vaccine (e.g. injection by syringe or mechanical device, coarse spray, oral) the duration and frequency of exposure to the vaccine the number, species and category of animals to be vaccinated (e.g. individual or mass vaccination, companion or food-producing animals) the time period of excretion from vaccinated animals. The risk of exposure to the vaccine is shared equally between both the person who administers the vaccine and people who assist in restraining the animal(s). In cases where the vaccine strain is excreted by the vaccinated animal(s), the animal owners or caretakers may in addition be exposed to the strain after vaccination.

Scope of the assessment


An assessment of the user safety of a product addresses only situations resulting from the normal conditions of use and from foreseeable accidents (including accidental selfinjection, oral ingestion or inhalation). It does not consider exposure resulting from deliberate misuse. For the assessment, the user is regarded as any person who: administers the vaccine comes into contact with the vaccine or components before its application to the animal (e.g. during storage or preparation of the product to be administered) comes into contact during its application may be exposed to the vaccine after its application (e.g. through contact with disposed of, unused or waste product, or with treated animals). This implies that the user can be, for example, a veterinarian, a farmer, a breeder, a pet-owner or any person who assists in restraining an animal during vaccination or who lives in the same environment. Consumers of products derived from vaccinated food-producing animals (such as meat and milk) are excluded from this definition (see Consumer safety). This discussion does not cover occupational safety during the manufacturing of vaccines or other risks, for example, due to injury from a mechanical vaccination device or from a contaminated needle.

Risk characterisation
In contrast to pharmaceuticals, where information on doseresponse relationships is available, in vaccines no quantitative risk assessment can be made. As an example, if hazards are identified for live vaccines, it must be assumed that the effects will occur at any exposure level.

Risk management and risk communication


Recommendations for managing risk include implementing appropriate precautionary measures such as

Rev. sci. tech. Off. int. Epiz., 26 (2)

345

the use of personal protective equipment. When the vaccine strain may be excreted, the period during which this is liable to occur should be stated on the package insert or label. In some cases, recommendations for appropriate action will be linked with particular characteristics of the user, such as immunocompromised persons and pregnant women. Guidance on remedial action to be taken following accidental contact is also given when necessary. If the advice of a physician is considered necessary, adequate information should be given on the nature of the identified risk, taking into account that it is not always possible for the physician in an emergency situation to identify the risk and take the full responsibility for risk assessment without proper information. Statements such as seek medical advice immediately and show the package insert or the label to the physician should be included only when a specific risk has been identified. When the assessment does not identify any specific risk, an appropriate statement could be included on a case-by-case basis (e.g. none, no specific risk identified). Guidance on standard warnings to be used in the EU has been published (3). A typical example is the following warning for vaccines containing oil adjuvants: To the user: This product contains mineral oil. Accidental injection/self injection may result in severe pain and swelling, particularly if injected into a joint or finger, and in rare cases could result in the loss of the affected finger if prompt medical attention is not given. If you are accidentally injected with this product, seek prompt medical advice even if only a very small amount is injected and take the package insert with you. If pain persists for more than 12 hours after medical examination, seek medical advice again. To the physician: This product contains mineral oil. Even if small amounts have been injected, accidental injection with this product can cause intense swelling, which may, for example, result in ischaemic necrosis and even in a loss of a digit. Expert and prompt surgical attention is required and may necessitate early incision and irrigation of the injected area, especially when there is involvement of finger pulp or tendon.

from those which arise naturally, and are considered not to affect consumers, consideration must be given to other substances contained in the vaccine, in particular excipients and adjuvants.

Scope of the assessment


The purpose of the assessment is to evaluate the known or potential adverse effects on human health of exposure to food-borne hazards or, more specifically, of exposure to residues of the chemical components of the immunological veterinary medicinal products used in food-producing animals. In the EU, such a safety-of-residue evaluation is required for all pharmacologically active substances which remain in foodstuffs obtained from animals to which the veterinary medicinal product in question has been administered. Active principles of biological origin used in an immunological veterinary medicinal product intended to produce active or passive immunity or to diagnose a state of immunity are excluded. In the EU the risk assessment of any chemical components of vaccines that may affect consumer safety is considered in a separate procedure prior to the marketing authorisation process. Council Regulation (EEC) 2377/90 lays down the procedure and lists all substances that are allowed to be used in veterinary medicinal products for food-producing animals. Appendix I lists all substances for which final MRLs have been established, Appendix II lists all substances for which the specification of MRLs was not considered necessary to protect consumer health, and Appendix III contains all substances with provisional MRLs (5). Adjuvants in vaccines, due to their very nature, are usually pharmacologically active and require a safety-of-residue evaluation. Depending on the nature of the excipient, these may be exempted following a case-by-case consideration based on appropriate data that has shown the absence of such activity at the dose at which the adjuvant is included in the veterinary medicinal product. Substances used in the manufacturing process of the active ingredients, which are not intended to be present in the final product but of which traces might be present, are not considered to require a safety-of-residue assessment. As mentioned before, medicinally induced immunological responses are usually indistinguishable from those which arise naturally, and are considered not to affect consumers. However, on a case-by-case basis, depending on the nature of the vaccine, e.g. in the case of live vaccines containing zoonotic organisms, a precautionary withdrawal period will be required to exclude risks to the consumer of food derived from the vaccinated animal.

Consumer safety
Problem statement
The use of a veterinary medicinal product in foodproducing animals may result in the presence of residues of the medicine in the animal body and in animal-derived foodstuffs such as meat, milk and eggs. This applies to pharmaceutical veterinary medicines as well as to immunological products. While medicinally induced immunological responses are usually indistinguishable

346

Rev. sci. tech. Off. int. Epiz., 26 (2)

Risk assessment
Hazard identification and characterisation
The purpose of hazard identification is to identify residues of a vaccine that may cause adverse effects on health and that may be present in any particular food derived from animals. In the hazard characterisation stage, the nature of the adverse effects associated with these residues is evaluated qualitatively and/or quantitatively on the basis of toxicological and pharmacological studies in laboratory animal species. Where available, observations in humans are taken into account. At this stage, where appropriate, the acceptable daily intake (ADI) for the substance under consideration is established.

potential exposure of the environment to the product, and the level of risk associated with any such exposure. Where it is concluded that the risk is low, there will generally be no need to proceed to Phase II and no further investigations will be required. In the majority of cases, the nature of vaccines is such that they will have a very low environmental risk potential. It can be expected that a Phase II assessment will be necessary only in exceptional circumstances, e.g. in vaccines containing live GMOs (3). The level of detail to be considered in a risk assessment will depend on the characteristics of the vaccine. Little detail will be required, for example, where it is immediately obvious that the hazards and hence the consequent risks are low, or that the proposed control measures are clearly adequate to limit contact between the product and the environment. For example, for inactivated vaccines to be administered by injection, the hazards and risks from the active ingredients are likely to be negligible.

Exposure assessment
The exposure assessment concerns vaccines intended for use in food-producing animals, and evaluates the likely intake of residues of a vaccine through food of animal origin. Data on absorption, pharmacokinetics, metabolism and residue depletion are usually necessary for the exposure assessment.

Risk assessment
Hazard identification and characterisation
In the context of the environmental risk assessment for vaccines, hazards are defined as those features of the substance which have the potential to cause harm to the environment either directly (as in the case of infection of a non-target-species by a vaccine virus) or through some form of possible event (such as infection by organisms excreted by the vaccinated animal). It is important to be exhaustive in the identification of possible hazards, which should aim to identify all possible factors contributing to adverse effects and should include the following: the capacity of live organisms to transmit to non-target species the shedding of live vaccine organisms from vaccinated animals (route, numbers, duration) the capacity to survive, establish and disseminate pathogenicity to other organisms

Risk characterisation
In a risk characterisation, an estimate is made of the risks to the consumer from any residues from vaccines that may be present in animal products. Regarding the chemical components identified as potential hazards for the consumer, the risk characterisation will decide whether MRLs would need to be established. Consideration is given to whether a withdrawal period would be necessary before food can be derived from the vaccinated animal.

Risk management and risk communication


The outcome of the assessment of risks to the consumer should be summarised and, where any risk is identified, appropriate advice should be given on mitigation measures. Such measures, e.g. a withdrawal period, should be incorporated in the SPC, package insert and label.

Environmental safety
Problem statement
A veterinary medicinal product may have undesirable effects on the environment and therefore, before any veterinary medicinal product can be authorised, any risk of harm it may pose to the environment should be assessed. On the basis of the assessment, specific risk mitigation measures may be considered.

potential for other effects of live product organisms the toxic effects of the product components the toxic effects of excreted metabolites. Regarding the two latter effects, in the case of products which are administered by injection, no detailed assessment of the potential risk of the excipients is likely to be required when the substances are of biological origin or form part of the animals normal diet.

Scope of the assessment


In the EU the environmental assessment is undertaken in two phases. Phase I is compulsory and should indicate the

Exposure assessment
In the first step, the likelihood (probability and frequency) of the hazard(s) is estimated. A key factor in determining

Rev. sci. tech. Off. int. Epiz., 26 (2)

347

this is the potential receiving environment. This includes the wider as well as the local environment in which the product is intended or likely to be used. Particular characteristics of the local environment that could contribute to the likelihood of the hazard e.g. climatic, geographical and soil conditions, demographic considerations, the types of fauna and flora in the potential receiving environment should be identified and assessed. Consideration should be given to any potential exposure of the environment to the product and the magnitude and duration of such exposure. The exposure assessment should consider: the type of packaging and procedures before and after administration the route of administration (parenteral vs. oral vs. oculonasal vs. spray) shedding of live product organisms (route, numbers, duration). For the hazard-survival capacity of living organisms, it is appropriate to assess the proportion of the organisms that are likely to survive. If the organism has pathogenic characteristics, the proportion of target species in the environment likely to be affected should be assessed, taking into consideration the likelihood that the organism will spread to or reach these species. It is recommended that the possibility of exposure and the likelihood of hazards occurring are expressed as high, medium, low or negligible, although it is recognised that this requires subjective judgment. Subsequently the consequences of possible hazards are assessed. This must be done for each identified hazard of the product being assessed. Whenever it is possible or probable that the components in the product will reach the environment, it must be considered whether that environment would cause or allow the hazard to happen. Thus, again, the characteristics of the potential receiving environment need to be considered.

For vaccinal organisms or excreted passage organisms, the main consideration will be the likely presence or absence of susceptible non-target species in the potentially affected environment.

Risk characterisation
Having identified any hazards and assessed the degree and likelihood of exposure and the consequences of that exposure, it is necessary to evaluate the risk associated with each hazard. Risk is generally held to be the product of exposure/likelihood and consequence. It is inevitably going to be difficult to multiply qualitative statements such as high and low, but Table I should help this process. The risk matrix is not definitive and there will always be some scope for flexible, case-by-case evaluation. In many cases, it will be necessary to decide between one of two outcomes and, as in the earlier parts of the process, some justification for the choice should be provided. In addition, a range of risks may be apparent if more than one hazard is being evaluated. There will therefore be a need to make an overall assessment of the risk, taking all factors into consideration. Once an overall assessment of the risk associated with each hazard has been produced, it will be necessary to evaluate the significance of the risk.

Risk management and risk communication


If the environmental risks are not as low as reasonably practicable, the process of risk assessment in relation to the hazard should be repeated to ascertain whether the use of additional management techniques could reduce the level of risk; consideration might be given, for example, to limiting the proposed routes of administration to those likely to lead to a lower level of risk. If it is considered that there is insufficient knowledge to come to a satisfactory conclusion, other studies and a Phase II assessment should be undertaken, addressing the specific issues that give rise to concern on a case-by-case basis. The outcome of the environmental risk assessment should be summarised and, where any risk is identified, appropriate warnings and advice on mitigation measures

Table I Environmental risk assessment: characterising the nature of a risk (high, medium, low, zero) by evaluating the likelihood of its occurrence and the severity of its consequences
High likelihood of hazard occurring Severe consequences Moderate consequences Minor consequences Negligible consequences High High Medium/low Effectively zero Moderate likelihood of hazard occurring High High Low Effectively zero Low likelihood of hazard occurring Medium Medium/low Low Effectively zero Negligible likelihood of hazard occurring Effectively zero Effectively zero Effectively zero Effectively Zero

348

Rev. sci. tech. Off. int. Epiz., 26 (2)

should be incorporated in the SPC, package insert and label.

Pharmacovigilance
Veterinary pharmacovigilance monitors the safety of veterinary medicines, including vaccines, used for the prophylaxis, diagnosis or treatment of disease in animals once they have reached the market after authorisation. The aim is to ensure that veterinary medicines are safe for the animals, for persons who come into contact with veterinary medicines, for the environment and, in the case of medicines used in food-producing animals, for consumers of the food derived from these animals. Under the EU pharmacovigilance scheme, suspected adverse reactions in animals and in human beings relating to the use of veterinary medicinal products under normal conditions are recorded. The EU system also takes into account any available information related to: lack of expected efficacy off-label use investigations into the validity of the withdrawal period potential environmental problems arising from the use of the product, which may have an impact on the benefit/risk balance of the product.

The responsibility for pharmacovigilance reporting lies with the marketing authorisation holders, who are required to collect and evaluate all reports of suspected adverse reactions to their veterinary medicinal products, and report to the responsible authority for the veterinary medicinal product concerned. The adverse reaction reports are submitted mainly by veterinarians, but also by animal owners. All suspected serious adverse reactions and human adverse reactions relating to the use of the veterinary medicinal product must be reported promptly by the company, usually within 15 or 30 days after it receives the report (e.g. 15 days for the EU), depending on the specific legislation of the country where the product is authorised. All adverse reports received, serious and non-serious, must be compiled in a so-called Periodic Safety Update Report (PSUR) or Periodic Summary Update (PSU), which includes a benefit/risk analysis of the product. The PSUR or PSU is to be submitted to the responsible authority at specified intervals. In the light of any adverse reports received, the benefit/risk analysis of the product is reviewed. If necessary, the conditions for the authorisation may be amended, e.g. by amending the claim or by adding new or revised warnings to the SPC, package insert and label. If the risks identified through pharmacovigilance cannot be overcome by risk management measures and the risk/benefit balance is no longer positive, the marketing authorisation of the product may even be suspended or withdrawn.

Utilisation sans risques de la vaccination et conformit des vaccins avec les exigences de la scurit sanitaire des aliments
K. Grein, O. Papadopoulos & M. Tollis
Rsum Grce aux volutions technologiques et rglementaires, les vaccins vtrinaires actuellement disponibles sont de grande qualit et prsentent un profil de scurit favorable en termes de risques potentiels pour les espces cibles, pour les personnes qui manipulent les vaccins, pour les consommateurs de produits alimentaires issus danimaux vaccins et pour lenvironnement. Le processus dautorisation exige quune srie dtudes soient ralises afin dvaluer linnocuit des produits. Lvaluation des vaccins au stade du dveloppement, de la production et de lutilisation sans risque se fait principalement au moyen

Rev. sci. tech. Off. int. Epiz., 26 (2)

349

dtudes approfondies conduites avant la commercialisation chez les espces cibles, ainsi que de tests de qualit spcifiques. Les garanties postrieures la commercialisation sont fournies par les bonnes pratiques de fabrication, par les essais de scurit par lots, par les inspections et par la pharmacovigilance. Aprs avoir identifi les dangers, une valuation exhaustive du rapport bnfice/risque doit tre ralise. Les rsultats de lvaluation mettent en avant certaines options de gestion du risque et dterminent les mesures rglementaires prendre en matire de scurit ; par exemple, des mises en garde spciales pourront tre exiges sur les emballages et les tiquettes des produits. Mots-cls Animal Bnfice Consommateur Environnement valuation Pharmacovigilance Risque Sant Scurit Utilisateur Vaccin.

Utilizacin segura de vacunas y observancia de las normas sobre higiene de los alimentos en el uso de vacunas
K. Grein, O. Papadopoulos & M. Tollis
Resumen Los adelantos tecnolgicos y los regmenes reglamentarios han ayudado a disponer de vacunas veterinarias que anan gran calidad y caractersticas de seguridad favorables desde el punto de vista de los posibles riesgos que presentan para los animales diana, las personas que entren en contacto con la vacuna, los consumidores de alimentos procedentes de animales vacunados y el medio ambiente. El proceso de autorizacin requiere que se presenten diversos estudios de seguridad para evaluar cada producto. Al evaluar la concepcin y fabricacin de vacunas y determinar el grado de seguridad que ofrece su empleo, se utilizan sobre todo datos procedentes de vastos estudios con animales diana, previos a la comercializacin, y de controles de calidad especficos. Una vez que un producto ha obtenido licencia de comercializacin tambin existen medidas de salvaguardia, que hoy en da consisten en aplicar buenas prcticas de fabricacin y pruebas de seguridad de los lotes, realizar inspecciones e instaurar un dispositivo de farmacovigilancia. Adems de determinar los peligros, es necesario proceder a una evaluacin completa de la relacin entre beneficios y riesgos. Los resultados de tal evaluacin determinarn la lnea que se adopte en cuanto a la gestin de riesgos e influirn en las decisiones normativas sobre el grado de seguridad que ofrece la vacuna. Cabra obligar al fabricante, por ejemplo, a incorporar un aviso especial en las etiquetas o encartes al embalar el producto. Palabras clave Animal Beneficio Consumidor Evaluacin Farmacovigilancia Medio ambiente Riesgo Salud Seguridad Usuario Vacuna.

350

Rev. sci. tech. Off. int. Epiz., 26 (2)

References
1. Clements D.N., Gear R.N., Tattersall J., Carmichael S. & Bennett D. (2004). Type I immune-mediated polyarthritis in dogs: 39 cases (1997-2002). J. Am. vet. med. Assoc., 224 (8), 1323-1327. 2. Committee for Medicinal Products for Veterinary Use (CVMP) (2003). CVMP advice on injection-site fibrosarcomas in cats. Vet. Rec., 152 (13), 381-382. 3. European Commission (2006). EudraLex: Volume 6 Notice to Applicants. Veterinary Medicinal Products. Available at: http://ec.europa.eu/enterprise/pharmaceuticals /eudralex/homev6.htm (accessed on 4 July 2007). 4. European Commission (2005). EudraLex: Volume 8 Notice to Applicants and Note for Guidance. Establishment of maximum residue limits (MRLs) for residues of veterinary medicinal products in foodstuffs of animal origin. Available at: http://ec.europa.eu/enterprise/pharmaceuticals/eudralex/ homev8.htm (accessed on 4 July 2007). 5. European Council (1990). Council Regulation (EEC) No. 2377/90 of 26 June 1990 laying down a Community procedure for the establishment of maximum residue limits for veterinary medicinal products in foodstuffs of animal origin Off. J. Eur. Communities, L 224, 1-8. 6. European Medicines Agency (EMEA) (2006). Series of guidelines regarding the data requirements and assessment of immunological veterinary medicinal products. Available at: http://www.emea.europa.eu/htms/vet/vetguidelines/immunol ogicals.htm (accessed on 2 June 2007). 7. European Parliament and Council (2004). Directive 2001/82/EC of the European Parliament and of the Council of 6 November 2001 (Off. J. Eur. Communities, L 311, 1-66), as amended by Directive 2004/28/EC of the European Parliament and of the Council of the 31 March 2004 amending Directive 2001/82/EC on the Community code relating to veterinary medical products. Off. J. Eur. Union, L 136, 58-84. Available at: http://ec.europa.eu/enterprise/ pharmaceuticals/eudralex/vol5/consol_2004/veterinary_code .pdf (accessed on 2 June 2007). 8. European Parliament and Council (2004). Regulation (EC) No. 726/2004 of the European Parliament and of the Council of 31 March 2004 laying down Community procedures for the authorisation and supervision of medicinal products for human and veterinary use and establishing a European Medicines Agency (Off. J. Eur. Union, L 136, 30.4.2004, 1-33. Available at: http://ec.europa.eu/enterprise/pharmaceuticals/ eudralex/vol-1/reg_2004_726/reg_2004_726_en.pdf (accessed on 2 June 2007). 9. European Pharmacopoeia (2004). European Pharmacopoeia 5th Edition & supplements. European Directorate for the Quality of Medicines and Healthcare (EDQM), Council of Europe, Strasbourg. 10. Falcone E., Tollis M. & Conti G. (1999). Bovine viral diarrhea disease associated with a contaminated vaccine. Vaccine, 18 (5-6), 387-388. 11. Hogenesch H., Azcona-Olivera J., Scott-Moncrieff C., Snyder P.W. & Glickman L.T. (1999). Vaccine-induced autoimmunity in the dog. In Veterinary vaccines and diagnostics (R. Schultz, ed.). Adv. vet. Med., 41, 733-747. 12. Kass P.H., Barnes W.G. Jr, Spangler W.L., Chomel B.B. & Culbertson M.R. (1993). Epidemiologic evidence for a causal relation between vaccination and fibrosarcoma tumorigenesis in cats. J. Am. vet. med. Assoc., 203 (7), 1046. 13. Martinod S. (1995). Risk assessment related to veterinary biologicals: side-effects in target animals. In Risk assessment for veterinary biologicals (E.G.S Osborne & J.W. Glosser, eds). Rev. sci. tech. Off. int. Epiz., 14 (4), 979-989. 14. Moore G.E., Guptill L.F., Ward M.P., Glickman N.W., Faunt K.K., Lewis H.B. & Glickman L.T. (2005). Adverse events diagnosed within three days of vaccine administration in dogs. J. Am. vet. med. Assoc., 227 (7), 1102-1108. 15. Newman S.J., Johnson R., Sears W. & Wilcock B. (2002). Investigation of repeated vaccination as a possible cause of glomerular disease in mink. Can. J. vet. Res., 66 (3), 158-164. 16. Spickler A.R. & Roth J.A. (2003). Adjuvants in veterinary vaccines: modes of action and adverse effects. J. vet. internal Med., 17 (3), 273-281. 17. Tllez S., Casimiro R., Vela A.I., Fernndez-Garayzbal J.F ., Ezquerra R., Latre M.V., Briones V., Goyache J., Bullido R., Arboix M. & Domnguez L. (2006). Unexpected inefficiency of the European Pharmacopoeia sterility test for detecting contamination in clostridial vaccines. Vaccine, 24 (18), 1710-1715.

Rev. sci. tech. Off. int. Epiz., 2007, 26 (2), 351-372

Marker vaccines and the impact of their use on diagnosis and prophylactic measures
P. Vannier (1), I. Capua (2), M.F. Le Potier (1), D.K.J. Mackay (3)*, B. Muylkens (4)**, S. Parida (5), D.J. Paton (5) & E. Thiry (4)
(1) Agence franaise de scurit sanitaire des aliments (AFSSA), BP 53, 22440 Ploufragan, France (2) Istituto Zooprofilattico Sperimentale delle Venezie, Legnaro, Padua, Italy (3) European Medicines Agency (EMEA), London, E14 4HB, United Kingdom (4) Virology, Department of Infectious and Parasitic Diseases, Faculty of Veterinary Medicine, University of Lige, B-4000 Lige, Belgium (5) Institute for Animal Health, Pirbright, GU24 0NF, United Kingdom After the first author, who coordinated the composition, the authors names are listed in alphabetical order. The different authors wrote on the following topics: I. Capua on avian influenza; S. Parida, D.J. Paton and D. Mackay on foot and mouth disease; M.F. Le Potier on classical swine fever; B. Muylkens and E. Thiry on infectious bovine rhinotracheitis and P. Vannier on Aujeszkys disease * The views expressed in this article are the personal views of the author and should not be taken to represent the position of the EMEA, the Committee for Medicinal Products for Veterinary Use or the European Commission ** Research fellow at the Fonds de la Recherche Scientifique FNRS in Belgium

Summary Molecular biology and technical advances in DNA recombination have ushered in a new era in vaccinology. This article examines the recent development of specific marker vaccines and examines the impact of their use on the diagnosis and prevention of major infectious diseases. Gene-deleted vaccines, DIVA strategies (differentiating infected from vaccinated animals) and similar methods have been successfully applied in the control and eradication of Aujeszkys disease, infectious bovine rhinotracheitis, classical swine fever, foot and mouth disease and, recently, avian influenza. The efficacy and performance of existing marker vaccines and their companion diagnostic tools (which should be assesed by an independent body) are discussed, as are the ways in which these tools are deployed by competent authorities. The limits and the advantages of the use of marker vaccines are carefully analysed in the light of practical experiences. Although these vaccines can limit the speed and the extent of virus dissemination and thus reduce the number of animals slaughtered, marker vaccines are no substitute for sanitary measures. Early detection and warning systems and the quick implementation of sanitary measures, including stamping out, remain key issues in the control of highly contagious diseases. Keywords Aujeszkys disease Avian influenza Classical swine fever Companion diagnostic tool Control DIVA (Differentiating infected from vaccinated animals) strategy Eradication Foot and mouth disease Latent infection Marker vaccine Sanitary measure Vaccination Viral excretion Virus carrier state.

352

Rev. sci. tech. Off. int. Epiz., 26 (2)

Introduction
Molecular biology and technological advances in deoxyribonucleic acid (DNA) recombination have ushered in a new era in vaccinology. In particular, deleted vaccines, used in conjunction with an appropriate diagnostic kit, have emerged over the past ten years, enabling infection-specific antibodies to be recognised regardless of an animals vaccination status. The first such vaccines were used to protect pigs against Aujeszkys disease (AD). The same principles were subsequently applied to the development of vaccines against infectious bovine rhinotracheitis (IBR). For classical swine fever (CSF), subunit proteins were obtained from baculovirus recombinant and the resulting vaccine obtained a European marketing authorisation, re-launching the debate on whether or not to use sanitary measures or medical prophylactic treatments. The same principle is also at work when the detection of antibodies to non-structural proteins (NSP) is used to identify animals infected with foot and mouth disease virus (FMDV), whether or not they have also been vaccinated. Furthermore, more recently, recombinant vaccines have been used to protect birds against avian influenza (AI).

These sequences, situated in enzymatic restriction fragment BamHI no. 7, code for two structural glycoproteins: gE and gI. Accordingly, the Bartha strain, when isolated under natural conditions, does not express gE, which makes it possible to distinguish vaccinated pigs from infected pigs, provided, of course, that the corresponding enzyme-linked immunosorbent assay (ELISA) kits are used. ELISA kits make it possible to detect anti-gE antibodies in the serum of pigs, by using monoclonal antibodies that are very specific to certain antigenic determinants of gE, as described by van Oirschot et al. (129, 131). Subsequently, knowledge about the molecular biology of mutants of the ADV led to a better understanding of the functions of the viral glycoproteins. The first factor of virulence that was identified in the herpes virus was the thymidine-kinase enzyme, which allows the virus to replicate itself in the central nervous system. Later, the virulence of strains of the ADV not expressing the glycoprotein membrane gE was seen to have diminished considerably compared with that of field viruses (9). This gE would therefore appear to play a major role in the spread of the virus within the nervous system, with the infection spreading both through the olfactory tract and trigeminal cavity (82). This knowledge has made it possible to develop new vaccines by means of genetic recombination, which modifies the genome of the vaccine strains in order to excise, remove or delete certain sequences that code for glycoproteins and prevents their expression. These proteins do not induce antibodies in vaccinated animals and so are used as serological markers for infection by wild-type viruses. The functions of these same proteins are often partially responsible for the virulence of field strains (such as gE); their non-expression helps to reduce or eliminate the pathogenicity of these vaccine strains, which always express the major glycoproteins (gB, gC, gD), thus inducing protective immune responses in vaccinated or infected pigs. Another generation of vaccines, not yet on the market, has appeared, which uses live vaccine strains of the genetically modified ADV as an expression vector of the gene coding for the immunogenic proteins of other viruses, such as CSF (135). These hybrid viruses protect the vaccinated animal against both AD and CSF Moreover, in-depth knowledge . of the molecular biology of the ADV has led to the creation of recombinants that cannot be shed by the vaccinated animal in an infectious form. Such recombinants can, however, spread from one cell to another in the inoculated organism, as do conventional live vaccine strains, but in rather limited sites (53). Finally, one should not overlook the considerable progress that has been made with immunological adjuvant technology, even though this is not directly linked with molecular biology. We have seen the emergence of vaccines

An historic example: marker vaccines used against Aujeszkys disease


Aujeszkys disease virus (ADV) belongs to the subfamily Alphaherpesviridae of the family Herpesviridae, which infect the central nervous system and other organs (such as the respiratory tract) in virtually all mammals, except humans and the tailless apes. It is associated primarily with pigs, the natural host, which remain latently infected following clinical recovery. After primary infection, most pigs develop clinical signs, depending on their age. In nave piglets, nervous signs are observed and the mortality can be very high. In sows, reproductive disorders are induced after infection. In fattening pigs, general clinical signs, such as fever and loss of appetite associated with respiratory disorders of varying severity, are observed. Silent infection can also occur.

Currently available Aujeszkys disease marker vaccines


Advances in molecular biology have contributed to better knowledge of the genome of existing vaccine strains. By studying conventional vaccine strains, it was found that certain coding sequences of the single-sequence short section of the Bartha strain of the ADV had been deleted.

Rev. sci. tech. Off. int. Epiz., 26 (2)

353

against AD that are produced by tank-mixing a live attenuated strain with an adjuvant comprised of mineral oils. At the same time, the nature of the oils used in adjuvant composition has evolved, as has emulsion technology, making the vaccines increasingly immunogenic and considerably reducing local reactions at the site of injection. ELISA kits which are available commercially use indirect or competitive techniques for measuring antibody levels. When marker deleted vaccines are used, several types of kits can be employed to detect specifically the infected vaccinated animals; the ELISA tests that detect gE antibodies are the most commonly used in the field in Europe, whereas tests that detect gG (NSP) or gC antibodies are used more frequently in the United States of America (USA) (89, 144). A combination of several kits is often used in the framework of control programmes against AD, such as gE kits associated with kits that detect gB antibodies or antibodies against the whole viral proteins. The latter two kits are used mainly in unvaccinated herds or regions where vaccination is not carried out; but they can also be used to interpret the herd status in regard to infection (gE+; gB+) or vaccination (gB+; gE). As well as testing sera, the ELISA can be adapted to test filter paper disks that have been moistened with a small quantity of blood obtained by puncturing a superficial vein (144). This technique is convenient for testing large numbers of pigs. The disks are air-dried before shipment to the laboratory. Moreover, muscular exudates can also be used as alternative biological samples as these kits have been evaluated taking serum samples as the reference; the individual sensitivity of the test was 93.2% and the individual specificity was 98.3% (64). Requirements for the detection of gE, gB or global viral antigen have been defined by several competent authorities. When comparing different serological tests that detect AD antibodies, the sensitivity of ELISA tests appears very good and the blocking ones often appear a little more sensitive than indirect ones, allowing earlier detection of more than 95% of infected pigs (88).

BoHV-1 is a pathogen that is found throughout the world and which displays significant differences in regional incidence and prevalence depending on geographical location and breeding management (1). BoHV-1 is responsible for significant financial losses incurred through disease and trading restrictions within the cattle industry, prompting the development of control programmes in North American herds. Based on serological surveys, several studies have aimed at identifying the risk factors for BoHV-1 seropositivity. Some of them are well characterised, e.g. age, sex (males are more frequently positive than females) and herd size (11, 107). Direct animal contacts, such as purchase of cattle and participation in cattle shows were also found to be important risk factors for the introduction of BoHV-1 (132, 133, 134). Other factors such as farm density or cattle density may increase the risk of BoHV-1 introduction (139). As reported for other diseases caused by herpesviruses in man and animals, the latency-reactivation cycle has a deep epidemiological impact since it is responsible for the maintenance of BoHV-1 in a cattle population. BoHV-1 infection of new generation cattle by latent carriers submitted to reactivation/re-excretion stimulus can occur at several different times, e.g. at birth (118), at mating, during transport (117) or following the introduction of heifers into a group of dairy cows. Therefore, the detection of BoHV-1 latent carriers is the main concern in the setting up of BoHV-1 control programmes. Moreover, sanitary measures must be taken to prevent the introduction of seropositive animals or even animals originating from a seropositive herd in order to improve vaccine programme efficacy. Depending on the seroprevalence of BoHV-1, eradication programmes are based either on the detection and the culling of seropositive animals, or on the repeated vaccination of infected herds. The use of vaccines to control BoHV-1 infections has evolved over the last few decades: previously they were used simply as an effective means of reducing the clinical impact of the disease, whereas nowadays vaccination programmes are implemented with the additional intent of preventing transmission, although this is not as readily achievable as a reduction in clinical signs. Indeed, BoHV-1 vaccines are not efficacious at preventing BoHV-1 infection and establishment of latency. Moreover, vaccine schemes must be accompanied by strict management measures to prevent the reintroduction of BoHV-1 into the cattle herd. Therefore, BoHV-1 control programmes may take a long time to eliminate this well-adapted virus infection of cattle.

Marker vaccines used against bovine herpesvirus-1


Bovine herpesvirus-1 (BoHV-1), classified as an alphaherpesvirus, is a major pathogen of cattle. Primary infection is accompanied by various clinical manifestations such as rhinotracheitis, pustular vulvovaginitis, abortion, and systemic infection in neonates. Following clinical recovery, a life-long latent infection is established in the nervous sensory ganglia of infected animals.

Currently available and future bovine herpesvirus-1 marker vaccines


In Europe, several countries have initiated control programmes aimed at BoHV-1 elimination and some

354

Rev. sci. tech. Off. int. Epiz., 26 (2)

countries are already BoHV-1-free (Finland, Sweden, Denmark, Switzerland and Austria). In this context, the ability to differentiate infected from vaccinated animals (known as the DIVA strategy) was critical for preventing trading restrictions in Europe. To set up the DIVA strategy, marker vaccines and reliable companion tests were developed. On the one hand, the marker vaccines must include BoHV-1 antigens able to induce a protective immune response. On the other hand, they consist either of subunit vaccines or of BoHV-1 strains from which a gene encoding a non-essential glycoprotein has been deleted. The deleted glycoprotein must be expressed by all BoHV-1 field strains and induce a detectable immune response. It should also be a virulence factor of BoHV-1 in order to ensure a further attenuation of the marker vaccine strain by the deletion of its gene. Non-essential BoHV-1 glycoproteins gC, gE, gI, gG and gM may be deleted to construct BoHV-1 marker vaccines. Four candidate deletion mutants (BoHV-1 gC-, gM-, gI- and gGnull mutants) do not correspond to the above-mentioned requirements. BoHV-1 gC-null mutants retain virulence in the natural host (56) and BoHV-1 gM-null mutants have never been tested in vivo (61). BoHV-1 gI-null mutants are not sufficiently immunogenic (56). BoHV-1 gG-null mutants are easily reactivated from latency (56) and gGspecific antibody tests are not available. Evidence of consistent results led to the selection of the BoHV-1 gE-null mutant as a candidate BoHV-1 deleted mutant for use as a marker vaccine. First, it was shown to be immunogenic (56) and to possess very little residual virulence (127). Moreover, BoHV-1 gE was shown to be expressed in a very large subset of BoHV-1 field strains (101) and a gE-specific antibody test has been developed as a companion test for the differentiation of vaccinated from infected animals (66, 130). The development of a BoHV-1 marker vaccine took advantage of the knowledge gained from ADV control and marker vaccines based on ADV gE-null mutants have successfully been developed for the control of ADV (137). Several studies have been conducted, and others are still in progress, to produce new generation vaccines against BoHV-1. Ideal marker vaccines should combine high levels of safety and efficacy. Several subunit vaccines have been tested. They consist mainly of glycoproteins B, C or D expressed in different systems such as transfected cell cultures (126), recombinant baculoviruses (125), recombinant adenoviruses (42, 43, 100, 148), or recombinant tobacco mosac viruses (96). The gD-based subunit vaccines are the most efficacious at reducing clinical disease and virus excretion when they are formulated with effective adjuvants. For example, chitosans (42) and CpG oligodeoxynucleotides (54, 86)

are new adjuvants that significantly enhance the protective immune response, as evidenced by increased neutralising antibody titres and reduced clinical disease and viral shedding following challenge. The latest vaccine approaches consist of plasmid DNA vaccines containing sequences encoding for the three major immunodominant BoHV-1 glycoproteins gC (50), gB (71) or gD (87, 120, 121). These constructs could potentially be administered by needle-free delivery methods which would help to prevent losses due to the tissue damage that classical vaccine delivery methods can cause (54, 124).

The biological properties of glycoprotein E (gE)-deleted bovine herpesvirus-1 marker vaccines


As for the majority of the BoHV-1 vaccines, marker vaccines are very effective at preventing clinical signs after challenge with highly virulent strains (55). However, none are able to fully prevent infection by the challenge strain, which establishes a latent infection, and might be reexcreted following a reactivation stimulus. New vaccine formulations and protocols have therefore been developed in order to improve the viral protection. Equivocal results were obtained when the two forms (inactivated and live attenuated) of the same marker vaccine were tested. When it was administered twice to seronegative cattle, the attenuated marker vaccine induced a better viral protection than the inactivated marker vaccine after challenge (12). However, the inactivated vaccine was more efficacious at reducing virus excretion after reactivation of latently infected calves than the live attenuated vaccine (13). An interesting approach was to combine the use of the attenuated vaccine as the priming dose and the inactivated vaccine as a booster injection to complete the primary course of vaccination. This kind of protocol was shown to be the most efficacious at reducing virus excretion following challenge (44, 58). The immune status of a BoHV-1 latent carrier is the key factor controlling viral reexcretion following a reactivation stimulus. Therefore, latent carriers must be repeatedly vaccinated at regular 6-month intervals in order to maximally decrease the risk of re-excretion (34). The efficacy of the DIVA vaccines was demonstrated in two field trials. In the first trial, a significant decrease in the number of gE seroconversions was observed in herds where the gE-deleted vaccines were used (77). The second study demonstrated that repeated vaccination using either inactivated or live attenuated gE-deleted BoHV-1 vaccines is efficacious at reducing the incidence of gE seroconversion in dairy cattle and consequently the herd prevalence of gE-positive animals (34). This study showed the superior efficacy of a protocol whereby all of the herd is vaccinated together at regular 6-month intervals

Rev. sci. tech. Off. int. Epiz., 26 (2)

355

compared to protocols where all of the herd is vaccinated, but not all at the same time (34). Three safety concerns about the live attenuated gE-deleted vaccine have to be addressed. First, gE-null BoHV-1 was demonstrated to establish a latent infection (67, 128) and to be re-excreted following experimental stimuli (67, 105) and in field conditions (35). However, there is so far no evidence that this deletion mutant can persist in the cattle population (78). A second concern rose from the production of seronegative BoHV-1 latent carriers following the use of BoHV-1 marker vaccines in passively immunised calves. Indeed, passively acquired colostral immunity interferes with an active antibody response following infection. It has been experimentally demonstrated that gE-negative BoHV-1 vaccine, when used in passively immunised calves, gives rise to seronegative vaccine virus carriers (67). The third concern is the potential for BoHV-1 recombinants to arise after co-infection of animals with a replicative gE-deleted BoHV-1 strain and a virulent BoHV1 field strain (115, 116). One field observation and two experimental findings underlie this concern: a) the isolation of a gE-deleted BoHV-1 vaccine strain in cows vaccinated eight months before (35) b) the frequent appearance of BoHV-1 recombinants in coinfected calves (105) c) the experimental isolation of a virulent gE-deleted BoHV-1 recombinant (84, 85).

BoHV-1 eradication programme in the Netherlands, has a very high sensitivity (> 99.0%) and a very high specificity (> 99.9%) (29). Finally, a useful approach for dairy cattle herd monitoring is the regular serological testing of milk tank samples (140). A positive result is obtained when 15% of the dairy cows are seropositive towards BoHV-1. This level of seropositivity is rapidly obtained in cases of BoHV-1 introduction into a previously negative herd.

Marker vaccines used against classical swine fever


Classical swine fever, previously known as hog cholera, is still a serious threat for the domestic pig population as it is a highly contagious viral disease of worldwide importance. Pigs and wild boars are the only natural reservoir of CSF virus (CSFV). CSFV, bovine viral diarrhoea virus (BVDV), and border disease belong to the genus Pestivirus of the Flaviviridae family. These are small, enveloped, positive single-stranded ribonucleic acid (RNA) viruses. The piglets develop more evident clinical signs than the adults. The usual clinical sign is hyperthermia, which is usually higher than 40C in piglets (which pile together in the corner) but in adults it can be lower (39.5C). The first usual signs of the acute form are anorexia, lethargy, conjunctivitis, respiratory signs and constipation followed by diarrhoea. The chronic form of the disease is generally fatal. Often the infected pigs present jaundice and cyanosis before death (65).

Properties of companion diagnostic tools


Even if the DIVA strategy has been demonstrated to be efficacious, it presents some weaknesses (7). Indeed, the strength of the tool is fully dependent on the capacity of the diagnostic test to detect BoHV-1 gE-specific antibody. But the sensitivity of the only available gE-specific ELISA is around 70% (62, 97). This rather low level of sensitivity has a 30% false-negative rate in individual tests but it remains sufficiently high for use at infected-herd level. The problem of the low sensitivity of this test is compounded by the weak level of the delayed immune response raised against BoHV-1 gE, which means that it can be as many as 42 days after infection before gE antibodies can be detected (7). The specificity of the gE-specific ELISA test is 92% (62). Although it is an acceptable level in the first steps of a control programme, this lack of specificity will be responsible for several misleading false positive results in herds where BoHV-1 has been eradicated. In these herds, a serial combination of serological tests should be performed. The Danish test system (consisting of a blocking and an indirect ELISA), which was used in the

Currently available classical swine fever vaccines


Two live attenuated vaccines have been used successfully for many years. The live vaccines include the Chinese strain also known as C strain vaccine, which was attenuated by serial passages in rabbits and later adapted to cell cultures (136), and the Thiverval strain derived from the virulent Alfort strain through more than 170 serial passages at 29C to 30C (4, 63). These traditional live vaccines induce a high level of protection against the development of clinical signs and neutralising antibodies at two weeks post challenge. Dewulf et al. (32) demonstrated that no virus transmission was detectable even when the pigs were challenged on the same day as the vaccination. The vaccinal protection lasts at least six to ten months whatever the immunisation route used: intramuscular or oronasal (57, 114). The main problem of using these live vaccines is that it is impossible to distinguish antibodies that are the result of vaccination from those that are due to natural infection. To clear up this difficulty, different teams have worked on the development of marker vaccines. CSFV envelope

356

Rev. sci. tech. Off. int. Epiz., 26 (2)

glycoprotein E2 is the major protective immunogenic protein responsible for eliciting neutralising antibodies and conferring protective immunity against the virus and it has been demonstrated to be a highly conformational dependent immunogenic protein (70, 149). Different types of subunit or marker vaccines have been developed as nonreplicative ADV expressing the E2 of CSFV (94), or live vectors such as porcine adenovirus. DNA vaccines have also been developed (3) but as yet these do not induce any real protection in pigs. Different vaccine schemes or combinations have been used such as: a prime-boost strategy (DNA-adenovirus) (52) or a co-administration with some interleukin (IL) recombinant protein such as IL-3, 12 or 18 or via a DNA vector (2, 141). At the present time, only two E2 recombinant protein subunit vaccines produced in the baculovirus expression system have been licensed for market use. The efficacy of these two commercially available E2 marker vaccines has been extensively assessed in different vaccinationchallenge and transmission trials. The results of these experiments were rather variable. A single vaccination with a vaccine dose of 32 g of E2 in a water-oil-water adjuvant prevented clinical signs and mortality following a CSFV challenge three weeks after vaccination (15). At least 14 days were needed to obtain clinical protection in growing pigs vaccinated with a single dose (14, 123), but in the case of earlier challenge, no protection against the disease and no reduction of virus shedding has been demonstrated (123). The ability of the two marker vaccines to prevent transplacental transmission of CSFV has also been evaluated. The results showed that with a double vaccination, virus spreading by transplacental infection under the conditions of emergency vaccination could not be prevented in most of the vaccinated animals and could lead to the carrier sow syndrome and, consequently, to the late onset form of CSF (30). Based on the results of using the double vaccination protocol on pregnant gilts challenged 46 days after the second immunisation, it was concluded that double vaccination with an E2 subunit marker vaccine protects pregnant gilts from the clinical course of the disease but does not prevent horizontal nor vertical spread of the CSFV (31). Despite the fact that these results indicated that the efficacy of these vaccines was not ideal, their use in an emergency vaccination protocol has not been banned by the European Commission (EC).

field sera were used as well as sera from the weaner and sow experiments carried out during the marker vaccine trial. Both discriminatory ELISAs were less sensitive than conventional CSF antibody ELISAs, although there was considerable variation between them. Neither discriminatory ELISA consistently detected the markervaccinated, CSF-challenged weaner pigs correctly as CSF positive, although CSF-challenged pregnant sows were identified correctly. The limitations of these discriminatory ELISAs would prevent the use of the two licensed marker vaccines under emergency field conditions (40). In 2003, the EC supported another large-scale interlaboratory trial to assess the performance of a new version of a companion Erns ELISA test. It was concluded that even if the specificity and the sensitivity of the test was better than the previous kits tested in 1999 (40), there was still a need for more reliable tests to be sure that a vaccinated pig has not been infected and is not a virus carrier.

Marker vaccines used against foot and mouth disease


Foot and mouth disease is a highly contagious disease of domestic and wild cloven-hoofed animals including cattle, sheep, goats and pigs. It is caused by a virus (FMDV) of the genus Aphthovirus, family Picornaviridae and exists as multiple serotypes and subtypes; it causes severe economic losses through decreased livestock productivity and trade restrictions. The virus is widely distributed and the disease is completely absent only in the European Union (EU) and in the Australasian and North American continents.

Current vaccines and their biological properties in the framework of eradication and control
In areas with endemic FMD, vaccination is commonly used in conjunction with zoosanitary measures to minimise losses and reduce virus circulation. In FMD-free countries, such as those of the EU and North America, the policy for controlling outbreaks has been primarily based upon stamping out, i.e. slaughtering of infected and contact animals together with restrictions on the movement of animals and animal products. Nevertheless, provision is retained to vaccinate under emergency circumstances where an outbreak is or threatens to become extensive. During and after the 2001 FMD outbreaks in Europe there was a growing desire to place more emphasis on a vaccinate-to-live policy to reduce reliance upon largescale slaughter of herds at risk of becoming infected. According to such a policy, the stamping out of infected premises and the imposition of movement restrictions would be accompanied by a limited period of emergency

Properties of companion diagnostic tools and their sensitivity and specificity limits
Discriminatory companion ELISA tests are based on the detection of antibodies to the Erns protein. In 1999, sixteen national swine fever laboratories participated in testing the discriminatory ELISAs. The two available kits were tested for sensitivity, specificity, reproducibility and practicability. Reference sera (CSFV and BVDV antibody positive) and

Rev. sci. tech. Off. int. Epiz., 26 (2)

357

vaccination of surrounding herds, followed by serosurveillance to detect and eliminate infected animals not identified on the basis of clinical disease or epidemiological tracing. In support of this policy, new regulations have been approved by the World Organisation for Animal Health (OIE) and the EU so that countries using this approach can now regain their FMD-free status six months after the last infection has been reported rather than one year later as was previously the case (39, 145). Current FMD vaccines are produced by infecting susceptible cell cultures (most frequently baby-hamster kidney cell lines) with virulent FMDV, followed by chemical inactivation with binary ethyleneimine and purification by ultra filtration. At formulation, antigen is mixed with either an aluminium hydroxide and saponin adjuvant to make an aqueous vaccine for use in ruminants or is emulsified in oil to make a vaccine for the immunisation of pigs and ruminants. These vaccines have been used successfully for decades to control FMD and regular mass vaccination, mainly of cattle, has helped in the eradication of the disease in some regions such as Europe and South America. Emergency vaccine banks have been established by national and international agencies, holding reserves of concentrated unformulated antigen frozen over liquid nitrogen (36). These may be formulated at higher doses than would be used for prophylactic vaccination so as to induce a rapid onset of immunity after administration as a single dose. Nevertheless, there are a number of concerns and limitations with the use of conventional vaccines in emergency control programmes. Their production requires the growth to high titre and subsequent complete inactivation of virulent strains of FMDV. Although this is conducted within high containment facilities there is still the potential for escape of live virus from these facilities or for inadequate inactivation of virus and these concerns have led some FMD-free countries to prohibit vaccine manufacture on their territory. Conventional FMD vaccines are more difficult to standardise than vaccines produced by synthetic or recombinant techniques and final product testing for safety and efficacy still requires in vivo testing in animals. As mentioned earlier, most vaccines are prepared from concentrated cell culture supernatants from FMDV infected cells and therefore contain variable amounts of viral NSP. Induction of antibody to NSP by vaccines contaminated with residual amounts of these proteins makes it difficult to identify infection in vaccinated populations by the use of NSP antibody tests (see ahead). In addition, a consistent cold chain is required in the field for the vaccine to remain efficacious. Although conventional vaccines can prevent clinical signs and spread of the disease in vaccinated animals, they do not induce a sterile immunity and therefore may not prevent virusexposed animals from becoming acutely infected, and a proportion of such animals will become persistently

infected virus carriers (25, 90, 91) whose presence jeopardises recovery of FMD-free status. Moreover, full protection takes time to develop and is short-lived without repeated booster doses (27, 37). Even when emergency vaccine was administered with a ten times greater antigen payload than the normal dose it could not fully protect vaccinated cattle from a severe challenge at ten days post vaccination (Cox and Barnett, unpublished results).

Possible future vaccines


Much work has been done on the development of alternative vaccines, including subunit vaccines based on highly immunogenic FMDV proteins or peptides and DNA vaccines (10, 26, 33, 41, 59, 60, 69, 99, 109), but their immunogenicity has been found to be much lower than that of conventional FMD vaccines. Efforts to produce attenuated FMD vaccines by the adaptation and further passage of FMDV in nonsusceptible hosts have been unsuccessful due to the reversion of the attenuated viruses to virulent forms (76). Targeted deletion of the Lpro gene, which is not essential to virus replication, produced a vaccine that induced a good FMD-specific neutralising antibody response, but could not protect fully (23, 79). Although a recombinant virus in which the RGD receptor binding site was deleted induced protection in natural hosts (74), with such a virus there is potential for selection of virus variants that could enter cells by utilising other receptors (5, 6). Another approach has been to produce a vaccine which expresses the entire virus capsid, and therefore all of the immunogenic sites present on intact virus, but lacks the infectious nucleic acids (8, 48, 49, 68, 102). Using this strategy, the Plum Island Animal Disease Center in the USA (47, 80, 83, 147) inserted the complete capsid coding sequences of FMDV into a live replication-defective human adenovirus vector, along with the FMDV 3C protease needed for capsid assembly. One parenteral inoculation with this vaccine induced antibody and clinical protection within seven days. The same adenovirus vector expressing the cytokine porcine interferon-alpha could protect animals from FMD for three to five days within one day of vaccination (22, 47).

Properties of companion diagnostic tools and their sensitivity and specificity limits
Conventional FMD vaccines are mainly comprised of viral structural proteins (SP) plus RNA and contain only trace amounts of viral NSP synthesised during viral replication. Furthermore, the amount of residual NSP present in vaccines can be reduced by additional purification steps during antigen preparation. Therefore, conventional vaccines elicit a mainly anti-SP antibody response, whereas

358

Rev. sci. tech. Off. int. Epiz., 26 (2)

infection with replicating virus elicits antibodies to both SP and NSP. Consequently, serological methods which detect antibodies to FMDV SP such as the liquid phase blocking ELISA (51), the solid phase competition ELISA (73) or the virus neutralisation test (45) cannot distinguish between infection and vaccination with conventional vaccines. However, ELISAs that measure antibodies to different NSP (3ABC, 3AB, 3A, 3B, 2A, 2B and 2C) can be used as marker tests to detect infection in conventionally vaccinated animals (17, 26, 28, 75, 98, 103, 106, 108). Incomplete purification of vaccine antigen as well as multiple vaccination increases the likelihood of inducing NSP antibodies, but the latter will not arise after emergency vaccination of previously nave animals. To date, the most promising NSP tests have been those that detect antibody to NSP 3ABC, and the OIE index method is an indirect 3ABC screening ELISA (first developed in South America [NCPanaftosa]) supported by a confirmatory immunoblotting test against NSP 3A, 3B, 2C, 3D and 3ABC (143). A number of commercial 3ABC ELISA test kits have recently become available and their sensitivity and specificity were compared to one another and to the index screening method at a workshop in Brescia (Italy) in 2004 (16). The specificity of the tests ranged between 97% and 98%, including when used to test cattle that had been given a single dose of European vaccine. All of the tests were highly sensitive for detection of infection in unvaccinated cattle, whereas the sensitivity of the tests to detect viral carriers in vaccinated and subsequently infected cattle ranged from 68% to 94% depending on the test used. The workshop concluded that two tests performed comparably to the OIE index method (Ceditest FMDV-NS, Cedi Diagnostics B.V. and 3ABC trapping-ELISA, IZS-Brescia) of which the Ceditest is the only one available as a commercial kit. Guidance on how to carry out post-outbreak serosurveillance for FMD in vaccinated populations is provided by the OIE Terrestrial Animal Health Code (Terrestrial Code) (145) and by the European Directive on FMD control (39). The Terrestrial Code requires that herds containing seroreactors must be followed up to determine whether these contain infected animals or not; finding evidence of infection at any stage automatically invalidates freedom from infection status. The European Directive on FMD control specifies that serosurveillance should be carried out at least one month after an outbreak has finished or one month after the last use of vaccine, whichever is the later. Further, it states that the entire vaccinated population should be sampled and tested or enough should be sampled and tested to give 95% confidence to detect a within-herd prevalence of infection of 5%. The problem of some vaccinated animals becoming carriers without seroconverting to NSP can be overcome by

interpreting NSP test results on a herd basis (104), although this still leaves a lack of certainty over freedom from infection in small herds (46, 92), since a test with 80% sensitivity at the individual level requires at least two infected animals in the herd to be sampled to have 95% confidence of detecting at least one of them (93). Imperfect test specificity can be partly overcome by retesting positive samples. For example, with the Ceditest at the Brescia workshop, discounting the positive results that were not confirmed on Cedi retest increased the specificity to 99.2% and decreased the sensitivity from 86.4% to 85.1% (93). A further increase in test specificity could be achieved by a second retesting of Ceditest-confirmed positive samples using another non-covariant, commercially available NSP assay (SVANOVIR FMDV 3ABC-Ab ELISA, Svanova, Upsala, Sweden), resulting in an overall specificity and sensitivity of 99.98% and 71.2% respectively (93). Foot and mouth disease is therefore a good example of where advances in vaccine production technology to reduce contamination of vaccines with NSP antigens, together with advances in diagnostic techniques to detect antibody to these antigens, have resulted in the development of marker vaccines and companion diagnostic tests that are sufficiently robust that they have resulted in amendments to FMD control policy such that a policy of vaccinate to live is now supported in appropriate circumstances. Improved FMD marker vaccines and tests may be available in the future. For example, the experimental adenovirus vectored vaccines described above express recombinant viral capsids that are devoid of NSP 3D, which is a protein that elicits a strong antibody response but cannot be eliminated from conventional vaccines by purification (81). This would enable use of a companion marker test for infection based on detection of anti-3D antibody (47). Testing of saliva for FMDV-specific IgA is also a promising tool for detection of infection in animals given conventional vaccine by the parenteral route (90).

Marker vaccines used against avian influenza


Avian influenza viruses all belong to the Influenzavirus A genus of the Orthomyxoviridae family. They are enveloped, negative-stranded RNA viruses with a segmented genome, consisting of 8 genes (PB1, PB2, PA, HA, NP, NA, MA and NS). AI viruses may be classified on the basis of the severity of the clinical signs they cause in susceptible birds. Low pathogenicity AI (LPAI), may be caused by viruses belonging to all 15 haemagglutinin types (H1-H15) and produce a mild disease in susceptible poultry characterised by respiratory and enteric signs that are often associated in breeders and laying hens with reproductive disorders. Some LPAI viruses are termed

Rev. sci. tech. Off. int. Epiz., 26 (2)

359

mildly pathogenic AI viruses. Highly pathogenic AI (HPAI) is, in contrast, a systemic viral disease of poultry with mortality that approaches 100% in many gallinaceous birds. The clinical disease HPAI is caused only by viruses of the H5 and H7 subtypes that contain multiple basic amino acids at the deduced sequence of the cleavage site of the precursor of the haemagglutinin molecule. The main clinical signs presented by HPAI-infected poultry are anorexia, depression, cessation of egg-laying, followed by complete reluctance to move and tremors of the head, paralysis of the wings and incoordination of the leg movements (19).

collected from natural outbreaks, selected without defined criteria. For this reason they may contain seed viruses of either high or low pathogenicity, although the OIE guidelines indicate that LPNAI strains should be used (146). In order to be efficacious, inactivated conventional vaccines must contain a seed virus of the same H subtype as the field strain against which vaccination is directed, while the subtype of the other surface antigen, the neuraminidase protein (N), is virtually irrelevant with regard to protection (111). Thus, vaccines may contain a seed virus of the same subtype as the field strain (H5N1 vaccine to combat an H5N1 field challenge) or may be of the same H subtype but of a different N (H5N9 vaccine to combat an H5N1 field challenge). The latter is known as heterologous vaccination. Currently there are two methods of detecting field exposure with this type of vaccine that have been evaluated in the field with satisfactory results. The introduction of unvaccinated sentinels into the shed has been used as a method of identifying field exposure within a vaccinated population, regardless of the strain used in the vaccine. This system requires the identification of unvaccinated birds and regular clinical inspections in conjunction with serological testing to detect LPNAI, HPNAI being clearly visible as clinical pathology in the sentinels. The system is deemed to be valid, but requires preparatory work and is more time consuming, especially when the number of herds to be vaccinated is very high, and particularly when birds that are not confined to cages need to be identified within the flock. Furthermore, the risk that sentinel birds could be substituted with nave birds in order to escape restriction policies exists. In the case of heterologous vaccination, it is possible to use the diversity between the N antigen contained in the vaccine and the one in the field as a natural marker system. This DIVA system was developed in Italy in 2001, and has been successfully used to combat multiple introductions of NAI (18). The system is based on the detection of antibodies to the N protein of the field virus, which represent evidence of field infection. Currently, only an indirect immunofluorescence antibody test has been fully validated (20, 21). An encouraging system that can be used as a companion test to vaccines containing homologous or heterologous seed strains, is based on the detection of anti-NS1 antibodies (122). This system is based on the fact that the NS1 protein is synthesised only during active viral replication and is therefore not present in significant amounts when inactivated vaccines, that do not replicate in the bird, are used. Birds that are vaccinated with such

Marker vaccines and companion diagnostic tests for notifiable avian influenza
Increased knowledge of AI occurrence and epidemiology has driven a revision process of the definition of AI for international trade regulations laid down by the OIE. The revised Chapter on AI now reads For the purposes of this Terrestrial Code, avian influenza in its notifiable form (NAI) is defined as an infection of poultry caused by any influenza A virus of the H5 or H7 subtypes or by any AI virus with an intravenous pathogenicity index (IVPI) greater than 1.2 (or as an alternative at least 75% mortality) as described below. NAI viruses can be divided into highly pathogenic notifiable AI (HPNAI) and low pathogenicity notifiable AI (LPNAI) (146). The aim of this section is to review currently available marker vaccines and companion diagnostic tests for NAI viruses of the H5 and H7 subtypes. Research in AI vaccinology has only recently become a field of interest for pharmaceutical companies and for research institutions, and for this reason the selection of products and performance of companion diagnostic tests are not adequate to cover fully all the complex field situations this infection may cause. Antigenic cross-reactivity between strains of the same H subtype is believed to occur even between strains belonging to different lineages. However, how crossprotection will be influenced by immunological pressure generated by the variety of seed strains is currently unknown (112). Similarly, the occurrence and extent of antigenic drift in this situation is impossible to predict, but could become a significant issue in the future.

Inactivated conventional vaccines and companion diagnostic tests or systems to reveal field exposure
These vaccines are based on a preparation containing inactivated virus grown in embryonated eggs. The seed strains that are currently being used are field isolates

360

Rev. sci. tech. Off. int. Epiz., 26 (2)

vaccines will develop antibodies to the NS1 only following field exposure (38). Full and field validation of this system under different circumstances is still in progress (H. Chen, personal communication) and should be made available before this system is recommended. No diagnostic kit has been validated to date.

on the one hand an excellent replicative efficiency which ensures high titred, consistent virus yields during production and on the other, suitable surface antigens. Since these vaccines have the same properties as conventional inactivated vaccines, the same companion tests apply. Recombinant protein-based vaccines are synthesised in the laboratory by expressing the haemagglutinin in a suitable system, for example baculoviruses, plants and yeasts (110). Several prototypes have been generated and have been tested in the laboratory with encouraging results, however, probably due to the cost of production, they have not attracted any commercial interest to date. The use of any of these engineered vaccines in the field would have several advantages, including the possibility of using a variety of companion diagnostic tests, as is the case with recombinant vectored vaccines and of course, of updating the haemagglutinin component should this be required. In summary, to date, only conventional inactivated (containing natural or synthetic strains) and recombinant live vectored vaccines are available for use and can be coupled with a suitable companion diagnostic test. Both categories have some advantages and limitations in their application in the field, and certainly, considering the complexities of these vaccines and the need to extend their use, more research is needed to optimise products and companion tests in order to tackle the current limitations to their use in the field.

Engineered vaccines and companion tests


Engineered vaccines include all vaccines that are not natural isolates or natural reassortants. These include: recombinant live vectored vaccines and vaccines based on reverse genetics and recombinant proteins. Recombinant live vectored vaccines are based on inserting an H5 or H7 haemagglutinin gene in a suitable, replicating vector which will, during its replication, induce the production of antibodies to the haemagglutinin of the influenza gene. Fowlpox-based recombinant viruses have obtained marketing authorisations and are being used currently in several countries. Newcastle disease virus (NDV)-based recombinants have been developed by several groups (H. Chen, personal communication; 113, 138), with a product developed by Chinese researchers being used currently in the field in the Peoples Republic of China. An infectious laryngotracheitis-based product has also been described (72). All these preparations, with the exception of the one developed by Swayne (113), offer protection from clinical signs and reduce shedding levels. However, for most of these products, their efficacy in the presence of preexisting antibodies due to natural infection with a field strain of the vector virus, e.g. NDV or fowlpox virus remains to be established (111, 113). The greatest advantage to the use of these products is that companion diagnostic tests directed to detect antibodies to any viral proteins other than the haemagglutinin may be used to identify field-exposed flocks. Thus, the agar gel immunodiffusion or ELISA tests directed to the detection of antibodies to the NP or M proteins, can be successfully used, and enable the detection of field challenge caused by any influenza A virus. Tests directed to the detection of antibodies to the N protein, identify field exposure only to viruses of known N subtype. Inactivated vaccines generated by reverse genetics have recently been developed in the USA and in the Peoples Republic of China (119). The seed strain contained in the vaccine is basically a synthetic virus, completely engineered in the laboratory. These viruses contain a backbone derived from a virus that has high replication capacities (A/Puerto Rico/8/XX) with the two genes encoding for surface antigens (H and N) derived from contemporary viruses. This combination of genes allows

Limitations and advantages of the use of marker vaccines


In spite of the major progress that has been made as a result of the development of marker vaccines, it would be a mistake to consider that their use could simply replace sanitary prophylactic measures. Indeed, past experience is very useful for assessing the limits and the advantages of the use of these marker vaccines, which could be a powerful tool in a set of measures to control and eradicate a contagious disease. However, the use of such vaccines has to be adapted to the epidemiological situation, the contagiousness of the disease concerned and to the presence or absence of conditions with the capacity to influence the spread of infection. To control a disease, the key point is to detect clinically inapparent infected animals (healthy carriers) which can infect in-contact susceptible animals. When vaccination is used, the critical stage of alert induced by the appearance of clinical signs is removed or suppressed. For this reason, such vaccines have to be as efficient as possible, not only to protect vaccinated animals against clinical signs, but also to

Rev. sci. tech. Off. int. Epiz., 26 (2)

361

prevent, as far as possible, the excretion of the virus by vaccinated and subsequently infected animals. Moreover, the sensitivity of the diagnostic kits should be as high as possible to reduce to the greatest possible extent, the probability of false negative results; indeed, in such a strategy, the epidemiological consequences of false positive results are less significant than false negative results, as positive results are generally confirmed (in a second, complimentary phase) by a reference laboratory using another diagnosis tool. The longest experience of using marker vaccines has been accumulated in relation to the control and eradication of AD. In this case, the use of deleted marker vaccines has represented a considerable advance in programmes to control the disease in several countries. First, these vaccines have made mass vaccination possible, whilst retaining the means for serological detection of infection. This has enabled vaccinated herds which subsequently become infected to be pinpointed so that the necessary measures can be applied to prevent the field virus from spreading further. Second, it has become possible to implement sanitary measures in a gradual manner in vaccinated, infected herds, by culling the infected sows at varying speeds, as required. These infected sows are detected through serological screening using the ELISA technique, which enables vaccinated pigs to be distinguished from those that have been vaccinated and then subsequently infected. This means that vaccination has a combined effect which allows a programme of prophylactic treatment to be carried out in total safety. Mass vaccination, conducted several years in succession, limits the quantity of virus shed into the air by the infected pigs, thereby considerably reducing the probability and scale of the air-borne spread of contagion between herds (95, 137). Furthermore, systematic vaccination avoids economic losses due to a poorly controlled infection. Consequently, after several years of vaccination in a country or region, and the introduction of sanitation measures into the infected herds

and the continual culling of the oldest infected sows, the prevalence of infection gradually diminishes; in addition, the incidence of infection remains very low and is kept under control. However, the cost of vaccination must be taken into account when calculating the total cost of a prophylactic treatment. Authors compared the cumulative costs, over ten years, of various measures for controlling AD in northern Germany, following the introduction of prophylactic treatment (Table I). Of the five possible strategies, the most economical is based on systematic vaccination, followed by screening of infected herds and the slaughter of sows presenting infection-specific antibodies. Of course, this is a cumulative cost which takes all costs into account: those of the State, those of trade organisations and those of breeders. The authors note that the prevalence of infection diminishes during the first two years, but that vaccination alone is not enough to eliminate infection; during the final years of the programme the ADV persisted in a small number of herds. After 42 months of vaccination, few herds still harboured infected breeding animals. The detection and elimination of these breeding animals lead to a sharp drop in the prevalence of infection in breeding herds, whilst the risk of infection in fattening farms (or of fattened animals in other farms) becomes zero (142). As a result of past and present experience, it has become possible to develop a strategy for using vaccines to control AD. In countries that have sufficient economic resources to envisage eradication of the infection, there are two possible options: where the prevalence of infection in a given territory is high, or there is a high density of pig herds, mass vaccination with effective deleted vaccines is the only means of reducing prevalence; however, although these measures are necessary, they are not in themselves sufficient to eradicate the infection. Identification, screening and culling of the infected breeding animals appear to be essential to successful eradication whilst

Table I Total costs of potential control strategies for Aujeszkys disease in Germany over the ten years following initiation of prophylactic treatment (147)
Strategy Vaccination of sows twice per year Vaccination of sows three times per year Vaccination of sows three times per year, and of pigs for consumption once per year Vaccination of sows three times per year, and of pigs for consumption once per year. Serological controls and slaughter of pigs presenting infectious antibodies (where prevalence is < 10%) Control and slaughter of pigs possessing infectious antibodies 9,907 19,342 Cumulated cost over ten years (in thousands of euros) 18,085 18,143 13,534

362

Rev. sci. tech. Off. int. Epiz., 26 (2)

continuing to systematically vaccinate the animals at least two years after elimination of the last infected pig. In the latter case, it is advisable to control the movements of piglets, pigs for consumption and breeding animals as much as possible; by contrast, in regions with a low herd density and low prevalence of AD, serological screening and the culling of infected breeders or total slaughter of certain herds, appear to be the most effective, and in some cases the most economical, measures for achieving eradication. Such measures have been successfully introduced, for example, in Denmark, the United Kingdom, Sweden and several regions of France. However, independently of the performances of the vaccines and the companion kits (which are key issues in determining the use of such tools), other examples show that the use of marker vaccines would not have had a significant impact on the control of the situation. When a serious CSF epizootic hit several European countries in 1997, many people believed that the use of these new generation serological marker vaccines could prevent a further animal health catastrophe. However, an analysis of the situation that existed when the first CSF outbreaks appeared in the Netherlands revealed that more than 22 herds were already infected when the primary outbreak was identified in the region of Venhorst on 4 February 1997. The situation rapidly became dramatic for the region because farmers had already sold piglets before the veterinary administration could isolate the infected zone. This led to a rapid spread of the infection in the south of the country. Under such circumstances, the use of a serological marker vaccine would not radically alter the basic nature of the problem, as it does not obviate the need to identify potentially infected animals and to take a sample of serum before any animals are transported, in other words, to strictly control the movement of pigs. Indeed, at the start of an epizootic, the success of control measures depends on their being rapidly implemented after the appearance of the first outbreak and before extensive, undetected spread has occurred. Vaccination is no substitute for basic measures to control contagious diseases. So, as a general rule, as long as CSF has not been eradicated in the world, there is still a great risk of reintroduction of the virus in free areas. Farmers and veterinarians are in the best position to detect a new introduction of CSFV, so they should receive training in the detection of clinical signs and remain extremely vigilant. A non-vaccination policy is logical in disease-free states but emergency vaccination may be considered in contingency plans to avoid destroying millions of pigs. When outbreaks occur, the use of the traditional live C strain vaccine is as

effective at preventing the spread of virus as culling the neighbouring herds. This strategy can be used after the start of an epizootic when there are too many outbreaks occurring at the same time, but this means that the pigs are seropositive and leads to their destruction. For this reason, the development of efficacious marker vaccines and reliable discriminatory tests should be encouraged. As it has been demonstrated that 14 days are necessary to induce good protection with the available subunit E2 vaccines, their use could be envisaged when several outbreaks occur at the same time and the use of strict sanitary prophylactic measures alone may not be enough to control the disease: vaccinating pigs in the zone around the outbreaks would allow the movement of pigs and prevent mass culling. As these E2 subunit vaccines do not prevent vertical transmission, their use must be limited to growing pigs. Simulation models will also be useful tools for choosing the best control measures to apply, depending on the epidemiological situation. At the start of an epizootic, in regions with a high density of pig herds, ring or zonal vaccination can also be envisaged in order to prevent the virus from replicating too rapidly and to limit the cost of preventive slaughter. However, in this case, transmission of the virus must be limited and control measures must be properly applied and effective. Such an approach is particularly pertinent for highly contagious diseases such as FMD in those circumstances under which the air-borne transmission is one of the main epidemiological factors in the spread of the virus. So, if the first outbreaks appear in an area with a high density of susceptible herds and under epidemiological conditions that favour air-borne spread, ring vaccination, implemented on the basis of the results of models and assessment to determine the risks and directions of spreading, could be useful in limiting the speed and the extent of the virus dissemination (24). However, due to the ability of vaccination to mask the appearance of clinical signs without preventing infection, vaccinated herds, even with a serological monitoring programme, represent a greater risk for undetected spread than unvaccinated herds, where monitoring can be based on clinical inspection alone. In the case of AI, the use of marker vaccines has proved very effective in controlling LPNAI infection in turkeys and poultry (18). Nevertheless, the difficulty of implementing such a strategy will depend on the species in which the vaccines are intended to be used and, consequently, on the efficiency of one type of vaccine in regard to one particular species. The efficiency of a vaccine and the performance of a companion diagnostic kit cannot be extrapolated from one avian species to another, so the use of marker vaccines will depend on the availability of validated data on the performance of the vaccine and companion diagnostic kit in the avian species that is to be vaccinated.

Rev. sci. tech. Off. int. Epiz., 26 (2)

363

In regard to the HPNAI strains, some Asian countries have implemented a massive vaccination campaign due to the high prevalence of infection in the domestic flocks. In some countries, such as Vietnam, this strategy seems to have given good results, but the lack of validated data prevents drawing definitive conclusions on the best strategies to use in the face of such complex and difficult animal health crises.

Conclusion
The power of the combination of a marker vaccine and a companion diagnostic kit for the control of an infectious disease depends not only on the performances of the vaccine and the kit themselves but also on the way in which these tools are deployed by the competent authority for use in the field. Ideally, both the vaccine and the diagnostic kit should be scientifically assessed by an independent body following submission of a dossier of data in support of an official application for a marketing authorisation. With respect to tests, at least for those used in the context of official control and eradication campaigns, national or international Reference Laboratories should control the quality of each batch released onto the market. If several diagnosis laboratories are involved in the diagnosis and the surveillance of the infection and the disease, a Reference Laboratory should organise proficiency tests, the results of which should be used to deliver an agreement to the diagnosis laboratories to allow them to carry out the diagnostic tests. Regulatory

systems already exist in most regions of the world (e.g. EU, USA, Japan) to control the quality of vaccines released onto the market. The situation is more varied with respect to companion diagnostic tests as not all regions or countries have legislation governing their marketing. Adequate validation of tests as fit for purpose and appropriate quality control, either by the manufacturer themselves or by official Reference Laboratories, are necessary to ensure that the test performs as claimed and that all batches reaching the market are of consistent, high quality. In addition, where testing is carried out by multiple laboratories, experience has shown a clear need for quality assurance of the testing performed in all participating laboratories. This can be achieved by a combination of laboratories working to recognised quality standards, backed up by external accreditation, and participation in regular proficiency tests conducted by an appropriate Reference Laboratory. Only those laboratories reaching the required standards should be certified as competent to be involved in national disease eradication campaigns. A successful programme can be based on vaccination, but should also include sanitary measures. Furthermore, when vaccination is part of a control programme, it should be implemented only for a certain period of time. Most of the time, when the prevalence of the infection decreases significantly and when the epidemiological unit is correctly protected from outside introduction of the agent, vaccination should be replaced by sanitary measures.

Les vaccins marqueurs et les consquences de leur utilisation sur le diagnostic et la prophylaxie
P. Vannier, I. Capua, M.F. Le Potier, D.K.J. Mackay, B. Muylkens, S. Parida, D.J. Paton & E. Thiry
Rsum La biologie molculaire et les avances techniques lies la recombinaison de lADN marquent le dbut dune nouvelle re en vaccinologie. Les auteurs examinent le dveloppement rcent de vaccins marqueurs spcifiques ainsi que les consquences de leur utilisation sur le diagnostic et la prvention des principales maladies infectieuses. Les vaccins obtenus par dltion de gne, les stratgies DIVA (visant diffrencier les animaux infects des animaux vaccins) et dautres mthodes similaires ont t appliqus avec succs pour

364

Rev. sci. tech. Off. int. Epiz., 26 (2)

contrler et radiquer la maladie dAujeszky, la rhinotrachite infectieuse bovine, la peste porcine classique, la fivre aphteuse et plus rcemment linfluenza aviaire. Les auteurs examinent lefficacit et les performances des vaccins marqueurs existants ainsi que des outils diagnostiques compagnons (dont lvaluation devrait tre conduite par un organisme indpendant) ; ils analysent galement la manire dont ces outils sont utiliss par les autorits comptentes. Ils tudient ensuite en dtail les avantages et les limites de lutilisation des vaccins marqueurs, la lumire des expriences pratiques en la matire. Bien que lutilisation de ces vaccins ait pour effets de freiner et de restreindre la dissmination virale, et partant de rduire le nombre danimaux abattus, elle na pas vocation remplacer les mesures sanitaires. Les systmes de dtection et dalerte prcoces et la mise en uvre rapide de mesures sanitaires, y compris labattage sanitaire, demeurent des mesures incontournables de la lutte contre les maladies trs contagieuses. Mots-cls radication Excrtion virale Fivre aphteuse Infection latente Influenza aviaire Maladie dAujeszky Mesure sanitaire Outil diagnostique compagnon Peste porcine classique Portage viral Prophylaxie Stratgie DIVA (diffrencier les animaux infects des animaux vaccins) Vaccin marqueur Vaccination.

Vacunas marcadoras y sus consecuencias sobre el diagnstico y medidas de profilaxis


P. Vannier, I. Capua, M.F. Le Potier, D.K.J. Mackay, B. Muylkens, S. Parida, D.J. Paton & E. Thiry
Resumen La biologa molecular y, en particular, los adelantos registrados en la tcnica de recombinacin de ADN, han marcado el comienzo de una nueva era para la vacunacin. En este artculo se examinan las vacunas marcadoras especficas, de reciente desarrollo, y las repercusiones de su administracin en el diagnstico y la prevencin de importantes enfermedades infecciosas. Se han obtenido resultados satisfactorios en materia de control y erradicacin de la enfermedad de Aujeszky, la rinotraquetis infecciosa bovina, la peste porcina clsica, la fiebre aftosa y, ms recientemente, la influenza aviar con vacunas producidas mediante delecin de genes, estrategias para diferenciar entre animales vacunados e infectados (estrategias DIVA) y otros mtodos similares. Los autores tambin analizan la eficacia y los resultados obtenidos con las vacunas marcadoras existentes y los equipos de diagnstico asociados (que debern ser evaluados por un organismo independiente), as como la forma en que las autoridades competentes las utilizarn. Asimismo, examinan detalladamente las limitaciones y las ventajas de la administracin de vacunas marcadoras a la luz de la experiencia adquirida en la prctica. Pese a que pueden limitar la velocidad y la importancia de la diseminacin del virus y, por consiguiente, reducir el nmero de animales sacrificados, las vacunas

Rev. sci. tech. Off. int. Epiz., 26 (2)

365

marcadoras no pueden sustituir a las medidas sanitarias. Los sistemas de deteccin y alerta rpidas y la aplicacin inmediata de medidas de profilaxis, incluido el sacrificio sanitario, siguen siendo decisivos para controlar las enfermedades altamente contagiosas. Palabras clave Control Enfermedad de Aujeszky Equipo de diagnstico asociado Erradicacin Estado portador de virus Estrategia DIVA (diferenciacin entre animales vacunados e infectados) Excrecin viral Fiebre aftosa Infeccin latente Influenza aviar Medida sanitaria Peste porcina clsica Vacuna marcadora Vacunacin.

References
1. Ackermann M. & Engels M. (2006). Pro and contra IBReradication. Vet. Microbiol., 113, 293-302. 2. Andrew M.E., Morris K., Coupar B., Sproat K., Oke P., Bruce M., Broadway M., Morrissy C. & Strom D. (2006). Porcine interleukin-3 enhances DNA vaccination against classical swine fever. Vaccine, 24, 3241-3247. 3. Andrew M.E., Morrissy C.J., Lenghaus C., Oke P.G., Sproat K.W., Hodgson A.L.M., Johnson M.A. & Coupar B.E.H. (2000). Protection of pigs against classical swine fever with DNA-delivered gp55. Vaccine, 18, 19321938. 4. Aynaud J.M., Lejolly J.C., Bibard C. & Galicher C. (1971). Studies of the properties of cold induced classical swine fever virus mutants. Application to vaccination. Bull. Off. int. Epiz., 75, 654-659. 5. Baranowski E., Ruiz-Jarabo C.M., Lim F & Domingo E. . (2001). Foot-and-mouth disease virus lacking the VP 1 G-H loop: the mutant spectrum uncovers interactions among antigenic sites for fitness gain. Virology, 288, 192-202. 6. Baranowski E., Ruiz-Jarabo C.M., Sevilla N., Andreu D., Beck E. & Domingo E. (2000). Cell recognition by footand-mouth disease virus that lacks the RGD integrin-binding motif: flexibility in aphthovirus receptor usage. J. Virol., 74, 1641-1647. 7. Beer M., Konig P., Schielke G. & Trapp S. (2003). Diagnostic markers in the prevention of bovine herpesvirus type 1: possibilities and limitations. Berl. Mnch. tier rztl. Wochenschr., 116, 183-191. a 8. Belsham G.J., Abrams C.C., King A.M., Roosien J. & Valk J.M. (1991). Myristoylation of foot-and-mouth disease virus capsid protein precursors is independent of other viral proteins and occurs in both mammalian and insect cells. J. gen. Virol., 72, 747-751. 9. Berns A., van den Ouweland A., Quint W., van Oirschot J. & Gielkens A. (1985). Presence of markers for virulence in the unique short region or repeat region or both of pseudorabies hybrid viruses. J. Virol., 53, 89-93. 10. Bittle J.L., Houghten R.A., Alexander H., Shinnick T.M., Sutcliffe J.G., Lerner R.A., Rowlands D.J. & Brown F (1982). . Protection against foot-and-mouth disease by immunization with a chemically synthesized peptide predicted from the viral nucleotide sequence. Nature, 298, 30-33. 11. Boelaert F., Speybroeck N., de Kruif A., Aerts M., Burzykowski T., Molenberghs G. & Berkvens D.L. (2005). Risk factors for bovine herpesvirus-1 seropositivity. Prev. vet. Med., 69, 285-295.

366

Rev. sci. tech. Off. int. Epiz., 26 (2)

12. Bosch J.C., Kaashoek M.J., Kroese A.H. & van Oirschot J.T. (1996). An attenuated bovine herpesvirus 1 marker vaccine induces a better protection than two inactivated marker vaccines. Vet. Microbiol., 52, 223-234. 13. Bosch J.C., Kaashoek M.J. & van Oirschot J.T. (1997). Inactivated bovine herpesvirus 1 marker vaccines are more efficacious in reducing virus excretion after reactivation than a live marker vaccine. Vaccine, 15, 1512-1517. 14. Bouma A., Desmit A.J., Dejong M.C.M., Dekluijver E.P. & Moormann R.J.M. (2000). Determination of the onset of the herd-immunity induced by the E2 sub-unit vaccine against classical swine fever virus. Vaccine, 18, 1374-1381. 15. Bouma A., Desmit A.J., Dekluijver E.P., Terpstra C. & Moormann R.J.M. (1999). Efficacy and stability of a subunit vaccine based on glycoprotein E2 of classical swine fever virus. Vet. Microbiol., 66, 101-114. 16. Brocchi E., Bergmann I., Dekker A., Paton D.J., Sammin D.J., Greiner M., Grazioli S., Desimone F Yadin H., Haas B., ., Bulut N., Malirat V., Neitzert E., Goris N., Parida S., Sorensen K. & De Clercq K. (2006). Comparative performance of six ELISAs for antibodies to the nonstructural proteins of foot-and-mouth disease. Vaccine, 24, 6966-6979. 17. Bruderer U., Swam H., Haas B., Visser N., Brocchi E., Grazioli S., Esterhuysen J.J., Vosloo W., Forsyth M., Aggarwal N., Cox S., Armstrong R. & Anderson J. (2004). Differentiating infection from vaccination in foot-and-mouthdisease: evaluation of an ELISA based on recombinant 3ABC. Vet. Microbiol., 101, 187-197. 18. Capua I. & Marangon S. (2006). The use of vaccination to combat multiple introduction of notifiable avian influenza viruses of the H5 an H7 subtypes between 2000 and 2006 in Italy. Vaccine, 25, 4987-4995. 19. Capua I. & Mutinelli F (2001). Aetiology of avian influenza. . In A colour atlas and text on avian influenza. Papi Editore, Bologna, 236. 20. Cattoli G., Milani A., Bettini F Beato M.S., Mancin M., ., Terregino C. & Capua I. (2006). Development and validation of an anti-N3 indirect immunofluorescent antibody test to be used as a companion diagnostic test in the framework of a DIVA vaccination strategy for avian influenza infections in poultry. Avian Pathol., 35 (2), 154-159. 21. Cattoli G., Terregino C., Brasola V., Rodriguez J.F & Capua I. . (2003). Development and preliminary validation of an ad hoc N1-N3 discriminatory test for the control of avian influenza in Italy. Avian Dis., 47 (3 Suppl.), 1060-1062. 22. Chinsangaram J., Moraes M.P., Koster M. & Grubman M.J. (2003). Novel viral disease control strategy: adenovirus expressing alpha interferon rapidly protects swine from foot and mouth disease. J. Virol., 77, 1621-1625.

23. Chinsangaram J., Mason P.W. & Grubman M.J. (1998). Protection of swine by live inactivated vaccines prepared from a leader proteinase-deficient serotype A12 foot-and-mouth disease virus. Vaccine, 16, 1516-1522. 24. Cox S., Voyce C., Parida S., Reid S., Hamblin P., Hutching G., Paton D.J. & Barnett P.V. (2006). Effect of antigen payload on protection, sub-clinical infection and persistence following direct contact challenge of cattle. Vaccine, 24, 3184-3190. 25. Cox S.J., Voyce C., Parida S., Reid S.M., Hamblin P.A., Paton D.J. & Barnett P.V. (2005). Protection against directcontact challenge following emergency FMD vaccination of cattle and the effect on virus excretion from the oropharynx. Vaccine, 23, 1106-1113. 26. Crowther J.R., Farias S., Carpenter W.C. & Samuel A.R. (1993). Identification of a fifth neutralizable site on type O foot-and-mouth disease virus following characterization of single and quintuple monoclonal antibody escape mutants. J. gen. Virol., 74, 1547-1553. 27. Cuncliffe H.R. (1964). Observations on the duration of immunity in cattle after experimental infection with foot-andmouth disease. Cornell. Vet., 54, 501. 28. De Diego M.E., Brocchi E., Mackay D.K.J. & Simone F .D. (1997). The non-structural polyprotein 3ABC of foot-andmouth disease virus as a diagnostic antigen in ELISA to differentiate infected from vaccinated cattle. Arch. Virol., 142, 2021-2033. 29. De Wit J.J., Hage J.J., Brinkhof J. & Westenbrink F (1998). . A comparative study of serological tests for use in the bovine herpesvirus 1 eradication programme in The Netherlands. Vet. Microbiol., 61, 153-163. 30. Depner K.R., Bouma A., Koenen F Klinkenberg D., Lange E., ., deSmit H. & Vanderhallen H. (2001). Classical swine fever (CSF) marker vaccine Trial II. Challenge study in pregnant sows. Vet. Microbiol., 83, 107-120. 31. Dewulf J., Laevens H., Koenen F Mintiens K. & deKruif A. ., (2001). An E2 sub-unit marker vaccine does not prevent horizontal or vertical transmission of classical swine fever virus. Vaccine, 20, 86-91. 32. Dewulf J., Laevens H., Koenen F Mintiens K. & deKruif A. ., (2004). Efficacy of E2 sub-unit marker and C-strain vaccines in reducing horizontal transmission of classical swine fever virus in weaner pigs. Prev. vet. Med., 65, 121-133. 33. DiMarchi R., Brooke G., Gale C., Cracknell V., Doel T. & Mowat N. (1986). Protection of cattle against foot-andmouth disease biosynthetic peptide. Science, 232, 639-641.

Rev. sci. tech. Off. int. Epiz., 26 (2)

367

34. Dispas M., Lemaire M., Speybroeck N., Berkvens D., Dupont A., Boelart F Dramaix M., Vanopdenbosch E., ., Kerkhofs P. & Thiry E. (2004). Deux protocoles dhyperimmunisation au moyen de vaccins marqus rduisent lincidence de sroconversion envers lherpsvirus bovin 1 en cheptels laitiers : rsultats dune tude sur le terrain. Ann. Md. vt., 148, 47-61. 35. Dispas M., Schynts F., Lemaire M., Letellier C., Vanopdenbosch E., Thiry E. & Kerkhofs P. (2003). Isolation of a glycoprotein E-deleted bovine herpesvirus type 1 strain in the field. Vet. Rec., 153, 209-212. 36. Doel T.R. (2003). FMD vaccines. Virus Res., 91, 81-99. 37. Doel T.R. (1999). Optimisation of the immune response to foot-and-mouth disease vaccines. Vaccine, 17, 13-14. 38. Dundon W., Maniero S., Toffan A., Capua I. & Cattoli G. (2006). Appearance of serum antibodies against the avian influenza non-structural 1 protein in experimentally infected chickens and turkeys. Avian Dis., 51, 209-212. 39. European Union (2003). Council Directive 2003/85/EC on Community measures for the control of foot-and-mouth disease repealing Directive 85/511/EEC and Decisions 89/531/EEC and 96/665/EEC and amending Directive 92/46/EEC. Off. J. Eur. Union, L306. 40. Floegel-Niesmann G. (2001). Classical swine fever (CSF) marker vaccine Trial III. Evaluation of discriminatory ELISAs. Vet. Microbiol., 83, 121-136. 41. Francis M.J., Hastings G.Z., Brown F McDermed J., Lu Y.A. ., & Tam J.P. (1991). Immunological evaluation of the multiple antigen peptide (MAP) system using the major immunogenic site of foot-and-mouth disease virus. Immunology, 73, 249-254. 42. Gogev S., de Fays K., Versali M.F Gautier S. & Thiry E. ., (2004). Glycol chitosan improves the efficacy of intranasally administrated replication defective human adenovirus type 5 expressing glycoprotein D of bovine herpesvirus 1. Vaccine, 22, 1946-1953. 43. Gogev S., Georgin J.P., Schynts F Vanderplasschen A. & ., Thiry E. (2004). Bovine herpesvirus 1 glycoprotein D expression in bovine upper respiratory tract mediated by a human adenovirus type 5. Vet. Res., 35, 715-721. 44. Gogev S., Vanderheijden N., Lemaire M., Schynts F., DOffay J., Deprez I., Adam M., Eloit M. & Thiry E. (2002). Induction of protective immunity to bovine herpesvirus type 1 in cattle by intranasal administration of replicationdefective human adenovirus type 5 expressing glycoprotein gC or gD. Vaccine, 20, 1451-1465. 45. Golding S.M., Hedger R.S. & Talbot P. (1976). Radial immuno-diffusion and serum-neutralisation techniques for the assay of antibodies to swine vesicular disease. Res. vet. Sci., 20, 142-147.

46. Greiner M. & Dekker A. (2005). On the issue of documenting small herds as free from disease. Prev. vet. Med., 70, 223-234. 47. Grubman M.J. (2005). Development of novel strategies to control foot-and-mouth disease: marker vaccine and antivirals. Biologicals, 33, 227-234. 48. Grubman M.J. & Mason P.W. (2002). Prospects, including time frames, for improved foot and mouth vaccines. In Foot and mouth disease: facing the new dilemmas (G.R. Thomson, ed.). Rev. sci. tech. Off. int. Epiz., 21 (3), 589-600. 49. Grubman M.J., Morgan D.O., Kendall J. & Baxt B. (1985). Capsid intermediates assembled in a foot-and-mouth disease virus genome RNA-programmed cell-free translation system and in infected cells. J. Virol., 56, 120-126. 50. Gupta P.K., Saini M., Gupta L.K., Rao V.D., Bandyopadhyay S.K., Butchaiah G., Garg G.K. & Garg S.K. (2001). Induction of immune responses in cattle with a DNA vaccine encoding glycoprotein C of bovine herpesvirus1. Vet. Microbiol., 78, 293-305. 51. Hamblin C., Barnett I.T. & Hedger R.S. (1986). A new enzyme-linked immunosorbent assay (ELISA) for the detection of antibodies against foot-and-mouth disease virus. I. Development and method of ELISA. J. immunol. Meth., 93, 115-121. 52. Hammond J.M., Jansen E.S., Morrissy C.J., Goff W.V., Meehan G.C., Williamson M.M., Lenghaus C., Sproat K.W., Andrew M.E., Coupar B.E.H. & Johnson M.A. (2001). A prime-boost vaccination strategy using naked DNA followed by recombinant porcine adenovirus protects pigs from classical swine fever. Vet. Microbiol., 80, 101-119. 53. Heffner S., Kovacs F Klupp B. & Mettenleiter T. (1993). ., Glycoprotein gp 50-negative pseudorabies virus: a novel approach toward a non-spreading live herpesvirus vaccine. J. Virol., 67, 1529-1537. 54. Ioannou X.P., Griebel P., Hecker R., Babiuk L.A. & van Drunen Littel-van den Hurk S. (2002). The immunogenicity and protective efficacy of bovine herpesvirus 1 glycoprotein D plus Emulsigen are increased by formulation with CpG oligodeoxynucleotides. J. Virol., 76, 9002-9010. 55. Kaashoek M.J., Moerman A., Madic J., Rijsewijk F Quak J., .A., Gielkens A.L. & van Oirschot J.T. (1994). A conventionally attenuated glycoprotein E-negative strain of bovine herpesvirus type 1 is an efficacious and safe vaccine. Vaccine, 12, 439-444. 56. Kaashoek M.J., Rijsewijk F Ruuls R.C., Keil G.M., Thiry E., .A., Pastoret P.-P. & van Oirschot J.T. (1998). Virulence, immunogenicity and reactivation of bovine herpesvirus 1 mutants with a deletion in the gC, gG, gI, gE, or in both the gI and gE gene. Vaccine, 16, 802-809.

368

Rev. sci. tech. Off. int. Epiz., 26 (2)

57. Kaden V. & Lange B. (2001). Oral immunisation against classical swine fever (CSF): onset and duration of immunity. Vet. Microbiol., 82, 301-310. 58. Kerkhofs P., Renjifo X., Toussaint J.F., Letellier C., Vanopdenbosch E. & Wellemans G. (2003). Enhancement of the immune response and virological protection of calves against bovine herpesvirus type 1 with an inactivated gE-deleted vaccine. Vet. Rec., 152, 681-686. 59. Kit M., Kit S., Little S.P., Di Marchi R.D. & Gale C. (1991). Bovine herpesvirus-1 (infectious bovine rhinotracheitis virus)-based viral vector which expresses foot-and-mouth disease epitopes. Vaccine, 9, 564-572. 60. Kitson J.D., Burke K.L., Pulle L.A., Belsham G.J. & Almond J.W. (1991). Chimeric polioviruses that include sequences derived from two independent antigenic sites of foot-and-mouth disease virus (FMDV) induce neutralising antibodies against FMDV in guinea pigs. J. Virol., 65, 3068-3075. 61. Konig P., Giesow K. & Keil G.M. (2002). Glycoprotein M of bovine herpesvirus 1 (BHV-1) is nonessential for replication in cell culture and is involved in inhibition of bovine respiratory syncytial virus F protein induced syncytium formation in recombinant BHV-1 infected cells. Vet. Microbiol., 86, 37-49. 62. Kramps J.A., Banks M., Beer M., Kerkhofs P., Perrin M., Wellenberg G.J. & van Oirschot J.T. (2004). Evaluation of tests for antibodies against bovine herpesvirus 1 performed in national reference laboratories in Europe. Vet. Microbiol., 102, 169-181. 63. Launais M., Aynaud J.M. & Corthier G. (1972). Peste porcine classique : proprits dun clone (souche thiverval) isol en culture cellulaire basse temprature. Application dans la vaccination. Rev. Md. vt., 123, 1537-1554. 64. Le Potier M.F., Fournier A., Houdayer C., Hutet E., Auvigne V., Hery D., Sanaa M. & Toma B. (1998). Use of muscle exudates for the detection of anti-gE antibodies to Aujeszkys disease virus. Vet. Rec., 143, 385-387. 65. Le Potier M.F Mesplde A. & Vannier P. (2006). Classical ., swine fever and other pestiviruses. In Diseases of swine (B.E. Straw, J.J. Zimmerman, S. DAllaire & D.J. Taylor, eds). Blackwell Publishing, 309-322. 66. Lehmann D., Sodoyer R., Leterme S. & Crevat D. (2002). Improvement of serological discrimination between herpesvirus-infected animals and animals vaccinated with marker vaccines. Vet. Microbiol., 86, 59-68. 67. Lemaire M., Schynts F Meyer G., Georgin J.P., Baranowski E., ., Gabriel A., Ros C., Belak S. & Thiry E. (2001). Latency and reactivation of a glycoprotein E negative bovine herpesvirus type 1 vaccine: influence of virus load and effect of specific maternal antibodies. Vaccine, 19, 4795-4804.

68. Lewis S.A., Morgan D.O. & Grubman M.J. (1991). Expression, processing and assembly of foot-and-mouth disease virus capsid structures in heterologous systems: induction of neutralising antibody response in Guinea pig. J. Virol., 65, 6572-6580. 69. Li Y., Aggarwal N., Takamatsu H.H., Sterling C.M., Voyce C. & Barnett P.V. (2006). Enhancing immune responses against a plasmid DNA vaccine encoding a FMDV empty capsid from serotype O. Vaccine, 24, 4602-4606. 70. Lin M., Lin F Mallory M. & Clavijo A. (2000). Deletions of ., structural glycoprotein E2 of classical swine fever virus strain Alfort/187 resolve a linear epitope of monoclonal antibody WH303 and the minimal N-terminal domain essential for binding immunoglobulin G antibodies of a pig hyperimmune serum. J. Virol., 74, 11619-11625. 71. Loehr B.I., Willson P., Babiuk L.A. & van Drunen Littelvan den Hurk S. (2000). Gene gun-mediated DNA immunization primes development of mucosal immunity against bovine herpesvirus 1 in cattle. J. Virol., 74, 6077-6086. 72. Luschow D., Werner O., Mettenleiter T.C. & Fuchs W. (2001). Protection of chickens from lethal avian influenza A virus infection by live-virus vaccination with infectious laryngotracheitis virus recombinants expressing the hemagglutinin (H5) gene. Vaccine, 19 (30), 4249-4259. 73. Mackay D.K., Bulut A.N., Rendle T., Davidson F & Ferris N.P. . (2001). A solid-phase competition ELISA for measuring antibody to foot-and-mouth disease virus. J. virol. Meth., 97, 33-48. 74. McKenna T.S., Lubroth J., Rieder E., Baxt B. & Mason P.W. (1995). Receptor binding site-deleted foot-and-mouth disease (FMD) virus protects cattle from FMD. J. Virol., 69, 5787-5790. 75. Malirat V., Neitzert E., Bergmann I.E., Maradei E. & Beck E. (1998). Detection of cattle exposed to foot-and-mouth disease virus by means of an indirect ELISA test using bioengineered non-structural polyprotein 3ABC. Vet. Q., 20, S24-26. 76. Margarita S., Nunez J.R., Jimenez-Clavero M.A., Baranowski E. & Sobrino F (2002). Foot-and-mouth . disease virus: biology and prospects for disease control. Microbes Infect., 4, 1183-1192. 77. Mars M.H., de Jong M.C., Franken P. & van Oirschot J.T. (2001). Efficacy of a live glycoprotein E-negative bovine herpesvirus 1 vaccine in cattle in the field. Vaccine, 19, 1924-1930. 78. Mars M.H., de Jong M.C. & van Oirschot J.T. (2000). A gE-negative BHV1 vaccine virus strain cannot perpetuate in cattle populations. Vaccine, 18, 2120-2124. 79. Mason P.W., Piccone M.E., McKenna P.S., Chinsangaram J. & Grubman M.J. (1997). Evaluation of a live- attenuated footand-mouth disease virus as a vaccine candidate. Virology, 227, 96-102.

Rev. sci. tech. Off. int. Epiz., 26 (2)

369

80. Mayr G.A., Chinsangaram J. & Grubman M.J. (1999). Development of replication-defective adenovirus serotype 5 containing the capsid and 3C protease coding regions of foot-and-mouth disease virus as a vaccine candidate. Virology, 263, 496-503. 81. Mayr G.A., ODonnell V., Chinsangaram J., Mason P.W. & Grubman M.J. (2001). Immune responses and protection against foot and mouth disease virus (FMDV) challenge in swine vaccinated with adenovirus FMDV constructs. Vaccine, 19, 2152-2162. 82. Mettenleiter T. (1994). Pseudorabies (Aujeszkys disease) virus. In Proc. OIE Symposium on Aujeszkys disease, 30 June-1 July, Bangkok, Thailand. OIE, Paris, 1-9. 83. Moraes M.P., Mayr G.A., Mason P.W. & Grubman M.J. (2002). Early protection against homologous challenge after a single dose of replication defective human adenovirus type 5 expressing capsid proteins of foot-and-mouth disease virus (FMDV) strain A24. Vaccine, 20, 1631-1639. 84. Muylkens B., Meurens F Schynts F de Fays K., Pourchet A., ., ., Thiry J., Vanderplasschen A., Antoine N. & Thiry E. (2006). Biological characterization of bovine herpesvirus 1 recombinants possessing the vaccine glycoprotein E negative phenotype. Vet. Microbiol., 113, 283-291. 85. Muylkens B., Meurens F Schynts F Farnir F Pourchet A., ., ., ., Bardiau M., Gogev S., Thiry J., Cuisenaire A., Vanderplasschen A. & Thiry E. (2006). Intraspecific bovine herpesvirus 1 recombinants carrying glycoprotein E deletion as a vaccine marker are virulent in cattle. J. gen. Virol., 87, 2149-2154. 86. Nichani A.K., Mena A., Kaushik R.S., Mutwiri G.K., Townsend H.G., Hecker R., Krieg A.M., Babiuk L.A. & Griebel P.J. (2006). Stimulation of innate immune responses by CpG oligodeoxynucleotide in newborn lambs can reduce bovine herpesvirus-1 shedding. Oligonucleotides, 16, 58-67. 87. Oliveira S.C., Harms J.S., Rosinha G.M., Rodarte R.S., Rech E.L. & Splitter G.A. (2000). Biolistic-mediated gene transfer using the bovine herpesvirus-1 glycoprotein D is an effective delivery system to induce neutralizing antibodies in its natural host. J. immunol. Meth., 245, 109-118. 88. Oren S.L., Swenson S.L., Kinker D.R., Hill H.T., Hu H.L. & Zimmerman J. (1993). Evaluation of serological pseudorabies tests for the detection of antibodies during early infection. J. vet. diagn. Invest., 5, 529-533. 89. Osorio F (1991). Diagnosis of pseudorabies (Aujeszkys .A. disease) virus infections. In Proc. 1st International Symposium: The eradication of pseudorabies (Aujeszkys) virus (R.B. Morrison, ed.), 19-22 May, Saint Paul, 7-32. 90. Parida S., Anderson J., Cox S.J., Barnett P.V. & Paton D.J. (2006). Secretory IgA as an indicator of oropharyngeal footand-mouth disease virus replication and as a tool for post vaccination surveillance. Vaccine, 24, 1107-1116.

91. Parida S., Cox S.J., Reid S.M., Hamblin P., Barnett P.V., Inoue T., Anderson J. & Paton D.J. (2005). The application of new techniques to the improved detection of persistently infected cattle after vaccination and contact exposure to footand-mouth disease. Vaccine, 23, 5186-5195. 92. Paton D.J., De Clercq K. & Dekker A. (2004). Post-vaccinal serosurveillance for FMD: a European perspective on progress and problems. In Report of the Session of the Research Group of the Standing Technical Committee of the European Commission for the Control of Foot-and-Mouth Disease, 12-15 October, Chania, Crete, Greece. Appendix 8. Food and Agriculture Organization, Rome, 68-71. 93. Paton D.J., De Clercq K., Greiner M., Deller A., Brocchi E., Bergmann I., Sammin D., Gubbins S. & Parida S. (2006). Application of non-structural protein antibody tests in substantiating freedom from foot-and-mouth disease virus infection after emergency vaccination of cattle. Vaccine, 24, 6503-6512. 94. Peeters B., Bienkowska-Szewczyk K., Hulst M., Gielkens A. & Kimman T. (1997). Biologically safe, non-transmissible pseudorabies virus vector vaccine protects pigs against both Aujeszkys disease and classical swine fever. J. gen. Virol., 78, 3311-3315. 95. Pensaert M.B., de Smet K. & de Waele K. (1990). Extent and duration of virulent virus excretion upon challenge of pigs vaccinated with different glycoprotein-deleted Aujeszkys disease vaccines. Vet. Microbiol., 22, 107-117. 96. Perez Filgueira D.M., Zamorano P.I., Dominguez M.G., Taboga O., Del Medico Zajac M.P., Puntel M., Romera S.A., Morris T.J., Borca M.V. & Sadir A.M. (2003). Bovine herpes virus gD protein produced in plants using a recombinant tobacco mosaic virus (TMV) vector possesses authentic antigenicity. Vaccine, 21, 4201-4209. 97. Perrin B., Perrin M., Moussa A. & Coudert M. (1996). Evaluation of a commercial gE blocking ELISA test for detection of antibodies to infectious bovine rhinotracheitis virus. Vet. Rec., 138, 520. 98. Persson K., Nordengrahn A., Decker C. & Merza M. (2004). The development of an indirect ELISA for the detection of antibodies to the non-structural protein 3ABC of the footand-mouth disease virus; the use of a polyclonal conjugate that allows a multi-species detection of antibodies. In Report of the Session of the Research Group of the Standing Technical Committee of the European Commission for the Control of Foot-and-Mouth Disease, 12-15 October, Chania, Crete, Greece. Appendix 79. Food and Agriculture Organization, Rome, 479-480. 99. Pfaff E., Mussgay M., Bohm H.O., Schulz G.E. & Schaller H. (1982). Antibodies against a preselected peptide recognize and neutralize foot and mouth disease virus. EMBO J., 1, 869-874.

370

Rev. sci. tech. Off. int. Epiz., 26 (2)

100. Reddy P.S., Idamakanti N., Chen Y., Whale T., Babiuk L.A., Mehtali M. & Tikoo S.K. (1999). Replication-defective bovine adenovirus type 3 as an expression vector. J. Virol., 73, 9137-9144. 101. Rijsewijk F.A., Kaashoek M.J., Langeveld J.P., Maris-Veldhuis M.A., Magdalena J., Verschuren S.B., Meloen R.H. & van Oirschot J.T. (2000). Epitopes on glycoprotein E and on the glycoprotein E/glycoprotein I complex of bovine herpesvirus 1 are expressed by all of 222 isolates and 11 vaccine strains. Arch. Virol., 145, 921-936. 102. Rousien J., Belsham G.J., Ryan M.D., King A.M. & Valk J.M. (1990). Synthesis of foot and mouth disease virus capsid proteins in insect cells using baculovirus expression vectors. J. gen. Virol., 71, 1703-1711. 103. Schalch L., Rebeski D.E., Samaras H., Lozana G., Thuer B. & Schelp C. (2002). Recently generated data with the CHEKIT-FMD-3ABC ELISA kit and methods to monitor the operational performance of a 3ABC ELISA. In Report of the Session of the Research Group of the Standing Technical Committee of the European Commission for the Control of Foot-and-Mouth Disease, 17-20 September, Cesme, Izmir, Turkey. Food and Agriculture Organization, Rome, 283302. 104. Schmitt B. (2005). Differential diagnostic test technology: sensitivity and specificity, an OIE validation perspective. Biologicals, 33, 211-213. 105. Schynts F Meurens F Detry B., Vanderplasschen A. & ., ., Thiry E. (2003). Rise and survival of bovine herpesvirus 1 recombinants after primary infection and reactivation from latency. J. Virol., 77, 12535-12542. 106. Shen P.D., Chen P.D., Walfield A.M., Ye J., House J., Brown F. & Wang C.Y. (1999). Differentiation of convalescent animals from those vaccinated against footand-mouth disease by a peptide ELISA. Vaccine, 17, 30393049. 107. Solis-Calderon J.J., Segura-Correa V.M., Segura-Correa J.C. & Alvarado-Islas A. (2003). Seroprevalence of and risk factors for infectious bovine rhinotracheitis in beef cattle herds of Yucatan, Mexico. Prev. vet. Med., 57, 199-208. 108. Sorensen K.J., Madsen K.G., Madsen E.S., Salt J.S., Nqindi J. & Mackay D.K. (1998). Differentiation of infection from vaccination in foot-and-mouth disease by the detection of antibodies to the non-structural proteins 3D, 3AB and 3ABC in ELISA using antigens expressed in baculovirus. Arch. Virol., 143, 1461-1476. 109. Strohmaier K., Franze R. & Adam K.H. (1982). Location and characterization of the antigenic portion of the FMDV immunizing protein. J. gen. Virol., 59, 295-306. 110. Swayne D.E. (2004). Application of new vaccine technologies for the control of transboundary diseases. Dev. Biol., 119, 219-228.

111. Swayne D.E., Beck J.R. & Kinney N. (2000). Failure of a recombinant fowl poxvirus vaccine containing an avian influenza hemagglutinin gene to provide consistent protection against influenza in chickens preimmunized with a fowl pox vaccine. Avian Dis., 44 (1), 132-137. 112. Swayne D.E., Perdue M.L., Beck J.R., Garcia M. & Suarez D.L. (2000). Vaccines protect chickens against H5 highly pathogenic avian influenza in the face of genetic changes in field viruses over multiple years. Vet. Microbiol., 74 (1-2), 165-172. 113. Swayne D.E., Suarez D.L., Schultz-Cherry S., Tumpey T.M., King D.J., Nakaya T., Palese P. & Garcia-Sastre A. (2003). Recombinant paramyxovirus type 1-avian influenza-H7 virus as a vaccine for protection of chickens against influenza and Newcastle disease. Avian Dis., 47 (3 Suppl.), 1047-1050. 114. Terpstra C., Woortmeyer R. & Barteling S.J. (1990). Development and properties of a cell culture produced vaccine for hog cholera based on the Chinese strain. Dtsch. tier rztl. Wochenschr., 97, 77-79. a 115. Thiry E., Meurens F Muylkens B., McVoy M., Gogev S., ., Thiry J., Vanderplasschen A., Epstein A., Keil G. & Schynts F (2005). Recombination in alphaherpesviruses. . Rev. Med. Virol., 15, 89-103. 116. Thiry E., Muylkens B., Meurens F Gogev S., Thiry J., ., Vanderplasschen A. & Schynts F (2006). Recombination . in the alphaherpesvirus bovine herpesvirus 1. Vet. Microbiol., 113, 171-177. 117. Thiry E., Saliki J., Bublot M. & Pastoret P.-P. (1987). Reactivation of infectious bovine rhinotracheitis virus by transport. Comp. Immunol. Microbiol. infect. Dis., 10, 59-63. 118. Thiry E., Saliki J., Schwers A. & Pastoret P.-P. (1985). Parturition as a stimulus of IBR virus reactivation. Vet. Rec., 116, 599-600. 119. Tian G., Zhang S., Li Y., Bu Z., Liu P., Zhou J., Li C., Shi J., Yu K. & Chen H. (2005). Protective efficacy in chickens, geese and ducks of an H5N1-inactivated vaccine developed by reverse genetics. Virology, 341 (1), 153-162. 120. Toussaint J.F Coen L., Letellier C., Dispas M., Gillet L., ., Vanderplasschen A. & Kerkhofs P. (2005). Genetic immunisation of cattle against bovine herpesvirus 1: glycoprotein gD confers higher protection than glycoprotein gC or tegument protein VP8. Vet. Res., 36, 529-544. 121. Toussaint J.F., Letellier C., Paquet D., Dispas M. & Kerkhofs P. (2005). Prime-boost strategies combining DNA and inactivated vaccines confer high immunity and protection in cattle against bovine herpesvirus-1. Vaccine, 23, 5073-5081.

Rev. sci. tech. Off. int. Epiz., 26 (2)

371

122. Tumpey T.M., Alvarez R., Swayne D.E. & Suarez D.L. (2005). Diagnostic approach for differentiating infected from vaccinated poultry on the basis of antibodies to NS1, the nonstructural protein of influenza A virus. J. clin. Microbiol., 43 (2), 676-683. 123. Uttenthal A., Le Potier M.F Romero L., DeMia G.M. & ., Floegel-Niesmann G. (2001). Classical swine fever (CSF) marker vaccine Trial I. Challenge studies in weaner pigs. Vet. Microbiol., 83, 85-106. 124. Van Drunen Littel-van den Hurk S. (2006). Rationale and perspectives on the success of vaccination against bovine herpesvirus-1. Vet. Microbiol., 113, 275-282. 125. Van Drunen Littel-van den Hurk S., Tikoo S.K., Liang X. & Babiuk L.A. (1993). Bovine herpesvirus-1 vaccines. Immunol. Cell Biol., 71 (Pt 5), 405-420. 126. Van Drunen Littel-van den Hurk S., van Donkersgoed J., Kowalski J., van den Hurk J.V., Harland R., Babiuk L.A. & Zamb T.J. (1994). A subunit gIV vaccine, produced by transfected mammalian cells in culture, induces mucosal immunity against bovine herpesvirus-1 in cattle. Vaccine, 12, 1295-1302. 127. Van Engelenburg F.A., Kaashoek M.J., Rijsewijk F.A., Moerman A., Gielkens A.L. & van Oirschot J.T. (1994). A glycoprotein E deletion mutant of bovine herpesvirus 1 is avirulent in calves. J. gen. Virol., 75, 2311-2318. 128. Van Engelenburg F .A., Kaashoek M.J., van Oirschot J.T. & Rijsewijk F (1995). A glycoprotein E deletion mutant of .A. bovine herpesvirus 1 infects the same limited number of tissues in calves as wild-type virus, but for a shorter period. J. gen. Virol., 76 (Pt 9), 2387-2392. 129. Van Oirschot J.T. (1998). Diva vaccines against bovine infectious diseases. European Centre for Pharmaceutical Information (ECPI) Conference, Rapid development of veterinary vaccines in Europe, 29-30 September, London. 130. Van Oirschot J.T., Kaashoek M.J., MarisVeldhuis M.A., Weerdmeester K. & Rijsewijk F .A.M. (1997). An enzymelinked immunosorbent assay to detect antibodies against glycoprotein gE of bovine herpesvirus 1 allows differentiation between infected and vaccinated cattle. J. virol. Meth., 67, 23-34. 131. Van Oirschot J.T., Rziha H.J., Moonen P.J., Pol J.M. & van Zaane D. (1986). Differentiation of serum antibodies from pigs vaccinated or infected with Aujeszkys disease virus by a competitive enzyme immunoassay. J. gen. Virol., 67, 1179-1182. 132. Van Schaik G. (2001). Risk and economics of disease introduction to dairy farms. Tijdschr. Diergeneeskd., 126, 414-418. 133. Van Schaik G., Schukken Y.H., Nielen M., Dijkhuizen A.A., Barkema H.W. & Benedictus G. (2002). Probability of and risk factors for introduction of infectious diseases into Dutch SPF dairy farms: a cohort study. Prev. vet. Med., 54, 279-289.

134. Van Schaik G., Schukken Y.H., Nielen M., Dijkhuizen A.A. & Benedictus G. (2001). Risk factors for introduction of BHV1 into BHV1-free Dutch dairy farms: a case-control study. Vet. Q., 23, 71-76. 135. Van Zijl M., Wensvoort G., de Kluyver E., Hulst M., van der Gulden H., Gielkens A., Berns A. & Moormann R. (1991). Live attenuated pseudorabies virus expressing envelope glycoprotein E1 of hog cholera virus protects swine against both pseudorabies and hog cholera. J. Virol., 65, 2761-2765. 136. Vandeputte J. & Chappuis G. (1999). Classical swine fever: the European experience and a guide for infected areas. Rev. sci. tech. Off. int. Epiz., 18 (3), 638-647. 137. Vannier P., Hutet E., Bourgueil E. & Cariolet R. (1991). Level of virulent virus excreted by infected pigs previously vaccinated with different glycoprotein deleted Aujeszkys disease vaccines. Vet. Microbiol., 29, 213-223. 138. Veits J., Wiesner D., Fuchs W., Hoffmann B., Granzow H., Starick E., Mundt E., Schirrmeier H., Mebatsion T., Mettenleiter T.C. & Romer-Oberdorfer A. (2006). Newcastle disease virus expressing H5 hemagglutinin gene protects chickens against Newcastle disease and avian influenza. Proc. natl Acad. Sci. USA, 103 (21), 8197-8202. 139. Vonk Noordegraaf A., Labrovic A., Frankena K., Pfeiffer D.U. & Nielen M. (2004). Simulated hazards of loosing infection-free status in a Dutch BHV1 model. Prev. vet. Med., 62, 51-58. 140. Wellenberg G.J., Verstraten E.R., Mars M.H. & van Oirschot J.T. (1998). ELISA detection of antibodies to glycoprotein E of bovine herpesvirus 1 in bulk milk samples. Vet. Rec., 142, 219-220. 141. Wienhold D., Armengol E., Marquardt A., Marquardt C., Voigt H., Buttner M., Saalmuller A. & Pfaff E. (2005). Immunomodulatory effect of plasmids co-expressing cytokines in classical swine fever virus subunit gp55/E2DNA vaccination. Vet. Res., 36, 571-587. 142. Willeberg P., Leontides L., Ewald C., Mortensen S., McInerney J.P., Howe H.S. & Kooij D. (1996). Effect of vaccination against Aujeszkys disease compared with test and slaughter programme: epidemiological and economical evaluations. Acta vet. scand. suppl., 90, 25-51. 143. World Organisation for Animal Health (OIE) (2004). Chapter 2.1.1. Foot and mouth disease. In Manual of Diagnostic Tests and Vaccines for Terrestrial Animals (mammals birds and bees), 5th Ed. OIE, Paris, 111-128. 144. World Organisation for Animal Health (OIE) (2004). Chapter 2.2.2. Aujeszkys disease. In Manual of Diagnostic Tests and Vaccines for Terrestrial Animals (mammals, birds and bees), 5th Ed. OIE, Paris, 295-307. 145. World Organisation for Animal Health (OIE) (2005). Chapter 2.2.10. Foot and mouth disease. In Terrestrial Animal Health Code, 14th Ed. OIE, Paris, 111-122.

372

Rev. sci. tech. Off. int. Epiz., 26 (2)

146. World Organisation for Animal Health (OIE) (2005). Chapter 2.7.12. Avian influenza. In Terrestrial Animal Health Code, 14th Ed. OIE, Paris, 294-300. Also available at: http://www.oie.int/eng/normes/mcode/en_chapter _2.7.12.htm (accessed on 25 June 2007). 147. Wu Q., Brum M.C.S., Caron I., Koster M. & Grubman M.J. (2003). Adenovirus-mediated type I interferon expression delays and reduces disease signs in cattle challenged with foot-and-mouth disease virus. J. Interferon Cytokine Res., 23, 359-368. 148. Zakhartchouk A.N., Pyne C., Mutwiri G.K., Papp Z., Baca-Estrada M.E., Griebel P., Babiuk L.A. & Tikoo S.K. (1999). Mucosal immunization of calves with recombinant bovine adenovirus-3: induction of protective immunity to bovine herpesvirus-1. J. gen. Virol., 80, 1263-1269.

149. Zhang F Yu M., Weiland E., Morrissy C., Zhang N., ., Westbury H. & Wang L.F (2006). Characterization of . epitopes for neutralizing monoclonal antibodies to classical swine fever virus E2 and Erns using phage-displayed random peptide library. Arch. Virol., 151, 37-54.

Rev. sci. tech. Off. int. Epiz., 2007, 26 (2), 373-378

OIE standards for vaccines and future trends


S. Edwards
Veterinary Laboratories Agency, New Haw, Addlestone, Surrey KT15 3NB, United Kingdom Professor Edwards is currently the President of the OIE Biological Standards Commission

Summary The World Organisation for Animal Health (OIE), sets out international standards for vaccines in the Manual of Diagnostic Tests and Vaccines for Terrestrial Animals (mammals, birds and bees). The texts are drafted by experts then sent for scientific peer review and for comment by all OIE Member Countries, thus achieving a consensus at the point of adoption. Introductory chapters in the Terrestrial Manual provide general principles of laboratory management and vaccine production, and disease-specific chapters provide detailed standards for vaccines for the OIE listed diseases. Issues that the OIE Biological Standards Commission are currently addressing or will address in the near future include the matching of vaccine strains to currently circulating infectious agents, the use of companion diagnostic tests to differentiate infected from vaccinated animals, the development of vaccine banks, and the application of DNA technology to vaccine design and production. Keywords Biotechnology Differentiating infected from vaccinated animals (DIVA) strategy International standard Quality control Safety Vaccine Vaccine bank.

Introduction
The World Organisation for Animal Health (OIE) is an intergovernmental organisation of 169 Member Countries. The Organisation is mandated under the Sanitary and Phytosanitary Agreement of the World Trade Organization to set standards that safeguard international trade in animals and animal products. It has six primary objectives, which include the promotion of international solidarity in the control of animal diseases, the publication of animal health standards, and provision of a better guarantee of safety in food of animal origin. One of the OIEs principal standard texts is the Manual of Diagnostic Tests and Vaccines for Terrestrial Animals (mammals, birds and bees) (hereafter referred to as the Terrestrial Manual) (13), currently in its 5th edition. This article will explain the process by which the Terrestrial Manual chapters are developed and approved, will outline the vaccine-related contents of the Terrestrial Manual, and will seek to put these in the context of other national and supra-national regulatory frameworks. The Terrestrial

Manual covers all the OIE listed diseases of terrestrial animals together with a number of other diseases of animal or public health concern. It has become widely adopted as a key reference book for veterinary laboratories around the world. Much of it is devoted to laboratory diagnostic tests, but this article will focus on the vaccine elements. There is a series of generic introductory chapters which give general guidelines, including six with specific relevance to vaccines. The rest of the Terrestrial Manual consists of disease-specific chapters that provide general background to: a) the disease, its diagnosis and control b) approved diagnostic techniques c) requirements for vaccines and diagnostic biologicals. A new updated edition of the Terrestrial Manual is published approximately every four years, but it has been agreed by the International Committee of the OIE that urgent updates, once approved by the Committee, may be incorporated in the online edition on the OIE website (www.oie.int) without waiting for the next full printed edition.

374

Rev. sci. tech. Off. int. Epiz., 26 (2)

The OIE Terrestrial Animal Health Code (14) also makes reference to use (or non-use) of vaccines, particularly in regard to the certification of animals or animal products for international trade, or to the surveillance and declaration of freedom from diseases or infections in specified populations. In such cases any vaccines should comply with the standards published in the Terrestrial Manual.

Seed lots used for production of the vaccine must be shown to be pure and free from contaminating strains. In addition, inactivated vaccines need to be shown to be fully inactivated. Again, appropriate methodologies are provided.

Safety in the veterinary microbiology laboratory

Development and approval of vaccine standards


The OIE Terrestrial Manual is produced under the aegis of the Biological Standards Commission, supported by an editorial team. The Commission appoints authors for each chapter who are recognised as international scientific experts on the particular disease or topic, and who produce a draft text, or modify and update an existing text. This is then submitted to scientific peer reviewers, and to the delegates of all OIE Member Countries, for comment. Comments are collated, incorporated into the chapter if straightforward, or where necessary referred back to the Commission for a decision. Finally, texts are presented by the President of the Commission to the OIE International Committee for approval and adoption. Thus, the OIE can rightly claim that the texts have consensus approval and support from its Member Countries, and that they therefore represent valid international standards. Chapters often make reference to new technologies under development, usually at the stage of ongoing research, but it should be clear that these are included for information and are not yet adopted as formal standards. Conversely, in some cases older vaccine technologies may still have a place in certain circumstances but may not be acceptable for use in all Member Countries.

Laboratories (including vaccine manufacturing plants) are intrinsically hazardous environments. The chapter sets out the principles of safe working practices taking into account physical and chemical hazards as well as the need to protect staff from pathogenic micro-organisms. Although many vaccines are derived from attenuated strains, this is not always the case and comprehensive risk assessments are essential before any work is started. The standards complement, and do not replace, those established by the World Health Organization or national authorities. In addition, veterinary laboratories and vaccine manufacturers need to avoid accidental release of animal pathogens, even where there is minimal risk to humans. An appendix to the chapter sets out the principles of such biocontainment specifying four containment levels, with the highest (level 4) applying to highly contagious organisms such as foot and mouth disease (FMD) virus.

Principles of veterinary vaccine production


The regulatory framework for vaccine production is complex and varies between countries and regions. Specific reference is made in the chapter to European Union Directive 2001/82/EC (as amended) (2), the European Pharmacopoeia (3), and the Code of Federal Regulations in the United States of America (USA) (9). The OIE is not a regulatory body, but sets out general principles that should be applied to any situation to ensure that authorised products are pure, safe, potent and effective. This provides a secure basis for the successful operation of control programmes for many animal diseases. The chapter deals with questions of nomenclature, types of vaccine, the requirements for a production plant, quality management, vaccine ingredients (including master seeds and other components), and the range of tests required to demonstrate safety, efficacy and stability, both during manufacture and in field use. Special reference is made to the use of vaccines derived from recombinant DNA technology, where guidelines are still at the stage of active development, and this is considered in more detail in the chapter on biotechnology (see below).

Generic chapters with relevance for vaccine development


Tests for sterility and freedom from contamination
It is essential to show that all vaccines are free from adventitious agents that may be accidentally introduced during various stages of the production process. This is a particular hazard with vaccines containing live attenuated organisms where a number of contaminants are recognised as high risk, notably mycoplasmas and noncytopathogenic viruses (particularly pestiviruses) in cell cultures used for viral vaccine production. The chapter provides methodologies for detection of these and other contaminants.

International standards for vaccine banks


This new guideline, adopted in 2005 and available online from the OIE website, recognises the importance for

Rev. sci. tech. Off. int. Epiz., 26 (2)

375

Member Countries of having rapid access to vaccines to deal with outbreaks of epizootic diseases. The manner in which such emergency vaccination is applied will vary with the disease, the epidemiological situation, and the disease control policies of the country concerned. Vaccine banks are designed to maintain stocks of vaccine or vaccine antigen for emergency use, often in countries where vaccination for the disease is not normally practised. The banks may be national or held on behalf of multi-country consortia. In the latter case clear legal agreements are required concerning access by individual countries. Arrangements need to take account of regulatory controls as well as lead time for reconstitution and filling to final product. The vaccine bank and its management should be an integral part of a countrys contingency plan for control of exotic diseases. Key decisions need to be made concerning the number of doses to be stored and the strains of organism to be included in the bank particularly for antigenically variable pathogens such as FMD virus, where the constituents of the bank may need to be regularly reviewed and updated to match currently circulating field strains. Although there may be special regulatory provisions for the use of emergency vaccination in Member Countries, the vaccine bank is not exempt from the general principles underlying safety and efficacy, and the facility must still comply with the quality principles of Good Manufacturing Practice.

virulence. A classic example is the vaccinia-vectored rabies vaccine that has been used successfully for disease control in wildlife (7). More recently, a number of vaccines based on avian pox viruses have been developed (8). When used in mammalian hosts these express the foreign vectored gene and stimulate protective immunity without replicating to produce progeny virus. Finally, the role of DNA vaccines is considered. Although not as yet a fully developed technology it shows considerable potential (6) and the current state of scientific development is described in the chapter.

The role of official bodies in vaccine regulation


This chapter (online version updated in 2005) outlines in some detail the regulatory procedures in the three regions that have made most progress in this field, namely Japan, the European Union, and the USA. In addition, the role of the appropriate pharmacopoeias is discussed, particularly the European Pharmacopoeia (3) (which has 35 signatory member states, as well as a further 17 with observer status). The procedural differences are described between the American and European systems, together with efforts being made to harmonise these with each other, and with regulations of other countries and regions. The International Cooperation on Harmonisation of Technical Requirements for Registration of Veterinary Medicinal Products (abbreviated to VICH) is a trilateral programme of the EU, Japan and the USA which has an ongoing commitment to develop harmonised technical standards for vaccine production. Unfortunately progress to date has been rather slow.

Biotechnology in vaccine development


A number of biotechnological approaches are now well established in vaccinology and authorised products are already in use for disease control in the field. Good examples are the deletion of genes from bacteria or viruses to reduce the virulence (e.g. Aro mutants of Salmonella spp. [4]), in contrast to traditional methods of attenuation through multiple passage in culture. The chapter provides several examples where such gene-deleted vaccines can be used in conjunction with a companion diagnostic test to differentiate infected from vaccinated animals (the DIVA strategy), e.g. Aujeszkys disease (10) and infectious bovine rhinotracheitis. Other technologies, which also offer the opportunity for DIVA strategies, include subunit viral vaccines, based on individual immunogenic proteins expressed in heterologous systems such as baculovirus or poxvirus (e.g. E2 protein of classical swine fever virus [14]). The DIVA approach allows the eradication of a disease in the presence of a vaccination strategy for disease control (11). Vaccines have also been developed in which a live virus vector is used to express an immunologically relevant protein of the pathogenic organism, giving the advantages of live virus vaccines (replication in the host leading to better immune responses) without the risk of reversion to

Disease-specific chapters
Vaccination as a disease control method is not applicable in all cases, but where there are useful vaccines available each disease-specific chapter provides a basic standard for their manufacture and usage. The standards follow a consistent format of: seed management method of manufacture in-process control batch control tests on the final product. Where there is more than one vaccine type available, an introductory account is given to enable users to make informed choices on the preferred approach. It is not

376

Rev. sci. tech. Off. int. Epiz., 26 (2)

possible in this short article to go into detail for all the listed diseases, for which reference should be made to the Terrestrial Manual. We shall briefly examine two specific aspects of particular interest, namely, vaccine matching tests for FMD and DIVA strategies.

recombinant vaccines expressing a single immunogenic viral protein (classical swine fever E2 glycoprotein).

Foot and mouth disease: vaccine matching tests


Vaccinal immunity to FMD virus is serotype-specific, but even within serotypes complete cross-protection between different isolates may not occur. A new text, adopted in May 2006 by the OIE International Committee, presents for the first time a detailed explanation of how to select circulating field viruses, and how to evaluate these using antisera specific to different vaccine strains for the best fit for cross-protection. Tests used may be based on complement fixation, enzyme-linked immunosorbent assay, or virus neutralisation technologies.

Future trends/concluding remarks


The need for new and improved vaccines will continue in the future because the demand for livestock and livestock products is expected to grow enormously in the next fifty years, particularly in developing countries. This will lead to more disease problems and emerging diseases. The demand for cheap, reliable vaccines will increase and general guidelines are essential, especially for the developing countries. There is a pressing need to encourage efforts towards international harmonisation, including technical procedures related to quality control, the evaluation of safety and efficacy of vaccines, and the regulatory protocols used to authorise products in different countries. In this respect the VICH initiative is to be welcomed. Meanwhile, the continual improvement of the chapters in the OIE Terrestrial Manual will provide internationally recognised standards that are of particular value to those countries which do not as yet have fully formed regulatory processes. Vaccine banks are expensive to set up and maintain, but this must be set against the enormous potential costs of non-vaccination strategies in the face of increasing global spread of diseases. Agreed standards for the maintenance and exploitation of such banks are therefore essential. Advances in biotechnology, immunology and proteomics will continue to generate novel approaches to vaccine design. It is essential that robust standards are in place and are applied to ensure that new products are adequately evaluated both for their safety and efficacy for the animal and the consumer, and for any environmental impacts that may ensue.

Strategies for differentiating infected from vaccinated animals


There are a number of diseases for which a DIVA approach may be used, allowing continued vaccination while still being able to detect infected animals through serological responses to proteins that are absent from the vaccine. So far this has been limited to certain virus infections, but the principle could be applied more widely. The details of the DIVA approach vary with the disease. Thus, for FMD, the vaccines used are conventional inactivated viral vaccines, but highly purified during manufacture so that they lack immunologically significant levels of non-structural proteins (NSP) (5). Animals naturally infected with FMD virus do develop antibodies to NSP, even if already vaccinated, so an NSP-specific antibody test can be used in a DIVA strategy. This has the additional advantage of not being serotype-specific. In the case of avian influenza, use can be made of naturally occurring strains for vaccine manufacture where the neuraminidase type is different from the circulating field virus, taking advantage of the fact that the main protective immune response is to the haemagglutinin protein. Thus, neuraminidase inhibition serological tests provide a means of detecting field infection in birds that have been vaccinated against a haemagglutinin type homologous to the field virus (1). Other examples of DIVA applications, as mentioned above include the use of gene-deleted vaccines for herpesviruses (Aujeszkys disease, infectious bovine rhinotracheitis), and

Acknowledgements
I am grateful to Christianne Bruschke for her insight and advice in the preparation of this article.

Rev. sci. tech. Off. int. Epiz., 26 (2)

377

Normes de lOIE relatives aux vaccins et tendances pour lavenir


S. Edwards
Rsum Les normes internationales prescrites par lOrganisation mondiale de la sant animale (OIE) en matire de vaccins sont dcrites dans le Manuel des tests de diagnostic et des vaccins pour les animaux terrestres (mammifres, oiseaux et abeilles). Ces normes sont prpares par des experts, soumises une valuation collgiale puis diffuses auprs des Pays Membres de lOIE afin de recueillir leurs commentaires ; au moment dtre adoptes, elles sont donc le fruit dun large consensus. Les premiers chapitres du Manuel terrestre exposent les principes gnraux rgissant la gestion des laboratoires et la production de vaccins, tandis que les chapitres sur les maladies dcrivent en dtail les normes relatives aux vaccins pour chaque maladie figurant sur la liste de lOIE. Plusieurs questions dintrt immdiat ou futur sont actuellement examines par la Commission des normes biologiques de lOIE : ladquation des souches vaccinales avec les souches pathognes en circulation ; le recours des tests diagnostiques compagnons, permettant de distinguer les animaux infects des animaux vaccins ; la cration de banques de vaccins et les technologies de lADN applicables au dveloppement et la production de vaccins. Mots-cls Banque de vaccins Biotechnologie Contrle de qualit Norme internationale Scurit Stratgie pour diffrencier les animaux infects des animaux vaccins (DIVA) Vaccin.

Normas de la OIE sobre vacunas y tendencias de cara al futuro


S. Edwards
Resumen La Organizacin Internacional de Sanidad Animal (OIE) fija normas internacionales sobre vacunas en su Manual de pruebas de diagnstico y vacunas para los animales terrestres (mamferos, aves y abejas). Tras una primera fase de redaccin, a cargo de especialistas, los textos se someten al examen de otros expertos de igual nivel y a las observaciones que puedan formular todos los Pases Miembros de la OIE, proceso que culmina con un consenso en el momento de la aprobacin. Los captulos introductorios del Manual terrestre sientan principios generales sobre procedimientos de laboratorio y fabricacin de vacunas, mientras que en los captulos siguientes, dedicado cada uno a una patologa, se fijan normas detalladas sobre las vacunas contra las enfermedades inscritas en la lista de la OIE. Entre los temas de los que se ocupa o va a ocuparse la Comisin de Normas Biolgicas de la OIE figuran la adecuacin de las cepas vacunales a los agentes infecciosos actualmente en circulacin, el uso de pruebas de diagnstico complementarias para distinguir entre animales infectados y vacunados, la creacin de bancos de vacunas o la aplicacin de la tecnologa del ADN a la concepcin y fabricacin de vacunas. Palabras clave Banco de vacunas Biotecnologa Control de calidad Estrategia de discriminacin entre animales infectados y vacunados (DIVA) Norma internacional Seguridad Vacuna.

378

Rev. sci. tech. Off. int. Epiz., 26 (2)

References
1. Capua I., Cattoli G., Marangon S., Bortolotti L. & Ortali G. (2002). Strategies for the control of avian influenza in Italy. Vet. Rec., 150, 223. 2. Commission of the European Communities (2001). Directive 2001/82/EC of the European Parliament and of the Council of 6 November 2001 on the Community code relating to veterinary medicinal products. Off. J. Eur. Communities, L 311, 1-66. 3. Council of Europe (COE) European Directorate for the Quality of Medicines (EDQM) (2005). European pharmacopoeia, 5th Ed. COE, Strasbourg. 4. Jones P.W., Dougan G., Howard C., Mackenzie N., Collins P. & Chatfield S.N. (1991). Oral vaccination of calves against experimental salmonellosis using a double aro mutant of Salmonella typhimurium. Vaccine, 9, 29-34. 5. Mackay D.K.J., Forsyth M.A., Davies P.R., Berlinzani A., Belsham G.J., Flint M. & Ryan M.D. (1997). Differentiating infection from vaccination in foot-and-mouth disease using a panel of recombinant, non-structural proteins in ELISA. Vaccine, 16, 446-459. 6. Nobiron I., Thompson I., Brownlie J. & Collins M.E. (2003). DNA vaccination against bovine viral diarrhoea virus induces humoral and cellular responses in cattle with evidence for protection against viral challenge. Vaccine, 21, 2091-2101. 7. Pastoret P.-P. & Brochier B. (1996). The development and use of vaccinia-rabies recombinant oral vaccine for the control of wildlife rabies: a link between Jenner and Pasteur. Epidemiol. Infect., 116, 235-240. 8. Taylor J., Trimarchi C., Weinberg R., Languet B., Guillemin F ., Desmettre P. & Paoletti E. (1991). Efficacy studies on a canary pox-rabies recombinant virus. Vaccine, 9, 190-193. 9. United States Department of Agriculture (USDA) (2001). Code of Federal Regulations, Title 9, Animals and animal products, Part 113, Standard requirements. US Government Printing Office, Washington, DC, USA. 10. Van Oirschot J.T., Terpstra C., Moormann R.J.M., Berns A.J.M. & Gielkens A.L.J. (1990). Safety of an Aujeszkys disease vaccine based on deletion mutant strain 783 which does not express thymidine kinase and glycoprotein I. Vet. Rec., 127, 443-446. 11. Van Oirschot J.T., Kaashoek M.J. & Rijsewijk F .A.M. (1996). Advances in the development and evaluation of bovine herpesvirus 1 vaccines. Vet. Microbiol., 53, 43-54. 12. Van Rijn P.A., Van Gennip H.G. & Moormann R.J. (1999). An experimental marker vaccine and accompanying serological diagnostic test both based on envelope glycoprotein E2 of classical swine fever virus (CSFV). Vaccine, 17, 433-440. 13. World Organisation for Animal Health (OIE) (2004). Manual of Diagnostic Tests and Vaccines for Terrestrial Animals, 5th Ed. OIE, Paris. 14. World Organisation for Animal Health (OIE) (2006). Terrestrial Animal Health Code, 15th Ed. OIE, Paris.

Rev. sci. tech. Off. int. Epiz., 2007, 26 (2), 379-393

Regulatory requirements for vaccine authorisation


P.G.H. Jones (1), G. Cowan (2), M. Gravendyck (3), T. Nagata (4), S. Robinson (5) & M. Waits (6)
(1) International Federation for Animal Health, rue Defacqz, 1, 1000 Brussels, Belgium (2) Pfizer Limited, Ramsgate Road, Sandwich, Kent, CT13 9NJ, United Kingdom (3) Intervet International BV, Wim de Korverstraat 35, PO Box 31, 5830 AA Boxmeer, the Netherlands (4) Merial Japan Ltd., Sanno Grand Building, 2-14-2 Nagata-cho, Chiyoda-ku, Tokyo 100-0014, Japan (5) Fort Dodge Animal Health, Asia Pacific Regional Technical Office, 1 Maitland Place, Baulkham Hills, NSW 2153, Australia (6) Merial Ltd., 115 Transtech Drive, Athens, GA 30601, United States of America

Summary Vaccines are one of the most important tools available in the prevention and control of diseases in animals. It is therefore of the utmost importance that when vaccines are used, such use should meet with the requirements of the World Organisation for Animal Health Terrestrial Animal Health Code and must be authorised by the recognised licensing body in the country/region where the vaccines are to be used, in accordance with the three key criteria of quality, safety and efficacy. This article provides a comprehensive and comparative description of the regulatory requirements in place for veterinary vaccines in major regions of the world, highlighting the similarities and pointing out also where there are differences. Recent advances in harmonisation of such testing requirements achieved through the International Cooperation on Harmonisation of Technical Requirements for Registration of Veterinary Medicinal Products (VICH) are also described. The contents will provide a valuable guide to those engaged in the research and development of vaccines globally, and reassure those involved in the prevention and control of animal diseases that veterinary vaccines, when fully authorised and used according to the label instructions, are safe and efficacious. Keywords Efficacy Harmonisation International Cooperation on Harmonisation of Technical Requirements for Registration of Veterinary Medicinal Products (VICH) Quality Regulatory requirements Safety Veterinary vaccine.

Introduction
Vaccines for animals, like all other veterinary medicines, have to be licensed by the relevant authorities charged with that responsibility in the country or region where the products are to be marketed. The authorisation process has to ensure that the medicinal product is of adequate quality and purity, that it is safe and that it works in the target species as claimed, for the indication/treatment for which it is intended.

Whilst society rightly demands that medicines, including vaccines, are licensed according to very high standards, the regulatory environment should not be so risk-averse in the demands made on sponsors of new products that innovation and investment in research and development are stifled, and the availability of adequate veterinary medicines compromised. Veterinary vaccines, like those produced for human use, are therefore authorised according to high standards of

380

Rev. sci. tech. Off. int. Epiz., 26 (2)

quality, safety and efficacy (taking account of the risk/benefit balance for each product under consideration) and these three major criteria are mostly defined in a similar way in the major markets around the world. However, there is variation between different countries, such that regulatory dossiers have often to be tailored for different markets. This adds to the already considerable expense which companies incur when researching and developing a new medicine, which can take over a decade and cost as much as 50 million euros. These high costs are often the cause of additional problems in the veterinary sector, which differs considerably from the human medicine sector. Because the investment needed to bring new products to the market place is so large, vaccines for so-called minor species, where the potential market size is small (e.g. rabbits, goats and some species of poultry) rarely get developed, because the return on investment is not attractive enough for the animal health companies involved in marketing them. As a result many vaccines needed for use in minor species are just not available and licensed vaccines for one species may not be suitable for use in another species, which can result in a potential conflict for practising veterinarians in their duty of care and welfare to their patients. During the development of a new vaccine the marketing authorisation holder must subject the vaccine to various tests defined in the legislation and guidelines, and these are sometimes revised. If such changes arise when a new product is well into its development phase, the consequences can be grave, with further costs being incurred to generate additional data to satisfy the newly imposed changes in requirements. Such unpredictability certainly drains reserves for investing in new products and curbs investment in new technology and innovation as well. Fortunately, regulatory authorities are now more aware of such constraints, and are prepared to collaborate with the animal health industry to set more realistic requirements that facilitate the development of new medicines, whilst at the same time removing hurdles that can delay the time it takes to bring badly needed new products to market.

will provide the reader with a good understanding of the rigorous regulatory systems in place to ensure that only high quality, safe and efficacious vaccines come onto the market place. Safety is of course of paramount importance. The determination of safety is fundamentally a determination that the benefits of the product outweigh any potential risks, not only to the target species being vaccinated, but also to the user/administrator of the vaccine, the environment, and in the case of animals from which food is derived, the consumer as well. The major regulatory authorities essentially have similar guidance on how to apply for a licence and a synopsis of the actual data required with reference to the differences between the various regions is provided in the next section. Applications are generally submitted to the authority in charge of licensing medicines and these can be separate veterinary regulatory departments within the Ministry of Agriculture, stand-alone agencies within governments or even parts of a joint regulatory department with responsibility for human medicines as well. The applicant, normally a pharmaceutical company, has to be legally established in the country/region concerned and constituted under civil or commercial law to ensure that obligations for compliance with the legal-regulatory pharmaceutical framework are fully adhered to. What follows is a summary of the systems in place, the authorities responsible and the type of information required.

Japan
Veterinary drugs are under the control of the Ministry of Agriculture, Forestry and Fisheries (MAFF), which issues the licence/market approval for each vaccine following a review of the application dossier by various committees charged with assessment of the data. The standard approval time for a new vaccine is up to one year, excluding any time required for responding to requests from the Ministry for additional data from the applicant. The approval of a conventional vaccine listed as a monograph in the Standards of Veterinary Vaccines for National Assay does not require renewal, whereas the approval of a vaccine containing a new class of antigen or new combination of antigens requires renewal after six years following the initial approval.

Procedures for licensing veterinary vaccines


This section describes the procedures which potential applicants wishing to license vaccines in the major animal health markets around the world need to follow. The conditions under which the different authorities will consider applications for licensing, and the requirements which have to be satisfied are also covered to a degree that

United States of America


Applications for licensing have to be submitted to the Center for Veterinary Biologics (CVB) which is part of

Rev. sci. tech. Off. int. Epiz., 26 (2)

381

Veterinary Services (VS), a division of the Animal Health and Plant Inspection Service (APHIS) of the United States Department of Agriculture (USDA). The agency allows phased filing, which allows for the submission of results from studies as they become available instead of having to assemble a complete dossier for submission. Documents needed for phased filing are described in Veterinary Services Memorandum No. 800.50 and consist of three major packages: Application for Product License, Supporting Data for Product License Application, and Requests to Conduct Field Studies. As stated in this memorandum, the applicants are encouraged to interact with CVB personnel as necessary, to facilitate submissions. The licence has no expiration date and does not need to be renewed as long as product is produced on a regular basis. The CVB supports a serial release system; each serial produced is submitted to the CVB for review, possible testing and official release.

market the product and these are all included in the assessment procedure from the outset, with one regulatory agency acting as the reference member state that leads the procedure. With mutual recognition procedures the dossier is submitted to the regulatory authority of one member state (the reference member state), who undertakes the assessment, and once approved, the authorisation to market is mutually recognised by the other member states where the company has filed the dossier (the concerned member states). Public Assessment Reports for vaccines authorised through this route are also soon to be made public.

Australia
All vaccines must be registered by the Australian Pesticides and Veterinary Medicines Authority (APVMA) in accordance with the Agricultural and Veterinary Chemicals Act 1994 and the Agricultural and Veterinary Chemicals Code Act 1994 and other supporting legislation. Once APVMA registration is granted, state by state registration is not required.

European Union
Potential applicants have a choice of routes to authorisation of vaccines in the European Union (EU). If the vaccine is derived from biotechnology then the application must be filed with the European Medicines Agency (EMEA) in London, through the so-called centralised procedure and the dossier is assessed by the Committee for Medicinal Products for Veterinary Use (CVMP) within a legislative time frame of 210 days, excluding the time taken for companies to provide additional data to the Committee if requested to do so. If the Committee is in favour of the application it issues a positive opinion which is then transferred into a Community marketing authorisation by the European Commission in Brussels, giving the company the commercial advantage of allowing it to market the vaccine in all 27 member states of the EU. This route to licensing is also open to non-biotech vaccines if they are innovative and/or can be shown to have particular advantages for animal health. Shortly after the Community Authorisation is published in the Official Journal of the EU, the EMEA publishes on its website a European Public Assessment Report (EPAR) which provides a detailed summary of the CVMP assessment, excluding proprietary information about the product considered to be confidential. EPARs of all vaccines authorised through this procedure are to be found on the EMEA website (www.emea.europa.eu). Alternatively, if the vaccine is not derived from biotechnology or is innovative, but the applicant only wishes to market the product in a few of the 27 EU member states, then the submission of the dossier can be made through a national procedure followed by the mutual recognition of this approval by other EU member states or through the new decentralised procedure, in which the applicant selects the member states in which they wish to

Presentation of dossiers
Details of the responsible authorities and the appropriate legislation for authorisation of vaccines in the various major markets are provided in Table I. With all these authorities, the dossier is presented and submitted according to the local requirements. However, the submission is generally required to be formatted in a structured manner and must include an administrative part which will include detailed information on the applicant company, the provision of samples if required, information on the manufacturing site, evidence of conformance to Good Manufacturing Practice (GMP), and details of marketing authorisations/licences granted elsewhere. Details on the product itself and its characteristics are included in this section, as is information on the proposed packaging and labelling of the vaccine. The main part of the dossier is the technical section consisting of manufacture and control, followed by safety, preclinical and clinical documentation, respectively.

Post-authorisation requirements
Regulation of vaccines does not, however, end with the issuing of a marketing authorisation. The postauthorisation phase is just as important in the products life cycle as that prior to its introduction: the requirement to monitor for any potential adverse reaction to a vaccine

382

Rev. sci. tech. Off. int. Epiz., 26 (2)

Table I Summary details of the responsible bodies and the legislation in place for authorising vaccines
Country Japan Department responsible National Veterinary Assay Laboratory, Ministry of Agriculture, Forestry and Fisheries, Tokyo United States of America Center for Veterinary Biologics, Animal Health and Plant Inspection Service, United States Department of Agriculture European Union Innovative and biotechnology vaccines: European Medicines Agency, Unit for Veterinary Medicines, London, United Kingdom Conventional vaccines: European Union Member State Competent Authorities* Australia Australian Pesticides and Veterinary Medicines Authority Agricultural and Veterinary Chemicals Act 1994 and the Agricultural and Veterinary Chemicals Code Act 1994
* see http://www.hevra.org/directory.asp

Pharmaceutical legislation Pharmaceutical Affairs Law No. 145 Series of 1960

Title 21 of the United States Code (Nos 151-159), with implementing regulations in Title 9 of the Code of Federal Regulations (Nos 101-122) Regulation No. 726/2004 of the European Parliament and of the Council

Directive 2004/28/EC of the European Parliament and of the Council

once licensed is an integral part of the regulatory system for most authorities. In most countries, therefore, the licence holder is required to have in place an appropriate means of monitoring the vaccine in the field, i.e. they must carry out pharmacovigilance/vaccinovigilance. With marketing and use of a product in large numbers of animals under normal field conditions, valuable information about the safety profile of the product is gathered. This post-marketing monitoring assists in the detection of adverse events that occur infrequently in large populations and would not ordinarily be recognised in pre-authorisation safety studies. If any potential safety problems are detected, the product sponsor and the regulatory authority collaborate on the appropriate measures to address identified issues. For this purpose the regulatory systems in force have very specific guidelines enshrined in law as to the obligations for reporting adverse drug reactions, the content and timing of such reports, and the actions incumbent on the licence holder in following up with the appropriate investigations.

framework of the systems for authorising vaccines in different parts of the world. It is important first of all however to draw attention to efforts that have been ongoing for a number of years to harmonise such requirements. The International Cooperation on Harmonisation of Technical Requirements for Registration of Veterinary Medicinal Products (VICH) was officially launched in April 1996. This is a trilateral programme between Japan, the USA and the EU aimed at establishing harmonised technical requirements which meet quality, safety and efficacy standards, minimising the use of test animals and reducing the costs of product development. A number of guidelines have been established or are in the process of being established to harmonise the requirements for biologicals, and details of these can be found on the VICH website (www.vichsec.org). Australia, Canada and New Zealand are observers to VICH and also implement the adopted guidelines in their regulatory systems.

Technical data requirements


What follows is a detailed presentation of the testing requirements in the major regions of the world for authorising vaccines, which enables a comparison to be made of the differences which may exist, and which are extremely important for companies who may be intending to register and commercialise products on a global basis. Details of the registration requirements are provided for the EU, the United States of America (USA), Japan and Australia, thus providing a global overview of the

Requirements for the registration of veterinary vaccines in the European Union


Categories of veterinary vaccines
Vaccines exist against viral, bacterial, fungal or parasitic infections. These can be based on live, attenuated, or inactivated agents. Within these categories complete causative agents (whole cell vaccines) or parts of an agent (subunit or vector vaccines) may be used to obtain protection. Because the vast majority of existing vaccines

Rev. sci. tech. Off. int. Epiz., 26 (2)

383

are used against viral or bacterial infections this article will mainly focus on the requirements for live and inactivated vaccines of these types.

Basic registration requirements


Although regulations may differ, in general, all authorities aim at licensing only those products that meet a number of basic requirements. Only vaccines whose quality, safety and efficacy have been proven obtain a marketing authorisation in the EU. A vaccine licence is initially issued with a five-year validity. After re-evaluation of the risk/benefit balance the marketing authorisation is renewed and is thereafter valid for an unlimited period of time. Apart from direct EU law, companies should also take into account other legislation that is applicable in Europe, e.g. the European Pharmacopoeia (Ph. Eur.). In the Ph. Eur. general requirements as well as specific tests for starting materials or the final product (vaccine monographs) are laid down. The current EU criteria which veterinary vaccines must meet are outlined below.

guarantee the quality of the material. The vaccine manufacturer must declare a specification for each starting material and ensure that each batch purchased meets the acceptance limits of the specification. Many starting materials are listed as monographs in the Ph. Eur. Even these must be tested to ensure compliance with the current monograph. Additionally, requirements exist for starting materials of biological origin which are commonly used for vaccine production (e.g. exclusion of extraneous agents). Seed stocks of the vaccine strain are laid down in a seed lot system, making passages from the master seed to establish a bank of working seed from which all production batches are produced. The history of the isolation and previous passages of the initial master seed must be known in order to minimise the risk of transmission of spongiform encephalopathies (TSE).

Testing
Purity A veterinary medicinal product must be sterile, i.e. free from any contamination with live microbiological agents. In the specific case of live vaccines the product should not contain live microbiological agents other than the vaccine strain(s). In order to achieve this, the master seed stock from which all subsequent vaccine batches will derive must be absolutely free from extraneous agents. For virus vaccines this means freedom from contaminating bacteria, fungi, Mycoplasma species, and extraneous viruses; for bacterial vaccines it means freedom from contaminating bacteria and fungi. The expression used to determine freedom from contamination for bacterial vaccines is purity. To confirm the absence of extraneous agents in virus vaccines, validated assays must be used, i.e. spiking of the seed virus with a series of extraneous agents should reveal positive test results. Identity In addition to purity, the vaccine strain must be tested to ensure it is identified as the correct strain. Identification is pursued beyond the strain and species to subtypes or serotypes as appropriate. In-process and finished product testing To ensure that each batch of a commercial vaccine is equivalent in quality and will therefore be safe and efficacious, the manufacturer must register all relevant inprocess tests as well as tests to be performed on the finished product, giving limits of acceptance that must be met before the batch can be released for sale. Once these test methods and limits have been approved by the regulatory agencies, they become mandatory tests for placing the vaccine on the market. The type of tests performed may include: sterility/purity tests for absence of contamination

Quality Manufacture
Since licensing of veterinary vaccines was formally introduced in the EU in 1981 it has been a requirement that both the active ingredient (antigen) and the finished product (vaccine) must be manufactured according to GMP. Even before that time, some national regulatory agencies already insisted on GMP for the manufacture of veterinary vaccines. In addition to a marketing authorisation for each country in which the product is to be marketed, a manufacturing licence must be obtained for each production facility where the vaccine (or part thereof) is being produced. Data must be presented to show that the manufacturer is able to produce the vaccine in a consistent manner. Regular internal and external audits aim at surveying the quality control procedures in place. Moreover, a vaccine producer must employ at least one qualified person who is, without prejudice to his relationship with the holder of the manufacturing authorisation, personally responsible for release of vaccine onto the market. The quality of starting materials used for production, whether bought from a commercial supplier or produced in-house, must be evidenced. In the case of ingredients purchased from external suppliers, the commercial supplier has his own quality assurance system. In addition, controls on incoming-goods are performed by the vaccine manufacturer (e.g. growth promotion assays for serum batches used to cultivate certain vaccine strains) to further

384

Rev. sci. tech. Off. int. Epiz., 26 (2)

antigen quantification tests (e.g. titre, cell count, optical density, enzyme-linked immunosorbent assay [ELISA], etc.) tests for complete inactivation (inactivated vaccines only) physico-chemical tests, e.g. pH, viscosity of emulsion, quantity of residual inactivant, etc. (mainly valid for inactivated vaccines) adjuvant content tests, if an adjuvant is included in the formulation tests to determine the titre (live vaccines) or potency (inactivated vaccines) of finished product (see the section on Potency below) tests to verify the safety of the product in the target species. Each batch of vaccine must be tested for absence of local and systemic effects in animals of the target species using an overdose (2 3 for inactivated vaccines and 10 3 for live vaccines) administered by a route recommended on the label. The marketing authorisation holder may apply for a variation to the licence to withdraw this test following satisfactory results from at least ten batches and satisfactory pharmacovigilance reporting. In addition, the manufacturer must prove that the quality of the vaccine is guaranteed until the end of its shelf life. To demonstrate this, stability testing must be performed on at least three batches of vaccine in the final container. The vaccine must be tested at regular intervals throughout the proposed shelf life. If a preservative is included in the vaccine (multidose containers only), its effectiveness during the shelf life must be tested using a method described in the Ph. Eur.

administration of the product will consist of monitoring the safety (e.g. injection site reaction, clinical symptoms) following three administrations of the vaccine with appropriate intervals between each administration. There are additional special requirements for live vaccines. A live vaccine strain must be stable, i.e. should not revert to virulence during consecutive passages. Depending on the agent, recombination or genomic reassortment needs to be evaluated. In general, vaccine is produced within a limited number of passages from the master seed stock. Usually this is limited to five passages for virus vaccines, however, there is no limit for bacterial vaccines. The safety of the vaccine strain at the lowest passage number is shown in animal studies using the most sensitive target animal/species. Issues such as the spread of the vaccine strain and dissemination in the vaccinated animal need to be taken into account where appropriate. In addition, studies on the immunological functions may have to be carried out where the vaccine might adversely affect the immune response. If the vaccine is intended for administration to pregnant animals, safety during pregnancy must be shown using a batch of vaccine at its highest potency or titre. It must be demonstrated that vaccination has no adverse effect on the reproductive performance of the target animal. Depending on the starting materials used for manufacturing the vaccines, studies may need to be carried out on possible residues if they are considered to be present at levels having pharmacological activity. Negative effects on the environment must also be assessed. Additional requirements have to be met for vaccines containing genetically modified organisms.

Efficacy
Data must be provided that support the efficacy claims. In other words: a product must be able to do what is claimed on the label (e.g. reduce virus shedding, control clinical signs, etc.). Preferably, these data are obtained from field trials performed under Good Clinical Practice-veterinary (GCPv) conditions and from laboratory studies in which, if possible, validated experimental challenge models are used. Data showing efficacy from laboratory studies must be provided using batches with the lowest potency or titre (see below).

Safety
Clearly, the product must be safe for the target animal, but it must also be documented that the product does not pose a danger to humans or to other animals that may come into contact with the product, or to the environment. Experimental data obtained with batches with the highest potency or titre (see below) must be generated in specially designed experiments, i.e. in accordance with European Good Laboratory Practice (GLP) standards. In addition to the safety of a single dose, the safety of an overdose and repeated doses of the vaccine must be shown. The rationale for demonstrating the safety of a repeated dose is because some vaccines, especially inactivated ones, have a primary vaccination course consisting of two doses, followed by a booster dose six or twelve months later. For a vaccine with this recommendation, demonstration of the safety of repeated

Potency
The manufacturer must provide data that guarantees the immunising capability and thereby protective effect of a product over the entire shelf life. This is a major hurdle in the development of inactivated vaccines. Confirmation of the protective effect of a specific immunogen is usually established by vaccination-challenge experiments.

Rev. sci. tech. Off. int. Epiz., 26 (2)

385

Such experiments are preferably not used as routine batch potency tests as these involve animal experimentation and are normally of several weeks duration. An alternative test should ideally be developed which is carried out on each batch of finished vaccine and has predictive value as to the efficacy of that particular batch. As the efficacy of the vaccine batch has to be guaranteed over its entire shelf life the batch potency test must provide assurance that the vaccine will remain potent throughout its shelf life. This assurance is provided by testing the vaccines stability over the period of the shelf life plus an additional three months beyond for a minimum of three batches. The pass level for batch release is set at the minimum level that was shown to be efficacious in the target animal. If the protective mechanism of immunity against a specific pathogen is not known, it may take years before an accurate potency test can be developed.

Onset and duration of immunity


Apart from the information requirements imposed by regulatory authorities, most of the label information relating to the onset and duration of immunity is included on the label because of market demand. All claims must be documented and supported, either by existing literature or by experimental data. This may take years of research, particularly if, for instance, one claims that a product has a shelf life of three years. Specific efficacy claims must also be supported by data. If, for example, the label claims that the duration of immunity is one year and that a yearly vaccination will sustain this level of immunity, data must be provided that show that one year after the primary vaccination course, animals are significantly protected (in some cases by experimental challenge infection) and also that animals that receive a single booster vaccination one year after initial vaccination are still protected one year later. This involves more than two years of experimentation.

Special requirements
Registration of the label claim
In the EU, each marketing authorisation is granted with an approved Summary of Product Characteristics (SPC). This document reflects the results of the supportive data provided in the registration dossier. It includes a description of the composition of the vaccine, the target species, the route of administration, the supported vaccination schedule, the claims, any contra-indications or adverse effects that might be seen, the shelf life and a description of the vaccine containers and storage conditions. If the marketing authorisation holder wishes to change any of the statements made on the SPC, this can be achieved by varying the licence with the appropriate supporting data.

Compatibility
If the label claims that the vaccination may be carried out within two weeks of vaccination with another product (concurrent use) then this must also be documented with supportive data. This can become a complicated task depending on the target animal. For example, the life span of the average broiler chicken is 6 to 7 weeks and these animals very often need to be vaccinated against a series of pathogens (amongst others, Mareks disease virus, Newcastle disease virus, infectious bronchitis virus, Gumboro disease virus) very early in life, i.e. before the age of 14 days. Compatibility of the new product with all of these vaccines must be shown if this use is claimed. This will involve safety studies as well as efficacy studies. Obviously, if it is claimed that a vaccine can be physically mixed with another product and subsequently administered, data from safety and efficacy studies must be presented to support such a claim.

Target animal
The animal species for which the product is intended must be specified and it should be clearly stated if the vaccine is designed for use in a specific category of animal, e.g. broiler chickens not breeder chickens. The suitability of the product for use in pregnant animals should also be stated. An important factor may be the presence of maternal immunity in young animals. If this may affect the induction and onset of vaccine-induced immunity then it must be studied. Results may prompt the manufacturer to recommend not vaccinating animals below a certain age.

Recommendations
Specific recommendations have to be provided that aid in the most efficient use of the product. Such recommendations vary with the particular product. Obvious recommendations are to only vaccinate healthy animals in the case of vaccines for prophylactic use. Apart from vaccines against cattle ringworm, no therapeutic vaccines are currently licensed, but they are envisaged for the future (e.g. for the treatment of leishmaniosis). In the case of live vaccines against pathogens that can be controlled by chemotherapeutics, it may be important to recommend a withdrawal period after chemotherapeutic treatment before administering the live vaccine. Likewise, it may be sensible to advise minimisation of the risk of concurrent infections during the vaccination period, as these could interfere with the induction of the proper immune response.

Route of administration
Some products can be administered through different routes, e.g. oral application or injection. For each of these routes of administration safety and efficacy experiments must be performed to support the claims made. These claims may differ depending on the route of administration, and this must be clearly stated in the dossier and on the label and leaflet (known as the circular in US regulations).

386

Rev. sci. tech. Off. int. Epiz., 26 (2)

Post-marketing requirements
Following the granting of a marketing authorisation in the EU, each batch of vaccine that has satisfactorily undergone all the finished product testing can be placed on the market in all European member states. In some countries, however, a batch may only be placed on the market after official release by the competent authority in the EU member state concerned. For a few vaccines, e.g. rabies vaccines, release onto the market is not authorised until an officially appointed laboratory has re-tested the batch and declared that it is in compliance with the registered specification. As with other regions of the world, pharmacovigilance is strictly applied in the EU and has become the acceptable replacement for the previous requirement to apply to renew a marketing authorisation every five years.

Contact with the CVB is via a person designated as the official (primary) liaison. The CVB sends all official mail to the official liaison. Alternate liaisons can be designated to assist the liaison in signing certain documents such as official correspondence and APHIS Form 2015, which is used for submission of labels, circulars, and Outlines of Production.

Quality Manufacture
Each Veterinary Licensed Establishment in the USA must submit and maintain the qualifications of supervisory personnel and facility documents, which include blueprints, plot plans, and legends. The manufacturer is required to review and update these documents annually. A manufacturer is required to submit and obtain CVB approval of an Outline of Production detailing key steps in the manufacturing process for each licensed product. Data on three consecutive pre-licensing serials must be presented to show that the manufacturer is able to produce the vaccine in a consistent manner. Consistency of production is monitored by the agency through the use of the approved Outline of Production, release tests, and unannounced inspections. Seed stocks of the vaccine strain are laid down in a seed lot system, making passages from the master seed to establish working seed stocks from which all production batches are produced. As stated in the section on the European Union, the history of the isolation and previous passages of the initial master seed must be known in order to minimise the risk of transmitting TSEs. Cell lines used in the production process must also follow the seed lot system. Both master seed stocks and master cell stocks must be approved by the CVB for use in production of a licensed product.

Requirements for the registration of veterinary vaccines in the United States of America
Who regulates veterinary vaccines?
The Center for Veterinary Biologics (CVB), under the umbrella of the USDA, is tasked with implementing the provisions of the Virus-Serum-Toxin Act passed in 1913. The CVB is divided into two sections: Policy, Evaluation, and Licensing (CVB-PEL) and Inspection and Compliance (CVB-IC). The CVB regulates vaccines, bacterins and bacterial extracts, antibody products, diagnostic products, antitoxins, toxoids, and other products of biological origin. As stated in the section on EU requirements, this article will focus mainly on the requirements for viral and bacterial live and inactivated vaccines.

Testing
Purity The Outline of Production for all veterinary biological products must include a description of the procedures that are to be followed in order to keep the product free from any viable contaminating microbiological agents. In the specific case of live vaccines, the product should not contain microbiological agents other than the vaccine strain(s). Consequently, it must be ensured that the master seed stock from which all subsequent vaccine serials will be derived is free from extraneous agents. Regulations require that master seed and final container testing be described in the Outline of Production. Final containers of each serial and subserial are tested as follows: for virus vaccines, demonstration of freedom from contaminating bacteria, fungi, mycoplasma, and, in some cases, specific extraneous viruses; for bacterial vaccines, demonstration of

Basic registration requirements


Veterinary biologics manufactured in the USA must have a US Veterinary Biologics Establishment License and a US Veterinary Biological Product License for each separate product. The requirements for labelling, for manufacturing processes, and for obtaining an establishment licence and a product licence, can all be found in Title 9 of the Code of Federal Regulations. Additional guidance can be found in published memorandums and notices. Veterinary biologics must be proven to be pure, safe, potent, and effective prior to licensure and upon release of each serial. Applicants are encouraged to interact with CVB-PEL personnel as necessary to facilitate submissions.

Rev. sci. tech. Off. int. Epiz., 26 (2)

387

freedom from contaminating bacteria and fungi. The word used to describe freedom from extraneous microorganisms or material is purity. Identity In addition to purity, master seeds for all product types and final container samples of vaccines of each serial or subserial must be tested to ensure it is identified as the correct strain. Subtypes and serotypes may also be tested if thought necessary. Identity of final container samples may be demonstrated in conjunction with potency testing by fluorescent antibody staining, serological methods, challenge of vaccinates, or other in vitro methods, such as ELISA techniques. Finished product testing To ensure that each serial (batch) and subserial of a commercial vaccine is equivalent in quality and will therefore be safe and efficacious, the manufacturer must file in the Outline of Production all relevant tests to be performed on the finished product, giving limits of acceptance that must be met before the serial or subserial can be released. Only the CVB can release a product for distribution and sale. In the US system, a manufacturer submits a summary of all relevant tests on an official form (APHIS Form 2008) for each serial and subserial. Once a serial or subserial is produced and representative samples are submitted, the serial or subserial may be randomly picked for confirmatory testing. After the firm submits APHIS Form 2008 at the conclusion of their testing, if the serial or subserial is not chosen for confirmatory testing or if the confirmatory testing has been completed satisfactorily, the product is released by the CVB for distribution and sale. The type of tests performed may include: sterility/purity tests for absence of contamination antigen quantification tests (e.g. ELISA and tests to determine titre, cell count, optical density, etc.) tests for complete inactivation (inactivated vaccines only) physico-chemical tests, e.g. pH, viscosity of emulsion, quantity of residual inactivant, etc. (mainly valid for inactivated vaccines) tests to establish titre/potency (titre mainly for live vaccines) of finished product (see section on Potency below) safety tests in target or laboratory animals.

In addition, the manufacturer must prove that the quality of the vaccine is guaranteed until the end of its shelf life. To demonstrate this, several serials of vaccine must be tested at regular intervals throughout the proposed shelf life.

Safety
It must be shown, of course, that the product is safe for the target animal, but documentation must also be provided which demonstrates that the product is safe for the environment, for other animals, and for humans that may come into contact with the product. Extensive literature searches into the background for each master seed candidate are required prior to CVB approval of that master seed. The manufacture may be required, for certain products, to conduct risk analysis to assess the potential effects of the product on the safety of animals, public health and the environment. An environmental assessment will be prepared to address the requirements of the National Environmental Policy Act of 1969. The manufacturer, in agreement with the CVB, may conduct additional tests if needed to provide additional proof of the safety of the strain used. Prior to licensure, a target animal field safety study following product label directions must be conducted. The studies are generally performed on at least two serials and in at least three geographical locations. Animal numbers should take into account label recommendations such as age, site of administration, sex, breed, pregnancy, protection claims in neonates, and any other distinguishing features. If the vaccine is intended for administration to pregnant animals, additional data may be required. A live vaccine strain must be stable, i.e. should not revert to virulence during consecutive passages. Depending on the agent, recombination or genomic reassortment needs to be evaluated. Vaccines are produced within a specified number of passages from the master seed stock. Usually this is limited to five passages for virus vaccines and higher for bacterial vaccines. The safety of the vaccine strain at the lowest passage number is shown in animal studies using the most sensitive target animal/species. Aspects such as spread of the vaccine strain and dissemination in the vaccinated animal need to be taken into account where appropriate. Also, studies on the immunological functions may have to be carried out where the vaccine might adversely affect the immune response.

Efficacy
All tests are described in Title 9 of the Code of Federal Regulations and are based on use of the final product in poultry or non-poultry, and on product type, i.e. live or inactivated (see the next section on Safety). Data must be provided that support the efficacy claims. There are four predefined claims supported by the CVB: prevention of infection, prevention of disease, aid in disease prevention, and aid in disease control.

388

Rev. sci. tech. Off. int. Epiz., 26 (2)

Manufacturers are encouraged to interact with CVB when planning the protocol for these studies, which are required to be laboratory controlled studies, not field performance studies. Experimental product used in the studies must be prepared from the highest passage from the master seed stock, and if cell culture is used in manufacturing it must be prepared at the highest passage from the master cell stock allowed in the Outline of Production. The studies must be conducted at or below the minimum antigen level needed for efficacy, which is also specified in the Outline of Production.

Route of administration
Just as in the EU, studies must be performed to support the safety and efficacy claims of each route of administration (e.g. oral application, injection) and the claims must be clearly stated in the Outline of Production and on the label and circular.

Onset and duration of immunity


Special efficacy trials are designed around claims for onset and duration of immunity. Trials to test duration of immunity claims take the form of efficacy studies in which the animals are vaccinated according to the directions for use, and then challenged at a specified time, i.e. one year or three years post vaccination. Data from non-challenge studies (publications, studies not used to support licensure, etc.) must be pre-approved through the CVB before a manufacturer is allowed to make such claims.

Potency
As in the EU, the manufacturer must provide data that guarantees the immunising capability and thereby protective effect of a product. In vivo or in vitro testing can be used for this purpose. Both the CVB and manufacturers are moving away from animal testing toward in vitro tests. For release of finished product, a pre-calculated titre value is added to the minimum antigen level demonstrated for efficacy. For live virus vaccines, the titre required through dating must be at least 0.7 log10 greater than the titre used in the efficacy study. For live bacterial vaccines, the bacterial counts required through dating must be twice those used in the efficacy study. Expected loss in stability or via steps in production should be added to these values, normally 0.5 log10 to the live viral products.

Compatibility
For those vaccines used in combination, additional supporting data must be submitted. Combination products have two or more antigens in one vial or are prepared by mixing two or more separately licensed products in the field. Of course, the efficacy of each fraction must be proven, but a lack of interference must also be established between fractions. Challenge or serology models may be used. These studies may be used to recalculate the minimum potency level at which the product will be released.

Special requirements Registration of the label claim


In the USA, the licence is granted with a CVB approved Outline of Production, label, and circular. The CVB assigns a true name and a product code number that is used to differentiate the biological product from others. This true name must be listed prominently on all packaging components. The Outline of Production includes the history and test methods used to support the master seed stocks. The Outline includes the passage details, source of media used, in-house testing as well as serial release testing, expiration dating confirmation, efficacy confirmation, release titres, and description of final product containers, use of the product, and storage conditions. Changes to the Outline of Production must be pre-approved by the CVB.

Requirements for the registration of veterinary vaccines in Japan


Quality
Manufacture
Both the antigen and the vaccine must be manufactured according to GMP. A manufacturing licence must be obtained for each production facility where the antigen or vaccine is being produced or stocked for sale. An overseas production facility which intends to export vaccines to Japan must be accredited by MAFF in Japan in advance. This accreditation is renewed every five years. A marketing authorisation must be obtained by a marketing licence holder prior to sale of the vaccine in Japan. As a precaution against the risk of contamination with bovine spongiform encephalopathy strict regulation is applied to the use of any raw materials of ruminant origin for production of veterinary vaccines regardless of the intended target animal species. For example, milk and

Target animal
As with the EU regulations, clearly, it must be specified for which animal species the product is intended. In addition, the category must be stated, e.g. whether the product can be used safely in pregnant animals or is intended for specific use, such as for broiler chickens as opposed to breeder chickens or layer flocks. The CVB also grants an autogenous licence for inactivated viral or bacterial products. This type of product has a very special use in the flock or herd of origin.

Rev. sci. tech. Off. int. Epiz., 26 (2)

389

dairy products from the United Kingdom and Portugal cannot be used, and bovine serum and bovine serum products from the USA have to be certified as not originating from a TSE-affected bovine herd in the USA.

National assay
In Japan, each batch of a veterinary vaccine is tested by the National Veterinary Assay Laboratory (NVAL) for conformance to the respective monographs published in the Standards of Veterinary Vaccines for National Assay prior to release of the batch on the market. This national assay process requires the sponsor of a veterinary vaccine application to propose those testing methods which are easily practicable by the NVAL in the registration dossier. The Standards also stipulate general test methods that are used in the national assay. While some of these test methods have been globally harmonised thanks to the VICH initiative, some other test methods remain unique to Japan and the sponsor often carries out the test according to the method used in Europe or the USA and then repeats the same test according to the Japanese method (sterility test for example) when exporting a vaccine to Japan from the USA or Europe. MAFF is currently planning to introduce a full seed lot system in the near future and the national assay process will be simplified or eliminated depending on the type of vaccine to be tested.

large animal vaccines (swine, cattle and horses). In the case of poultry vaccines, reporting of non-specific mortality in the safety tests is not accepted unless the cause of death is shown to be irrelevant to the vaccine, whereas a certain number of non-specific deaths are accepted in the safety test in chickens in the USA and Europe.

Physico-chemical and biological properties


Physico-chemical properties such as morphology of the vaccine strain and biological properties such as virulence of the vaccine strain in various cell lines or laboratory animal species, and comparison with a standard reference strain should be included in the registration dossier. In addition, data on immunogenicity, growth characteristics and, where applicable, interference between antigens should be submitted. For a live vaccine, data on identification of the marker of attenuation and the strain as well as the marker stability, shed and spread, and reversion to virulence should be submitted. An outline of production is also required. Three pilot batches should be manufactured and tested according to the proposed specification and test methods.

Stability
In the stability test, three pilot batches are stored under the proposed storage conditions for the proposed shelf life period. Each individual vaccine sample should be within the proposed specification at all time points throughout the storage time, whereas a statistical approach is often used in the interpretation of the stability test in the USA and Europe. Stability after reconstitution of the vaccine, where applicable, and changes at room temperature should also be investigated.

Potency test
In Japan, traditionally an in vivo potency test has been required as part of the batch release control procedure of practically all veterinary vaccines, including live vaccines. This is usually performed by a serological technique such as neutralisation, haemagglutination inhibition or ELISA, but sometimes a challenge test is also performed. The potency test has to be correlated with the efficacy of the vaccine. Since this is not a requirement for a live vaccine in the USA or Europe, a sponsor usually has to develop a validated in vivo potency test for registration of a live vaccine in Japan.

Safety
In Japan, a target animal safety study requires that a group be given a recommended dose and another group be given an overdose equivalent to 100 times the recommended dose for live vaccines or 10 times the recommended dose for inactivated vaccines, administered in divided doses to avoid causing physical harm to the test animals. The study should normally use the final vaccine, i.e. the vaccine that is to be registered. Use of target animal safety data obtained with a maximum combination vaccine for the registration of a combination vaccine containing fewer antigens is generally not accepted, unlike in the USA or Europe. For a vaccine for food-producing animals containing adjuvant, depletion of adjuvant must be investigated in addition to normal histopathology. An appropriate withdrawal period is established based on depletion of the adjuvant and resolution of the lesions at the injection site. All the target animal safety studies must be conducted in accordance with the GLP guidelines.

Abnormal toxicity test


The abnormal toxicity test in mice and guinea pigs, or a toxicity limit test in either of these species, which is a modified version of the abnormal toxicity test, is required as part of the batch release control of all mammalian vaccines. The abnormal toxicity test is not routinely conducted in the USA or Europe.

Safety test
Safety testing in the target animal species is required in the batch release control of all veterinary vaccines except for

390

Rev. sci. tech. Off. int. Epiz., 26 (2)

Efficacy
Efficacy studies consist of basic studies supporting efficacy and field trials. Basic studies supporting efficacy should normally include: establishment of minimum effective antigen dose establishment of minimum protective antibody titre against the target disease determination of duration of immunity establishment of the relationship between the antigen dose and the antibody titre analysis of the relationship between local immunity and the protection induced comparison of response between different ages, breeds and routes of administration analysis of protective mechanism determination of the relationship between the maternally derived antibody and the protection induced. In addition, data must be presented on onset of immunity, the effect of booster vaccination and the effect of other vaccines that are likely to be administered concurrently. Field trials, usually at two or more locations in Japan, are required to provide safety and efficacy data from a minimum of 60 animals for a mammalian vaccine or 200 birds for a poultry vaccine. Usually, all animals, or just certain individual animals, have to be monitored periodically for serological responses to each antigen contained in the vaccine. Field trial data collected in foreign countries are accepted if the epidemiology and the trial protocol are similar, but even in this case, at least one local field trial is required. Field trials must be conducted in accordance with the GCP guidelines.

(FSC), which reports to the Prime Ministers Office on the possible impact on human health. The review by the FSC consists of several steps, including an invitation for public comment, and it usually lengthens the regulatory approval process by 6 to 12 months.

Recombinant vaccine
Detailed requirements for registration of a recombinant vaccine in Japan remain somewhat vague and the guidelines for application of the Cartagena Protocol to veterinary vaccines are still awaited. To date, no recombinant vaccine has been approved in Japan.

Requirements for the registration of veterinary vaccines in Australia


Australia is considered an advanced country from the point of view of registration of vaccines. Vaccine products are registered and their accompanying labels are approved by the government regulator prior to marketing. The basis of approval of vaccines is essentially similar to that in the EU or USA, but the process of registering differs in some respects.

Pre-marketing requirements
Australia maintains a national registration scheme for veterinary products which is administered by the federal government on behalf of the Australian States and Territories. All vaccines must be registered by the Australian Pesticides and Veterinary Medicines Authority (APVMA) in accordance with the Agricultural and Veterinary Chemicals Act 1994 and the Agricultural and Veterinary Chemicals Code Act 1994 and other supporting legislation. Once APVMA registration is granted, state by state registration is not required. All registered vaccines must be manufactured in a facility approved by the APVMA under the Manufacturers Licensing Scheme. Mutual Recognition Agreements exist between the APVMA and some other countries and others are recognised under the Overseas GMP Scheme. Although the APVMA has its own format for the presentation of data for the registration of vaccines, the content is essentially the same as that for other advanced countries (it is particularly similar to the data required in the EU). Data presented in the format of another country is generally acceptable so long as effective cross-referencing is provided in an Australian format dossier. The APVMA does not accept phased filing in the same way as the USDA. Australia has so far adopted all VICH guidelines without

Special requirements
Generic vaccine
In the case of a vaccine that is considered equivalent to those vaccines that are already registered in Japan (e.g. a vaccine containing a different strain but having similar biological properties, similar composition, similar dosage and administration instructions and the same indication) a sample of the vaccine has to be submitted to the NVAL for confirmation of equivalence. Once such confirmation is obtained from the NVAL, the registration process subsequently becomes much simpler compared with that of a new vaccine.

Food animal vaccine


The dossier on a new food animal vaccine is reviewed not only by MAFF but also by the Food Safety Commission

Rev. sci. tech. Off. int. Epiz., 26 (2)

391

any modification in the case of vaccines. It is a default requirement that local efficacy data is presented for vaccines, but this is negotiable with the APVMA on the basis of scientific argument; this requirement is more strongly enforced for economic animals than for companion animals. A summary of each application for registration is published on the APVMA website at the start of the review process together with, for some applications, a list of supporting trial data for the purposes of establishing data protection. All new active ingredients (including new antigens) are subject to public comment before registration is finalised. At the time of registration, a summary is published of advice on the application received from other authorities or external assessors. Imported vaccines or locally produced vaccines containing animal-origin materials of overseas origin require prior clearance by the Australian Quarantine and Inspection Service (AQIS) (see the section on Biosecurity below). Vaccines derived from biotechnology require prior clearance or exemption by the Office of the Gene Technology Regulator. The APVMA publishes guidelines for the time frame for registration review to which it aims to adhere. The final act in the registration process is the submission and approval of the final product label, which must conform to local guidelines.

All registrants are required to maintain a pharmacovigilance programme and lodge annual or, in some cases, real-time reports. An annual renewal fee for each registered vaccine is payable to the APVMA, together with a levy on sales. Periodic re-submission of data is not a requirement unless requested by the APVMA under its Existing Chemical Review Program.

Biosecurity
Australia enjoys a fortunate freedom from a number of infectious animal diseases and guards this status jealously. Its strict quarantine regulations and policies are established by Biosecurity Australia and policed by AQIS. An in vivo permit must be obtained from AQIS for each vaccine, or animal-derived raw material intended for use in a vaccine, imported to Australia. All imports are checked at the border for correct documentation. Application for such a permit requires submission of a substantial body of biosecurity data and a potentially prolonged review period (no published time frame). Permits issued for the import of vaccines are generally for a finite period and usually require a compliance audit to be conducted by AQIS on each serial imported. The stringency of these requirements should not be under-estimated.

Acknowledgements
The authors would like to thank Dr Kent MacClure, Animal Health Institute, Washington DC, who kindly undertook the proof-reading of the text of this paper.

Post-marketing requirements
Finished-product testing and release is carried out by the licensed manufacturer. The maintenance of appropriate records is a requirement of GMP compliance and is subject to periodic inspection. There is no requirement for batch release by the regulatory authority (but see Biosecurity below).

392

Rev. sci. tech. Off. int. Epiz., 26 (2)

Exigences rglementaires lies lagrment des vaccins


P.G.H. Jones, G. Cowan, M. Gravendyck, T. Nagata, S. Robinson & M. Waits
Rsum Les vaccins sont lun des principaux outils pour prvenir et matriser les maladies animales. Ds lors, il est dune importance capitale que les vaccins soient utiliss conformment aux prescriptions du Code sanitaire pour les animaux terrestres (Code terrestre) de lOrganisation mondiale de la sant animale (OIE) aprs avoir t avaliss par lorganisme charg de dlivrer les autorisations de mise sur le march dans le pays/la rgion o les vaccins doivent tre utiliss, en respectant les critres fondamentaux de qualit, de scurit et defficacit. Les auteurs examinent et comparent les exigences rglementaires en vigueur pour les vaccins vtrinaires dans les principales rgions du monde, en soulignant les similitudes ainsi que les diffrences constates. Ils dcrivent galement les derniers progrs accomplis dans le domaine de lharmonisation des exigences relatives aux tests, grce la Coopration internationale sur lharmonisation des exigences techniques applicables lenregistrement des mdicaments vtrinaires (VICH). Cet article fournit des orientations utiles pour tous ceux qui soccupent de recherche et de dveloppement de vaccins dans le monde, tout en apportant aux personnes charges de la prvention et de la lutte contre les maladies animales la garantie que les vaccins vtrinaires, ds lors quils sont dment autoriss et utiliss suivant le mode demploi prescrit, sont srs et efficaces. Mots-cls Coopration internationale sur lharmonisation des exigences techniques applicables lenregistrement des mdicaments vtrinaires (VICH) Dispositions rglementaires Efficacit Harmonisation Qualit Scurit Vaccin vtrinaire.

Rev. sci. tech. Off. int. Epiz., 26 (2)

393

Requisitos de las reglamentaciones relativas a la autorizacin de comercializacin de vacunas


P.G.H. Jones, G. Cowan, M. Gravendyck, T. Nagata, S. Robinson & M. Waits
Resumen La vacunacin es uno de los instrumentos existentes ms eficaces para prevenir y controlar las enfermedades animales. Por consiguiente, es indispensable que su administracin se conforme a las disposiciones del Cdigo Sanitario para los Animales Terrestres (denominado tambin Cdigo Terrestre) de la Organizacin Mundial de Sanidad Animal (OIE) y haya sido autorizada por el organismo habilitado para la concesin de licencias de comercializacin del pas o regin interesados, de conformidad con los criterios clave relativos a la calidad, inocuidad y eficacia. Los autores exponen las exigencias sobre las vacunas veterinarias de las reglamentaciones en vigor en las principales regiones del mundo de manera pormenorizada y comparada, destacando sus similitudes y diferencias. Tambin resean los ltimos avances realizados por la Cooperacin Internacional para la Armonizacin de los Requisitos Tcnicos relativos al Registro de Medicamentos de Uso Veterinario (VICH) en la armonizacin de los requisitos que han de atenderse durante las pruebas. Este artculo, que constituye una valiosa gua para quienes trabajan en la investigacin y el desarrollo de vacunas en todas partes del mundo, demostrar a quienes participan en la prevencin y el control de las enfermedades animales que las vacunas de uso veterinario, una vez aprobadas y administradas conforme a las instrucciones del fabricante, son inocuas y eficaces. Palabras clave Armonizacin Calidad Cooperacin Internacional para la Armonizacin de los Requisitos Tcnicos relativos al Registro de Medicamentos de Uso Veterinario (VICH) Eficacia Inocuidad Requisito reglamentario Vacuna veterinaria.

Rev. sci. tech. Off. int. Epiz., 2007, 26 (2), 395-414

Regulatory issues surrounding the temporary authorisation of animal vaccination in emergency situations: the example of bluetongue in Europe
C. Saegerman (1), M. Hubaux (2), B. Urbain (3), L. Lengel (2) & D. Berkvens (4)
(1) Department of Infectious and Parasitic Diseases, Epidemiology and Risk Analysis Applied to Veterinary Sciences, Faculty of Veterinary Medicine, University of Lige, Boulevard de Colonster, 20, B42, B-4000 Lige, Belgium (2) Directorate-General for Animals, Plants and Foodstuffs, Federal Public Service of Public Health, Food Chain Safety and Environment, Eurostation II 7th floor, Place Victor Horta 40 box 10, B-1060 Brussels, Belgium (3) Marketing Authorisation Veterinary Unit, Federal Agency for Medicinal and Health Products, Eurostation II 8th floor, Place Victor Horta 40 box 40, B-1060 Brussels, Belgium (4) Department of Animal Health, Unit of Epidemiology and Applied Statistics, Prince Leopold Institute of Tropical Medicine, Nationalestraat 155, B-2000 Antwerp, Belgium

Summary A marketing authorisation for a veterinary vaccine is granted after the quality, safety and efficacy of the product have been assessed in accordance with legal standards. The assessment includes complete characterisation and identification of seed material and ingredients, laboratory and host animal safety and efficacy studies, stability studies, and post-licensing monitoring of field performance. This assessment may not be possible during the emergence of a new animal disease, but several mechanisms exist to allow for the availability of products in an emergency animal health situation, e.g. autogenous biologics, conditional licences, experimental and emergency use authorisations, the importation of products in use elsewhere in the world and pre-approved vaccine banks. Using the emergence of bluetongue in northern Europe as an example, the regulatory issues regarding the temporary authorisation of animal vaccination are described. Several conditions must be fulfilled before a temporary authorisation can be granted, e.g. inactivated vaccines should be used in order to exclude reversion to virulence and reassortment between vaccine viruses and/or field strains of the bluetongue virus; decision-making must be supported by scientific evidence and risk analysis; there must be a complete census of the susceptible animals that were vaccinated; vaccination protocols must be adhered to and there must be a scheme allowing for registration, delivery and follow-up of vaccination, and monitoring, analysis and, possibly, adjustment of field use of the vaccination. This temporary authorisation must be replaced by a full authorisation as quickly as possible. Keywords Animal health Emerging disease Regulatory issues Temporary authorisation Vaccination Vaccine.

396

Rev. sci. tech. Off. int. Epiz., 26 (2)

Introduction
Each and every animal disease control and/or eradication programme is based on two general principles: a) the control of infected herds through the application of corrective control measures b) the protection of infection-free herds through preventive measures. The identification of individual animals and herd registers are two essential prerequisites before these principles can be applied. The herd is the epidemiological unit of interest in disease control programmes. The above indicates that different strategic options can be adopted, separately or conjointly, firstly to decrease the prevalence to an acceptable level (disease control) and secondly to eliminate the remaining clinical or sub-clinical infection foci (disease eradication). The overall strategy consists of one or more of the following steps: a) generalised compulsory vaccination of the entire susceptible population (medical prophylaxis); b) slaughter of known infected animals (test and slaughter) combined with a selective vaccination programme, including either only the subclass of animals at risk (e.g. young naive animals) or all animals in a limited area depending on the prevalence (medico-sanitary prophylaxis) c) stamping out of animals known to be infected or exposed (sanitary prophylaxis). Adequate compensation for the animals slaughtered is an unavoidable requirement for success of the programme (86). The choice of the different steps to be undertaken depends on a certain number of considerations, amongst them: infection prevalence in the different susceptible animal species (and human clinical incidence in the case of a zoonosis) the structure and management of the livestock sector the availability of a national reference laboratory and regional laboratories the capacity of the Veterinary Services to follow up the programme and to control livestock movements and the cooperation between the Veterinary Services and the private sector in the implementation of such a programme the involvement of political decision-makers and their willingness to support an uninterrupted effort over many years, sometimes several decades the availability of financial resources and the capacity to mobilise extra funds the coordination and collaboration between the Health and Agriculture Ministries when planning the programme

the level of involvement of farmers organisations, which ideally should be convinced of the benefits of the exercise before the start of the programme (5, 86). Each party concerned must participate in developing and implementing disease control programmes, in financing these programmes and in taking responsibility for their implementation, and each party must be rewarded for their achievements. This is the principle of joint decisionmaking, joint financing and joint accountability (44). A prerequisite for any programme is the implementation of an efficient animal disease surveillance network, capable of monitoring the progress made, and being able to adjust the strategy when required (55). Every programme must therefore be evaluated regularly and performance indicators must be set beforehand (87). Moreover, the strategy must be adjusted not only according to the epidemiological situation, but also in the light of new scientific knowledge (86). In the case of countries that are free of a disease vaccination is usually not practised, but the option to use vaccination in an emerging disease situation is still available (101). Moreover, policies involving mass slaughtering being less and less popular, there is a tendency to use so-called marker vaccines together with a companion diagnostic test (33, 76). A marketing authorisation for a veterinary vaccine is granted after the quality, safety and efficacy of the product have been assessed in accordance with legal standards (18, 22, 48). Even when using an accelerated assessment procedure it can still take a long time before authorisation is granted. However, Member States are authorised to use vaccines without a marketing authorisation in an emergency situation. Using the example of the emergence of bluetongue (BT) in Europe, the advantages and disadvantages of this temporary authorisation permit are discussed and solutions to improve this unusual situation are proposed.

Emergency situations
Several definitions of an emerging disease coexist (9, 13, 69, 77) but they all have a common denominator. An emerging disease is a disease of which the true incidence increases in a significant way in a given population, in a given area and during a given period, in comparison with the usual epidemiological situation of this disease (103). This increase in true incidence is due to several factors, such as the evolution or the modification of a pathogenic agent or an existing parasite, which results in a change of host, of vector, of pathogenicity or strain (68, 116). Specific social, ecological, climatic, environmental and

Rev. sci. tech. Off. int. Epiz., 26 (2)

397

demographic factors precipitate the emergence of a disease (69, 97, 113, 114), but it is difficult to establish a ranking of causes or of mechanisms (81). There are several models for understanding emerging risks, they include: a) the convergence model for zoonotic diseases (53) b) the pan European pro-active identification of emerging risks in the field of food production model (PERIAPT) concerning emerging risk in the food chain (108) c) the generalised model for rare events (85, 87). The example of the recent BT epidemic in northern Europe is treated more in detail hereafter (24, 43, 102, 104). Bluetongue virus (BTV) is the prototype species of the genus Orbivirus in the family Reoviridae. The viral genome consists of 10 double-stranded RNA segments that encode for four non-structural (NS1, NS2, NS3 and NS3A) and seven structural (VP1-VP7) proteins (82, 111). Currently, there are 24 known serotypes of BTV worldwide (63). However, a genetic diversity of BTV exists and it is a consequence of both drift (i.e. point mutations) and shift (i.e. reassortment of individual BTV gene segments). Serotypes 1, 2, 3, 4, 6 and 10 are known to have a high pathogenic index and high epidemic potential (18). Bluetongue is a notifiable disease of the World Organisation for Animal Health (OIE), and is thus of serious socio-economic concern and of major importance in the international trade of animals and animal products (18). The BTV is transmitted between its ruminant hosts exclusively by bites of Culicoides midges (60, 61). Transmission is limited to seasons during which adult insects are active. Bluetongue is thought to infect all known ruminant species. However, severe disease usually occurs only in certain breeds of sheep and in some species of deer (57, 74, 100). Both cattle and goats suffer from unapparent infection, but probably serve as important reservoirs of the virus for sheep and wild ruminants (57, 109). Common clinical signs include fever, salivation, abundant nasal discharge, oedema (particularly in the head region), congestion, ulcerations of the oral mucosa, lameness, depression and, sometimes, cyanosis of the tongue (45, 57). The mortality rate and the severity of the clinical signs seem to be dependant on factors such as the breed and age of the animal infected (older age groups being more susceptible) and the type and strain of the virus (62). Because BTV infection is not contagious, meat and dairy products pose no hazard for the spread of the pathogen in ruminants. However, blood and other biologicals for cell culture or in vivo use are a potential risk for spread (20, 118). The genus Culicoides comprises 1,260 species, of which 30, to a greater or lesser extent, are involved in the

transmission of orbiviral diseases injurious to livestock almost worldwide. Until recently, Culicoides imicola (Kieffer) was believed to be the only vector of BTV in both Africa and southern Europe (43), but it is now known that other newly identified (and as yet unidentified) vectors are involved. Other possible factors that have contributed to the spread of BTV include animal migration and importation, extension in the distribution of its major vector, Culicoides spp., the apparent ability of the virus to overwinter in the absence of adult vectors, and its occurrence in healthy reservoir hosts, such as cattle and some wild ruminants (18, 79, 99). In 2004, bluetongue was reported in nine countries in the world (115) (Fig. 1). Between 1998 and 2004, when there were incursions of BTV into the Mediterranean Basin, the Balkans and beyond (Table I), the disease penetrated into areas where C. imicola does not occur, thus incriminating novel vectors (3, 8, 12, 42, 62, 64, 119). This was confirmed subsequently when the causative virus was isolated from mixed pools of the two species Culicoides obsoletus (Meigen) and Culicoides scoticus (Downes and Kettle) collected in central Italy (92, 93) and from Culicoides pulicaris (Linnaeus) in Sicily (11).

Fig. 1 Bluetongue throughout the world in 2004 Source: Handistatus II (http://www.oie.int/hs2/report.asp?lang=en) (115)

Bluetongue appeared for the first time in the north of Europe (Fig. 2) following a heat wave and strong rains, and can be defined as being an emergent disease in this zone (116). After the first notification on 17 August 2006 (19), more than 1,200 cases of BT were entered in the European Commissions Animal Disease Notification System (http://ec.europa.eu/food/animal/diseases/adns/index_en. htm). In this area, BTV was isolated from Culicoides dewulfi (Goetghebuer) (58), an indigenous European Culicoides species (Fig. 3) thereby increasing the risk of transmission over larger geographical regions (42, 79).

398

Rev. sci. tech. Off. int. Epiz., 26 (2)

Table I Outbreaks of bluetongue in Europe in the period 1998-2004 (7, 62, 75, 115)
Country Albania Bosnia-Herzegovina Bulgaria Croatia Cyprus Former Yugoslav Republic of Macedonia France (Corsica) Greece Italy Kosovo Portugal Serbia and Montenegro Spain Turkey
ND: no data recorded a) Probably serotype 4

Year of first outbreak 2002 2002 1999 2001 2003 2001 2000 1998 2001 2004 2001 2000 1999

Serotype(s) of bluetongue virus 9 9 9 9, 16 16 9 2 1, 4, 9, 16 2, 9, 16 9 ND 9 2 9, 16


(a)

Main vector(s) identified Culicoides obsoletus, C. pulicaris ND C. obsoletus, C. pulicaris C. obsoletus, C. scoticus C. obsoletus, C. pulicaris ND C. imicola, C. pulicaris, C. obsoletus C. imicola, C. obsoletus C. imicola, C. obsoletus, C. pulicaris ND C. imicola, C. obsoletus, C. pulicaris ND C. imicola, C. obsoletus, C. pulicaris C. imicola

Black circle (Belgium; n = 460) Red circle (Germany; n = 457) Yellow circle (Netherlands; n = 306) Blue circle (France; n = 5)

Fig. 2 Bluetongue outbreaks (serotype 8) in northern Europe between 17 August and 25 October 2006 Source: European Commissions Animal Disease Notification System, 2006

Rev. sci. tech. Off. int. Epiz., 26 (2)

399

Vaccination as a strategic option


Strategies for controlling BT range from control measures in areas free from the disease to systematic vaccination in BT endemic areas. In areas free from BT but where Culicoides spp. are present, any introduction of live animals from infected countries should be controlled bearing in mind the maximum duration of viraemia (e.g. quarantine for at least 60 days is recommended by the OIE). When importing ruminant semen or embryos/ova from suspected infected areas, donors are kept in quarantine and protected from potential vectors for at least 60 days prior to collection, or donors are tested according to the protocol outlined in the OIE Terrestrial Animal Health Code (118). In recently infected areas, eradication of BTV is theoretically possible by adopting a range of methods, including some or all of the following: animal movement restrictions This recent finding is an important new epidemiological event, because previously most BT outbreaks were linked to C. imicola. Culicoides dewulfi has been reported throughout the Palaearctic area (52, 105). This indicates that the disease could now remain in the rest of Europe where cattle and horses are kept (C. dewulfi is known to breed in cattle dung and horse dung), with the risk of more cases of orbiviral diseases such as BT occurring during the following spring when the vector activity is high (58). The global distribution of the BTV endemic areas has traditionally been accepted to be between the latitudes of approximately 50N and 35S (17, 54, 61, 80). There is now enough scientific evidence to show that it has spread further north, to 53N (58, 117). serological/virological surveys and slaughtering of infected and potentially infected animals vaccination and/or vector control (e.g. use of insecticide or keeping animals indoor during periods of high vector activity) (Table II). In areas where BTV is enzootic, controlling the vectors is usually considered to be impossible and any reduction in their numbers is transient (56). Nevertheless, attempts to control vector populations have been implemented using chemical repellents and/or insecticide treatment (6, 91, 98, 112). With regard to prophylaxis, possibly the best way to control clinical BT outbreaks in endemic areas is to

Fig. 3 Gravid female of Culicoides dewulfi isolated near outbreaks in Belgium in 2006 (Reginald De Deken and Maxime Madder, Institute of Tropical Medicine, Antwerpen, Belgium)

Table II Control, safeguard and preventive measures against bluetongue applied in Eastern Europe, 1998-2004 (adapted from 62, 75)
Control measures Modified stamping out policy, i.e. killing and destroying clinically affected animals (viraemic animals) to prevent them acting as a source of virus for vector insects Vector control measures by means of insecticides and/or insect repellents Intensive clinical surveillance Serological surveillance Virological surveillance Entomological surveillance Safeguard measures Protection zone Surveillance zone Restricting the movement of animals and germinal products to prevent new foci or infection Preventive measures Vaccination

400

Rev. sci. tech. Off. int. Epiz., 26 (2)

Table III Available bluetongue vaccines (according to 15, 18, 34, 94, 119)
Type (form) Inactivated (suspension) Species Sheep (a) Valences Monovalent Polyvalent Sheep (b) Monovalent Strains 2&4 16 & 9 in development 2 Istituto Zooprofilattico Sperimentale dellAbruzzo e del Molise, G. Caporale, Teramo, Italy Attenuated (freeze-dried) Sheep (c) Monovalent Series of three separate injections with different serotypes in each bottle (d) Polyvalent Sheep Sheep Sheep and goats Monovalent Monovalent Monovalent (Can be produced on request (e)) 4 4 10 Central Veterinary Control & Research Institute, Ankara, Turkey Biopharma, Rabat, Morocco Colorado Serum Co., Denver, Colorado, Unites States of America
a) Indications for efficacy in cattle (94) b) Immunogenicity was assessed by subcutaneously inoculating sheep, goats and bovines but only vaccinated sheep were challenged (15) c) Indications for efficacy in cattle (65) d) The vaccine is presented as: A (1, 4, 6, 12 & 14 serotypes); B (3, 8, 9, 10 &11 serotypes) and C (2, 5, 7, 13, & 19 serotypes) e) Production of these vaccines requires two months for production and quality control (45)

Producer Mrial SAS, Lyons, France

Onderstepoort Biological Products, Onderstepoort, South Africa

Table IV Advantages and disadvantages of a bluetongue vaccination strategy using attenuated vaccines
Advantages Provision of an alternative to slaughter policies, which are becoming less and less popular Reduction of the intensity and duration of viraemia following contact between vaccinated animals and wild-type strains of the bluetongue virus (progressive reduction of BTV transmission) Decrease in direct losses (number of outbreaks and diseased animals, mortality) Decrease in indirect losses (exportation of live animals) 39 39, 41, 45 39, 41 Reference 76 Disadvantages Risk of disease Risk of reversion to virulence Risk of reassortment of genome segments between vaccine and field viruses Risk of foetal abnormalities (a) Risk of introducing exotic serotypes into an ecosystem if using polyvalent vaccines (b) Risk of inappropriate coverage in susceptible species (at least 80% of the population) (c) Risk of inappropriate coverage of all susceptible domestic ruminant species (not only sheep) Costs of the implementation and monitoring of the vaccination programme (serological, virological and entomological surveillance)
a) This disadvantage is easily resolved because the manufacturer does not recommend this vaccine for use in pregnant animals (18) b) The most commonly used vaccines are those produced by Onderstepoort Biological Products Ltd, South Africa. A monovalent bluetongue vaccine is only produced on special request and its production and quality control take two months (45) c) This disadvantage is easily resolved by proper implementation and monitoring of the vaccination programme BTV: bluetongue virus

Reference 110 18 90, 95, 96 32 59 39 40, 59 119

Rev. sci. tech. Off. int. Epiz., 26 (2)

401

vaccinate susceptible animals and limit contact between susceptible hosts and insect vectors (18). Live attenuated vaccines have been available for many years (18, 94, 110). Inactivated vaccines have been developed, but currently are less used (16) and recombinant vaccines (83) are still under development (56) (Table III).

Attenuated vaccines
In South Africa, around eight million doses of attenuated vaccines are used annually (18, 20). These vaccines are cheap, easy to produce and effective in controlling clinical outbreaks of BT in areas of endemic disease (18, 20). They replicate in sheep without causing significant clinical effects and provide protection (humoral and cellular responses) against challenge with virulent virus of the same serotype (70). The vaccines most commonly used are those produced by Onderstepoort Biological Products Ltd, South Africa (45). Sheep are vaccinated three times every year at three-week intervals with vaccines containing five serotypes each. As one of the side effects of these vaccines is abortion, the last vaccination round has to be carried out three weeks before the mating period (18). Very few solid data have been published on the safety and efficacy of these vaccines (18, 45, 50, 65, 66, 67). Potential safety issues associated with attenuated vaccines, although probably not likely to occur often (no quantitative studies), are the following: risk of foetal abnormalities risk of reassortment risk of reversion to virulence risk of transmission of vaccine strains by vector midges (Table IV). Some attenuated BT vaccine strains were shown to be teratogenic in sheep when administered during the first half of pregnancy (32). Attenuated BTV vaccine strains may be responsible for spontaneous cases of BTV-induced malformation in both sheep and cattle, but it is noted that different serotypes of BTV differ in their pathogenesis, transmissibility and growth characteristics (18). This risk must be avoided by clear indication on the product label that pregnant ewes must not be vaccinated in the first half of pregnancy. The reassortment of genome segments between vaccine and field viruses has been demonstrated in the laboratory, but only rarely has it been reported to occur in the field (14, 46, 71, 89, 90, 95, 96). The reassortment often involves segments of different serotypes of the vaccine (110). The effect of such reasortmens in dually infected vectors and/or hosts could result in altered virus characteristics and virulence (110). However, the risk of

reassortment in the vertebrate and/or invertebrate hosts will be minimised if the interval between the recommended vaccination period (late winter, early spring) and the BT season (especially summer) is respected; this would make the incidence of co-circulating vaccine and virulent wild-type viruses unlikely. The number of possible reassortments in the case of BTV, which has ten segments, increases with the number of cocirculating serotypes (e.g. 1,024 for two serotypes [210] and 59,049 for three serotypes [310]) (18). Reassortment occurred during the recent 1998-2005 BTV outbreaks in Europe (56). The release and transmission of attenuated virus into the environment may also result in a reversion to virulence through reassortment with a wild-type strain (18). The possibility that insects could acquire vaccine virus by feeding on vaccinated animals and transmit it to sheep or cattle cannot be eliminated (18, 70). The minimum viraemia in infected animals necessary for successful vaccine virus dissemination by biting midges remains to be determined, although Fu et al. (37) have shown that only 12% of competent vector Culicoides sonorensis that were fed on blood containing 106-7 TCID50/ml were able to become persistently infected and to transmit the virus through saliva (42). However, the bite of only one infected competent midge vector is sufficient to induce transmission to susceptible ruminants (72, 73). Moreover, the vaccine virus serotype-2 was also detected recently in areas not included in the vaccination campaign (30). The possible circulation of this vaccine virus poses problems that have to be considered if vaccination with attenuated strains is the strategy of choice, particulary if the zone where vaccination is carried out is bordered by territories free from disease/infection (30). Lastly, attenuated vaccines were traditionally produced using eggs, so allergy cases could not be excluded (2, 88). Currently they are produced in cell culture (18, 20) and this risk decreases.

Inactivated virus vaccines


To avoid the problems listed above, inactivated vaccines have been developed (16). Recently, two new inactivated vaccines against BTV2 and BTV4 were developed and similar vaccines against other BTV serotypes are in the final stages of development (94). However, limited reports are currently available on the effectiveness of these new vaccines in the field (15, 119).

Recombinant vaccines
Recombinant DNA technology has provided novel approaches to developing intrinsically safe vaccines. This

402

Rev. sci. tech. Off. int. Epiz., 26 (2)

technology involves the synthesis of immunogenic proteins and particles that elicit highly protective immune responses. Protein engineering systems have been utilised to synthesise individual bluetongue virus proteins and core- (single coat) and viral-like (double coat) multiprotein structures (CLPs, VLPs). These engineered particles mimic the virus particles, but do not contain any genetic materials (35, 36, 78). These recombinant vaccines have been tested successfully in vitro and under experimental conditions (83, 84, 107). Together with a suitable diagnostic test to detect antibodies against viral proteins not present in the vaccine a new BTV vaccine would allow the distinction between vaccinated and infected animals (marker vaccines) (20). In addition, deliberate release of any organism containing recombinant DNA into the environment should be subject to review and approval by competent authorities (33). It is necessary to develop an inactivated vaccine or a vaccine based on more advanced technology that would not only have the potential to effectively control the disease but that would also facilitate trade, because the virus circulating in live animals can be detected (117). Because of the small commercial market, vaccine manufacturers may need some public funding to achieve this objective (20).

an emergence (e.g. the considerable cost of the quantitative real-time reverse transcription polymerase chain reaction [Rt-RT-PCR] for the detection of the BTV).

Regulatory issues surrounding the temporary authorisation of animal vaccination


A marketing authorisation for a veterinary vaccine is granted after an assessment of the quality, safety and efficacy of the product has been carried out in accordance with legal standards (18, 27, 29, 48). In order to fulfil the necessary criteria, data from all phases of the product development are evaluated against these key elements. Under the standard licensing process, the evaluation includes: complete characterisation and identification of seed material and ingredients laboratory and host animal safety and efficacy studies stability studies post-licensing monitoring of field performance (48).

Creating and developing an early warning system for emerging animal diseases
One objective of the vaccination strategy in endemic BT areas is to reduce the intensity and duration of viraemia in susceptible animal populations (39). To resort to vaccination requires a fast, argued and concerted decision. Any delay in the detection of the disease reduces the anticipated vaccine protection. Indeed, without early detection, emergence can pass unperceived unless amplification of viral multiplication and transmission reveals it. Detection is often too late from the point of view of risk control, particularly if the disease, before expressing itself clinically, passes through an incubation period allowing transmission to the sensitive population by means of vectors (known or unknown) present in the environment (4) and via the sale of animals outside contaminated areas. The improvement of identification tools and an increase in the speed of detection are essential (25). The clinical approach to the detection of emergence must be favoured because for a new disease no laboratory test is available to ensure its detection at the moment of emergence (87). In addition, for exotic diseases, the cost of specific analyses can be prohibitive in view of the need for immediate detection of

This comprehensive evaluation may not be possible during the emergence of a new animal disease, but several mechanisms exist to allow for the availability of products in an emergency animal health situation, including: autogenous biologics conditional licenses experimental and emergency use authorisations the importation of products in use elsewhere in the world pre-approved vaccine banks (Table V) (27, 29, 31, 47, 48, 49). Even when using the accelerated assessment procedure and conditional marketing authorisation procedure for a new medical product it can take up to five months to obtain an opinion concerning the granting of a European authorisation (26). Even in an emergency situation it can still take a long time. However, Member States are still authorised to use vaccines without a marketing authorisation in an emergency situation and, in this case, the European Commission must be informed (27, 29). This procedure has been used successfully for bluetongue in the Mediterranean endemic area (8, 21, 39, 119). The vaccination strategy was mainly based on the use of the monovalent or polyvalent attenuated vaccines produced by

Rev. sci. tech. Off. int. Epiz., 26 (2)

403

Table V Legislation governing the mechanisms which allow for the use of vaccines without marketing authorisation in the United States of America and in the European Union in the case of an emerging disease (adapted from 48)
Mechanism Approval of experimental products (a) United States of America Title 9 of the Code of Federal Regulations (b) Part 103.3 (porcine reproductive and respiratory syndrome) Use of autogenous biologics Attenuated Inactivated No specific legislation Title 9 of the Code of Federal Regulations Part 113.113 (c) (infectious salmon anaemia virus) Conditional Product Licences
(d)

European Union No specific legislation

Directive 2001/82/EC, article 4 (27) No specific legislation

Title 9 of the Code of Federal Regulations Part 102.6 (West Nile virus)

Regulation 2004/726/EC, article 39 (centralised procedure) (29) Directive 2001/82/EC, article 26 (other procedure) (27)

Importation of products in use elsewhere in the world Vaccine bank (f) Other (h)
(e)

Title 9 of the Code of Federal Regulations Part 104 (rabbit calicivirus)

Directive 2004/28/EC, article 8 (28)

No specific regulation (foot and mouth disease) Title 9 of the Code of Federal Regulations Part 106.1

Decision 2001/433/EC, article 3 (g) (bluetongue) (21) Directive 2004/28/EC, article 8 (28)

a) Experimental production, distribution and evaluation of biological products may be authorised prior to licensing under very specific circumstances b) Title 9 of the Code of Federal Regulations (http://www.aphis.usda.gov/vs/cvb/html/cfr.html) (106) c) According to this act, they are prepared from cultures of micro-organisms which have been inactivated and are non-toxic under certain circumstances d) Conditional licences are authorised under very specialised circumstances to meet an emergency condition, limited market, local situation, or other special circumstance. Licences are issued under an accelerated procedure which assures purity, safety and efficacy of the products involved e) Permits for biological products imported into the considered country may be authorised for a variety of purposes. In each case, a separate veterinary biological product permit is required for each shipment of biological product to be imported f) No specific regulations exist for the creation and maintenance of a vaccine or seed bank g) Under very specific circumstances, biological products may be exempted from one or more of the requirements. These circumstances are warranted if products will be used by or under the supervision or control of the competent authority in the prevention, control or eradication of animal diseases in connection with an official programme or an emergency animal disease situation, or an experimental use of the product by the Authority h) In general, European legislation concerning the disease must specify the use of vaccination. In this case, the European Commission can propose the constitution of a vaccine bank with a proposal for a European decision

Onderstepoort Biological Products Ltd (South Africa) and to a lesser extent on the inactivated vaccine produced by Mrial SAS in Lyons, France (8, 39, 119). Epidemiological surveillance of BT in this vaccination context requires the following tasks: a) clear communication all along the process

h) monitoring, analysis and possible adjustment of field implementation of the vaccination. Each of these points is described in more detail below. This information is also useful for replacing the temporary authorisation by a complete authorisation as quickly as possible.

Clear communication throughout the process


b) a description of the prevalent epidemiological situation c) the choice of an appropriate national response strategy d) the choice of an appropriate vaccine e) verification of the quality of the vaccine used f) strict adherence to the vaccination protocol and initiation and maintenance of a system allowing registration, delivery and follow-up of vaccination g) evaluation of the necessary level of vaccination coverage Communication is the rational process of conveying a representative picture of objects or situations when having to perform a concerted action with several partners (51). Several levels of communication should be maintained (i.e. inside and outside of the epidemiological network). Concerning international trade, clear and immediate communication in the framework of animal disease notification is essential to limit the spread of the disease (e.g. the OIE World Animal Health Information System) and to ensure mutual trust between countries. Clear communication about uncertainties in all phases of the decision process is also important.

404

Rev. sci. tech. Off. int. Epiz., 26 (2)

Describing the prevalent epidemiological situation


A description of the true epidemiological situation is a prerequisite before any action can be undertaken, because this knowledge is needed to choose the appropriate strategic option(s), including vaccination. This description requires the incorporation of detailed information concerning the following: suspected and confirmed clinical cases of BT (e.g. reason for the visit, date of suspicion, date of first infection, animal species involved, clinical signs, laboratory diagnosis) outbreaks (e.g. species concerned, number and size of herds, location of herds, mortality rate, morbidity rate) vector (e.g. date and location of trapping, number of Culicoides identified) virus activity (e.g. date and location of sampling, number of animals sampled, number of animals at risk and number of animals seroconverted) boundaries and subdivision boundaries geography and climate (e.g. vegetative index, temperature, rainfall) (10, 41, 119).

product, its availability and cost, and consequences of vaccination. If a vaccinated animal cannot be differentiated from an infected animal, further animal movement controls may need to be considered (33).

Choosing an appropriate vaccine


The choice of vaccine should be made taking into account the epidemiological situation of BT in the country or territory concerned (e.g. BTV serotype(s)) and in accordance with current knowledge about the advantages and disadvantages of the bluetongue vaccination strategy (Table IV) and the availability on the world market of the vaccines being considered (94) (Table III). Information concerning the quantity of BT vaccines deemed necessary in the case of emergency vaccination should be available in the contingency plan of each Member State (23).

Verifying the quality of the vaccine used


The quality of the vaccine used must be verified. Results of experimental infection with the chosen vaccine and subsequent challenge with the field strain BTV should be collected. Information concerning the safety and efficacy of the vaccine is required and help in the interpretation of serology and virology results after vaccination may be necessary (45, 67).

Choosing an appropriate national response strategy


Risk assessment is a tool advocated by the World Trade Organization in the context of trade policy (Agreement on the Application of Sanitary and Phytosanitary Measures). The methodology can also be used to assist in the choice of an appropriate national response strategy following an incursion of bluetongue (40). The choice of strategy should be made after an independent, scientific and collective assessment in which the range and magnitude of consequences of implementing or not implementing a vaccination programme of all susceptible domestic livestock in the affected regions is considered (scenario analysis). This methodology was used in Italy before initiating vaccination of all susceptible domestic ruminant species in infected areas and areas surrounding them (protection zone). Under the relevant Italian conditions, the model predicted that at least 80% of susceptible livestock should be immunised for vaccination to be effective (39). Such risk assessment should be conducted taking into account current scientific knowledge, the particular local situation and uncertainties about the parameters used in the model. In addition, decision trees are often used to clarify the path to appropriate vaccine usage. Analysis includes evaluation of several parameters, such as the probability of exposure to the infectious agent, the efficacy and safety of the

Adhering to the vaccination protocol and maintaining a system allowing registration, delivery and follow-up of vaccination
It is vital to strictly follow the manufacturers instructions and to quickly administer the doses after reconstitution of the freeze-dried vaccine. The storage conditions of vaccine are very important (isotherm box): a rapid decrease of infectious virus titre is observed after a few hours at ambient temperature (45). This observation could explain some apparent vaccine failure in the field. As with every veterinary medical product the traceability of vaccine doses must be assured at local, regional and national level. The following details must be entered into a vaccine management information system, as without them it is not possible to correctly estimate vaccine coverage (39): vaccine serotype(s) vaccine batch numbers farm codes total numbers of animals on the farm numbers of target animals numbers of vaccinated animals by species vaccination date.

Rev. sci. tech. Off. int. Epiz., 26 (2)

405

Evaluating the necessary level of vaccination coverage


It is important to evaluate the level of vaccination coverage that should be attained to eradicate the disease. An empirical evaluation gives a herd immunity threshold (HIT) value of 75% (Charles Nicolles law) but it has been shown that HIT calculation should be based on the evaluation of the rate of spread of the disease measured by the basic reproductive number R0, i.e. the average number of secondary infections produced by one infected individual introduced into a fully susceptible population (1). Basically, if R0 <1, the disease cannot persist in a population. It can be shown that the proportion of animals to be vaccinated in a nave population must exceed 11/ R0 in order to reduce R0 to a value below 1. It is noted that for vector-borne diseases, several factors attributable to the vector will change the classical approach (e.g. biting rate, trophic preferences, survival rate, incubation period and vectorial competency, i.e. capacity for the virus to develop in the vector). Consequently, even with high vaccination coverage, it is still possible that the virus will persist in small areas or in reservoir animals. There is then a risk of re-emergence when coverage declines (38). The relationship between vaccination coverage of the target populations and animal losses due to disease and virus circulation must be analysed.

herds in the risk area is performed. Because vaccinated animals are seropositive, subpopulations that are included in the study must be unvaccinated animals (e.g. young animals sampled when prophylactic treatment is administered or in slaughterhouses). Blood samples from sentinel herds should be collected using a statistically based sampling frame, with the frequency of collection depending upon the season and infection occurrence in the area. Entomological surveillance is performed by trapping midges in areas of ecological importance in order to monitor the spread and seasonal dynamics of Culicoides spp. (39, 119). The presence of field BTV and vaccine virus (when using attenuated vaccine) must be investigated. The possibility that midges could acquire vaccine virus by feeding on vaccinated animals and transmit it to unvaccinated animals should also be investigated, in particular, with regards to territories that are free from disease/infection but border zones where vaccination is carried out (30).

Conclusions and recommendations


In the case of an emerging disease (e.g. bluetongue), the decision by national authorities to include temporary authorisation of a non-registered animal vaccine in disease control strategies requires scientific, economic, technical, social and risk analysis support. Advantages and disadvantages of the vaccination option strategy (Table IV) should be taken into account, as well the regulatory aspects (Table V), the biology and the epidemiology of the disease, the structure and efficacy of the epidemiological network and the economic and social impacts. Moreover, in the case of a zoonotic disease, factors affecting humans become a priority. Use of decision trees should also be encouraged. For safety reasons inactivated or other more technologically advanced vaccines should be preferred because they have the potential not only to effectively control the disease but also to allow trade due to detection of virus circulation in live animals. The development of marker vaccines is the correct choice for manufacturers, but national authorities and supra-national organisations should stimulate early development of these new vaccines (including bank vaccines) with the aim of delivering them quickly to reduce pathogen circulation in an emerging situation. Listing and prioritisation of diseases of major concern (i.e. diseases which can have a severe impact on human/animal health or on the economy) should be the first step, for which an understanding of the accuracy of the estimation of the true prevalence of disease is essential.

Monitoring, analysis and possible adjustment of field vaccination


In order to monitor the progress of the vaccination campaign in the infected zone and verify the immune status of vaccinated herds (coverage), a random sample of vaccinated animals in each grid cell should be tested for antibodies (39). All clinical suspicion must be investigated to confirm whether or not BTV is present. In the case of viral detection and isolation, serotyping of BTV is performed (e.g. by seroneutralisation test) to verify possible new strain introductions into the territory. In this case, the vaccination programme must be promptly adapted (119). If possible, an RT-PCR test should be carried out to differentiate between the wild type and vaccine viruses (8). In addition, strain genotyping permits the study of the molecular epidemiology of BT and the monitoring of possible reassortment of genome segments between vaccine and field viruses (7). The vaccination programme must be maintained until there is proof that the virus is no longer circulating. Once the clinical signs disappear the absence of BTV circulation in the vaccinated populations must be verified. In order to do this, a longitudinal study of representative sentinel

406

Rev. sci. tech. Off. int. Epiz., 26 (2)

The latter depends on the presence and quality of the epidemiologic network and on clearly and swiftly notifying the disease to the appropriate authorities (e.g. information promptly entered into the OIE World Animal Health Information System and into the Centralized Information System for Infectious Diseases of the World Health Organization). The second step should be to find out about existing patents and to develop a map showing the locations of research laboratories, technology facilities and manufacturers to be used by competent authorities in case of emerging diseases. The third step should be to identify gaps and important weaknesses in available tools and sources. The fourth step should be to develop a proposition to focus research on priority animal disease so as to increase the knowledge base and product availability for these diseases (94). When applying this vaccination strategy several factors should be evaluated collectively, including: communication prevalent epidemiological situation response strategy to the disease appropriateness of the vaccine used adherence to vaccination protocol and maintenance of a system allowing registration, delivery and follow-up of vaccine

level of vaccination coverage analysis and possible adjustment of field vaccination. This information is useful for replacing the temporary authorisation with a complete authorisation. It is also necessary to continuously adapt the vaccination programme to the epidemiological situation. This adaptation requires the collaboration of all stakeholders and needs to include independent and collective scientific advice. Finally, the concept of temporary authorisation can easily be transposed for use with emerging zoonotic diseases and may assist in improving relationships between different Veterinary Services worldwide and it is an excellent way to build the necessary capacity to respond to emerging diseases.

Acknowledgements
The authors wish to thank P. Houdart (Federal Agency for the Safety of the Food Chain) for help in accessing data originating from the European Commissions Animal Disease Notification System and R. De Deken and M. Madder (Institute for Tropical Medicine) for use of the photo of C. dewulfi.

Questions rglementaires lies lautorisation temporaire de vacciner les animaux en situation durgence : lexemple de la fivre catarrhale du mouton en Europe
C. Saegerman, M. Hubaux, B. Urbain, L. Lengel & D. Berkvens
Rsum Lautorisation de mise sur le march des vaccins vtrinaires est accorde ds lors que la qualit, linnocuit et lefficacit du produit ont t values conformment aux normes rglementaires. Cette valuation comprend la caractrisation exhaustive suivie de lidentification des semences et des ingrdients utiliss, lvaluation de la scurit pour le laboratoire et pour lespce hte, ainsi que des tudes defficacit, un suivi aprs la mise sur le march et un contrle des performances sur le terrain. Cette valuation nest pas toujours ralisable en cas dmergence dune nouvelle maladie animale, mais il existe plusieurs mcanismes permettant de disposer de produits dans des

Rev. sci. tech. Off. int. Epiz., 26 (2)

407

situations durgence zoosanitaire : autovaccins, autorisations de mise sur le march sous condition, autorisations limites aux exprimentations ou aux situations durgence, importation de produits utiliss en dautres endroits du monde, banques de vaccins pr-enregistrs. Les auteurs dcrivent les questions rglementaires lies lautorisation temporaire de vacciner les animaux, en prenant lexemple de lmergence de la fivre catarrhale du mouton en Europe. Plusieurs conditions doivent tre runies avant de dlivrer une autorisation temporaire : le vaccin doit tre virus inactiv afin dempcher tout risque de rversion vers la virulence ou de rassortiment entre les souches vaccinales et/ou les souches sauvages du virus de la fivre catarrhale du mouton ; les dcisions en la matire doivent tre justifies scientifiquement et se fonder sur une analyse du risque ; les animaux susceptibles ayant t vaccins doivent tre rigoureusement recenss ; les protocoles de vaccination doivent tre connus et respects ; un dispositif doit tre en place pour lenregistrement, ladministration et le suivi de la vaccination, ainsi que pour le suivi, lanalyse et dventuelles corrections dans lutilisation de la vaccination sur le terrain. Cette autorisation temporaire doit tre remplace aussi rapidement que possible par une autorisation rgulire. Mots-cls Autorisation temporaire Maladie mergente Question rglementaire Sant animale Vaccin Vaccination.

Cuestiones de reglamentacin en torno a la autorizacin temporal de vacunacin de animales en situaciones de emergencia: el ejemplo de la lengua azul en Europa
C. Saegerman, M. Hubaux, B. Urbain, L. Lengel & D. Berkvens
Resumen Para autorizar la comercializacin de una vacuna veterinaria es preciso que previamente se hayan evaluado su calidad, inocuidad y eficacia conforme a la normativa vigente. Ello comprende la descripcin y caracterizacin completa del material y los ingredientes de partida, los estudios de inocuidad para los animales hospedadores y de laboratorio y los estudios de eficacia y estabilidad, as como el control del rendimiento del producto sobre el terreno una vez obtenida la licencia. Aunque todo este proceso puede resultar impracticable en el momento en que se manifiesta una nueva enfermedad animal, existen varios mecanismos para asegurar el suministro de productos en el curso de una emergencia zoosanitaria, por ejemplo productos biolgicos autgenos, licencias condicionales, autorizaciones de uso experimental y de emergencia, importacin de productos utilizados en otras partes del mundo y bancos de vacunas preaprobadas. Utilizando el ejemplo de la aparicin de la lengua azul en el Norte de Europa, los autores exponen las cuestiones de reglamentacin que se plantean respecto a la autorizacin temporal de vacunacin de animales.

408

Rev. sci. tech. Off. int. Epiz., 26 (2)

Para otorgar una autorizacin temporal se exige el cumplimiento de varias condiciones, por ejemplo: el uso de vacunas inactivadas para evitar la posibilidad de reversin a la virulencia o de reordenamiento entre virus vacunales y/o cepas salvajes del virus de la lengua azul; la existencia de pruebas cientficas y anlisis de riesgos que respalden la adopcin de decisiones; la existencia de un censo completo de los animales sensibles que hayan sido vacunados; el cumplimiento de los protocolos de vacunacin; y la existencia de un mecanismo que permita registrar y aplicar las vacunaciones, supervisar sus efectos y controlar, analizar y eventualmente ajustar la administracin de vacunas sobre el terreno. Esta autorizacin temporal debe ser sustituida lo antes posible por una autorizacin completa. Palabras clave Autorizacin temporal Enfermedad emergente Problema de reglamentacin Sanidad animal Vacuna Vacunacin.

References
1. Anderson R.M. & May R.M. (1991). Infectious diseases of humans: dynamics and control. Oxford University Press. 2. Anon. (1996). Pharmacovigilantie: anaphylaxie bij runderen. Onverenigbaarheid tussen vaccins en antibiotica: mogelijke aanleiding tot ernstige shockreacties [Pharmacovigilance. Anaphylaxis in bovines incompatibility of vaccines and antibiotics: a possible reason for severe allergic reactions]. Press release from Mycofarm, Boehringer, Pfizer and Vtoquinol. Mycofarm, Turnhout, 3 pp. 3. Anon. (1998). Report on the incursion and evolution of bluetongue in Greece in 1998. Ministry of Agriculture, DG of Veterinary Services, Department of Infectious Diseases, Greece. 4. Barnouin J. & Vourch G. (2004). Les maladies mergentes : un dfi pour le dveloppement durable des productions animales. INRA Prod. anim., 17 (5), 355-364. 5. Benkirane A. (2001). Surveillance pidmiologique et prophylaxie de la brucellose des ruminants : lexemple de la rgion Afrique du Nord et Proche-Orient. Rev. sci. tech. Off. int. Epiz., 20 (3), 757-767. 6. Braverman Y., Chizov-Ginzburg A., Pener H. & Wilamowski A. (2004). Susceptibility and repellency of Culicoides imicola and Culex pipiens to lambda-cyhalothrin. Vet. ital., 40 (3), 336-339. 7. Brard E., Hamblin C., Hammoumi S., Sailleau C., Dauphin G. & Zientara S. (2004). The epidemiology and diagnosis of bluetongue with particular reference to Corsica. Res. vet. Sci., 77, 1-8. 8. Brard E., Sailleau C., Coupier H., Mure-Ravaud K., Hammoumi S., Gicquel B., Hamblin C., Dubourget P. & Zientara S. (2003). Comparison of genome segments 2, 7 and 10 of bluetongue viruses serotype 2 for differentiation between field isolates and the vaccine strain. Vet. Res., 34, 777-789. 9. Brown C. (2001). Importance des maladies mergentes pour la sant publique et animale et pour les changes commerciaux. 69th General Session of the International Committee of the World Organisation for Animal Health (OIE), 27 May-1 June, Paris, document 69 SG/9. OIE, Paris, 6 pp.

Rev. sci. tech. Off. int. Epiz., 26 (2)

409

10. Cameron A.R. (2004). The impact of current and proposed changes to general guidelines on bluetongue surveillance of the Office International des Epizooties. Vet. ital., 40 (4), 693-696. 11. Caracappa S., Torina A., Guercio A., Vitale F Calabro A., ., Purpari G., Ferrantelli V., Vitale M. & Mellor P.S. (2003). Identification of a novel bluetongue virus vector species of Culicoides in Sicily. Vet. Rec., 153 (3), 71-74. 12. Center for Veterinary Biologics (CVB) (2006). Code of Federal Regulations, Title 9 Animals and Animal Products, Parts 101 to 123. Ames, Iowa, United States of America. Available at http://www.aphis.usda.gov/vs/cvb/html/cfr.html (accessed on 28 October 2006). 13. Centers for Disease Control and Prevention (CDC) (1998). Preventing emerging infectious diseases. A strategy for the 21st Century. Center for disease control and prevention, US Department of health and human services, Atlanta, Georgia, October. 14. De Mattos C.C., de Mattos C.A., Osburn B.I. & MacLachlan N.J. (1994). Heterogeneity of the L2 gene of field isolates of bluetongue virus serotype 17 from the San Joaquin Valley of California. Virus Res., 31, 67-87. 15. Di Emidio B., Nicolussi P., Patta C., Ronchi G.F Monaco F ., ., Savini G., Ciarelli A. & Caporale V. (2004). Efficacy and safety studies on an inactivated vaccine against bluetongue virus serotype 2. Vet. ital., 40 (4), 640-644. 16. Dubourget P. & Bolon A. (2005). Inactivated bluetongue vaccine. Workshop: EU funded research in bluetongue, 10-11 March, Brussels, Belgium. 17. Dulac G.C., Dubuc C., Myers D.J., Afshar A. & Taylor E.A. (1989). Incursion of bluetongue virus type 11 and epizootic haemorrhagic disease of deer type 2 for two consecutive years in the Okanagan Valley. Can. vet. J., 30, 351. 18. Dungu B., Gerdes T. & Smit T. (2004). The use of vaccination in the control of bluetongue in southern Africa. Vet. ital., 40 (4), 616-622. 19. Enserink M. (2006). Emerging infectious diseases. During a hot summer, bluetongue virus invades northern Europe. Science, 313 (5791), 1218-1219. 20. European Commission (2000). Possible use of vaccination against bluetongue in Europe. Report of the Scientific Committee on Animal Health and Animal Welfare. Health & Consumer Protection Directorate-General. Adopted 27 June. Doc. Sanco/C3/AH/R19/2000. 21. European Commission (2001). Decision 2001/433/EC of 21 May 2001 for purchase by the Community of bluetongue vaccine and restocking of the Community bank. Off. J. Eur. Communities, L 154, 61-62. 22. European Council (1981). Council Directive 81/852/ECC on the approximation of the laws of the Member States relating to analytical, pharmacotoxicological and clinical standards and protocols in respect of the testing of veterinary medicinal products. Off. J. Eur. Communities, L 317, 53-98.

23. European Council (2000). Directive 2000/75/EC of 20 November, laying down specific provisions for the control and eradication of bluetongue. Off. J. Eur. Communities, L 327, 74-83. 24. European Food Safety Authority (EFSA) (2006). Bluetongue serotype 8 epidemic bulletin by EFSA epidemiology working group. Available at: http://www.efsa. europa.eu/en/in_focus/bluetongue/outbreak_overview.html (accessed on 28 October 2006). 25. European Food Safety Authority (EFSA) (2006). Opinion of the Scientific Committee on a request from EFSA related to the early identification of emerging risks. EFSA J., 375, 1-14. 26. European Medicines Agency (2006). Guideline on the procedure for accelerated assessment pursuant to article 39 (8) of regulation (EC) No. 726/2004. Committee for Medicinal Products for Veterinary Use, London, United Kingdom, 18 May. Doc. Ref. EMEA/CVMP/32995/2006. 27. European Parliament and the European Council (2001). Directive 2001/82/EC of 6 November 2001 on the Community code relating to veterinary medicinal products. Off. J. Eur. Communities, L 311, 1-66. 28. European Parliament and the European Council (2004). Directive 2004/28/EC of 31 March 2004 amending Directive 2001/82/EC on the Community code relating to veterinary medicinal products. Off. J. Eur. Communities, L 136, 58-84. 29. European Parliament and the European Council (2004). European Regulation 726/2004/EC of 31 March 2004 laying down Community procedures for the authorisation and supervision of medicinal products for human and veterinary use and establishing a European Medicines Agency. Off. J. Eur. Communities, L 136, 58-84. 30. Ferrari G., De Liberato C., Scavia G., Lorenzetti R., Zini M., Farina F Magliano A., Cardeti G., Scholl F Guidoni M., ., ., Scicluna M.T., Amaddeo D., Scaramozzino P. & Autorino G.L. (2005). Active circulation of bluetongue vaccine virus serotype 2 among unvaccinated cattle in central Italy. Prev. vet. Med., 68 (2-4), 103-113. 31. Fvrier J. (2004). European Union policy for the control and eradication of bluetongue. Vet. ital., 40 (4), 682. 32. Flanagan M. & Johnson S.J. (1995). The effects of vaccination of Merino ewes with an attenuated Australian BTV serotype 23 at different stages of gestation. Aust. vet. J., 72, 455-457. 33. Foley P.L. & Hill R.E. Jr (2005). Regulatory considerations for marker vaccines and diagnostic tests in the US. Biologicals, 33 (4), 253-256. 34. Food and Agriculture Organization of the United Nations (FAO) (2000). Preliminary data on veterinary vaccine production/needs in some countries of the Near East. FAO Regional Office for the Near East, Cairo, Egypt.

410

Rev. sci. tech. Off. int. Epiz., 26 (2)

35. French T.J., Marshall J.J. & Roy P. (1990). Assembly of double-shelled, viruslike particles of bluetongue virus by the simultaneous expression of four structural proteins. J. Virol., 64 (12), 5695-5700. 36. French T.J. & Roy P. (1990). Synthesis of bluetongue virus (BTV) corelike particles by a recombinant baculovirus expressing the two major structural core proteins of BTV. J. Virol., 64 (4), 1530-1536. 37. Fu H., Leake C.J., Mertens P.P.C. & Mellor P.S. (1999). The barriers to bluetongue virus infection, dissemination and transmission in the vector, Culicoides variipennis (Diptera: Ceratopogonidae). Arch. Virol., 144, 747-761. 38. Gerbier G., Hendrikx P., Roger F Zientara S., Biteau-Coroller ., F Grillet C., Badler T. & Albina E. (2004). Bluetongue ., control using vaccines: experience of the Mediterranean islands. Vet. ital., 40 (4), 611-615. 39. Giovannini A., Calistri P., Nannini D., Paladini C., Santucci U., Patta C. & Caporale V. (2004). Bluetongue in Italy: Part II. Vet. ital., 40 (3), 252-259. 40. Giovannini A., MacDiarmid S., Calistri P., Conte A., Savini L., Nannini D. & Weber S. (2004). The use of risk assessment to decide the control strategy for bluetongue in Italian ruminant populations. J. Risk Anal., 24 (6), 1737-1753. 41. Giovannini A., Paladini C., Calistri P., Conte A., Colangeli P., Santucci U., Nannini D. & Caporale V. (2004). Surveillance system of bluetongue in Italy. Vet. ital., 40 (3), 369-384. 42. Gomulski L.M., Meiswinkel R., Delecolle J.C., Goffredo M. & Gasperi G. (2005). Phylogeny of the subgenus Culicoides and related species in Italy, inferred from internal transcribed spacer 2 ribosomal DNA sequences. Med. vet. Entomol., 20 (2), 229-238. 43. Guyot H., Mauroy A., Thiry E., Losson B., Bodmer M., Kirten P., Rollin F & Saegerman C. (2007). Fivre catarrhale . ovine chez les ruminants. Description clinique des cas vcus dans le Nord de lEurope durant lt et lautomne 2006. Bulletin GTV, 39, 89-96. 44. Hallet L. (2003). Les modes de collaboration entre vtrinaires officiels, vtrinaires privs et organisations dleveurs. In Veterinary Services: organisation, quality assurance, evaluation (E. Correa Melo & F Gerster, eds). . Rev. sci. tech. Off. int. Epiz., 22 (2), 523-532. 45. Hammoumi S., Brard E., Sailleau C., Russo P., Grillet C., Cetre-Sossah C., Albina E., Sanchis R., Pepin M., Guibert J.M. & Zientara S. (2003). Studies on the safety and immunogenicity of the South African bluetongue virus serotype 2 monovalent vaccine: specific detection of the vaccine strain genome by RT-PCR. J. vet. Med., B, infect. Dis. vet. public Hlth, 50 (7), 316-321. 46. Heidner H.W., Iezzi L.G., Osburn B.I. & MacLachlan N.J. (1991). Genetic variation and evolutionary relationships amongst bluetongue viruses endemic in the United States. Virus Res., 21, 91-109.

47. Hill R.E. (2004). Regulation of conditionally licensed biologicals in the United States. Dev. Biol. (Basel), 117, 147-152. 48. Hill R.E., Foley P.L., Carr M.Y., Elsken L.A., Gatewood D.M., Ludeman L.R. & Wilbur L.A. (2003). Regulatory considerations for emergency use of non-USDA licensed vaccines in the United States. Dev. Biol. (Basel), 114, 31-52. 49. Hill R.E., Foley P.L., Carr M.Y., Elsken L.A., Gatewood D.M., Ludeman L.R. & Wilbur L.A. (2005). Regulatory considerations for emergency use of non-USDA licensed vaccines in the United States. Workshop: EU funded research in bluetongue, 10-11 March, Brussels, Belgium. 50. Hunter P. & Modumo J. (2001). A monovalent attenuated serotype 2 bluetongue virus vaccine confers homologous protection in sheep. Onderstepoort J. vet. Res., 68, 331-333. 51. Hupet P. (2002). La communication relative au risque : principes gnraux. pidmiol. Sant anim., 41, 189-200. 52. Kettle D.S. (1962). The bionomics and control of Culicoides and Leptoconops (Diptera, Ceratopogonidae Heleidae). Annu. Rev. Entomol., 7, 401-418. 53. King L.J. (2004). Emerging and re-emerging zoonotic diseases: challenges and opportunities. Technical Item I. In Proc. 72nd General Session of the International Committee of the World Organisation for Animal Health (OIE), Paris, France, 23-28 May. OIE, Paris, Doc. 72 SG/9. 54. Kirkland P.D., Zhang N., Hawkes R.A., Li Z., Zhang F ., Davis R.J., Sanders D.A., Li H., Zhang K., Ben J., He G.F ., Hornitzky C.L. & Hunt N.T. (2002). Studies on the epidemiology of bluetongue virus in China. Epidemiol. Infect., 128, 257-263. 55. Le Gall F (2006). Economic and social justification of .G. investment in animal health and zoonoses. Technical Item II. In Proc. 74th General Session of the International Committee of the World Organisation for Animal Health (OIE), Paris, France, 21-26 May 2006. OIE, Paris, Doc. 74 SG/10. 56. Lefvre P.-C. & Mellor P.S. (2007). Bluetongue. In Infectious and parasitic disease of livestock. Lavoisier, Provigny, France, in press. 57. MacLachlan N.L. (1994). The pathogenesis and immunology of bluetongue virus infection of ruminants. Comp. Immunol. Microbiol. infect. Dis., 17, 197-206. 58. Meiswinkel R. (2006). The Culicoides vector of bluetongue disease in Limburg, the Netherlands. International Society for Infectious Diseases. Available at: www.promedmail.org (accessed on 28 October 2006). 59. Mellor P.S. (2005). Development of a safe, efficacious bluetongue virus vaccination strategy for Europe. Workshop: EU funded research in bluetongue, 10-11 March, Brussels, Belgium.

Rev. sci. tech. Off. int. Epiz., 26 (2)

411

60. Mellor P.S. & Boorman J. (1995). The transmission and geographical spread of African horse sickness and bluetongue viruses. Ann. trop. Med. Parasitol., 89 (1), 1-15. 61. Mellor P.S., Boorman J. & Baylis M. (2000). Culicoides biting midges: their role as arbovirus vectors. Annu. Rev. Entomol., 45, 307-340. 62. Mellor P.S. & Wittmann E. (2002). Bluetongue virus in the Mediterranean basin 1998-2001. Vet. J., 164, 20. 63. Mertens P.P.C., Attoui H. & Bamford D.H. (eds) (2006). The RNAs and proteins of dsRNA viruses. Institute for Animal Health, Pirbright. Available at: www.iah.bbsrc.ac.uk/dsRNA_ virus_proteins/Orbivirus.htm (accessed on 28 October 2006). 64. Miranda M.A., Borras D., Rincon C. & Alemany A. (2003). Presence in the Balearic Islands (Spain) of the midges Culicoides imicola and Culicoides obsoletus group. Med. vet. Entomol., 17, 52-54. 65. Monaco F Bonfini B., Zaghini M., Antonucci D., Pini A. & ., Savini G. (2004). Vaccination of cattle using monovalent modified-live vaccine against bluetongue virus serotype 2: innocuity, immunogenicity and effect on pregnancy. Vet. ital., 40 (4), 671-675. 66. Monaco F De Luca N., Morelli D., Pisciella M., Palmarini S., ., Di Giandomenico M. & Savini G. (2004). Field vaccination of cattle using a bivalent modified-live vaccine against bluetongue virus serotypes 2 and 9: effect on milk production. Vet. ital., 40 (4), 661-663. 67. Monaco F De Luca N., Spina P., Morelli D., Liberatore I., ., Citarella R., Conte A. & Savini G. (2004). Virological and serological response of cattle following field vaccination with bivalent modified-live vaccine against bluetongue virus serotypes 2 and 9. Vet. ital., 40 (4), 657-660. 68. Morse S.S. (1995). Factors in the emergence of infectious diseases. Emerg. infect. Dis., 1, 7-15. 69. Morse S.S. (2004). Factors and determinants of disease emergence. In Emerging zoonoses and pathogens of public health concern (L.J. King, ed.). Rev. sci. tech. Off. int. Epiz., 23 (2), 443-451. 70. Murray P.K. & Eaton B.T. (1996). Vaccines for bluetongue. Aust. vet. J., 73 (6), 207-210. 71. Oberst R.D., Stott J.L., Blanchard-Channell M. & Osburn B.I. (1987). Genetic reassortment of bluetongue virus serotype 11 strain in the bovine. Vet. Microbiol., 15, 11-18. 72. OConnell L. (2001). Transmission rates of bluetongue virus by Culicoides biting midges. In Proc. of the Royal Entomological Society International Symposium on insects and disease, 10-12 September, University of Aberdeen. Abstracts book.

73. OConnell L. (2002). Entomological aspects of the transmission of arboviral diseases by Culicoides biting midges. Ph.D. thesis of the University of Bristol, 176 pp. 74. Osburn B.I. (1994). The impact of bluetongue virus on reproduction. Comp. Immunol. Microbiol. infect. Dis., 17, 189-196. 75. Panagiotatos D.E. (2005). Review of bluetongue virus incursions, vectors, surveillance and epidemiological features in Eastern Europe in the period 1998-2004. Workshop: EU funded research in bluetongue, 10-11 March, Brussels, Belgium. 76. Pastoret P.-P. & Mackay D. (2002). Regulatory considerations for emergency use of vaccines in the European Union. Vaccines for OIE list A and emerging animal diseases, Scheman Conference Center, Ames, Iowa, USA, 16-18 September. 77. Pattison J. (1998). The emergence of bovine spongiform encephalopathy and related diseases. Emerg. infect. Dis., 4 (3), 390-394. 78. Pearson L.D. & Roy P. (1993). Genetically engineered multi-component virus-like particles as veterinary vaccines. Immunol. Cell Biol., 71 (5), 381-389. 79. Purse B.V., Mellor P.S., Rogers D.J., Samuel A.R., Mertens P.P. & Baylis M. (2005). Climate change and the recent emergence of bluetongue in Europe. Nat. Rev. Microbiol., 3 (2), 171-181. 80. Qin Q., Tai Z., Wang L., Luo Z., Hu J. & Lin H. (1996). Bluetongue epidemiological survey and virus isolation in Xinjiang, China. In Bluetongue disease in Southeast Asia and the Pacific. Australian Centre for International Agricultural Research, Canberra, 67-71. 81. Rodhain F (2003). Emergences de maladies transmission . vectorielle. pidmiol. Sant anim., 43, 33-49. 82. Roy P. (1992). Bluetongue virus proteins. J. gen. Virol., 73, 3051-3064. 83. Roy P. (1996). Multiple gene expression in baculovirus system. Third generation vaccines for bluetongue disease and African horse sickness disease. Ann. N.Y. Acad. Sci., 23, 318-332. 84. Roy P., Bishop D.H.L., LeBois H. & Erasmus B.J. (1994). Long-lasting protection of sheep against bluetongue challenge after vaccination with virus-like particles: evidence for homologous and partial heterologous protection. Vaccine, 12, 805-811. 85. Saegerman C. (2006). Epidmiosurveillance des vnements rares chez les bovins en Belgique. Ann. Md. vt., 150 (S), 4-9. 86. Saegerman C., Berkvens D., Godfroid J. & Walravens K. (2007). Bovine brucellosis. In Infectious and Parasitic Disease of Livestock. Lavoisier, Provigny, France, in press.

412

Rev. sci. tech. Off. int. Epiz., 26 (2)

87. Saegerman C., Berkvens D., Speybroeck N., Roels S., Vanopdenbosch E. & Thiry E. (2003). Amlioration de la dtection dune maladie mergente : exemple de lencphalopathie spongiforme bovine. In pidmiologie des maladies mergentes (special issue). pidmiol. Sant anim., 44, 61-77. 88. Saegerman C., Claes L., Dewaele A., Desmecht D., Rollin F ., Hamoir J., Gustin P., Czaplicki G., Bughin J., Wullepit J., Laureyns J., Roels S., Berkvens D., Vanopdenbosch E. & Thiry E. (2003). Differential diagnosis of neurologically expressed disorders in Western European cattle. In Risk analysis of prion diseases in animals (C.I. Lasmzas & D.B. Adams, eds). Rev. sci. tech. Off. int. Epiz., 22 (1), 83-102. 89. Samal S.K., Elhussein A., Holbrook F .R., Beaty B.J. & Ramig R.F (1987). Mixed infection of Culicoides-variipennis . with bluetongue virus serotype 10 and serotype 17 evidence for high-frequency reassortment in the vector. J. gen. Virol., 68, 2319-2329. 90. Samal S.K., Livingstone C.W. Jr, McConnell S. & Ramig R.F . (1987). Analysis of mixed infection of sheep with bluetongue serotype 10 and 17: evidence for genetic reassortment in the vertebrate host. J. Virol., 61, 1086-1091. 91. Satta G., Goffredo M., Sanna S., Vento L., Cubeddu G.P. & Mascherpa E. (2004). Field disinfestation trials against Culicoides in north-west Sardinia. Vet. ital., 40 (3), 329-335. 92. Savini G., Goffredo M., Monaco F de Santis P. & Meiswinkel ., R. (2003). Transmission of bluetongue virus in Italy. Vet. Rec., 152, 119. 93. Savini G., Goffredo M., Monaco F., Di Gennaro A., Cafiero M.A., Baldi L., De Santis P., Meiswinkel R. & Caporale V. (2005). Bluetongue virus isolations from midges belonging to the Obsoletus complex (Culicoides, Diptera: Ceratopogonidae) in Italy. Vet. Rec., 157, 133-139. 94. Schumacher C.L. (2005). Available products, not licensed in Europe. European Technology Platform for Animal Health. Working Group 2, Brussels, October 12, 2005. Available at: http://www.ifahsec.org/Europe/EUPlatform/ Minutes/WG2/WEB-C.%20Schumacher.ppt (accessed on 18 June 2006). 95. Scott J.L., Oberst R.D., Channell M.B. & Osburn B.I. (1987). Genome segment reassortment between two serotypes of bluetongue virus in a natural host. J. Virol., 61, 2670-2674. 96. Scott J.L., Osburn B.I. & Barber T.L. (1982). Recovery of dual serotypes of bluetongue virus from infected sheep and cattle. Vet. Microbiol., 7 (3), 197-207. 97. Slingenbergh J., Gilbert M., de Balogh K. & Wint W. (2004). Ecological sources of zoonotic diseases. In Emerging zoonoses and pathogens of public health concern (L.J. King, ed.). Rev. sci. tech. Off. int. Epiz., 23 (2), 467-484.

98. Standfast H.A., Muller M.J. & Wilson D.D. (1985). Mortality of Culicoides brevitarsis fed on cattle treated with Ivermectin. In Bluetongue and related Orbiviruses (T.L. Barber & M.M. Jochim, eds). Alan R. Liss Inc., New York, 519-525. 99. Takamatsu H., Mellor P.S., Mertens P.P.C., Kirkham P.A., Burroughs J.N. & Parkhouse R.M.E. (2003). A possible overwintering mechanism for bluetongue virus in the absence of the insect vector. J. gen. Virol., 84, 227-235. 100. Taylor W.P. (1986). The epidemiology of bluetongue. Rev. sci. tech. Off. int. Epiz., 5 (2), 351-356. 101. Thiry E., De Clercq K., Saegerman C. & Vermeersch J.P. (2002). La vaccination contre la fivre aphteuse : perspectives. Bull. GTV, 14, 117-121. 102. Thiry E., Saegerman C., Guyot H., Bodmer M., Kirten P., Losson B., Rollin F Czaplicki G., Toussaint J.F ., ., De Clercq K., Dochy J.M., Dufey J., Gilleman J.L. & Messeman K. (2006). Clinical cases of bovine bluetongue in Belgium. Vet. Rec., 159 (10), 327. 103. Toma B. & Thiry E. (1999). Quest-ce quune maladie mergente ? pidmiol. Sant anim., 44, 1-11. 104. Toussaint J.-F., Sailleau C., Mast J., Houdart Ph., Czaplicki G., Demeestere L., VandenBussche F., Van Dessel W., Goris N., Brard E., Bounaadja L., Thiry E., Zientara S. & De Clercq K. (2007). Bluetongue in Belgium, 2006. Emerg. infect. Dis., 13 (4), 614-616. 105. Townley P., Baker K.P. & Quinn P.J. (1984). Preferential landing and engorging sites of Culicoides species on a horse in Ireland. Equine vet. J., 16, 117-120. 106. United States Department of Agriculture (USDA) (2005). Overview of the Mediterranean basin bluetongue disease outbreak, 1998-2004. Center for Emerging Issues, Animal and Plant Health Inspection Service, USDA. Available at: http://www.aphis.usda.gov/vs/ceah/cei/taf/emergingdisea senotice_files/bluetongue_medbasin_ednotice_sep_04_files /bluetongue_med_basin_jan_05_update.htm (accessed on 28 October 2006). 107. Urakawa T., French T.J., Adachi Y., Fukusho A., LeBlois H., Flamand M., Mertens P. & Roy P. (1994). Synthesis of recombinant baculoviruses expressing the outer capsid protein VP2 of five BTV serotypes and the induction of neutralizing antibodies to homologous and heterologous BTV serotypes. Virus Res., 31, 149-161. 108. Van Boxstael S., Vereecken K., Huyghebaert A., Noteborn H. & Saegerman C. (2005). Development of a system for pro-active identification of food-related emerging risks. In Proc. Tenth Conference on Food Microbiology, University of Lige, 23-24 June, 102-103. 109. Venter G.J., Gerdes G.H., Mellor P.S. & Paweska J.T. (2004). Transmission potential of South African Culicoides species for live-attenuated bluetongue virus. Vet. ital., 40 (3), 198203.

Rev. sci. tech. Off. int. Epiz., 26 (2)

413

110. Veronesi E., Hamblin C. & Mellor P.S. (2005). Live attenuated bluetongue vaccine viruses in Dorset Poll sheep, before and after passage in vector midges (Diptera: Ceratopogonidae). Vaccine, 23, 5509-5516. 111. Verwoerd D.W., Louw H. & Oellermann R.A. (1970). Characterization of bluetongue virus ribonucleic acid. J. Virol., 5, 10-17. 112. Webster W.R., Gard G.P., St George T.D. & Kirkland P.D. (1992). The Australian bluetongue control strategy. In Bluetongue, African horse sickness and related Orbivirus (T.E. Walton & B.I. Osburn, eds). CRC Press, 843-850. 113. Weiss R.A. & McMichael A.J. (2004). Social and environmental risk factors in the emergence of infectious diseases. Nature Med., 10 (12), suppl., 70-76. 114. Wittmann E.J., Mellor P.S. & Baylis M. (2001). Using climate data to map the potential distribution of Culicoides imicola (Diptera: Ceratopogonidae) in Europe. Rev. sci. tech. Off. int. Epiz., 20 (3), 731-740. 115. World Organisation for Animal Health (OIE) (2006). Handistatus II. Multiannual animal disease status for bluetongue. Available at: http://www.oie.int/hs2/report.asp (accessed on 28 October 2006).

116. World Organisation for Animal Health (OIE) (2006). Bluetongue detected for the first time in Northern Europe. Press release, 23 August. Available at: http://www.oie.int/ eng/press/en_060823.htm (accessed on 28 October 2006). 117. World Organisation for Animal Health (OIE) (2006). Bluetongue in Northern Europe: an OIE Reference Laboratory makes a breakthrough in identifying the vector causing the disease. Press release, 23 October. Available at: http://www.oie.int/eng/press/en_061023.htm (accessed on 28 October 2006). 118. World Organisation for Animal Health (OIE) (2006). Bluetongue. Chapter 2.2.13. In Terrestrial Animal Health Code, 15th Ed. OIE, Paris. Available at: http://www.oie.int/ eng/normes/mcode/en_chapitre_2.2.13.htm (accessed on 28 October 2006). 119. Zientara S. (2005). Epidmiosurveillance des maladies transmission vectorielle. Deux exemples : West Nile et fivre catarrhale des ovins. In Proceedings of the Symposium of the Epidemiology and Animal Health Association [AESA: Association dEpidmiologie et de Sant Animale], Faculty of Veterinary Medicine, University of Lige, 12 May, 22-33.

Rev. sci. tech. Off. int. Epiz., 2007, 26 (2), 415-420

International harmonisation of regulatory requirements


M. Holmes (1) & R.E. Hill (2)
(1) Australian Pesticides and Veterinary Medicines Authority, PO Box E240, Kingston ACT 2604, Australia (2) Center For Veterinary Biologics, Animal and Plant Health Inspection Service, United States Department of Agriculture, 510 S. 17th Street, Suite 104, Ames, IA 50010, United States of America

Summary The International Cooperation on Harmonisation of Technical Requirements for Registration of Veterinary Medicinal Products (VICH) was formed in April 1996 and is a programme of collaboration between regulatory authorities and the animal health industries of three world regions: the European Union, Japan and the United States of America. Two other regions, Canada and Australia/New Zealand, have observer status. The principal goal of VICH is to harmonise technical data requirements of participating regulatory authorities before granting marketing authorisation or registration. VICH has finalised six guidelines on the technical requirements for marketing authorisation/registration of biological products. These guidelines have been fully implemented in the regions. Three more technical guidelines are under development by two expert working groups. VICH has also finalised a guideline which specifically deals with pharmacovigilance and veterinary medicinal products, including biological products. A further four guidelines relating to pharmacovigilance are under development by an expert working group. Keywords Harmonisation Mycoplasma contamination Pharmacovigilance Regulatory requirements Residual formaldehyde Residual moisture Reversion to virulence Specification Stability testing Vaccine VICH.

Introduction
Under the auspices of the World Organisation for Animal Health (OIE), three main regions (the European Union [EU], the United States of America [USA], Japan) and two observer regions (Canada and Australia/New Zealand) seek to harmonise regulatory data requirements for marketing authorisation for pharmaceutical and biological veterinary medicinal products (VMPs). This article gives an update on the present situation for biologicals and outlines future perspectives. Many of the worlds veterinary authorities require VMPs (biologicals and pharmaceuticals) to be granted marketing authorisation before the products can be distributed in their countries. In some countries the alternate term registration is used. This degree of regulatory control is consistent with one of the missions of the OIE: To improve

the legal framework and resources of national Veterinary Services. International harmonisation objectives are consistent with improving this framework.

The role of the International Cooperation on Harmonisation of Technical Requirements for Registration of Veterinary Medicinal Products
The International Cooperation on Harmonisation of Technical Requirements for Registration of Veterinary Medicinal Products (VICH) was formed in April 1996 and

416

Rev. sci. tech. Off. int. Epiz., 26 (2)

is a programme of collaboration between the regulatory authorities and the animal health industry of the EU, Japan and the USA. Canada and Australia/New Zealand have observer status at VICH, and participate at both the Steering Committee and Expert Working Group levels. The OIE participates as an associate member in the VICH process, and assists by supporting and disseminating the outcomes worldwide. VICH provides a forum for a constructive dialogue between regulatory authorities and the animal health industry on harmonising the regulatory technical requirements for VMPs within the VICH regions. The objectives of VICH are to: establish and implement harmonised regulatory requirements for VMPs in the VICH regions which meet high quality, safety and efficacy standards and minimise the use of test animals and the costs of product development provide a basis for wider international harmonisation of registration requirements monitor and maintain existing VICH guidelines, taking particular note of the International Conference on Harmonisation of Technical Requirements for the Registration of Pharmaceuticals for Human Use (ICH) work programme, and where necessary, update these VICH guidelines ensure efficient processes for maintaining and monitoring consistent interpretation of data requirements following the implementation of VICH guidelines provide technical guidance (by means of a constructive dialogue between regulatory authorities and industry) that enables an effective response to be made to significant emerging global issues and scientific developments that impact on regulatory requirements within the VICH regions. VICH also provides guidance to members on the technical requirements for authorisation/registration of VMPs in order to protect public health, animal health, animal welfare and the environment. VICH achieves its objectives through the development of guidelines by expert working groups. These groups are composed of recognised experts on the topic and are taskoriented, with their primary objective being the development of a draft guideline that will be released for consultation by the Steering Committee, and subsequently implemented by the regulatory authorities in each of the regions. Members of expert working groups and the VICH Steering Committee work towards the harmonisation of technical requirements by way of consensus in accordance with

established procedures. All guidelines are published on the VICH website (http://www.vichsec.org). To finalise guidelines, VICH uses a nine-step process which includes: formation of an expert working group to prepare drafts of the guideline approval by the VICH Steering Committee for public release of draft guidelines for comment release of the final guideline to regulatory authorities for implementation revision of guidelines. To date, VICH has finalised 36 guidelines. Eight guidelines are at advanced stages of development and consultation, six guidelines are at an early stage of development and a further five topics are potential future guidelines. VICH currently categorises its working groups into five broad areas: quality pharmacovigilance target animal safety biologicals quality monitoring metabolism and residue kinetics. Each working group may work on one or several VICH guidelines.

Guidelines for veterinary biological products


Five VICH guidelines (GL), all of which are available from the VICH website, are relevant to the technical requirements for marketing authorisation/registration of VMPs that have been developed and implemented in each of the member and observer regions. One more technical guideline is under development by an expert working group. One guideline relevant to pharmacovigilance has been finalised and an additional four pharmacovigilance guidelines are under development by an expert working group.

Stability testing: GL3 and GL17


The stability of vaccines and biologicals must be determined to ensure that the storage conditions under which they are designed to be held are appropriate.

Rev. sci. tech. Off. int. Epiz., 26 (2)

417

The aim is to determine the optimum storage conditions (such as ideal temperature and protection from light) which will maintain the efficacy of the product up to the end of its shelf life.

guideline provides public assurance that the clinical study data are reliable and that due regard has been given to animal welfare and to the protection of the personnel involved in the study, the environment and the human and animal food chains.

GL3: Stability testing of new veterinary drug substances and medicinal products
GL3 is a generic guideline for all VMPs. This guideline describes the core stability data package required for new drug substances and products. The guideline provides a general indication of the requirements for stability testing, but leaves sufficient flexibility to encompass the variety of different practical requirements necessary for specific scientific situations and for the particular characteristics of the materials being evaluated.

Testing for residual formaldehyde: GL25


Many inactivated veterinary vaccines, particularly bacterins, contain residual levels of formaldehyde. It is important to determine the residual level of formaldehyde to: help assure product safety ensure that the product will not inactivate other products used in combination help assure that the product remains active throughout its shelf life ensure that any clostridial toxoids will be antigenic and safe. GL25 is a guideline for the general requirements for residual formaldehyde testing. The guideline allows for flexibility for other testing methods based on specific scientific situations or the characteristics of the target material.

GL17: Stability testing of new biotechnological/biological veterinary medicinal products


While GL3 applies in general to new biotechnological/ biological products, GL17 recognises that specific biotechnological/biological products may have distinguishing characteristics, the stability of which should be evaluated using a well-defined testing programme that can confirm that the products efficacy would be maintained during the intended storage period. Thus, GL17 covers the generation and submission of stability data for products such as cytokines, growth hormones and growth factors, insulins, monoclonal antibodies, and those vaccines which consist of well-characterised proteins or polypeptides even when chemically synthesised. GL17 provides guidance on: selection of batches for the generation of stability data for submission to the regulatory authority selection of the assay or parameter that profiles the stability characteristics of the product storage conditions for stability studies, such as temperature and humidity studies on stability under conditions of heat stress, such as may occur during transport stability after reconstitution and in-use stability of multiple-use vials.

Testing for residual moisture: GL26


Freeze-dried veterinary vaccines always contain some water, commonly known as residual moisture (RM). It is important to determine the level of RM in final products because a satisfactory test gives assurance of an adequate shelf life and that the manufacturers freeze-dry cycle was properly controlled. The RM test should confirm that moisture level is consistently within the manufacturers specification. GL26 is a guideline on the general requirements for RM testing. The guideline allows for flexibility for other test methods based on specific scientific situations or the characteristics of the target material.

Guidelines relevant to pharmacovigilance: GL24, 29, 30, 35, 42


Pharmacovigilance is the process of monitoring the ongoing safety and efficacy of marketed products after they have received marketing authorisation/registration. Pharmacovigilance provides essential feedback to product manufacturers and also to regulatory authorities. Manufacturers need pharmacovigilance data for purposes of product warranty, product improvement and future

Good clinical practice: GL9


This VICH guideline is intended to be an international ethical and scientific quality standard for designing, conducting, monitoring, recording, auditing, analysing and reporting clinical studies that evaluate veterinary products, including biological products. Adherence to this

418

Rev. sci. tech. Off. int. Epiz., 26 (2)

product development. Regulatory authorities need pharmacovigilance data to provide feedback on the integrity of the regulatory process, and more importantly, to provide ongoing assurance that the products are safe for animals and humans. Four of these guidelines are currently in the advanced stages of final drafting and consultation before being released for adoption by regulatory authorities. Together, the five guidelines form a suite which provides a comprehensive framework for submitting, receiving and analysing adverse event reports (AERs), using uniform terminology.

Specifications are one part of a total control strategy designed to ensure product quality and consistency. Other parts of this strategy include thorough product characterisation during development (upon which many of the specifications are based), adherence to good manufacturing practices, a validated manufacturing process, raw materials testing, in-process testing, stability testing, etc. Specifications are chosen to confirm the quality of the drug substance and medicinal product rather than to establish full characterisation and should focus on those molecular and biological characteristics found to be useful in ensuring the safety and efficacy of the product.

Testing for Mycoplasma spp. contamination: GL34


It is important that biological products for veterinary use are free of contamination with Mycoplasma spp. to help assure consistency of production and final product safety. Mycoplasma contaminants may be introduced into cell culture and in ovo-origin biological products through the master seeds, the master cell seed (stock) or starting materials of animal origin, and in processing of biological materials during passage and product assembly. Therefore, it is necessary to demonstrate through testing that Mycoplasma are not present in the final product, working seeds and cells and harvests, or starting materials such as the master seed, master cell seed, or ingredients of animal origin. GL34 gives guidance on the stages of manufacture to be tested and test procedures to detect the presence of Mycoplasma contamination. GL34 is currently in an advanced stage of final drafting and consultation before being released for adoption by regulatory authorities.

Examination of live veterinary vaccines in target animals for absence of reversion to virulence: GL41
The absence of reversion to, or increase in, virulence is a fundamental requirement for all live vaccines. Live vaccines replicate in the animal and stimulate a useful immune response. Live vaccines generally cannot be completely characterised by chemical and physical tests alone. For these reasons, a test for absence of reversion to virulence is of critical importance. GL41 is a draft guideline which gives guidance for studies to be conducted on the master seed, by passaging in suitable animals, to check for reversion to virulence. If available data or assessment indicate a substantial risk that the test organism may revert to or increase in virulence, additional studies may be required to provide further information on the organism.

Target animal safety testing for live and inactivated vaccines: GL44 Specifications test procedures and acceptance criteria for new biotechnological/biological veterinary medicinal products: GL40
A specification is defined as a list of tests, references to analytical procedures, and appropriate acceptance criteria such as numerical limits, ranges, or other criteria for the tests described. A specification establishes the set of criteria to which a veterinary product should conform to be considered acceptable for its intended use. Conformance to specification means that the product, when tested according to the listed analytical procedures, will meet the acceptance criteria. Specifications are critical quality standards that are proposed and justified by the manufacturer and approved by regulatory authorities as conditions of approval. This important guideline is currently under development by an expert working group. It is a contribution towards international harmonisation and standardisation of methods used to evaluate the target animal safety of new veterinary vaccines. The guideline is designed to aid sponsors in preparing protocols for target animal safety studies conducted under laboratory conditions and in related field studies.

Conclusion
The regulation by national veterinary authorities of the quality and safety of veterinary vaccines and biologicals makes a significant contribution to the ongoing usefulness of vaccines and biologicals in the prevention and treatment

Rev. sci. tech. Off. int. Epiz., 26 (2)

419

of animal disease. However, regulation by itself has limited effect unless the manufacturing companies manufacture product to the highest quality standards, and provide comprehensive data dossiers for evaluation by the regulatory authorities. The value of the VICH guidelines lies in their joint development by technical experts from industry, academia and regulatory authorities. This collaboration provides guidance documents consistent with current scientific

methods, and methods recognised worldwide as the standard for testing and evaluating VMPs. The VICH guidelines have made a significant contribution towards internationally harmonised technical standards for veterinary vaccines and biologicals.

Harmonisation internationale des dispositions rglementaires


M. Holmes & R.E. Hill
Rsum La Coopration internationale sur lharmonisation des exigences techniques applicables lenregistrement des mdicaments vtrinaires (VICH) a t cre en avril 1996 afin dorganiser la collaboration entre les autorits rglementaires et lindustrie pharmaceutique en sant animale dans trois rgions du monde : lUnion europenne, le Japon et les tats-Unis dAmrique. Deux autres rgions ont un statut dobservateur : le Canada et lAustralie/Nouvelle-Zlande. La VICH a pour principal objectif dharmoniser les cahiers des charges techniques imposs par les autorits rglementaires en vue de lautorisation de mise sur le march ou de lenregistrement dun mdicament vtrinaire. La VICH a prpar six lignes directrices sur les exigences techniques relatives lautorisation de mise sur le march/enregistrement des produits biologiques. Ces lignes directrices sont intgralement appliques dans les trois rgions. Trois autres lignes directrices sont en prparation, sous la responsabilit de deux groupes de travail spcialiss. La VICH a galement produit une ligne directrice sur la pharmacovigilance et les mdicaments vtrinaires, y compris les produits biologiques. Quatre nouvelles lignes directrices lies au thme de la pharmacovigilance sont en prparation sous la conduite dun groupe dexperts. Mots-cls Contamination par Mycoplasma Dispositions rglementaires preuve de stabilit Harmonisation Humidit rsiduelle Pharmacovigilance Rsidu de formaldhyde Rversion vers la virulence Spcification Vaccin VICH.

420

Rev. sci. tech. Off. int. Epiz., 26 (2)

Armonizacin internacional de los requisitos reglamentarios


M. Holmes & R.E. Hill
Resumen La Cooperacin Internacional para la Armonizacin de los Requisitos Tcnicos para el Registro de Medicamentos Veterinarios (VICH), que ech a andar en abril de 1996, es un programa de colaboracin entre las instancias reglamentarias y la industria zoosanitaria de tres regiones del mundo: la Unin Europea, Japn y los Estados Unidos de Amrica, ms otras dos regiones, Canad y Australia/Nueva Zelanda, que participan en calidad de observadoras. La VICH tiene por objetivo primordial armonizar los requisitos de datos tcnicos que imponen las instancias normativas participantes para registrar un producto u otorgar licencia de comercializacin. La VICH ha elaborado seis directrices sobre requisitos tcnicos para autorizar la comercializacin o registrar productos biolgicos, y todas ellas se han aplicado integralmente en las tres regiones citadas. Ahora mismo hay dos grupos de expertos que estn elaborando otras tres directrices tcnicas. La VICH tambin tiene ya ultimada una directriz dedicada especficamente a la farmacovigilancia y los medicamentos veterinarios includos los productos biolgicos. Un grupo de expertos trabaja actualmente para elaborar otras cuatro directrices sobre farmacovigilancia. Palabras clave Armonizacin Contaminacin por micoplasma Especificacin Farmacovigilancia Formaldehdo residual Humedad residual Prueba de estabilidad Requisitos reglamentarios Reversin a la virulencia Vacuna VICH.

Rev. sci. tech. Off. int. Epiz., 2007, 26 (2), 421-428

Authorisation within the European Union of vaccines against antigenically variable viruses responsible for major epizootic diseases
D.K.J. Mackay
European Medicines Agency (EMEA), 7 Westferry Circus, London E14 4HB. Email: david.mackay@emea.europa.eu The views expressed in this article are the personal views of the author and should not be taken to represent the position of the EMEA, the Committee for Medicinal Products for Veterinary Use, or the European Commission

Summary Antigenically variable viruses are responsible for some of the most contagious and economically important diseases that affect domestic livestock. The serious consequences of such diseases in terms of economic loss, and human and animal health, were clearly demonstrated by recent epizootics of foot and mouth disease, and outbreaks of avian influenza and bluetongue in the European Union (EU). For such diseases, government authorities need to be able to respond, if appropriate, by making use of vaccines that are suited to the epidemiological situation. The current EU regulatory framework is not well adapted for approval and maintenance of vaccines where the antigens included have to be chosen to reflect the epidemiological need. An extensive revision of the technical requirements for authorisation of veterinary medicinal products within the EU is currently underway. Additionally, a major revision of the regulations that control how such authorisations are kept up-to-date is about to start. This provides an ideal opportunity to introduce into EU legislation the concept of the multistrain dossier whereby a potentially large number of approved strains may be included within a marketing authorisation and the final vaccines may be blended to include strains according to need. In addition, new strains may be added onto the marketing authorisation by means of a rapid regulatory procedure should new antigenic variants actually or potentially threaten the EU. Keywords Authorisation Avian influenza Bluetongue European Union Foot and mouth disease Licensing Vaccine.

Introduction
The recent experience of Europe with respect to foot and mouth disease (FMD), avian influenza (AI) and bluetongue (BT) has raised the issue of vaccination against these antigenically variable viruses in the mind of the general public and of those responsible for controlling these highly contagious animal diseases. This article discusses the reasons why there is a need for such vaccines to be

authorised, the factors that currently act as a disincentive to authorisation and the proposals currently under discussion aimed at promoting their authorisation. The latter part of the 20th Century saw the introduction of regulatory systems to ensure the consistent quality of medicines in all of the major developed markets of the world, first for human medicines and subsequently for veterinary medicines. The main driver for developing these

422

Rev. sci. tech. Off. int. Epiz., 26 (2)

regulatory requirements was the increasing recognition by governments and consumers of the need for medicines to be safe, of high and consistent quality, and efficacious. These requirements inevitably lead to increased costs for manufacturers both to meet the required standards and to put in place suitable systems of quality assurance. These costs can fairly readily be recouped in the case of products with a substantial and predictable market. In contrast, in the case of diseases which are exotic to the country or region concerned, or are subject to successful eradication programmes, there is little incentive for industry to invest in meeting the costs of authorising a vaccine for which the use and financial return are uncertain. On the other hand, in the case of such diseases, the consequences of not vaccinating are frequently not acceptable due to the risk of uncontrolled spread. Therefore, veterinary authorities will often seek to obtain and use an appropriate vaccine, whether or not it has met the usual regulatory requirements of the country or region concerned. In addition to the financial disincentives, the highly complex antigenic nature of the viruses responsible for FMD, AI and BT presents particular problems in terms of authorising vaccines. Each of these viruses exist as multiple serotypes wherein the definition of a serotype is that vaccination against, or previous infection with, one serotype does not provide protection against other serotypes. Furthermore, in the case of FMD and AI, the extent of variation within each serotype is sufficiently great that not all strains within a serotype will cross-protect against each other. Each virus has its own particular mechanism by which it maintains antigenic diversity. In the case of FMD virus, this phenotypic diversity is the result of the quasispecies nature of small ribonucleic acid (RNA) viruses. The inherent imprecision of the replication mechanism of the genome results in the constant production of progeny viruses that are slightly different from their parent. Added to this, simultaneous infection with more than one strain and/or serotype of FMD virus is not uncommon and can lead to recombination. In the case of BT virus there are 24 serotypes and the virus has a segmented genome. Again, co-circulation of different serotypes is normal in endemic areas, thereby creating ideal conditions for reassortment. In the case of AI, as with other influenza viruses, antigenic drift results in the constant evolution of the antigenic nature of the prevalent strains in a population and antigen shift, as a result of reassortment, can result in the production of antigenically novel viruses with the consequent potential for pandemic spread. It is not the intention of this paper to cover in detail the mechanisms whereby these three viruses maintain antigenic diversity. What is relevant is to emphasise the following features which these viruses have in common that present particular challenges from a regulatory perspective:

they are antigenically complex and there is therefore a need to have in place a wide range of authorised vaccine strains there is a constant need to monitor the field situation to ensure that vaccine strains remain relevant and to have in place a mechanism to change strains rapidly when required in each case there is the potential for the rapid introduction, or development, of antigenically novel variants against which existing vaccine strains are ineffective and which therefore require the development, validation, authorisation and introduction into manufacturing of new master seed viruses in as short a space of time as possible.

Why authorise vaccines?


In view of the expense and administrative complexity that authorisation of vaccines introduces, it is important to understand why it is desirable that vaccines are authorised. As indicated in the introduction, authorisation provides assurance to the customer as to the quality, safety and efficacy of the product. In the case of vaccines used under national authority control, the customer can be said to be the national authority that purchases the vaccine from the manufacturer. National authorities see authorisation as a means of assuring the efficacy of the product without themselves having to conduct the necessary trials. In the case of all three diseases under consideration, experimental trials involving challenge with live viruses can only be conducted under highly contained conditions. The exact category of containment depends on the nature of the agent concerned but is the highest level for animals in the case of FMD virus and the highest level for both man and animals in the case of H5 highly pathogenic avian influenza viruses. Such facilities are extremely expensive to build and maintain and access to them is therefore limited on both logistical and financial grounds. For these reasons, as well as to reduce the need for testing in animals, it is highly advantageous if vaccines are authorised by an independent body based on compliance to agreed standards resulting from accumulated scientific data, thereby reducing or replacing the need for exhaustive and expensive tests in animals. In the case of compulsory vaccination campaigns, national authorities have an obligation to provide the best possible assurance of the quality of the products used, since the stockholders have no choice as to whether or not their animals are vaccinated. In this respect, the additional assurance that is provided by independent assessment of the quality and safety of the products is extremely helpful in terms of reassurance both to the stockholders and to the

Rev. sci. tech. Off. int. Epiz., 26 (2)

423

consumers of products from vaccinated animals. Recent experience with both FMD and AI has shown a reluctance on the part of some wholesalers and retailers to accept products from vaccinated animals, resulting in them opting to source produce from other, unvaccinated animals. Much of this reluctance results from ignorance of common agricultural practices and can be countered by education campaigns. However, the use of authorised vaccines provides an added level of assurance as to the safety of the vaccines themselves, and of products from animals treated with them, which is useful when seeking the support of retail organisations, stakeholder groups and independent food standards agencies.

The challenges of authorising vaccines against antigenically variable viruses


Authorisation of conventional vaccines provides a high degree of regulatory certainty by permitting manufacturers to produce a product in approved premises according to Good Manufacturing Practice (GMP) and in line with the terms of a marketing authorisation (the EU term marketing authorisation is used throughout this paper, but in other regulatory areas such an approval may be termed a product licence or permit). The marketing authorisation specifies the raw materials that are to be used, the manufacturing method to be followed and the inprocess and final tests that must be performed to demonstrate the quality and consistency of the final product. A variation procedure must be approved by the licensing authority before any changes can be made to the terms of the authorisation. Thus, in the case of a vaccine, the authorisation defines a fixed formulation consisting of specified amounts, or limits, of one or more antigens derived from pre-determined master seeds blended with specified excipients and/or adjuvants. This definition is not suited to vaccines against highly antigenically variable viruses for a number of reasons. Manufacturers will generally blend vaccines according to the epidemiological situation of the region in which they are to be used. Thus, vaccines may contain antigens derived from one or more of a wide range of master seed viruses. Under normal regulatory requirements, manufacturers would be required to demonstrate the safety and efficacy of each antigen, both individually and in combination, something which is clearly impractical when manufacturers may use any combination of up to 20 or more master seeds. Testing the safety, efficacy, onset and duration of immunity for each of 20 strains as monovalent vaccines would occupy a high containment unit for several years. Add to this trials for bi-, tri- or quadrivalent vaccines, and the myriad combinations of possible

antigens, and it is immediately apparent that a new approach is required to the authorisation of such vaccines. Under existing legislation, inclusion or substitution of a new strain into an existing vaccine would require a new authorisation which could take up to 210 days for approval. Again, a new approach is required as this is clearly unacceptable for diseases such as FMD, BT and AI where there is the potential for pandemic spread. The Committee for Medicinal Products for Veterinary Use (CVMP) of the European Medicines Agency started considering this issue following the outbreak of FMD in the United Kingdom (UK) and several other European Union (EU) Member States in 2001, ultimately resulting in the publication of the Position Paper on Requirements for Vaccines against Foot-and-Mouth Disease (EMEA/CVMP/ 775/02 adopted June 2004) (3) and the Guideline on Requirements for Vaccines for Use in Birds against Avian Influenza (4).

Exceptional authorisation and use of vaccines without an authorisation


To date, the solution to these problems has been sought on a Member State level by using unauthorised vaccines or vaccines authorised under exceptional circumstances. Article 8 of Directive 2001/82/EC (as amended by Directive 2004/28/EC) (5) permits Member States to use immunological medicinal products without an authorisation in the event of serious epizootic disease. Article 7 of the same Directive permits Member States to use products authorised in another Member State where the health situation so requires, but this is of little use where the necessary vaccines are not authorised in any Member State. Article 26 (3) of Directive 2001/82/EC for nationally authorised products, and Article 39 (7) of Regulation 726/2004 for centrally authorised products (6), provide for authorisation under exceptional circumstances for objective and verifiable reasons, such as the need to combat outbreaks of contagious, exotic disease and where it is not possible to conduct all of the usual tests (see below). In such circumstances some of the usual requirements for authorisation can be made into specific obligations to be carried out by the marketing authorisation holder as a condition of receiving an authorisation. These take the form of commitments to conduct studies or provide additional data to an agreed timetable after the authorisation has been issued, thereby allowing a more rapid authorisation procedure. However, such authorisations do not represent a real solution to the problem as they are reactive by nature and, due to the necessity for an annual review, burdensome in administrative terms, making them expensive both for national authorities and manufacturers.

424

Rev. sci. tech. Off. int. Epiz., 26 (2)

Routes to authorisation of medicinal products within the European Union


There are two main routes to authorisation of medicinal products within the EU. In the centralised procedure, applicants apply to the European Medicines Agency (EMEA) for an application to be considered by the CVMP. Following a positive opinion from the CVMP, the European Commission issues a decision granting a marketing authorisation that is valid in all 27 Member States of the EU as well as Norway, Iceland and Liechtenstein. In the national, mutual recognition or decentralised procedures, national marketing authorisations are issued in each Member State in which authorisations are sought and the Coordination Group for Mutual Recognition and Decentralised Procedures (veterinary) (CMDv) ensures that the authorisations issued are harmonised. Following the review of EU pharmaceutical legislation in 2001, several amendments were introduced to promote authorisation of vaccines against major epizootic diseases through the centralised procedure. Commission Regulation (EC) 726/2004 (6) opens up the scope of the centralised procedure to any immunological veterinary medicinal product that is used for the treatment of animal diseases that are subject to Community prophylactic measures. This therefore includes conventional, inactivated vaccines against FMD, BT and AI. In addition, it is obligatory for any vaccine produced using recombinant deoxyribonucleic acid (DNA) technology to be authorised through the centralised procedures, including recombinant or vector vaccines against these three diseases. Authorisation of vaccines to be used as part of Community campaigns through the centralised procedure has many advantages from an EU perspective in that a single authorisation permits an identical product of proven quality to be available for use in all Member States. As mentioned above, vaccines may be authorised under exceptional circumstances through both the centralised and national procedures. An authorisation under exceptional circumstances permits Member States and the Commission to issue authorisations subject to specific obligations. This provision is necessary, for example, to exempt the manufacturer from having to conduct field trials before authorisation in view of the fact that community legislation prohibits vaccination against exotic diseases except as part of an approved vaccination campaign. Instead, the marketing authorisation holder will be required to put in place a system for enhanced pharmacovigilance (i.e. detection of side effects and adverse reactions) in the event that the product is used within the EU. In addition, the revised legislation provides for an accelerated assessment as part of the centralised

procedure of up to a maximum of 150 days for products of major importance in terms of animal health. The provisions for accelerated assessment and exceptional authorisation were both used successfully in 2006 to grant central authorisations within a total period of four months for two vaccines against AI so that authorised vaccines were available throughout the Community in time for the period of increased risk in the autumn.

The need for a specific approach to antigenically variable viruses: the concept of the multistrain dossier
Whilst all of these measures are certainly helpful in promoting the authorisation and use of vaccines against diseases such as FMD, BT and AI within the EU, they are not, in themselves, likely to act as a major incentive to authorisation. The regulatory model of one authorisation = one fixed formulation does not adequately take into account the complex and varied requirements for vaccines against these diseases. The technical requirements that must be met for a marketing authorisation to be initially granted are defined in Annex 1 to Directive 2001/82/EC (5). The requirements for varying a marketing authorisation once granted are defined in the variations regulations (Commission Regulations (EC) No. 1084/2003 & 1085/2003) (1, 2). Both of these sets of requirements are currently being revised by the Commission in consultation with Member States and this provides an ideal opportunity to introduce a more flexible approach to authorisation of antigenically variable viruses. This approach has been termed the Multistrain Dossier concept. The detailed guidance on how such a concept could operate in practice has yet to be elaborated. The following proposals develop the approach outlined in the CVMP Position Paper on Requirements for Vaccines against Foot-andMouth Disease and offer one potential solution. In this proposal, the authorisation for a multistrain dossier would include a number of defined, approved master seed viruses and would specify the maximum number of antigens derived from these master seeds that may be incorporated into a vaccine blend. Within these parameters, the marketing authorisation holder may then choose the actual number and type of antigens to be blended in a particular batch for a particular epidemiological situation. The amount of each antigen blended would be constrained within limits agreed with the regulator based on safety and efficacy data submitted at the time of approval of the master seed for the authorisation. This approach presupposes that all of the antigens on the authorisation are

Rev. sci. tech. Off. int. Epiz., 26 (2)

425

essentially similar in all respects other than their antigenic nature. This assumption is only valid where manufacturers operate to strict standards of GMP, thereby tightly controlling all aspects of the manufacturing process such that there is a high reproducibility of production, both between different batches of the same antigen and between different antigens. This approach is only likely to be feasible for modern generation vaccines in which the antigen is well defined and, preferably, purified before blending. Provided this assumption is valid, it is then possible to extrapolate certain parameters between antigens, reducing or removing the need to repeat studies for all antigens, and all combinations of antigens, included within the authorisation. These parameters include safety, in terms of inducing adverse local and systemic reactions, and immunogenicity, in terms of onset, extent and duration of immunity. To demonstrate that extrapolation is valid, manufacturers need to supply appropriate data on batch production and testing records. These data should show the expected consistency of production and of results for in-process and final testing of stability, antigen content, batch safety and potency. Further work is required within the regulatory community to define more precisely what data is required to accept a new master seed virus, and antigen produced from that master seed, onto the authorisation for a multistrain dossier, both at the time of authorisation and subsequently. Different requirements may apply, particularly in relation to testing for the presence of extraneous agents, depending on the urgency of the situation and therefore whether or not authorisation is sought under exceptional circumstances (see section on Exceptional authorisation and use of vaccines without an authorisation above). In technical terms, to add a new master seed, the authorisation holder should demonstrate that antigen from the new seed can be produced using the method of production described in the dossier. The results of inprocess and final product tests should demonstrate equivalence of the new antigen to already approved antigens. In order for the multistrain dossier approach to work in practice there needs to be a legislative mechanism that allows new strains to be added to an authorisation more quickly than is possible under the current line extension procedure, which can take up to 210 days. This could either take the form of a shortened line extension procedure or, preferably, an amendment to the legislation which allows the addition of a strain onto a multistrain

dossier through a rapid variation procedure. An analogous situation already exists for human influenza vaccines whereby a change of strain to reflect the prevailing epidemiological situation is completed by means of a variation procedure within a maximum of 60 days. Furthermore, there is a legal provision for immediate acceptance of a variation in pandemic situations for influenza or other pandemic human viruses (Articles 7 and 8 of Commission Regulations (EC) No. 1084/2003 and 1085/2003) (1, 2). There is clearly value in adopting a similar approach on the veterinary side as part of the measures necessary to make the multistrain dossier approach a feasible option. In the case of vaccines for human influenza, approval of new strains is based largely on an assessment that the analytical section of the dossier demonstrates that the new antigen is of appropriate quality, together with some limited serology data. As a condition of approval, marketing authorisation holders enter into commitments to provide confirmatory evidence of efficacy in the form of data from clinical trials conducted postapproval. A similar approach would be helpful on the veterinary side in order to rapidly approve new strains in the event of epizootic situations.

Conclusion
Recent experiences with FMD, BT and AI in the EU have emphasised the need for national authorities to have access to authorised vaccines to be used as part of the control strategy for these major epizootic diseases. Amendments to the regulatory requirements would act as a significant incentive to manufacturers to authorise their vaccines. The current review of the technical requirements for obtaining and maintaining marketing authorisations within the EU presents an ideal opportunity to introduce the concept of the multistrain dossier. This approach would more closely suit the particular requirements of these antigenically variable viruses and thereby promote the authorisation of high quality vaccines.

Acknowledgements
The author is grateful to Dr Nikolaus Kriz, EMEA, for review of this manuscript.

426

Rev. sci. tech. Off. int. Epiz., 26 (2)

Lagrment au sein de lUnion europenne des vaccins dirigs contre des virus pizootiques majeurs prsentant une variabilit antignique
D.K.J. Mackay
Rsum Les virus prsentant une variabilit antignique sont lorigine de maladies parmi les plus contagieuses et les plus importantes du point de vue conomique qui affectent le btail domestique. Les rcentes pizooties de fivre aphteuse et les foyers dinfluenza aviaire et de fivre catarrhale du mouton survenus dans lUnion europenne (UE) tmoignent des pertes conomiques que ces maladies peuvent occasionner ainsi que de leurs consquences nfastes pour la sant humaine et animale. Face ces maladies, les pouvoirs publics doivent tre mme dapporter une rponse approprie, notamment en recourant des vaccins adapts chaque situation pidmiologique. lheure actuelle, le cadre rglementaire de lUE ne permet pas dautoriser ni de maintenir des vaccins lorsque le choix de lantigne doit se faire ultrieurement, en fonction des ncessits pidmiologiques. Lensemble des exigences techniques requises pour lautorisation de mise sur le march des mdicaments vtrinaires sont actuellement en cours de rvision au sein de lUE. En outre, les rglementations applicables la mise jour de ces autorisations sont galement sur le point dtre examines. La possibilit est ainsi offerte dintroduire dans la lgislation communautaire le concept de demande dautorisation multisouches , permettant aux autorisations de mise sur le march de couvrir un nombre potentiellement important de souches approuves de sorte que le vaccin final puisse tre formul en utilisant les souches ncessaires en fonction des besoins. De plus, grce une procdure rglementaire simplifie, de nouvelles souches pourront tre ajoutes aux autorisations de mise sur le march si de nouveaux variants antigniques viennent menacer lUE. Mots-cls Autorisation Autorisation de mise sur le march Fivre aphteuse Fivre catarrhale du mouton Influenza aviaire Union europenne Vaccin.

Rev. sci. tech. Off. int. Epiz., 26 (2)

427

Autorizacin de vacunas contra los virus con variaciones antignicas responsables de las principales enfermedades epizoticas en la Unin Europea
D.K.J. Mackay
Resumen Algunas de las enfermedades ms contagiosas y con mayores consecuencias econmicas que afectan a la ganadera de la Unin Europea (UE) pueden imputarse a virus que presentan variaciones antignicas. Las graves prdidas comerciales que ocasionan esas enfermedades, as como sus importantes repercusiones en la salud humana y animal, quedaron demostradas tras las epizootias de fiebre aftosa y los focos de influenza aviar y lengua azul que se produjeron en los ltimos tiempos. De ser preciso, las autoridades nacionales han de poder enfrentarlas mediante medidas de inmunizacin adaptadas a la situacin epidemiolgica. El marco reglamentario en vigor en la UE no es el ms conveniente para la aprobacin y el mantenimiento de vacunas cuyos antgenos deben poder seleccionarse en funcin del pronstico epidemiolgico. En la actualidad se estn examinando minuciosamente los requisitos tcnicos para la autorizacin de medicamentos de uso veterinario en la UE. Adems, est por iniciarse una profunda revisin de la reglamentacin sobre la modificacin de los trminos de esas autorizaciones. Se trata, pues, de una ocasin que convendra aprovechar para introducir en la UE el concepto de expedientes de multicepas, en virtud del cual se podra incluir un nmero potencialmente elevado de cepas aprobadas en las autorizaciones de comercializacin, y las vacunas finales podran combinarse de modo que comprendieran las cepas correspondientes a las necesidades. Adems, para prever la amenaza real o potencial de nuevas variantes antignicas en la UE, podra autorizarse la introduccin de nuevas cepas en esas autorizaciones mediante un mecanismo reglamentario rpido. Palabras clave Autorizacin Fiebre aftosa Influenza aviar Lengua azul Registro Unin Europea Vacuna.

428

Rev. sci. tech. Off. int. Epiz., 26 (2)

References
1. European Commission (2003). Commission Regulation (EC) No. 1084/2003 of 3 June 2003 concerning the examination of variations to the terms of a marketing authorisation for medicinal products for human use and veterinary medicinal products granted by a competent authority of a Member State. Off. J. Eur. Union, L 159, 1-23. Available at: http://ec.europa. eu/enterprise/pharmaceuticals/ eudralex/vol-1/reg_2003_ 1084/reg_2003_1084_en.pdf (accessed on 14 January 2007). 2. European Commission (2003). Commission Regulation (EC) No. 1085/2003 of 3 June 2003 concerning the examination of variations to the terms of a marketing authorisation for medicinal products for human use and veterinary medicinal products falling within the scope of Council Regulation (EEC) No. 2309/93. Off. J. Eur. Union, L 159, 24-45. Available at: http://ec.europa.eu/enterprise/ pharmaceuticals/eudralex/vol-1/reg_2003_1085/reg_2003_ 1085_en.pdf (accessed on 14 January 2007). 3. European Medicines Agency (EMEA) (2004). Position Paper on Requirements for Vaccines against Foot-and-Mouth Disease (adopted by the Committee for Medicinal Products for Veterinary Use [CVMP] June 2004). EMEA/CVMP /775/02. Available at: http://www.emea.europa.eu/pdfs/ vet/press/pp/077502en.pdf (accessed on 14 January 2007). 4. European Medicines Agency (EMEA) (2004). Guideline on Requirements for Vaccines for Use in Birds against Avian Influenza. EMEA/CVMP/222624/06. Available at: http:// www.emea.europa.eu/pdfs/vet/iwp/22262406en.pdf (accessed on 14 January 2007). 5. European Parliament (2001). Consolidated Directive 2001/82/EC of the European Parliament and of the Council of 6 November 2001 on the Community code relating to veterinary medicinal products (Off. J. Eur. Communities, L 311, 1-66) as amended by Directive 2004/28/EC (Off. J. Eur. Union, L 136, 58-84). Available at: http://ec.europa.eu/enterprise/pharmaceuticals/eudralex/vol5/consol_2004/veterinary_code.pdf (accessed on 14 January 2007). 6. European Parliament (2004). Regulation (EC) No. 726/2004 of the European Parliament and of the Council of 31 March 2004 laying down Community procedures for the authorisation and supervision of medicinal products for human and veterinary use and establishing a European Medicines Agency. Off. J. Eur. Union, L 136, 1-33. Available at: http://ec.europa.eu/enterprise/pharmaceuticals/eudralex/ vol1/reg_2004_726/reg_2004_726_en.pdf (accessed on 14 January 2007).

Rev. sci. tech. Off. int. Epiz., 2007, 26 (2), 429-441

Regulations for vaccines against emerging infections and agrobioterrorism in the United States of America
L.A. Elsken (1), M.Y. Carr (1), T.S. Frana (1), D.A. Brake (2), T. Garland (2), K. Smith (2) & P.L. Foley (1)
(1) United States Department of Agriculture (USDA), Animal and Plant Health Inspection Service, Veterinary Services, Center for Veterinary Biologics, 510 S. 17th St., Ames, Iowa, 50010, United States of America (2) United States Department of Homeland Security, Science and Technology Directorate, Washington, DC, United States of America

Summary The Virus-Serum-Toxin Act of 1913, as amended in 1985, provides the legal basis for the regulation of veterinary vaccines and related biological products in the United States of America (USA). The regulatory authority for the issuance of licences and permits that allow the shipment or importation of pure, safe, potent, and effective veterinary biological products lies with the Center for Veterinary Biologics (CVB), an agency of the United States Department of Agriculture (USDA). Under the standard licensing or permitting process, a manufacturer must develop and completely characterise and evaluate a product prior to licensure, and the CVB must review and evaluate the submitted information, audit and inspect the manufacturing facilities and methods of production and testing, and confirm key product test results through independent testing of product. This complete and comprehensive evaluation may not be possible in emergency situations, so processes and mechanisms are in place that allow for the more rapid availability of veterinary vaccines. Next generation vaccine development against foreign animal diseases such as foot and mouth disease is actively in progress in the USA and the authorities must ensure that there is an adequate supply of these vaccines in the National Veterinary Stockpile. Keywords Bioterrorism Disease Emergency Emerging Licensing Regulatory Vaccine Veterinary.

Introduction
Regulation of veterinary biological products in the United States of America (USA) began in 1913 with the passage of the Virus-Serum-Toxin Act by the United States Congress. The United States Department of Agriculture (USDA) is responsible for regulating veterinary biological products,

including, but not limited to, vaccines, bacterins, toxoids, antibodies, and antitoxins which are intended for use in the treatment of animals and which act primarily through the stimulation, supplementation, enhancement, or modulation of the immune system or the immune response. Because the term treatment, by definition, includes the prevention, diagnosis, management, or cure of diseases of animals, diagnostic test kits are also under

430

Rev. sci. tech. Off. int. Epiz., 26 (2)

regulatory control by the USDA. The current USDA regulatory programme consists of: review of all data developed by manufacturers in support of each product and product claim inspection of manufacturing processes and practices including equipment, facilities, materials, personnel, production, quality control, and records confirmatory testing of manufacturers biological seeds, cells, and product a post-licensing monitoring system of inspection and random testing of product post-marketing epidemiological surveillance of product performance under normal conditions of use. This combination of regulatory oversight before and after licensure assures the availability of pure, safe, potent, and effective veterinary biologics to veterinarians and animal owners. Under the standard licensing process, this spectrum of evaluation and review includes the complete characterisation, identification and purity testing of seed materials, product ingredients and the final product; environmental, laboratory, and host animal safety and efficacy studies; stability studies, and post-licensing monitoring of field performance. All aspects of this comprehensive conventional evaluation may not be possible during the emergence of a new animal disease agent or in an animal disease emergency such as in the intentional (agrobioterrorism) or unintentional introduction of a significant exotic animal disease agent. In these situations, the USDA has various mechanisms for expedited product approval. In addition, the USDA may also provide exemption of products from some or all of the regulatory requirements for conventional product approval. Implementation of these existing mechanisms and perhaps, the identification of other novel avenues for product approval will be critical for the acceleration and timely development of next generation foreign animal disease (FAD) vaccines and immune-based biotherapeutics. Recently, to expedite emergency disease response capabilities, the Center for Veterinary Biologics (CVB), an agency of the USDA, has partnered with other authorities in the USA, such as the USDAs National Veterinary Stockpile (NVS), and foreign regulatory authorities, such as the Tripartite Canada-Mexico-USA North American Foot and Mouth Disease Vaccine Bank, to purchase vaccine antigen concentrates and/or finished emergency use vaccines. In addition, the NVS is entering into contractual agreements with biologics manufacturers to ensure immediate access to existing stocks of licensed emergencyuse vaccines. These emergency preparedness products may be licensed and distributed under standard processes or

with exemption from some or all of the normal regulatory requirements.

Regulatory framework
The Virus-Serum-Toxin Act of 1913 (Title 21 of the United States Code Parts 151-159) provides the legal basis for the regulation of veterinary biologicals in the USA; the CVB has the regulatory authority for the issuance of licences and permits for such products. The law was amended in 1985 by the Food Security Act to include the distribution of all veterinary biologics (both interstate and intrastate) in the USA as well as those intended for export. Administrative regulations and standards appear in Title 9 of the Code of Federal Regulations (9 CFR) Parts 101-118, with additional programme guidance found in CVB Notices, Veterinary Services Memoranda, General Licensing Considerations and other guidance documents. Veterinary biologic products are defined in the regulations as all viruses, serums, toxins (excluding substances that are selectively toxic to microorganisms, e.g. antibiotics), or analogous products at any stage of production, shipment, distribution, or sale, which are intended for use in the treatment of animals and which act primarily through the direct stimulation, supplementation, enhancement, or modulation of the immune system or immune response. This includes, but is not limited to, vaccines, bacterins, allergens, antibodies, antitoxins, toxoids, immunostimulants, certain cytokines, antigenic or immunising components of live organisms, and diagnostic components that are of natural or synthetic origin or that are derived from synthesising or altering various substances or components of substances such as microorganisms, genes or genetic sequences, carbohydrates, proteins, antigens, allergens, or antibodies. The USDA is authorised to issue licences to veterinary biologics manufacturers in the USA. In addition to what are variously referred to as the conventional, regular or full veterinary biological product licences in the USA, conditional biologics licences and autogenous biologics licences may be issued for use in specific situations or under certain conditions. Thus, some consideration should be given to use these identified mechanisms for licence approval of next generation vaccines and immune-based biotherapeutics in order to reduce their overall time to operational readiness. The USDA is further authorised to issue three types of permits to allow the importation of veterinary biologics into the USA. A Permit for Transit Shipment Only is required for a biological product shipped from one non-US country to another non-US country by way of the USA. A biological product issued a Permit for Transit Shipment Only may not be used in the USA. A Permit for Research and Evaluation

Rev. sci. tech. Off. int. Epiz., 26 (2)

431

may be issued if the manufacturer provides adequate information for the USDA to assess the product and the products impact on the environment and the product investigator (user) demonstrates scientific capabilities adequate to safeguard domestic animals (all animals, other than man, including poultry) and protect public health, interest, or safety from any deleterious effects which might result from the use of such product. For this permit type, the user could be the Department of Homeland Security (DHS) Science and Technology (S&T) Directorate Foreign Animal Disease Biological Countermeasure Program at Plum Island Animal Disease Center (PIADC). This government facility, located off the north-eastern tip of New Yorks Long Island, is specifically designed to conduct research and development studies on FADs and thus is in a unique position to further evaluate experimental biologicals that may be of value for further development and licensure. A Permit for Distribution and Sale may only be issued if the manufacturer and the permittee can insure that the product is pure, safe, potent, and effective. A Permit for Distribution and Sale is essentially equivalent to a conventional Veterinary Biological Product License in the USA. For this permit type, DHS S&T could coordinate activities between the foreign vaccine manufacturer and biological company in the USA with a veterinary establishment licence. The USDAs Animal and Plant Health Inspection Service (APHIS) Administrator may also specifically exempt any veterinary biological product from one or more of the normal regulatory requirements under certain circumstances, e.g. if the product is to be used experimentally (see section entitled Exemption of biological products from licensing requirements) or if this product will be used by or under the supervision of the USDA in the prevention, control, or eradication of animal diseases in conjunction with: a) an official USDA programme, or b) an emergency animal disease situation, or c) a USDA experimental use of the product. DHS S&T can play an integral role in this exemption policy by leading in the testing and evaluation of experimental vaccine product candidates for FADs. The scientific data generated can be used, for example, to better inform foot and mouth disease (FMD) modelling studies. In addition, DHS S&T can directly test or facilitate the testing of FAD vaccines currently licensed and manufactured in foreign countries. Examples include licensed veterinary vaccines for FMD, highly pathogenic avian influenza, rinderpest, Rift Valley fever, and classical swine fever. These current products can be characterised to yield new scientific data (e.g. days to onset of protection, virus shed/spread, vaccine dose sparing, etc.) that are

important in a decision process immediately following outbreak diagnosis. All licences and permits issued by the CVB may include restrictions on the distribution and use of the product (this also applies to products that have been exempted from certain other requirements by the APHIS Administrator). In an animal disease emergency such as in the intentional (bioterrorism) or unintentional introduction of a significant exotic animal disease agent, these restrictions would include, but not be limited to, the following: Domestic distribution and use shall be under the supervision or control of USDA, APHIS, Veterinary Services, as part of an official USDA animal disease control program. Distribution in each State shall be limited to authorized recipients designated by proper State officialsunder such additional conditions as these authorities may require. DHS S&T can serve a coordinating role in obtaining scientific data that can be used by APHIS-Veterinary Services to help underpin decisions on the specific restrictions under which these exempted products would be used. Examples include data on the ability of test material to reduce pathogen shed (including the duration of reduction) and the ability of the product to be used in a Differentiating Infected from Vaccinated Animals (DIVA) diagnostic recovery phase programme. The CVBs Inspection and Compliance (CVB-IC) section is responsible for determining whether licensed and permitted companies are being properly inspected and whether the products they manufacture are being prepared, tested, and distributed according to all applicable regulations and requirements. Major CVB-IC activities include on-site inspections of manufacturing and quarantine facilities, control and release of product batches (serials), and post-licensing product monitoring (pharmacovigilance). Licensed and permitted manufacturing establishments are subject to comprehensive in-depth inspections at one to three year intervals. In-depth inspections include 14 categories of inspection, each of which includes numerous items suggesting records to audit and items for observation. The categories of inspection considered in all in-depth inspections include licences, personnel, facilities, equipment, sanitation, research, seeds and cells, production, final production, labels, testing, animals, distribution, and miscellaneous (including pharmacovigilance). Special inspections of establishments are also utilised to address issues such as: observation of pivotal pre-licence efficacy or safety studies auditing of production, quality control, or animal test records

432

Rev. sci. tech. Off. int. Epiz., 26 (2)

inspection of new facilities or equipment review of product distribution or pharmacovigilance records. Each licensee or permittee is required to furnish the CVB with representative samples of every vaccine master seed organism and master cell stock proposed for use in the preparation of veterinary biological products, and samples of each serial or subserial of finished veterinary biological product manufactured in or imported into the USA. The APHIS Administrator is authorised to cause these samples to be examined and tested for purity, safety, potency, and efficacy. A master seed organism or master cell stock found unsatisfactory by any test may not be used to prepare veterinary biological products. A serial or subserial found to be unsatisfactory by a required test prescribed in an approved Outline of Production or Standard Requirement is not in compliance with the regulations, and may not be released for distribution.

use in an animal disease emergency. DHS S&T PIADC can provide a proactive, coordinating role in the testing of master seed, cell stocks, and formulated vaccines for the presence of foreign animal disease agents. Eggs used in production of biological products must be acquired from specific-pathogen-free flocks. Purity and identification of master seed and master cell are confirmed by testing at the CVB. In addition to the first serials (batches of completed product) prepared under licence, a random sample of serials are subjected to pre-release purity testing at the CVB to verify manufacturers quality assurance/quality control on final product.

Safety
Products must be shown to be safe through a combination of safety evaluations. Master seeds and master cell stocks must be fully identified and characterised. Production passage levels (limits) are established for both seeds and cells. Master seeds for live products are tested for shed, spread, and reversion to virulence through backpassage studies in the host animal. Following a minimum of five passages in the host, recovered isolates are fully characterised using the same procedures used for the master seed. Demonstration of an acceptable level of attenuation must be shown. Other safety studies are required as appropriate (e.g. safe use in pregnant animals, environmental safety, safety of adjuvants in products for food-producing animals). Field safety studies designed to detect unexpected reactions that may not have been detected in product development are required before licensure. The CVB and DHS S&T must decide if the next generation FAD vaccines currently under development will be exempt from this requirement or if field safety vaccine trials in foreign countries with reported FAD will be required. Host animal tests are conducted at a variety of geographical sites using large numbers of susceptible animals representing all ages and husbandry practices for which the product is intended. Final product serials are subjected to safety testing primarily through in vivo animal tests.

Data and review requirements for conventional product licences or permits for distribution and sale
All regulations pertaining to product licences or permits for distribution and sale (data requirements, confirmatory testing by the CVB, inspection and compliance requirements see following sections) are contained in 9 CFR Parts 101-118. Pre-licensing data evaluation and review procedures are designed to assess the purity, safety, potency, and effectiveness of each product and support all product label claims. In order to fulfil these criteria, data from all phases of product development are evaluated against these key elements. This spectrum of evaluation includes complete characterisation and identification of seed material and ingredients, laboratory and host animal safety and efficacy studies, demonstration of stability, and monitoring of field performance. Specific purity, safety, potency, and efficacy requirements are described in the following paragraphs.

Efficacy and potency


All products must be shown to be effective according to the claims indicated on the label, and each batch (serial) of each product must demonstrate potency at least equal to that of the reference serial(s). As defined by regulations in the USA, efficacy is a product characteristic, demonstrated at least once prior to licensure, while potency is a batch measure, and is intended to confirm that a serial will be at least as effective as a known immunogenic serial. The two terms can be distinguished as follows: efficacy is the specific ability or capacity of a biological product to effect the result for which it is produced, when

Purity
All product components and ingredients must meet standards of purity and quality. Master seed, master cell stock, primary cells, ingredients of animal origin, and final products must be tested and shown to be free of extraneous microorganisms. This requirement is especially important in a scenario in which veterinary vaccines licensed and produced overseas would be considered for

Rev. sci. tech. Off. int. Epiz., 26 (2)

433

used under the conditions recommended by the manufacturer potency is the relative strength of a biological product as defined by test methods or procedures established by APHIS in the 9 CFR Standard Requirements or in the approved Outline of Production for the product. Efficacy is generally demonstrated by statistically valid host animal vaccination-challenge studies and must be correlated to the product potency assay. The following general considerations are applied to efficacy studies: immunogenicity studies must be conducted using minimum levels of antigen at the highest passage level from the master seed that is permitted for production; product must be prepared in production facilities on a scale representative of normal production; challenge methods and criteria for evaluating protection will vary with the immunising agent, but in general, tests are conducted under controlled conditions using seronegative animals of the youngest age recommended on the label; duration of immunity data is required for some existing products (e.g. for rabies) and for all newly licensed antigens; field efficacy studies may be considered where laboratory animal challenge models are not well established. Similarly, serologic data may be used to establish efficacy only when serology is indicative of protection; data is required for each species for which the product is recommended and for each route, dose, and regimen of administration; for products with two or more fractions (components), data demonstrating no antigenic interference is required; stability studies are required to set the expiration date on the label; potency tests correlated to host animal vaccination and designed to measure the relative strength of each serial must be developed prior to full licensure. In addition, each serial must be formulated and tested prior to marketing to ensure effectiveness and reproducibility of activity (potency) according to standards set at the time of licensing. Generally, this is accomplished through an established immune-mediated animal or in vitro assay or by using microbiological counts or virus titrations for live bacterial or viral products.

vaccines and biotechnology-derived veterinary biologics, and to assess proposals to import veterinary biologics into the USA. To facilitate the preparation of scientifically valid and credible risk analyses, the CVB has developed Summary Information Formats (SIFs) and Risk Assessment (RA) outlines to provide guidance to interested parties. The SIFs and RA outlines identify the relevant information that should be evaluated in veterinary biologics risk analysis, and may be downloaded from the CVB website. The SIF for conventionally-derived live vaccines is designed to identify the appropriate information that should be provided to properly characterise the vaccine microorganism, based on its microbiological and biological properties, and those of the parental microorganism from which the vaccine strain was derived. The SIF for Category I biotechnology-derived veterinary biologics is designed to identify the appropriate information that should be provided to properly characterise the recombinant microorganisms for inactivated biotechnology-derived veterinary biologics. Subcategories for inactivated biotechnology-derived veterinary biologics are as follows: I-A-1, bacterins, killed virus vaccines, and subunit vaccines I-A-2, recombinant antigens for use in diagnostic test kits I-B-1, monoclonal antibodies for therapeutic or prophylactic use I-B-2, monoclonal antibodies for use in diagnostic test kits I-C-1, synthetic peptides for therapeutic or prophylactic use I-C-2, synthetic peptides for use in diagnostic test kits I-D-1, nucleic acid-mediated vaccines I-D-2, nucleic acid-mediated diagnostic test kits. Inactivated biotechnology-derived microorganisms can be used in the manufacture of killed vaccines, subunit vaccines, and diagnostic kits. The characterisation of the inactivated microorganism centres on its molecular properties, as well as on those of the parental microorganism and on any deleted or donor genes. Obviously, it is not anticipated that inactivated microorganisms will pose a threat to the environment. Accordingly, for inactivated biotechnology-derived products, the veterinary biologics risk analysis process is used to ensure that the biotechnology-derived microorganism is properly characterised and inactivated. Risk analyses to evaluate proposed environmental releases of inactivated products are not conducted.

Risk analysis
The CVB uses risk analysis procedures to evaluate licence applications for all new live conventionally-derived

434

Rev. sci. tech. Off. int. Epiz., 26 (2)

The SIF for Category II biotechnology-derived veterinary biologics is designed to identify information needed to properly characterise microorganisms for biotechnologyderived live vaccines containing gene deletions and\or heterologous marker genes. The characterisation of the gene-deleted microorganism includes the molecular and biological properties of the vaccine microorganism, and of the parental microorganism receiving the genetic modifications. The SIF for Category III biotechnology-derived veterinary biologics is designed to identify information needed to properly characterise the vaccine microorganisms for biotechnology-derived live vector vaccines containing heterologous genes encoding immunising antigens and\or other immune stimulants. The characterisation of biotechnology-derived vector vaccines includes the molecular and biological properties of the recipient, the donor, and the recombinant master seed microorganism. The characterisation of the donor microorganism includes the properties of both the donated structural genes and their regulatory elements. It is anticipated that the majority of next generation molecular vaccines and/or immunebased biotherapeutics for FADs currently under development by DHS S&T will fall under this risk classification. A risk analysis prepared by the licensee or permittee must be submitted to the CVB for pre-licence evaluation of every Category I, II, or III biotechnology-derived product. The risk analysis should contain the most current version of the SIF and a risk assessment based on safety characteristics of the vaccine. A risk assessment outline for use in the preparation of risk analyses for biotechnology-derived products is available from the CVB. Risk analyses for environmental release must be prepared by licensees or permittees for new live conventionally derived and biotechnology-derived veterinary vaccines. The risk analysis needs to include environmental release assessments, which evaluate the safety characteristics of the vaccine microorganism within the context of the target environment. A SIF which identifies the information that should be included in release assessments for proposed environmental releases is available from the CVB. Prior to the first release of a live biotechnology-derived veterinary vaccine determined not to have a significant impact on the quality of the human environment, the CVB prepares an environmental assessment and advises the public of plans for field testing the vaccine. The CVB also provides public notice of their intent to issue a product licence or permit for the vaccine, provided the field test data support the conclusions of the environmental assessment and the issuance of a finding of no significant impact and the product meets all other requirements for licensing. The veterinary biologics risk analysis model for importing veterinary biologics centres on the risk of introducing

FADs into the USA through the importation of a contaminated veterinary biological product. This requires a thorough evaluation of all potential sources of contamination during the development and manufacture of the product. The SIF for importation of veterinary biological products identifies the information regarding the facilities, reagents, production procedures, and testing procedures that should be evaluated when preparing risk analyses for proposals to import veterinary biological products into the USA. As previously discussed, DHS S&T PIADC can play an important coordinating role in generating additional, scientific data that can be used in the risk analysis model, such as BSL3 Ag (biosafety level 3 for agricultural hazards) safety testing in animals of veterinary biologics that may be candidates for importation. Licensing data should be developed and submitted for licensure in a logical sequence representing successive steps in product development (e.g. master seed purity and identity, laboratory safety, immunogenicity, initial production serials [usually three], purity, safety, potency testing, and field safety). All data generated in product development is required to be reported to the CVB.

Confirmatory testing by the Center for Veterinary Biologics laboratory for conventional product licences or permits for distribution and sale
The laboratory staff within the CVB conduct assays on veterinary biological products and key biologics manufacturing materials (master seed bacteria, master seed virus, and master cell stocks) as required in 9 CFR parts 101-118. Primary activities include: pre-licence testing test development and standardisation (including reagent activities) post-licence quality control monitoring. Pre-licence testing by the CVB laboratory for conventional product licences or Permits for Distribution and Sale includes assaying both parent materials (master seeds and cells) and final product. Master seeds and master cell stocks are evaluated for purity and identity (including identity of construct and expressed antigen for genetically engineered products). Prior to licensure or permitting of a product, a manufacturer is required to demonstrate their ability to consistently produce pure, safe, and potent product. This is usually accomplished through the

Rev. sci. tech. Off. int. Epiz., 26 (2)

435

satisfactory production of three consecutive, independent pre-licence serials. Before a licence or permit is issued, the purity, safety, and potency of pre-licence serials will be confirmed by testing in the CVB laboratory. This testing assures that the product tests are appropriate and transferable and that the manufacturer is able to consistently reproduce quality product. It is important to note that although all tested master seeds, master cells, and pre-licence serials have been tested by the manufacturer and submitted to the CVB as satisfactory, the CVB laboratory regularly finds a small percentage of seeds, cells, and products that are not pure, safe, potent, or effective.

a) the regulations require that manufacturing establishments be operated under the direct supervision of persons competent by education and experience to handle all matters pertaining to the preparation and testing of biological products. Summaries of the relevant qualifications for each supervisory employee responsible for essential steps in production, testing, and initial distribution of products must be provided to and accepted by the CVB; b) facility documents, including plot plans for all buildings, blueprints for each building used in preparing biological products, and blueprint legends which provide a brief description of all activities in each room or area of a production facility must be submitted to and accepted by the CVB; c) for each room, the blueprint legend should include: all activities conducted in a room, for example, inoculation, harvest, concentration, filling, etc. all microorganisms prepared, tested, or stored in the room, including an indication of whether the microorganism is viable or killed a listing of stationary or other essential equipment such as mills, centrifuges, mixing tanks, bottling and sealing equipment, etc. for rooms where products are exposed to the surroundings, a description of decontamination procedures and other precautions against crosscontamination; d) the regulations require buildings to be constructed so that: the floors, walls, ceilings, partitions, posts, doors, and all other parts of all structures, rooms, or facilities used in the preparation of biologicals can be readily and thoroughly cleaned all rooms are located and arranged to prevent crosscontamination of products there are adequate air handling systems to ensure sanitary and hygienic conditions for the protection of products and personnel there are separate rooms or compartments for preparing, handling, or storing virulent or dangerous microorganisms adequate hot and cold water supplies and efficient draining and plumbing systems are provided for animals and equipment; e) equipment cleaning, pasteurisation, sterilisation, temperature recording devices or other records of sterilisation must be acceptable; f) all animals used in the preparation or testing of biologicals must be healthy. Animals admitted into, used in, and disposed of from biologics production areas must

Inspection and compliance requirements for conventional product licences or permits for distribution and sale
According to Title 9 of the CFR, parts 101-118, for a US manufacturer to distribute veterinary biological products, the producer must hold valid licences of two types: a United States Veterinary Biologics Establishment License one or more Veterinary Biological Product Licenses. In order to distribute veterinary biological products manufactured outside the USA, a permittee (defined as any individual, firm, partnership, corporation, company, association, educational institution, state or local government who resides in the USA or operates a business establishment within the USA) must hold a United States Veterinary Biologics Establishment License and a United States Veterinary Biological Product Permit, For Distribution and Sale. Thus, the distribution of FAD imported vaccines that may be of interest to the National Veterinary Stockpile will require the identification of a company, individual or government authority that is willing to apply for these licences and to serve as the permittee. Partnerships between DHS S&T and private industry will most likely be required to ensure programme success, since the customer target for these imported products will be USDA APHIS and the National Veterinary Stockpile, rather than traditional meat, milk and egg producers. Prior to the issuance of an establishment licence or permit for distribution and sale, an applicant for biologics manufacturing must address the following items:

436

Rev. sci. tech. Off. int. Epiz., 26 (2)

meet the standards in the regulations for biologics and the Animal Welfare Act; g) records and record-keeping systems must be adequate to give a complete accounting of all activities within each establishment, including all procedures used in all steps in the preparation, testing, and disposition of products; h) if, at any time, there are indications that raise questions regarding the purity, safety, potency, or efficacy of a product, or if it appears that there may be a problem regarding the preparation, testing, or distribution of a product released for marketing, the manufacturer must agree to immediately notify the CVB concerning the circumstances and the action taken; i) written assurance must be filed with the CVB that the licensed or permitted products will not be advertised so as to mislead or deceive the purchaser and that the packages or containers used in marketing the product will not bear any statement, design, or device which is false or misleading in any particular; j) for biologics manufacturers in the USA, the entire premises of an establishment are subject to inspection, at any time, without prior notification. The regulations specifically authorise inspection of all buildings, compartments, and other places, all biological products, and organisms and vectors in the establishment, and all materials and equipment, such as chemicals, instruments, apparatus, and the like, and the methods used in the manufacture of, and all records maintained relative to, biological products produced at such establishments; k) for non-US manufacturing facilities, the producer and permittee must agree to submit to periodic inspections of the production facilities, and the permittee must agree to be responsible for all costs associated with the inspections. Before a United States Veterinary Biologics Establishment License or a United States Veterinary Biological Product Permit, For Distribution and Sale is issued, a pre-licence inspection by the CVB must confirm whether the condition, equipment, facilities, methods, etc. used to prepare biological products conform with the above items and all other requirements in the US regulations.

aid in a response to an intentional (bioterrorism) or unintentional introduction of a significant animal disease agent not normally found in the USA. If not, the processes and mechanisms described in this section have been used in the USA to allow for the more rapid availability of veterinary vaccines in an emerging or emergency animal health situation.

Conditional product licences


Conditional licences (regulated by 9 CFR Part 102.6) are authorised under very specialised circumstances to meet an emergency condition, limited market, local situation, or other special circumstance. Licences are issued under an expedited procedure which assures purity and safety, and provides a reasonable expectation of efficacy and/or potency for the vaccines involved. The data generated by a manufacturer to provide this reasonable expectation varies, and is evaluated on a case-by-case basis, but licences have been issued in the following situations: a) efficacy data was adequate, but correlation to the proposed potency assay was not determined or validated b) clinical or experimental efficacy studies suggested a protective effect against disease, but did not provide definitive data c) a scientifically accepted correlate of efficacy, such as a virus, toxin or similar neutralising antibody titre or level was available. It is important to note that approvals allowing the preparation of vaccines under conditional licences are time-limited, and manufacturers must actively pursue data to support full licensure. It is also important to note that the CVB may not issue Conditional Permits. Conditional licences may only be issued to biologics manufacturers in the USA. Vaccine prepared under a conditional licence must be in compliance with all other applicable licensing regulations and standards. This will involve, for example, conducting pre-licence field safety studies, following data and risk analysis procedures, advising the public prior to release of live biotechnology-derived vaccines, obtaining satisfactory results from confirmatory testing of seeds, cells, and prelicence serials by the CVB laboratory, and complying with facility and other CVB inspection requirements. Use of conditional licences for new FAD vaccines should be contemplated in strategic countermeasure programmes in which funds for full licensing may be limited and/or the specific advanced price or purchase commitments by the National Veterinary Stockpile are not well established. This will ultimately translate to biologics manufacturing capacity incentives.

Expedited processes for veterinary biologics in the United States of America


Ideally, a fully licensed or permitted vaccine is available for use in the USA to aid in the control of a new or emerging animal disease or when vaccine is immediately required to

Rev. sci. tech. Off. int. Epiz., 26 (2)

437

Autogenous product licences or permits for distribution and sale


Regulations concerning autogenous product licences and permits are contained in 9 CFR Part 113.113. Autogenous biologics are prepared from cultures of microorganisms which have been inactivated and are non-toxic. Such products are to be used only by or under the direction of a veterinarian within a veterinarian-client-patient relationship. The microorganisms used as seed to prepare autogenous biologics must be isolated from sick or dead animals in the herd of origin and there must be reason to believe they are the causative agent(s) of the current disease affecting such animals. Autogenous isolates may not be modified by biotechnology methods. Normally, microorganisms from one herd are not to be used to prepare an autogenous biologic for another herd. However, under certain circumstances, preparation of an autogenous biologic for use in herds other than the herd of origin, when those herds are considered to be at risk and have an epidemiologic link, may be authorised. In general, the microorganism(s) used for the production of autogenous biologics must be used within 15 months of the date of isolation, or within 12 months of the date of harvest of the first serial of product produced from the microorganism(s), whichever comes first. Testing requirements for autogenous products include testing of final container samples for purity (sterility) and mouse and/or guinea pig and/or host animal safety tests. Master seed testing by the manufacturer is not required, nor is any CVB laboratory confirmatory testing of the seeds or cells conducted. Products must include a label precaution that potency and efficacy have not been established. A licence to produce, distribute or ship autogenous products includes the restriction that the licence does not authorise production, distribution, or shipment of autogenous vaccine/bacterin for the following: foot and mouth disease rinderpest any H5 or H7 subtype of avian influenza any subtype of avian influenza if the vaccine is intended for use in chickens swine vesicular disease Newcastle disease African swine fever classical swine fever Brucella abortus vesicular stomatitis rabbit haemorrhagic disease any other disease that the USDA determines may pose a risk to animal or public health.

Autogenous FAD vaccines are not an attractive option for a countermeasure programme due to the high risk involved in manufacturing the live pathogen prior to inactivation and the increased investment that would be required to build separate, dedicated manufacturing facilities to produce these vaccines. Vaccine prepared under an autogenous licence or permit must be in compliance with all other applicable licensing regulations and standards, including risk analysis procedures. Manufacturers purity and safety test results may be confirmed by testing in the CVB laboratory. All manufacturing establishments are thoroughly inspected (personnel, facilities, production processes, recordkeeping) as CVB compliance requirements must be met.

Experimental product approvals


Regulations pertaining to experimental product approvals are contained in 9 CFR Parts 103.3 and 104.4. Under very specific circumstances the experimental production, distribution, and evaluation of biological products by manufacturers in the USA may be authorised prior to full licensing or permitting. For the benefit of licence applicants and to permit and encourage research, a person may be authorised to ship unlicensed biological products for the purpose of evaluating such experimental products by treating limited numbers of animals. Conditions must exist to ensure the experiment is conducted in a manner to prevent the spread of disease, with special restrictions imposed in the case of products containing live organisms. Requests for authorisation to ship an unlicensed biological product for experimental study and evaluation shall be accompanied by certain information including the following: a permit or letter of permission from the animal health authorities of each US state or foreign country involved; a tentative list of the names of the proposed recipients and quantity of experimental product that is to be shipped to each individual; a description of the product, recommendations for use, and results of preliminary research work; labels or label sketches which show the name or identification of the product and bear a statement, Notice! For Experimental Use Only Not For Sale, or equivalent. The US Veterinary License legend shall not appear on such labels; a general plan covering the methods and procedures for evaluating the product and for maintaining records of the quantities of experimental product prepared, shipped and used; data demonstrating that use of the experimental biological product in meat animals is not likely to result in

438

Rev. sci. tech. Off. int. Epiz., 26 (2)

the presence of any unwholesome condition in the edible parts of animals subsequently presented for slaughter; a statement from the research investigator or research sponsor agreeing to furnish, upon the Administrators request, additional information concerning each group of meat animals involved prior to their movement from the premises where the test is to be conducted. Such information must include the owners name and address; number, species, class and location of animals involved; date the shipment is anticipated; name and address of consignee, buyer, commission firm or abattoir; information in order to assess the products impact on the environment. Permits are not issued for biological products from countries known to have exotic diseases including, but not limited to, FMD, rinderpest, highly pathogenic avian influenza, swine vesicular disease, Newcastle disease, and African swine fever if such products may endanger livestock or poultry in the USA. Recently, DHS S&T has partnered with the CVB and industry to secure permission for the shipment of experimental FMD vaccine candidates to PIADC for efficacy studies in livestock. Continuation of this partnership will be important to allow for the timely identification of lead candidates for further development.

would be dramatic. In the event of such an outbreak, the lost export markets for live animals and meat products and the multiplier effect of lost production would be felt throughout the US economy. On 30 January 2004, Homeland Security Presidential Directive 9 (HSPD-9) established a national policy to defend the agriculture and food system in the USA against terrorist attacks, major disasters, and other emergencies. This directive noted that this would be done by: identifying and prioritising sector-critical infrastructure and key resources for establishing protection requirements developing awareness and early warning capabilities to recognise threats mitigating vulnerabilities at critical production and processing nodes enhancing screening procedures for domestic and imported products enhancing response and recovery procedures. As a part of response planning and recovery, HSPD-9 directed the creation of a National Veterinary Stockpile (NVS) containing sufficient amounts of animal vaccine, antiviral, diagnostic, or therapeutic products to appropriately respond to the most damaging animal diseases affecting human health and the economy. The NVS is mandated to be capable of deployment within 24 hours of an outbreak. The ability to respond to the intentional, simultaneous introduction of disease agents in multiple locations (i.e. an act of terrorism) is a particular focus for the NVS. The NVS is the national repository of vaccines, personal protective equipment, and other critical veterinary products. It exists to augment state and local resources in the fight against dangerous animal diseases that could potentially devastate American agriculture, seriously affect the economy, and threaten public health. The NVS reflects two significant changes in the way APHIS is able to respond to and eradicate animal diseases, as follows: a) previously, functional groups of specialists responding to an outbreak managed their own logistics support. This fragmentation has caused several problems: groups duplicate the efforts of others planning before an event is more complex and potentially incomplete coordination of resources during an event is more difficult when products come from multiple sources managed by multiple groups

Exemption of biological products from licensing requirements


The APHIS Administrator may exempt any veterinary biological product from any or all of the licensing or permitting requirements (9 CFR Part 106.1) if the products will be used by the USDA, or under the control and supervision of the USDA, for the prevention, control, or eradication of animal disease in any of the following circumstances: a) as part of an official USDA programme b) in an emergency animal disease situation c) as part of a USDA experimental trial. DHS S&T can provide value-added information for decisions regarding product exemption by conducting scientific studies designed specifically to characterise the product in the context of its effectiveness as an emergencyuse countermeasure tool.

Homeland Security Presidential Directive 9 and the National Veterinary Stockpile


The economic and social impacts of an outbreak of a highly contagious animal disease in the USA, such as FMD,

Rev. sci. tech. Off. int. Epiz., 26 (2)

439

costs of responding are higher because each group purchases supplies in smaller quantities and at high prices b) in the past, APHIS eradicated disease outbreaks primarily by destroying infected and potentially exposed animals. However, given both changes in agricultural practices in the USA (e.g. herd sizes are much larger than in the past), and the availability of animal disease control models and scenarios involving multifocal disease outbreaks, planning disease control options beyond quarantine and depopulation is prudent. Vaccine in the NVS gives APHIS another option. Within five years, the NVS intends to acquire vaccine, diagnostic testing capabilities, and therapeutic countermeasures against a variety of the most significant animal diseases, including highly pathogenic avian influenza, FMD, Rift Valley fever, exotic Newcastle disease, and classical swine fever. Within ten years, NVS plans to acquire countermeasures against all of the most dangerous diseases of animals. In order to rapidly deliver large quantities of critical veterinary supplies and equipment to the right place, at the right time, for as long as is necessary, the NVS is: directly obtaining, stockpiling and managing finished vaccines and other ready-to-use supplies for delivery by the NVS within 24 hours contracting, for delivery within 24 hours, for stocks of vendor-managed vaccines, diagnostic tests and reagents, and other perishable supplies developing both government and vendor-managed biologics precursors suitable for long-term storage and rapid formulation into needed products (e.g. vaccine antigen concentrates for highly pathogenic avian influenza and FMD). Veterinary biologics acquired by or contracted to the NVS may be any licensed, permitted, experimental, or exempted product, as is deemed prudent by risk analysis. DHS S&T can function as the single point of authority for either leading or coordinating the development of veterinary biodefence countermeasures for procurement

by the NVS. DHS S&T can provide a permanent funding source through which the federal government can codevelop next generation vaccines or novel immune-based biotherapeutics with private industry. Industry development costs can be off-set and minimised by forming strategic partnerships with DHS S&T. This approach is currently being used to establish a pipeline of next generation molecular vaccines for FMD. Expansion of this approach for other high priority foreign disease agents of threat to agriculture will be critical to enable the NVS to reach its five-year and ten-year objectives.

Conclusion
The procedures reviewed above provide a regulatory framework and outline general purity, safety, potency, and efficacy requirements for licensing of products for traditional animal diseases, emerging animal diseases, and the accidental or intentional introduction of animal diseases. Supplementary procedures may be required for certain products. These regulatory processes demonstrate the flexibility of the current regulatory system to accommodate a variety of animal health situations while providing the data-driven, performance-based oversight necessary to assure that only quality biologicals are available for use. DHS S&T, USDA-CVB and other government agencies of the USA must work together in a coordinated fashion to ensure the successful development and availability of highly efficacious, biological-based countermeasures for the US National Veterinary Stockpile.

440

Rev. sci. tech. Off. int. Epiz., 26 (2)

Rglementation applicable aux vaccins contre les maladies infectieuses mergentes et lagro-bioterrorisme aux tats-Unis dAmrique
L.A. Elsken, M.Y. Carr, T.S. Frana, D.A. Brake, T. Garland, K. Smith & P.L. Foley
Rsum Aux tats-Unis dAmrique, la loi de 1913 sur les virus, les srums et les toxines (Virus-Serum-Toxin Act) telle quamende en 1985, constitue le fondement juridique de la rglementation relative aux vaccins vtrinaires et aux produits biologiques usage vtrinaire. Lautorit rglementaire charge de dlivrer les autorisations de mise sur le march et les autorisations dexpdition ou dimportation de mdicaments vtrinaires purs, sans danger, puissants et efficaces est une agence sous tutelle du Dpartement amricain de lAgriculture, le Center for Veterinary Biologics (CVB). Conformment la procdure normalise dautorisation, les fabricants doivent dvelopper, caractriser intgralement et valuer les produits candidats avant de prsenter une demande dautorisation ; ces informations sont ensuite examines et values par le CVB, puis celui-ci procde un audit et une inspection des tablissements du fabricant et de ses mthodes de production et de test ; les principaux rsultats des tests relatifs au produit sont ensuite vrifis lors dun examen indpendant. Cette valuation exhaustive ntant pas toujours possible dans les situations durgence, des procdures et des mcanismes particuliers sont prvus afin que des vaccins vtrinaires soient rapidement disponibles dans ces situations. Des vaccins de nouvelle gnration contre des maladies animales exotiques telles que la fivre aphteuse sont en cours de dveloppement aux tats-Unis, et il appartient aux autorits comptentes de sassurer que la rserve nationale de vaccins vtrinaires en contienne un stock suffisant. Mots-cls Autorisation de mise sur le march Bioterrorisme Maladie Maladie mergente Rglementation Urgence sanitaire Vaccin Vaccin vtrinaire.

Rev. sci. tech. Off. int. Epiz., 26 (2)

441

Reglamentacin sobre vacunas contra infecciones emergentes y bioterrorismo en los Estados Unidos de Amrica
L.A. Elsken, M.Y. Carr, T.S. Frana, D.A. Brake, T. Garland, K. Smith & P.L. Foley
Resumen En los Estados Unidos de Amrica (EE.UU.), la ley relativa a los virus, sueros y toxinas (Virus-Serum-Toxin Act) de 1913, modificada en 1985, constituye la base legal de la reglamentacin sobre las vacunas de uso veterinario y los productos biolgicos conexos. El Centro de Productos Biolgicos de Uso Veterinario (CVB, Center for Veterinary Biologics), un organismo del Departamento de Agricultura de ese pas, es el ente regulador de la concesin de autorizaciones de exportacin e importacin de productos biolgicos de uso veterinario puros, inocuos, potentes y eficaces. De conformidad con el procedimiento normalizado de otorgamiento de licencias y permisos, es preciso que los fabricantes hayan desarrollado, caracterizado y evaluado totalmente un producto antes de que se le otorgue la homologacin. A su vez, el CVB ha de revisar y evaluar la informacin presentada, examinar los mtodos de produccin y prueba, realizar una inspeccin de las plantas de elaboracin, y someter los resultados de los ensayos de los productos clave a un organismo independiente para su verificacin. En situaciones de emergencia puede carecerse de tiempo suficiente para efectuar esa evaluacin global y exhaustiva; por consiguiente se han establecido procedimientos y mecanismos para disponer de vacunas de uso veterinario con mayor rapidez. El desarrollo de la nueva generacin de vacunas contra enfermedades animales forneas, como la fiebre aftosa, avanza con rapidez en los EE.UU. Las autoridades deben asegurarse de que se suministren cantidades suficientes de esas vacunas a la Reserva Nacional Veterinaria. Palabras clave Autorizacin de comercializacin Bioterrorismo Emergencia Emergente Enfermedad Regulacin Vacuna Veterinaria.

References
1. Bunn T.O. (1992). Testing of veterinary biologics in the United States. Dev. biol. Standard., 79, 187-192. 2. Espeseth D.A. (1995). Present systems and future needs for risk assessment of biologicals in the United States of America: the perspective of the regulator. In Risk assessment for veterinary biologicals (E.G.S Osborne & J.W. Glosser, eds). Rev. sci. tech. Off. int. Epiz., 14 (4), 1143-1155. 3. Espeseth D.A. & Myers T.J. (1999). Authorities and procedures for licensing veterinary biological products in the United States. Adv. vet. Med., 41, 585-593. 4. Foley P.L. & Hill R.E. (2005). Regulatory considerations for marker vaccines and diagnostic tests in the US. Biologicals, 33 (4), 253-256. 5. Henderson L.M. (2005). Overview of marker vaccine and differential diagnostic test technology. Biologicals, 33 (4), 203-209. 6. Hill R.E., Foley P.L., Carr M.Y., Elsken L.A., Gatewood D.M., Ludemann L.R. & Wilbur L.A. (2003). Regulatory considerations for emergency use of non-USDA licensed vaccines in the United States. Dev. Biol. (Basel), 114, 31-52. 7. Randall D.C. (1992). Inspection of veterinary biologics in the United States. Dev. biol. Standard., 79, 163-170. 8. United States Code of Federal Regulations (2006). Title 9 Animals and Animal Products.

Rev. sci. tech. Off. int. Epiz., 2007, 26 (2), 443-449

Lexprimentation animale dans la dcouverte et la production de vaccins usage vtrinaire


J.-Ch. Audonnet (1), J. Lechenet (2) & B. Verschuere (3)
(1) Dpartement de recherche exploratoire, Mrial, 254, rue Marcel-Mrieux, 69007 Lyon, France (2) Dpartement des affaires rglementaires Europe, enregistrements biologiques, Mrial, 29, avenue Tony-Garnier, 69007 Lyon, France (3) Groupe interprofessionnel de rflexion et de communication sur la recherche (GIRCOR), 15 rue Rieux, 92100 Boulogne, France

Rsum Les tablissements de recherche, de dveloppement et de production des vaccins vtrinaires doivent rpondre aux questions du public, aux exigences de la rglementation et aux objectifs damlioration de la bientraitance animale, tout en assurant leur mission de production de vaccins toujours plus efficaces et plus inoffensifs. Le recours lanimal pour la mise au point et le dveloppement de nouveaux vaccins vtrinaires est invitable, aucun modle in vitro ne permettant de prdire la capacit dun candidat vaccin induire une protection chez lespce cible. Face aux contraintes thiques et rglementaires, les progrs pour la mise en uvre des meilleures conditions possibles pour lutilisation des animaux dexprience sont permanents. Lvolution constante dans le domaine de lthique animale impose un effort particulier lindustrie pharmaceutique qui doit modifier avec rigueur les documents lappui de lenregistrement en fonction de chaque nouveau dveloppement. Mots cls Dveloppement Embryon Exprimentation animale Modle in vivo Recherche Vaccin usage vtrinaire.

Introduction
Depuis plusieurs annes, l exprimentation animale est un thme de dbat social. Partage entre les arguments des chercheurs qui expliquent et prouvent rgulirement la ncessit du recours l animal pour le progrs scientifique et mdical, et la compassion pour les animaux qui seront tus et qui souffriront peut-tre de ces expriences, l opinion publique ne peut donner une rponse simple la question : tes-vous pour ou contre l exprimentation animale ? . Selon le contexte dans lequel la question est pose, la rponse pourra tre je suis pour, mais il faudrait que les animaux ne souffrent pas, ou souffrent le moins possible ou je suis contre, sauf si cela doit sauver des vies humaines ou dautres vies animales plus nombreuses . Les sondages dopinion raliss rgulirement sur ce thme rvlent invariablement que la position du public est conditionne par le pourquoi ? et le comment ? .

Selon la raison pour laquelle on a recours l exprimentation animale (le pourquoi ? ), cette exprimentation sera plus ou moins accepte. Dans ce domaine, la sant humaine et la sant animale sont les raisons qui entranent clairement la plus forte adhsion du public. Les conditions dans lesquelles l exprimentation animale est ralise (le comment ? ), sont aussi un lment cl sur lequel sappuiera la perception (positive ou ngative) du public : le respect de la condition animale au travers du respect absolu de la rglementation et des normes professionnelles, la formation des chercheurs, la qualit des installations danimaleries, les comits dthique et la prsence de vtrinaires, sont des lments cls de lapprciation. Face ces interrogations de la socit, le lgislateur a logiquement mis en place un systme de textes qui encadrent l exprimentation animale conformment aux vux des citoyens. On voit dans tous les pays occidentaux

444

Rev. sci. tech. Off. int. Epiz., 26 (2)

des rglementations de plus en plus convergentes qui demandent que lexprimentation animale rponde obligatoirement des ncessits de recherche, de contrle, de scurit du consommateur ou denseignement, quelle soit ralise uniquement dans des tablissements rfrencs et contrls par une autorit dsigne, et enfin que des comits dthique examinent et apprcient de faon indpendante le bien-fond de la recherche et les conditions de sa ralisation. Des centres nationaux ou internationaux subventionns par des fonds publics ou privs ont t crs dans le but de promouvoir ou dvelopper des mthodes scientifiques qui permettent dutiliser moins danimaux ou en induisant moins de douleur, de stress ou dinconfort, le cas chant. Ces centres (par exemple le European Centre for the Validation of Alternative Methods [ECVAM] : http://ecvam.jrc.it/ en Italie ; le Interagency Coordinating Committee on the Validation of Alternative Methods [ICCVAM] : http:// iccvam.niehs.nih.gov/ aux tats-Unis dAmrique ; le European Consensus-Platform for Alternatives [ECOPA] : http://www.ecopa.eu/ en Belgique ; et le National Centre for the Replacement, Refinement and Reduction of Animals in Research [NC3R] : http://www.nc3rs.org.uk/ au Royaume-Uni) visent appliquer au plus prs du chercheur et de lanimal de laboratoire la rgle dite des trois R (remplacement, rduction, raffinement) promue ds 1959 par Russell et Burch (7). Trs tt, l industrie du vaccin vtrinaire a pris en compte les exigences rglementaires et thiques lies l utilisation des animaux en recherche et dveloppement (R&D). L objet de cet article est de prsenter comment les tablissements de recherche, de dveloppement et de production des vaccins vtrinaires rpondent aux questions du public, aux exigences de la rglementation et aux objectifs de la rgle des trois R tout en assurant leur mission qui est de protger toujours plus efficacement les animaux contre les maladies l aide de vaccins les plus inoffensifs possibles.

rage ( destination de l homme) a t ainsi prcd par la mise au point de plusieurs vaccins vtrinaires (charbon, cholra de la poule) qui ont confirm la possibilit dinduire une protection solide contre les maladies infectieuses par l immunisation avec des vaccins . Si les principes gnraux des contrles de qualit ntaient bien sr pas encore connus au XIXe sicle, il nest pas moins difiant de savoir que la rigueur scientifique de Pasteur lavait dj pouss contrler les lots de vaccin contre la rage avant de les utiliser chez l homme en toute scurit. La Directive europenne 2001/82/EC (amende le 31 mars 2004) (4) dfinit les produits immunologiques comme suit : un mdicament vtrinaire administr aux animaux en vue de provoquer une immunit active ou passive ou de diagnostiquer l tat dimmunit . L immunit active (vaccins classiques) induit des tats de protection plus ou moins complets qui sont dtaills dans les indications reconnues officiellement par les autorits denregistrement. De nos jours, des vaccins vtrinaires existent pour toutes les espces et permettent de combattre toutes les grandes maladies. Ce sont des outils essentiels pour le maintien de l tat sanitaire de nos levages et pour le bien-tre des animaux de rente et de compagnie.

Particularits de la recherche et du dveloppement pour les vaccins vtrinaires


Si les outils qui aident la slection des vaccins vtrinaires et leur production montrent une convergence inluctable en technicit et en qualit avec les vaccins humains, un grand avantage pour les vaccins vtrinaires est que leur valuation se ralise sur l espce cible (bnficiaire directe du vaccin dvelopp). Mais l art de cette valuation reste complexe, particulirement dans le domaine vtrinaire o les quipes de R&D sont confrontes de nombreuses espces diffrentes et des individus non standardiss quant aux niveaux de rponse immunitaire et aux spcificits de cette rponse. La plupart des maladies tant strictement spcifiques despce, il est malheureusement impossible dextrapoler facilement une autre espce les rsultats obtenus sur une espce donne. Les travaux de R&D pour les vaccins vtrinaires requirent un trs grand savoir-faire dans la matrise des modles animaux (sur espces cibles) qui nest en gnral obtenu quaprs de longues annes de pratique et une capacit dduire des conclusions solides dun ensemble de donnes caractrises par une grande variabilit. Le corollaire de lutilisation importante des espces cibles pour la R&D des vaccins vtrinaires est que ces animaux,

Historique de lexprimentation animale pour les vaccins et cadre rglementaire


Les animaux domestiques ont t associs au dveloppement des vaccins ds lorigine du concept mme de vaccin. En effet, cest la production en grande quantit, par scarification de flancs de gnisses, du virus de la vaccine de Jenner que la vaccination a pu rapidement diffuser dabord en Europe puis dans le monde entier. Mais ce sont ensuite Pasteur et ses disciples (dont de nombreux vtrinaires) qui ont ralis les premires exprimentations animales raisonnes des fins de recherche et de dveloppement de vaccins. Le vaccin de la

Rev. sci. tech. Off. int. Epiz., 26 (2)

445

bien que considrs comme des animaux dexprimentation animale ne sont pas forcment perus par le public comme des animaux dexprience ou des animaux de laboratoire. Les carnivores (chiens, chats, furets, etc.) excepts, les animaux de rente (bovins, ovins, caprins, porcs, poulets, dindes, oies, canards, etc.) ou les chevaux sont en effet rarement assimils spontanment par le public des animaux de laboratoire .

tudes, compte tenu de tailles des marchs beaucoup plus petites que pour les vaccins humains. La gense dun vaccin vtrinaire sorganise autour de plusieurs phases successives : recherche, dveloppement, enregistrement, commercialisation et suivi post-lancement.

Premire phase : recherche


La facult de pouvoir intervenir chez lespce cible ds le dbut de la phase de recherche est une chance pour la vaccinologie vtrinaire car elle permet davoir des rponses rapides quant la faisabilit du candidat vaccin. Mais cest condition de disposer dun modle dpreuve fiable et reproductible. Cest dans cette tape de mise au point du modle infectieux que lon utilise le plus danimaux, car de nombreuses variables sont mal matrises au dpart. Toutefois le nombre total danimaux utiliss ce stade reste limit. Les contraintes thiques, conomiques et pratiques (lorsque lon travaille par exemple avec de grandes espces comme le cheval) sont importantes et contribuent, outre les encadrements rglementaires et thiques, optimiser au mieux la dmarche exprimentale. Dans ces tapes, les nombres minima danimaux par groupe sont valids par les biostatisticiens en fonction de la variabilit connue du modle, de manire garantir que les rsultats seront interprtables (et donc dviter de faire des expriences inutiles aux plans scientifique et thique). Pour lespce cible, on utilise en moyenne 100 200 animaux, selon quil est ncessaire ou non de mettre au point le modle dpreuve (dans le cas dune nouvelle maladie) et que la pathologie infectieuse laquelle on sadresse est simple ou non. En effet, si la reproduction exprimentale de la maladie est bien matrise, quelques tudes seulement permettront un tri trs pertinent et efficace des diffrentes options qui existent lors de la phase recherche . Dans le cas contraire, la dfinition dun modle suffisamment reproductible pour donner des informations fiables peut tre longue et consommatrice danimaux. Au cours de la phase recherche , un nombre assez limit de rongeurs et lapins (quelques dizaines au plus) peut servir galement la prparation de ractifs srologiques spcifiques aux antignes du vaccin tudi.

Utilisation des animaux dexprimentation au cours des tapes du dveloppement des vaccins vtrinaires
La phase pr-clinique , classique dans le dveloppement des vaccins humains, nexiste pas pour les vaccins vtrinaires. Ceci est d plusieurs raisons : des animaux de l espce cible (dits dexprience ) peuvent tre utiliss directement, et trs en amont, dans des tudes dinnocuit et defficacit ; les rsultats obtenus nen sont que plus pertinents pour le vaccin tudi ; cest une contrainte rglementaire imposant de reproduire la maladie chez les animaux en exprience pour dmontrer visiblement la protection attendue et de raliser la dmonstration de toutes les qualits du nouveau vaccin dans un environnement matris (tude en laboratoire ; partie C des 7e et 8e parties de lAnnexe 1 de la Directive europenne 2001/82/CE) (4) ; enfin, le temps de dveloppement est raccourci. L un des bnfices, inconnus du grand public, du recours direct lespce cible est que cette dmarche, trs pragmatique et habituelle pour le monde vtrinaire, vite lutilisation des nombreux rongeurs qui sont traditionnellement associs aux phases pr-cliniques des vaccins humains. Une autre particularit est le fait que les vaccins vtrinaires qui sadressent aux animaux de production (bovins, ovins, porcs, poulets, dindes, etc.) sont des vaccins conomiques . Ce facteur pousse une optimisation (en nombre de protocoles et en nombre danimaux par protocole) des cots de R&D et limite donc les recours inutiles. La conjonction de ces particularits fait que le processus de R&D pour les vaccins vtrinaires tend fortement sautolimiter pour ce qui concerne le nombre dtudes et le nombre des animaux engags dans ces

Deuxime phase : dveloppement et enregistrement


Comme pour la phase prcdente, cette phase va concerner essentiellement les animaux de lespce cible afin de vrifier les caractristiques intrinsques du vaccin et de gnrer les donnes cliniques du dossier denregistrement. Au total, moins dune centaine

446

Rev. sci. tech. Off. int. Epiz., 26 (2)

danimaux sont impliqus en gnral pour un dossier concernant un vaccin pour un mammifre, mais les vaccins pour volailles peuvent ncessiter de plus grands nombres, plus pour des raisons historiques ou de pharmacope que par relle ncessit scientifique ou technique. Une exception notable est le cas des vaccins recombins vivants ( vaccins vecteur ) pour lesquels il faut en plus valuer l innocuit dans dautres espces (vrification de labsence de modification du tropisme et de labsence de virulence) afin dtudier son comportement dans des environnements variables. Cette phase est trs encadre par les exigences rglementaires : Directives europennes 90/219/CE (utilisation en milieu confin) (2) et 2001/18/CE (dissmination dans lenvironnement, cest-dire hors confinement) (3). Le dveloppement est aussi le moment o diffrents tests de contrle doivent tre mis au point pour que le fabricant puisse suivre et garantir la rgularit et la qualit des lots de vaccins lors des oprations de production commerciale. Les tests dactivit sont traditionnellement raliss sur des petits rongeurs de laboratoire et l tablissement dune corrlation solide entre la rponse srologique des rongeurs et celle de l espce destinataire nest parfois pas vident. Cest souvent cette mise au point qui entrane les plus grandes consommations danimaux (jusqu plusieurs centaines voire quelques milliers dans des cas extrmes) (6).

Production in vivo (sur animaux ou embryons)


De nos jours trs peu de vaccins utilisent des animaux pour leur production. Sont prfrs de trs loin pour des raisons de reproductibilit industrielle, de bio-scurit et de cot, les productions sur cellules de ligne. Les lapins et certaines volailles sont les seuls animaux encore utiliss pour de trs rares produits. La raison est souvent l absence dun systme in vitro de culture de l agent pathogne, l animal tant malheureusement le seul support possible. L utilisation dembryons de poulets (sous forme dufs embryonns) est par contre beaucoup plus frquente, luf tant un systme industriel trs rpandu pour la production de certains vaccins (l exemple le plus connu et le plus important en volume est celui du vaccin de la grippe humaine). Nous sommes ici la frontire entre ce quil est convenu de considrer comme un animal ou pas. De nombreux comits dthique tablissent dailleurs des limites prcises (ge dincubation) pour la frontire entre l embryon de poulet non animal et animal (5).

Production in vitro (sur cellules primaires, cellules de ligne, milieux de culture pour bactries)
Cette production in vitro ne met pas en uvre l utilisation danimaux.

Les animaux utiliss lors de lessai terrain


Les essais cliniques sur le terrain ont pour but de vrifier en conditions relles l innocuit du vaccin sur un grand nombre dindividus tout venant . Le but et la conception de l essai terrain pour les vaccins vtrinaires diffrent clairement de la phase III des essais cliniques pour les vaccins humains qui ont pour but de dmontrer l efficacit statistique du vaccin. Nous avons vu plus haut que lefficacit du vaccin vtrinaire est dmontre trs tt par preuve exprimentale sur lespce cible, ds la phase de recherche, puis confirme lors de la phase de dveloppement. ce stade, les essais nont donc pour but que de confirmer les rsultats grande chelle. Le principal point retenir ici est que les essais terrain sont soumis autorisation rglementaire et que les animaux engags dans ces essais le sont aprs consentement de leurs propritaires et nont donc pas le statut danimaux dexprimentation.

Libration des lots et mise en circulation des vaccins


Si les oprations de production proprement dite nimpliquent quasiment plus l usage danimaux, des techniques de contrle sur animaux (principalement des rongeurs) sont obligatoires afin de pouvoir librer les lots de vaccins conformment aux exigences rglementaires du dossier dautorisation de mise sur le march (AMM). Ces contrles sont : des tests dinnocuit spcifique (sur espce cible) et non spcifique (sur petits rongeurs) ; ces derniers tests ont disparu des obligations rglementaires alors que les premiers tendent disparatre. En effet, aprs dmonstration dune bonne innocuit sur un certain nombre de lots diffrents de production, et si le procd de fabrication est jug robuste, il est possible de saffranchir de ce test (5) ; des test dactivit dans le cas des vaccins inactivs seulement (test gnralement fait sur rongeurs) : un nombre limit danimaux est utilis pour chaque lot (gnralement une dizaine), mais le nombre total est fonction du nombre de lots de vaccins tester. Il faut souligner que la mise en uvre gnralise des normes de bonnes pratiques de fabrication a fortement contribu la diminution du nombre danimaux ncessaires au contrle des vaccins via diffrents facteurs. Par exemple,

Troisime phase : mise sur le march (production et vente, tests de contrle qualit)
Il faut distinguer dans ce paragraphe l utilisation des animaux comme support direct pour la production des vaccins (exemples dsormais rarissimes) de l utilisation des animaux pour les contrles rglementaires de qualit des vaccins commerciaux.

Rev. sci. tech. Off. int. Epiz., 26 (2)

447

l augmentation de la taille des lots industriels, suite la fiabilisation des procds, a un impact direct trs concret sur le nombre danimaux utiliss.

Quatrime phase : soutien du produit


L histoire dun vaccin ne sarrte pas l enregistrement et il est parfois important pour le producteur de gnrer des donnes complmentaires au dossier (mais non ncessaires l enregistrement pharmaceutique) qui sont utiles pour soutenir les ventes du vaccin et pour renforcer sa position concurrentielle. Ces essais complmentaires vont concerner bien sr les animaux de l espce cible et engager quelques dizaines danimaux au maximum. Selon les cas, les essais seront faits soit en station exprimentale (animaleries de R&D, donc animaux dexprimentation), soit sur le terrain, aprs autorisation administrative, avec des animaux de statut normal (on ne parlera alors pas danimaux dexprimentation). Il peut tre prcis que parfois l environnement rglementaire volue et les conclusions scientifiques sont une nouvelle fois questionnes par les autorits, non pas parce quelles ne sont plus fondes, mais parce que le protocole de l tude nest plus conforme aux dernires exigences, obligeant ainsi de refaire cette tude voire une partie du dveloppement (Directive europenne n 2001/82/CE, article 27) (4).

Cette tendance la diminution des animaux pour les contrles de qualit a surtout t rendue possible du fait de lintroduction des bonnes pratiques de fabrication (BPF) et dun nombre grandissant doutils sophistiqus pour le contrle de la qualit du vaccin. De trs nombreux paramtres sont dsormais suivis et mis sous contrle , permettant de mieux garantir la reproductibilit dun lot l autre. Les amliorations de procd sont mises en place aprs validation complte et aprs autorisation des autorits denregistrement. La qualit de production est plus prdictible, la qualit se construit durant la production, ce qui rduit ainsi les besoins de contrle. Cest un des facteurs importants ayant permis la disparition du test de toxicit anormale demand par la Pharmacope europenne, qui tait trs consommateur de cobayes et souris. Un suivi plus rapproch du mdicament aprs la vente a t galement instaur : la pharmacovigilance. Tout problme vu lors de lutilisation du vaccin sur le terrain peut tre rapport aux autorits et aux laboratoires producteurs. Aujourdhui, les conclusions sur les observations du terrain confirment que les manques defficacit ou les rels problmes dinnocuit sont peu nombreux (validant ainsi la qualit du dveloppement sur espce cible mme avec un faible nombre danimaux). De mme une bonne innocuit en gnral des vaccins est confirme. Ce nouvel outil a donn plus confiance l ensemble des acteurs pour quen association avec la mise en place des BPF une rduction des tests (donc des , animaux utiliss) soit entreprise. Lutilisation trs directe de lespce cible pour la recherche et le dveloppement des vaccins vtrinaires est accueillie plutt favorablement par le public quand il est averti, car les rsultats pertinents gnrs par cette dmarche contribuent limiter trs fortement le nombre d'animaux utiliss.

Progrs dans le domaine rglementaire


L utilisation des animaux, mme pour des raisons de contrle de qualit de production se doit de rpondre aux attentes du public. Les textes de base (Directive europenne 86/609/CE) (1) ont aid les autorits accepter une moindre utilisation et llimination de duplications inutiles. La mise en place de ces textes a galement favoris la modification des esprits qui a conduit des cadres rglementaires encourageant cette diminution. Par exemple la Pharmacope europenne (6), dans sa partie introductive sur le vaccin (Monographie n 62) mentionne : Essais sur animaux. Conformment aux dispositions de la Convention europenne sur la protection des animaux vertbrs utiliss des fins exprimentales et dautres fins scientifiques, les essais doivent tre raliss de telle faon quils utilisent le moins danimaux possible et quils rduisent au minimum toute douleur, souffrance, dtresse ou nuisance durable .

Conclusion
Le recours l animal dexprimentation pour la mise au point et le dveloppement de nouveaux vaccins vtrinaires est malheureusement invitable. Aucun modle thorique ou in vitro ne permet en effet de prdire ou dcrire la capacit dun candidat vaccin induire une protection chez l espce cible. Par ailleurs, le vaccin est un mdicament, produit fortement rglement pour assurer un niveau de qualit irrprochable. Face ces contraintes thiques et rglementaires, les chercheurs ont dvelopp des stratgies pour limiter au maximum le nombre danimaux utiliss. Les vtrinaires et les chercheurs de l industrie au contact des animaux sont trs conscients que la mise en uvre de conditions visant un bien-tre optimal des animaux en exprimentation est un facteur essentiel de qualit pour les tudes. Tous ces personnels sont engags dans une dmarche permanente de progrs pour amliorer sans cesse les conditions dexprimentation.

448

Rev. sci. tech. Off. int. Epiz., 26 (2)

Les progrs pour la mise en uvre des meilleures conditions possibles dans l utilisation des animaux dexprimentation sont permanents et constants dans le domaine de la R&D des vaccins vtrinaires. Ces progrs correspondent aux principes des trois R, dclins espce par espce et modle infectieux par modle infectieux. La perception du public vis--vis de l exprimentation animale change au cours du temps, de mme que celle des exprimentateurs, car ceux-ci font aussi partie du

public . La prise en compte dune volution constante dans le domaine de lthique animale demande un effort particulier dans l environnement pharmaceutique du fait de la rigueur observer pour la documentation des changements dans les documents de rfrence. Cet effort est toutefois lgitime et il est directement rcompens par la fiert de raliser des amliorations au bnfice de l animal.

Animal experimentation in the discovery and production of veterinary vaccines


J.-Ch. Audonnet, J. Lechenet & B. Verschuere
Summary Veterinary vaccine research, development and production facilities must aim to improve animal welfare, respond to public concerns and meet regulatory requirements, while at the same time fulfilling their objective of producing evermore effective and safer vaccines. The use of animal experimentation for the development of new veterinary vaccines is inevitable, as no in vitro model can predict a candidate vaccines ability to induce protection in the target species. Against the backdrop of ethical and regulatory constraints, constant progress is being made in creating the best possible conditions for animal experimentation. Keeping up to date with the constant changes in the field of animal ethics requires a particular effort on the part of the pharmaceutical industry, which must make careful changes to product registration documentation in accordance with each new development. Keywords Animal experimentation Development Embryo In vivo model Research Veterinary vaccine.

La experimentacin con animales en relacin con el descubrimiento y la produccin de vacunas de uso veterinario
J.-Ch. Audonnet, J. Lechenet & B. Verschuere
Resumen Los establecimientos de investigacin, desarrollo y produccin de vacunas veterinarias deben responder a las demandas del gran pblico, a los requisitos reglamentarios y a los objetivos de mejora del bienestar de los animales, y al mismo tiempo cumplir su funcin de elaborar vacunas cada vez ms eficaces e

Rev. sci. tech. Off. int. Epiz., 26 (2)

449

inocuas. El recurso a los animales para poner a punto y desarrollar vacunas es inevitable, pues ningn modelo in vitro permite predecir la capacidad de una eventual vacuna para inducir proteccin en la especie destinataria. Ante las exigencias de orden tico y reglamentario, constantemente se vienen mejorando las condiciones en que se utilizan animales con fines de experimentacin. El continuo progreso de la tica en este campo exige un particular esfuerzo por parte del sector farmacutico, que debe observar un gran rigor para modificar los documentos de referencia utilizados para el registro de las vacunas en funcin de cada nuevo desarrollo. Palabras clave Desarrollo Embrin Experimentacin con animales Investigacin Modelo in vivo Vacuna de uso veterinario.

Bibliographie
1. Communauts europennes (1986). Council Directive 86/609/EEC of 24 November 1986 on the approximation of laws, regulations and administrative provisions of the Member States regarding the protection of animals used for experimental and other scientific purposes. Off. J. European Communities, L 358, 18/12/1986, 1-28 (page web: http:// europa.eu.int/eur-lex/lex/LexUriServ/Lex UriServ. do?uri= CELEX:31986L0609:EN:HTML, consulte le 30 mai 2007). 2. Communauts europennes (1990). Directive 90/219/CEE du Conseil, du 23 avril 1990, relative l utilisation confine de micro-organismes gntiquement modifis. J. off. Union eur., L 117, 08/05/1990, 0001-0014 (page web : http://eurlex. europa.eu/LexUriServ/LexUriServ.do?uri=CELEX:31990 L0219:FR:HTML, consulte le 30 mai 2007). 3. Communauts europennes (2001). Directive 2001/18/CE du Parlement Europen et du Conseil du 12 mars 2001 relative la dissmination volontaire dorganismes gntiquement modifis dans lenvironnement et abrogeant la directive 90/220/CEE du Conseil. J. off. Communauts eur., L 106/1, 17/4/2001, 1-38 (page web : europa.eu.int/eurlex/pri/fr/oj/dat/2001/l_106/l_10620010417fr00010038.pdf, consulte le 30 mai 2007). 4. Communauts europennes (2001). Directive 2001/82/CE du Parlement europen et du Conseil du 6 novembre 2001 instituant un code communautaire relatif aux mdicaments vtrinaires [amende par la Directive 2004/28/CE du Parlement europen et du Conseil du 31 mars 2004 modifiant la directive 2001/82/CE instituant un code communautaire relatif aux mdicaments vtrinaires (Texte prsentant de l intrt pour l EEE)]. J. off. Union eur., L 311, 28/11/2001, 1-6 (pages Web : http://eur-lex.europa.eu/ LexUriServ/LexUriServ.do?uri=CELEX:32001L0082:FR:HTM L ; directive amende : http://eur-lex.europa.eu/LexUriServ/ LexUriSer v.do?uri=CELEX:32004L0028:FR:HTML, consultes le 30 mai 2007). 5. Institutional Animal Care and Use Committee (2005). Policy on the use of avian embryos. Page web : http:// casemed.case.edu/ora/iacuc/policies/avian_embryos.cfm (consulte le 30 mai 2007). 6. Pharmacope europenne (2005). Vaccins pour usage vtrinaire. Monographie n 62. Pharmacope europenne. Site web : http://online.pheur.org/entry.htm (consulte le 30 mai 2007). 7. Russell W.M.S. & Burch R.L. (1959). The principles of humane experimental technique. Methuen & Co., Londres.

Rev. sci. tech. Off. int. Epiz., 2007, 26 (2), 451-459

Consumer attitudes to vaccination of food-producing animals


J.M. Scudamore
University of Liverpool, Faculty of Veterinary Science, Leahurst, Neston, South Wirral, CH64 7TE, United Kingdom

Summary The 2001 outbreak of foot and mouth disease in the United Kingdom was unprecedented, with the need to develop a vaccination policy at the height of the epidemic. The extent of consumer concerns about eating products derived from vaccinated animals was unknown as survey results were equivocal. A recent survey on avian influenza reveals that the European public are well informed about the disease and its control, but over 50% of respondents would be reluctant to consume meat from vaccinated birds. There is little specific information available on consumer views about routine vaccination for other diseases. Their concerns appear to increase in an emergency situation when there is heightened awareness through the media. With the development of newer types of vaccines consumers will need more assurance about the safety and use of these products. This article examines these issues and makes practical recommendations for ensuring public confidence when emergency vaccination for disease control is proposed. Keywords Avian influenza Consumer attitudes Consumer concern Food safety Foot and mouth disease Lessons learned Public confidence Vaccination.

Introduction
It is difficult to obtain detailed information on consumer attitudes to eating products from food-producing animals which have been vaccinated, especially as there is limited information in the literature. A compounding problem is that consumers often only consider the issue when there is heightened media interest and emergency vaccination is to be used in the face of an outbreak of an epizootic disease such as foot and mouth disease (FMD) or avian influenza (AI). The FMD outbreak in the United Kingdom (UK) in 2001 and the current situation with respect to AI along with a number of recent surveys provide some indication of consumer attitudes.

Foot and mouth disease vaccination


The 2001 outbreak
The UK outbreak of FMD began in February 2001. The final case occurred on 30 September 2001, just over seven months after the commencement of the outbreak. The last remaining FMD-infected area was pronounced clear on the 4 December 2001. In January 2002, the World Organisation for Animal Health (OIE) declared the UK free of FMD without vaccination. The UK approach prior to the FMD outbreak in 2001 was to rely on a traditional

452

Rev. sci. tech. Off. int. Epiz., 26 (2)

stamping out policy. Field instructions for vaccination were available prior to the outbreak but only as an outline procedure which had not been publicised or discussed with stakeholders. Vaccination was considered during the first weeks of the outbreak but was thought to be impractical due to the widespread dissemination of infection throughout the country. Vaccination was subsequently considered throughout the epidemic and on at least ten specific occasions. Apart from a proposal to vaccinate cattle in Cumbria and Devon (which was accepted but ultimately not carried out see next paragraph) vaccination was ruled out for a number of reasons. It was recognised that the eradication of FMD in most areas could be achieved quickly and most effectively with minimal disruption through culling and tighter biosecurity. The vaccination of cattle in Cumbria and possibly Devon was under active consideration from the end of March. These were areas where the concentration of disease and the local farm structure made it difficult to control the disease without large-scale slaughter of exposed animals. The proposed vaccination was intended to protect cattle which were due to move out of the winter housing onto spring pasture. The potential loss of particularly large numbers of animals and the difficulties in disposing of carcasses made vaccination an important option in these two areas. The government accepted the case for an emergency vaccination programme and on 28 March 2001, the UK obtained authority (Decision 2001/257/EC) from the European Commission in Brussels to apply emergency vaccination in Cumbria and Devon. It was clear, however, that any vaccination programme could only succeed with substantial support from the key stakeholders, including farmers, veterinarians, consumers, retailers and food manufacturers. By the end of April it was evident that the level of support for vaccination was insufficient and with the reducing number of cases in Cumbria and Devon the justification for vaccination was less compelling.

The food industry was concerned over the sale of produce from vaccinated animals, particularly the marketing of the produce post vaccination. The overall message from the food retailers was that they would expect to continue stocking meat and milk from pooled milk supplies which included product from vaccinated herds provided public confidence remained. Retailers provided assurances about the marketing of products from vaccinated animals but indicated they would need to reconsider their position if consumer resistance was encountered. The key consideration for retailers was public confidence. If that were to disappear and customers were to demand that product be labelled to identify whether it was from vaccinated or unvaccinated animals the retailers would need to respond. In these circumstances retailers might decide to source their milk and meat only from unvaccinated herds. Many considered these fears to be exaggerated but possible consumer reactions in the midst of an outbreak are difficult to predict so there could be no certainty about the results.

Food safety issues


Early in the outbreak the UK Food Standards Agency (FSA) issued guidance on the safety of eating meat and milk from animals vaccinated against FMD. This guidance referred to the UK Veterinary Products Committee (VPC) safety assessments of the O1 Manisa vaccine (available from the International Vaccine Bank), which already had a UK marketing authorisation at the time of the outbreak. A commercially available FMD vaccine was purchased during the outbreak as a precautionary measure in the event that large-scale vaccination was required at a later stage. This vaccine had been assessed by the VPC for safety, quality and efficacy and had received a UK marketing authorisation from the Veterinary Medicines Directorate (VMD). Unfortunately, due to time constraints the final potency testing required under the terms of the licence were not conducted in the early stages of the outbreak. If the vaccine had been used in the earlier stages of the epidemic it would have been released as an unauthorised medicine. Legally it would not have been the same product as that holding the marketing authorisation and it could not have been labelled with the same trade name. In the event, potency testing was completed later in the year and a full marketing authorisation was issued. In the case of an unauthorised product, assurances would need to be sought from the manufacturer that the manufacturing process was the same as for the authorised product and that for all practical purposes the unauthorised product would be the same as the authorised product. Provided appropriate assurances were received from the manufacturer the use of the vaccine would pose no problems.

Stakeholder concerns
Stakeholders were consulted throughout the discussions on vaccination but attempting to obtain their support at the height of the epidemic was difficult. Food safety was not considered to be a specific issue but it was recognised that vaccination could result in problems if consumer perceptions about the safety of milk and meat changed after vaccination was introduced. If consumer fears about vaccination gained any credence some sections of the food industry considered that this could lead to a two-tier market in meat and dairy products.

Rev. sci. tech. Off. int. Epiz., 26 (2)

453

Vaccination against other diseases


Avian influenza
The UK has recently ordered 10 million doses of AI vaccine for potential use in poultry and other captive birds. The vaccines could be used against both the H5 and H7 strains. These vaccines have a provisional marketing authorisation from the VMD which confirms their safety and quality. Purchasing these vaccines is a precautionary measure taken as part of the contingency plan that has been established in the light of the uncertainties which exist about the future spread and nature of the virus. Vaccination would only be used if risk assessment and scientific evidence indicated that it would help to prevent the spread of disease. Stakeholders have agreed that preventative vaccination of poultry is not the most effective defence against AI because currently available vaccines have a number of disadvantages. The European Medicines Agency has recently recommended that three AI vaccines should be given marketing authorisations under exceptional circumstances. This provides for an accelerated assessment because with the current concerns about the AI situation in both birds and humans there is an urgent need for authorised vaccines to be available as part of disease control campaigns. Their use entails specific obligations (which will be reviewed annually) that are intended to provide additional assurance in relation to the products and to ensure a programme of active pharmacovigilance should the vaccines be used in the field. These vaccines will only be used as part of disease control programmes in compliance with European Union (EU) legislation. Authorisation of these products will provide assurances to national authorities of the quality of the vaccines.

Authorisation of vaccines
Emergency vaccination strategies must be acceptable to stakeholders, who will want assurances that the vaccines to be used at the very least meet regulatory requirements. European legislation requires that veterinary medicinal products must be authorised by means of a marketing authorisation. Minimum requirements (in terms of quality, safety and efficacy) that medicines must meet to obtain an authorisation are specified. The existence of a marketing authorisation confirms that the vaccine is safe in terms of animal and human health and that it works. If there is no authorised vaccine for use against the disease concerned there is an exemption in European legislation from the requirement for an authorisation when a product is to be used in the event of a serious disease epidemic. The European Commission must be informed of the detailed conditions of use. The term serious disease epidemic is not defined in the legislation but clearly applies to outbreaks of FMD and other epizootic diseases. In these circumstances stakeholders will require assurances that the product is safe and of high quality and that there are no risks to consumers from products derived from vaccinated animals.

Consumer attitudes in general


Public concern
There has been a dramatic increase in public concern over food safety during the past decade, partly due to the number of food safety crises that have occurred, such as those surrounding BSE, salmonella, Escherichia coli O157 and dioxin. In the EU there is concern over the use of genetically modified products, eating food from animals treated with antibiotics and the impact on human health of the development of resistance to antibiotics and anthelmintics used in animals. As a consequence there is increasing public awareness and demand for high standards, with consumers expecting food to be safe and free of toxic substances, contaminants, additives, pesticide residues and veterinary medicinal residues. As awareness of food safety grows amongst the public there is a greater need to provide assurances about safety and quality. Perceptions of the extent and prevalence of food safety hazards are constantly changing. Many food safety hazards are well defined, but there remain differences of opinion and a lack of understanding about the degree of risk posed by specific situations, such as emergency vaccination, or by possible new hazards, especially those related to new and advanced vaccines. It is natural for consumers to feel some scepticism towards new and unfamiliar vaccines or situations when the perceived risk may be different from the actual risk.

Other diseases
There are increasing threats from other diseases, many of which pose a risk to humans as well as animals. From a public health perspective AI poses a major risk, but with climate change and globalisation other potential zoonoses such as Rift Valley fever may also constitute a threat to Europe. Foot and mouth disease, classical swine fever (CSF), African swine fever, and swine vesicular disease, all have the potential to create economic problems. The recent incursion of bluetongue into northern Europe (France, Germany, the Netherlands and Belgium) demonstrates how a disease situation can change rapidly. For some of these diseases no vaccines exist, whilst in other cases they exist but have not been licensed or authorised for use.

454

Rev. sci. tech. Off. int. Epiz., 26 (2)

Special Eurobarometer on risk issues


In 2005 the EU Directorate-General for Health and Consumer Protection and the European Food Safety Authority (EFSA) commissioned a survey in all EU Member States to assess how people perceive risk, focusing in particular on food safety. The survey results, published as a Special Eurobarometer on risk issues in 2006 (2) indicate that when people are asked to specifically cite any problems or risks associated with food, many things spontaneously come to mind but without any sense of unanimity. Food poisoning comes to mind most often (16%), followed by chemicals (14%) and obesity (13%). For 7% of respondents food did not present any risks or problems at all. The report notes, however, that attitudes changed when consumers were reminded of the possible risks associated with food: concerns then appeared to be quite widespread. The main finding in the report is that people do not differentiate greatly between the various types of risks, although they are more likely to worry about risks caused by external factors over which they have no control. At the top end of the worry scale, consumers express concern regarding external factors that are clearly identified as dangerous (pesticide residues, new viruses such as AI, residues in meats, contamination of food by bacteria, unhygienic conditions outside the home). In the midrange, one finds other external factors such as environmental pollutants (e.g. mercury), genetically manipulated organisms, food additives, animal welfare and bovine spongiform encephalopathy (BSE). The fear of being endangered by food contaminated by toxic substances, viruses, bacteria, and to a lesser extent by prions, is widespread among Europeans. On average, around a quarter of EU citizens are very worried about food contamination. The level of worry does not vary much between the food contamination issues included in the survey. However, it is interesting to note from the report that close to three out of ten Europeans state that they are very worried about new viruses like AI and a further 38% are fairly worried about this. The report notes that survey participants gave these answers in response to a question that did not specifically focus on the way new viruses were transmitted, but talked about emerging viruses in general. This suggests that it is not a particular method of transmission that worries the public, it is simply that they feel threatened by the existence of new unknown viruses, whatever the means of transmission. The report does not contain specific comments about the vaccination of food-producing animals but it provides background information to explain why there could be consumer concern about vaccination in certain circumstances. The report indicates that when confronted

with possible risks associated with food, consumers identify a wide range of concerns and tend to worry most about those factors which they cannot themselves control. It is clear that European consumers would be concerned about the potential impact of vaccination if they had no control over the products and particularly if products from vaccinated animals were not labelled as such, thereby leaving them without the choice of whether or not to eat them. To overcome these fears consumers need information which is reliable and from a trusted source.

Consumer attitudes to vaccination


Survey in 2001
The decision on whether to vaccinate in 2001 was linked to the attitude of consumers to food products from FMD vaccinated animals. Various surveys conducted throughout the 2001 crisis provided equivocal answers. Only by testing consumer behaviour in the market place could a definitive view be obtained. The FSA made it clear by its announcement in late April that food products from FMD vaccinated animals posed no additional risks to food safety. Consumer organisations also concluded that it was unlikely to be an issue for consumer choice and that special labelling was not required. The Soil Association (a UK environmental organisation promoting sustainable, organic farming) and organic producers supported this view. Small-scale surveys into public opinion in the UK during 2001 indicated that most respondents considered that an alternative to extensive culling in the case of an FMD outbreak was needed. There was a widespread feeling that consideration should be given to a vaccination strategy if it was practical and could be made to work. Whilst most consumers preferred vaccination as a method of avoiding extensive and widespread culling it would be necessary to convince people that it could work and to overcome their reservations about eating meat and other products from vaccinated animals. Most consumers interviewed had some reservations about the idea of eating meat from vaccinated animals and needed assurances that meat would be safe to eat. Even so some retained doubts about the long-term safety of meat from vaccinated animals, which is due partly to the impact of the BSE crisis and the long incubation periods for prion disease in animals and humans. It was mainly mothers with younger children who were most resistant to eating products from vaccinated animals. Most indicated that given the option they would take meat from unvaccinated animals if the meat were labelled vaccinated or

Rev. sci. tech. Off. int. Epiz., 26 (2)

455

non-vaccinated. To overcome these doubts it would be necessary to demonstrate safety and also put the vaccination in context, i.e. remind consumers that vaccines are regularly used to control endemic diseases. The advice from the FSA emphasised that produce from animals routinely vaccinated against other diseases could be safely eaten and that FMD vaccination was no different. Some of the respondents accepted that meat from Argentina probably originated from vaccinated animals and that it was consumed in the UK. Some consumers believed that produce from FMD vaccinated animals was safe but preferred not to consume them if given the choice. Consumer nervousness is a real risk when vaccination is introduced with full media coverage in an emergency situation. This in turn creates fears that retailers would not purchase products of vaccinated livestock, resulting in the animals becoming valueless.

public would reject food products from vaccinated animals. It may be that the public are more likely to suppose that they would accept such food after the event but they may be more cautious during a major outbreak. A range of general conclusions and lessons were described in the study report. Those in relation to vaccination are listed below: explaining complex scientific arguments to the public or to special interest groups in the middle of a crisis is not feasible. This should be anticipated and the foundations of understanding the rationale for, and limitations of, vaccination should be established with the public prior to the next epidemic; informing the public requires their active engagement in the process of gaining knowledge; there was evidence that the lay public interprets issues concerning animal vaccination in terms of understandings of how human vaccination is used. This accounts for some of the difficulties in understanding the vaccinate to kill option. The anchoring of understanding of animal vaccination in human vaccination could be used as the basis for developing future information provision; there was no evidence from the focus groups that the public would be unwilling to purchase or consume meat products from vaccinated animals. The press coverage echoed the FSA advice that such food stuffs would be safe to eat.

Scoping study into public perceptions


A small scoping study (1) was conducted in 2003 into public perceptions concerning animal vaccination, using the 2001 FMD outbreak as a case study. The study had three components: an examination of press coverage of the issue of vaccination during the epidemic data collection from subsets of the lay public likely to represent differing viewpoints on animal vaccination an examination of the concerns expressed by food manufacturers (including farmers) and distributors about the use of vaccination and how these relate to what they believe their customers, both domestic and commercial, would want or would reject. The study established seven focus groups whose members were drawn from different backgrounds and were predicted to have different reactions to the epidemic. The author of the report recognised that focus group methodology typically does not permit tests of the representativeness of the findings for the population as a whole or discrete subsets of it, but the report indicated that the object of a focus group methodology was to capture some of the diversity and complexity of views surrounding the issue. It was clear from the focus groups that there was considerable diversity in public understanding of vaccination. Interestingly, consumers in the focus groups did not indicate that they would have rejected products from vaccinated animals (and food manufacturers did not mention consumer rejection as a possible reason for not vaccinating). This contrasts with the consumer survey carried out during the epidemic, which suggested that the

Eurobarometer survey into avian influenza


The Eurobarometer survey conducted in March and April 2006 (3) questioned over 25,000 EU citizens on AI, in particular the risks it could pose to human health and how it spreads. The study revealed that most EU citizens are well informed about AI but when it came to food safety those surveyed were less sure. Whilst over 60% knew that properly cooked poultry meat and eggs could not transmit the virus, only 47% believed it was true that meat from vaccinated birds was not dangerous. It is of concern that over 50% of those questioned did not believe that eating meat from vaccinated birds carries no risk to human health. There was a marked difference between consumers in different countries, with those showing the highest awareness of the true facts coming from countries where outbreaks have occurred.

Research requirements
The European Technology Platform for Global Animal Health published a strategic research agenda (SRA) in 2006 (4). The SRA identified many of the consumer concerns about technology and the use of veterinary medicines

456

Rev. sci. tech. Off. int. Epiz., 26 (2)

(including vaccines) that have already been discussed in this paper. It was considered important to investigate the background of these concerns by developing an understanding of public perception and societal views on a range of issues such as risk, benefits, products based on new technology, new control measures and ethics. There appears to be a seemingly large information gap between public perception and the scientific situation in reality. It is important to be aware of how society will view advances in technology and the use of new technologies. Not addressing these issues will lead to misunderstanding and mistrust. The actual rather than the perceived risk for new technologies or existing technologies to control animal diseases must be discussed with the wider public to ensure social acceptance. A number of studies into public engagement and understanding have been carried out in the past. The SRA recognised that societal studies were needed to assess the impact of new technologies or alternative eradication programmes which use veterinary medicines. The SRA identified a need to evaluate the most effective ways to present new technology and new programmes to the public. An evaluation of the risk communication and science strategies available to present new information would be of considerable assistance in the development of publicly acceptable evidence-based policies to control epizootic diseases.

The statement records that discussion with retailers has confirmed that meat and milk from vaccinated animals would not be separately identified, indeed there is no reason to do so. Vaccination is now accepted as one of the important options available in fighting an FMD outbreak and its implications are now seen as practical ones regarding the handling and marketing of products from vaccinated animals rather than ones of the acceptability of those products to retailers and consumer groups.

Conclusions
A review of the available literature does not provide a great deal of specific information on consumer attitudes to vaccination of food-producing animals. Experiences in the FMD crisis and subsequently with AI indicate that there is potential public concern over the consumption of products from animals vaccinated as a control measure during a major outbreak of disease. This contrasts with the apparent lack of concern about routine vaccination against a wide range of diseases in most food-producing species. This is not unexpected as there is little public discussion on vaccination of food-producing animals during periods when there are no active national vaccination campaigns. Furthermore, the public generally identifies with vaccination in a favourable manner due to the benefits to human health and the positive impact of vaccination in companion animals. An analysis of a number of linked surveys indicates that consumers are most concerned when actions take place outside their own control and where they have no choice. It would also appear that in the height of an epidemic there is increased sensitivity to the potential impact of the control measures. Even though the media often reflects the advice from the food safety authorities, the fact that the subject of food safety is constantly raised impacts on public consciousness and often leads to increased concern of the unknown. Whilst many aspects of the safety of vaccines are a scientific issue, public perception is equally important in the policy-making process, especially if the issue has a high political impact. The discussions around vaccination against FMD, CSF and AI show that there are potentially non-scientific concerns over vaccination. Many agree on the value of existing or new vaccines but the main obstacle to their success may be the issue of public acceptance. Lack of societal acceptance can also be a barrier to the development and use of new technologies to control disease. It is important to anticipate this type of debate and to ensure that understanding and agreement is reached before vaccination is used to control outbreaks of epizootic

Current developments in the United Kingdom


Many lessons have been learned since 2001, not least of which is that the issue of vaccination should be settled at an early stage in the contingency planning process. Since January 2003, there have been extensive discussions and regular meetings with UK stakeholders to discuss the implications of emergency vaccination and to obtain support from the whole of the food chain should emergency vaccination be required in future. Initially these discussions related to FMD, but a similar process is now in place for AI. There are understandable concerns about the marketing of products post vaccination. A statement summarising the situation on the use of FMD vaccination as part of disease control strategies has been produced by the UK government in cooperation with consumer organisations. In the statement the FSA confirmed that the consumption of products from animals treated with authorised FMD vaccines would not have any implications for food safety. Furthermore BEUC, the European Consumers Organisation, also stated in December 2004 that, from the perspective of consumer organisations there is no safety concern with products from emergency vaccinated animals. Consumer organisations in the UK have confirmed that they support this view.

Rev. sci. tech. Off. int. Epiz., 26 (2)

457

disease. A number of important steps can be taken to maintain public confidence. These include: developing a vaccination policy for inclusion in the contingency plans before an outbreak occurs and identifying the circumstances in which vaccine will be used discussing the vaccination policy with all stakeholders obtaining public support for the control policy ensuring that vaccines to be used have a licence or marketing authorisation for use in the country concerned providing and discussing safety information with all stakeholders if an unauthorised vaccine needs to be used in an emergency avoiding a two-tier system by not separately identifying products from vaccinated and unvaccinated sources and issuing clear statements that products from vaccinated animals will be used as part of the general food supply providing unequivocal and authoritative assurance that vaccination poses no threat to human health and that products from vaccinated animals are safe to eat ensuring that national and international independent bodies that are respected by the consumer issue statements to reassure consumers that the consumption of products from vaccinated animals poses no risk to human health ensuring that any statements are endorsed by the producer, retailer and consumer organisations

developing a communication strategy involving all stakeholders to ensure that consistent messages on vaccination are provided by all stakeholders before and during an outbreak in which emergency vaccination may be used implementing a concerted campaign organised by the government and the industry to convey the safety messages whenever emergency vaccination is to be used to control a disease outbreak. Public disapproval of control measures such as mass slaughter to control epizootic diseases will continue to contribute to a drive for vaccination as an alternative measure. As vaccination is seen as ethically and morally acceptable public acceptance of such a measure would be high. However, the results of surveys are equivocal and it is not always clear that consumers are willing to eat products from vaccinated animals. It is essential to have a clear and agreed policy on the use of vaccines against FMD and other diseases that can cause serious economic problems. Clear advice to consumers by respected independent bodies such as the FSA in the UK and EFSA is critical to ensure a successful vaccination policy. Consumer confidence is essential for any emergency vaccination programme and a major public relations programme would be needed.

Comportement des consommateurs lgard de la vaccination des animaux destins lalimentation humaine
J.M. Scudamore
Rsum Lpisode de fivre aphteuse survenu en 2001 au Royaume-Uni a mis en vidence la ncessit de mettre en uvre une politique de vaccination lacm de lpizootie. En raison de lambigut des rsultats des enqutes dopinion ralises jusqualors, il tait impossible dapprcier le degr de mfiance des consommateurs lgard des aliments issus danimaux vaccins. Une enqute rcente sur linfluenza aviaire a rvl que si les consommateurs europens sont bien informs au sujet de la maladie et des mthodes de lutte applicables, plus de 50 % des personnes interroges ne sont pas disposes consommer de la viande issue de volailles vaccines. Il ny a pas dinformations prcises sur lopinion des consommateurs lgard de la vaccination rgulire visant dautres maladies. Les proccupations du public semblent saccentuer pendant les priodes de crise, qui ont une plus forte couverture mdiatique. Le dveloppement de nouveaux types de vaccins doit saccompagner de mesures

458

Rev. sci. tech. Off. int. Epiz., 26 (2)

visant rassurer les consommateurs quant linnocuit et lutilisation de ces produits. Lauteur examine ces questions et fait quelques recommandations pratiques sur les moyens de rassurer le public en cas dapplication de la vaccination durgence pour lutter contre les maladies animales. Mots-cls Attitude des consommateurs Confiance du public Influenza aviaire Leon de lexprience Proccupation des consommateurs Scurit sanitaire des aliments Vaccination.

Actitud del consumidor frente a la vacunacin de animales destinados al consumo humano


J.M. Scudamore
Resumen El brote de fiebre aftosa que en 2001 asol el Reino Unido fue un episodio sin precedentes, frente al cual hubo que obtener una vacuna justo en el momento lgido de la epidemia. Dados los resultados poco claros de las encuestas, no fue posible determinar el grado de preocupacin de los consumidores por el hecho de ingerir alimentos procedentes de animales vacunados. De una reciente encuesta relativa a la influenza aviar se desprende que el gran pblico europeo est bien informado sobre la enfermedad y su control, aunque ms del 50% de los encuestados seran reacios a consumir carne procedente de aves vacunadas. No existe mucha informacin especfica sobre la opinin de los consumidores acerca de la vacunacin sistemtica contra otras enfermedades. Su inquietud parece acrecentarse en las situaciones de emergencia, cuando los medios de comunicacin ofrecen abundante informacin sobre el tema. Con la aparicin de nuevos tipos de vacuna, ser preciso ofrecer al consumidor ms garantas sobre el uso y la inocuidad de tales productos. Adems de examinar todas estas cuestiones, el autor formula recomendaciones prcticas para ofrecer al gran pblico las debidas garantas a la hora de proponer una vacunacin de emergencia con fines zoosanitarios. Palabras clave Actitud del consumidor Confianza del gran pblico Enseanzas extradas Fiebre aftosa Influenza aviar Inocuidad de los alimentos Preocupacin del consumidor Vacunacin.

Rev. sci. tech. Off. int. Epiz., 26 (2)

459

References
1. Breakwell G.M (2003). Public perceptions concerning animal vaccination: a case study of foot and mouth 2001. Available at: http://www.defra.gov.uk/science/documents/ publications/mp0140.pdf (accessed on 12 September 2006). 3. European Commission (2006). Special Eurobarometer report number 257 on avian influenza. Available at: http://ec.europa.eu/public_opinion/archives/eb_special_en. htm (accessed on 12 September 2006). 4. European Technology Platform for Global Animal Health (2006). Strategic Research Agenda: societal acceptance of technology, 44-45. Available at: http://www.ifah.be/Europe/ euplatform/SRA_May06.pdf (accessed on 12 September 2006).

2. European Commission (2006). Special Eurobarometer report number 238 on Risk issues. Available at: http://ec.europa.eu/public_opinion/archives/eb_special_en. htm (accessed on 12 September 2006).

Rev. sci. tech. Off. int. Epiz., 2007, 26 (2), 461-470

Vaccines for immunological control of fertility in animals


C.M. Hardy & A.L. Braid
Commonwealth Scientific and Industrial Research Organisation GPO Box 1700, Canberra, ACT, 2601, Australia, and Invasive Animals Cooperative Research Centre, 3D1 University of Canberra, ACT 2601, Australia. E-mail: chris.hardy@csiro.au

Summary Fertility control has gained considerable momentum as a management tool to regulate populations of captive and wild animals and to control aggressive behaviour or improve meat quality in livestock. Anti-fertility vaccination (immunocontraception and immunocastration) is a humane alternative to methods that rely on surgical or chemical sterilisation and lethal control. Two types of experimental immunocontraceptive vaccine have been registered for field use in animals. They contain either porcine zona pellucida (PZP) proteins extracted from pig ovaries or synthetic conjugated gonadotrophin releasing hormone (GnRH) peptides. These vaccines require repeated injections and are limited to captive or small populations of free-ranging wild animals. Alternative immunocontraceptive vaccines are actively being developed either to improve efficacy or enable large numbers of wild animals to be targeted. Some employ live genetically modified viruses to deliver immunocontraception and have proved successful under laboratory conditions. The relative merits, risks, social acceptability and regulations controlling the use of existing and novel animal immunocontraceptives are reviewed. Keywords Animal contraception Ethics Genetically modified organisms Legislation Risk.

Introduction
Fertility control, either by surgical, chemical or immunological means has been demonstrated to be a viable alternative to lethal methods of controlling animal populations (9, 21, 38). The appropriate approaches to controlling fertility depend on the target species and intended outcome and include consideration of factors such as relative efficacy and safety, economic value, delivery mechanism, non-target and environmental effects and social acceptability (5, 9, 25, 58). Each approach raises ethical (48) and social acceptability issues (11). In contrast to vaccines that protect against infectious diseases, anti-fertility vaccines aim to induce immune responses (antibodies or cellular immunity) against molecules naturally present in an individual. These include

proteins or chemicals involved in the production of gametes and sex steroids, release of viable sperm or eggs, fertilisation, implantation and subsequent embryo development (20). Immunological approaches to controlling fertility lead to three different outcomes, depending on the reproductive functions that are affected. These outcomes are contraception, castration or interference with gestation (5, 25, 60). Immunocontraception describes the prevention of fertilisation, either by blocking the release of sperm or eggs or their subsequent interactions in the female reproductive tract. It can also be considered to include immunological interference with receptiveness of the uterine environment to embryo implantation in females. Immunocastration on the other hand describes autoimmune responses that directly affect the structure and function of the gonads in either sex, thus interfering with their ability to produce gametes and sex steroids. The third target for

462

Rev. sci. tech. Off. int. Epiz., 26 (2)

immunological interference with reproduction is postimplantation embryo development and survival (maintenance of pregnancy). Immunocontraception and immunocastration can be applied to both sexes. Immunocontraception ideally is reversible once vaccination ceases and should not affect levels of circulating sex hormones or libido in any discernable way (1). Immunocastration, apart from infertility, causes a reduction in sex hormone levels and alterations to metabolism. It is generally applied in cases where modification to behavior such as aggression control is required (19, 43, 65). Interference with pregnancy applies only to females and is linked to health risks for the mother and fetus, particularly late in gestation. It can also appear as an undesired side effect in some other contraceptive approaches and raises ethical issues comparable to those associated with chemical or surgical termination of pregnancy (2, 60). Anti-fertility vaccines are intended to break immune tolerance to self antigens and so pose a real risk of inducing unintended long-term autoimmune disease (69). Many potential target molecules are either directly required for physiological activities other than reproduction or have features in common with molecules with different functions. Vaccines against fertility must therefore selectively target antigens to minimise the risk of immunological cross-reactions and avoid unintended interference with other physiological processes (61).

reversible method for animal applications (5) and oral delivery of chemical contragestatives has also been used with mixed success to control populations of overabundant bird species (87). In humans, the technology for safe female chemical contraception is well advanced (7), but remains elusive for males (29). Chemical contraception can be associated with metabolic side effects and severe pathology in some species following prolonged use (18, 55). In livestock, the presence of steroid metabolites in the animals can affect meat quality (40, 71, 76) or growth rates (34). The barrier method and intrauterine devices are effective and have been used with high-value animals (dogs, monkeys and camels) but have not been widely adopted (82). It has been recognised that alternative forms of contraception need to be made more widely available both for human (58) and animal use (24).

Immunological fertility control


Adjuvant vaccines
Most vaccines that have been used for routine fertility regulation in captive or small groups of free-ranging animals are comprised of various formulations of proteins or peptide antigens mixed with adjuvants. The most effective antigens fall into three main groups: those derived from the gonadotrophin releasing hormone (GnRH), the luteinizing hormone releasing hormone (LHRH) and the follicle stimulating hormone (FSH) the structural glycoproteins forming the zona pellucida (ZP) that surrounds the oocyte the ovary and/or placental hormones, e.g. luteinizing hormone (LH) and human chorionic gonadotrophin (hCG) (19, 27, 30). Many other antigens (natural and recombinant proteins and synthetic peptides), most notably derived from sperm, also have immunocontraceptive potential based on a large number of experimental studies, but so far appear less efficacious than hormonal or ZP antigens (31, 60). Vaccines based on GnRH and pig ZP (PZP) have proved practical to use in a range of captive and wild animals where direct injection or darting is feasible, whereas hCG vaccines are restricted to primates (25). Immunisation against GnRH prevents LH and FSH synthesis and leads to atrophy of the ovaries and testes. This generally induces reversible castration in males and females, although it can lead to irreversible infertility in rats (49) and lesions in the hypothalamus in pigs (54). However, immunising against the beta subunit of hCG or PZP also causes reversible contraception by preventing fertilisation and gamete development in the case of PZP (8) or early embryo

Non-immunological fertility control


Conventional methods of regulating fertility in a wide range of animals include physical separation, chemical contraception, surgical sterilisation and barrier methods (5, 9, 21, 42). All these approaches have certain advantages, but also suffer from drawbacks (24, 57). Physical separation, whilst highly effective, can affect the behaviour of animals, particularly males. Surgical sterilisation (castration and hysterectomy) and contraception (tubal ligation) are highly effective for captive animals when irreversible infertility is required. However, this approach generally requires veterinary skills and since it involves invasive procedures, causes pain, discomfort and carries the risk of infection in the short term. They are not readily applicable to wildlife. Long term, castration prevents sex steroid production and affects metabolism. Tubal ligation is highly effective where there is a need to retain hormonally-dependent social hierarchies, but is not effective for controlling undesirable sex-related behaviour. Chemical contraception or castration (using oral or injectable implants) can be a highly effective and

Rev. sci. tech. Off. int. Epiz., 26 (2)

463

survival in the case of hCG (3, 73, 78). All these selfantigens require potent adjuvants and repeated immunisations to generate their effect and overcome immune tolerance. The antigens and some adjuvants in current use (particularly Freunds complete, Freunds modified and other oil-based formulations) are known to be irritant and are associated with side effects that vary significantly in their nature and intensity, depending on the species and sex targeted (27, 33, 56).

Other immunological approaches


Several studies have explored the use of bacteria as delivery vectors for immunocontraception (13, 50, 75, 88), although efficacy has been disappointing. DNA vaccines (14, 59, 62, 86), recombinant plant viruses (26) and recombinant bacteriophages (1) have also been trialed in mice and primates with some success.

Risks of exposure to anti-fertility vaccines for human handlers Virally vectored immunocontraception
A relatively recent development is the use of genetically modified viral vectors to produce anti-fertility biological control agents. In this approach, termed virally vectored immunocontraception (VVIC), the gene for a fertility antigen is inserted into the genome of a replication competent virus. The recombinant virus then expresses the antigen in infected animals and induces a contraceptive autoimmune response (80). The viral vectors can either be used for the direct inoculation of individuals or they can be administered in a way that allows the vectors to be transmitted (self-dissemination) among free-ranging wildlife and pest animals. These vaccines are intended to supplement current control practices of shooting, trapping and poisoning for wide-scale problem species. VVIC has yet to be tested outside laboratory and quarantine containment, but it shows considerable promise as a biological control for wildlife (31, 32). Virally vectored immunocontraception has been tested with varying degrees of success in laboratory trials in a range of species; vaccines tested include recombinant vaccinia virus in rats and foxes, myxoma virus in rabbits, canine herpesvirus in foxes, and ectromelia virus and murine cytomegalovirus in mice and rats (32). The approach has proven highly successful in the laboratory and infertility rates approaching 100% have been achieved in nave mice infected with recombinant viruses expressing mouse ZP3 after only a single inoculation (35, 44, 67). Sterility, as well as reversible long- and short-term, single shot contraception, without the need for adjuvants, appear feasible using this approach. The long-term safety and reversibility of hCG, FSH and GnRH immunocontraceptive vaccines have been directly assessed in humans. Various formulations of hCG and FSH vaccines have also undergone fertility trials in humans (19, 60), although clinical human trials with a GnRH vaccine were restricted to male patients with advanced carcinoma of the prostate (77). The potential safety issues for humans at risk from unintentional exposure to most contraceptive vaccines being used in animals can only be inferred from primate or other animal studies. As some immunocontraceptive antigens (particularly those based on FSH and GnRH) have the potential to affect fertility in both males and females, precautions should be taken to minimise the risk of accidental exposure to these agents when administering them to animals. However, repeated or prolonged exposure by accidental injection would be required in most cases to induce long-term effects in human handlers. A recent comprehensive coverage of the relative merits of currently available chemical and immunological agents for animal contraception, including the procedures and precautions that should be taken when handling each of these products has been made available on the website of the AZA Wildlife Contraception Center at Saint Louis Zoo in the United States of America (USA) (http://www.stlzoo.org/downloads/CAGrecs2006final.htm). The use of PZP antigen may pose an additional risk to human handlers as they must be given using either Freunds complete or modified adjuvants (27). Adjuvants such as these contain a combination of mycobacteria and mineral oil that have been reported to cause severe localised lesions in people following accidental selfinoculation (84). The PZP antigens themselves are also associated with the appearance of adverse side effects in some species, including T-cell mediated ovarian damage (oophoritis) and inhibition of steroid production (6, 64, 68, 74, 81). In other species, there do not appear to be any significant PZP-related side effects in somatic tissues after PZP immunisation (8).

Nematode-vectored immunocontraception
New Zealand Landcare Research and AgResearch are developing a parasitic nematode (Parastrongyloides trichosuri) for biological control of possums in New Zealand (17). The ability to produce transgenic P trichosuri . has been established (28) and it appears amenable for use as an immunocontraceptive vaccine vector in the field as the parasite is specific to possums and is able to establish rapidly in wild possum populations (17).

Regulatory issues
The various national regulatory frameworks that govern the use of animal contraceptives are complex (39). Each

464

Rev. sci. tech. Off. int. Epiz., 26 (2)

country has its own set of regulations and legislations, although many international arrangements are also in place, and researchers or practitioners are required to adhere to their own jurisdictions guidelines. Vaccines against GnRH (VaxstrateTM and ImprovacTM) have been registered for control of libido and sex steroid production as well as increased growth in livestock such as pigs, sheep and cattle in Australia (19, 25). Another GnRH vaccine (GonaConTM) is registered as an experimental wildlife vaccine by the United States Food and Drugs Administration (FDA). It is undergoing a number of field studies in the USA, principally in white-tailed deer, by the National Wildlife Research Center (NWRC) in the United States Department of Agriculture (USDA) Wildlife Services (20, 25, 51, 52). A vaccine derived from whole solubilised PZP (SpayVacTM) is also registered as an investigational new animal drug by the FDA for use in captive zoo animals and certain wildlife applications (27). There are considerable regulatory and social acceptability issues linked with the use of live genetically modified immunocontraceptive virus vaccines. These include the perceived risks associated generally with the use of genetically modified organisms and concerns about species-specificity, efficacy, safety, choice and international implications for trade and biodiversity (4, 45). The presence of strict regulatory controls governing any intended registration of VVIC products has meant that this technology has not yet been approved for use in any country outside containment facilities for handling genetically modified organisms.

with GnRH in bison is being explored as a possible means of controlling the transmission of brucellosis to adjoining populations of cattle in the USA (52).

Social and ethical issues


The ethical principles underpinning the rights, welfare and fertility control of captive and free-ranging animals have been debated (53, 85). It is beyond the scope of this paper to address the rights of animals to live their lives without human interference (66), whether animals should have freedom to express normal behaviour (23), or whether animals suffer from imposed castration or contraception (83). It is, however, understood that separation of males from females is the preferable means for controlling fertility in livestock and zoos. However, animal welfare issues and the ethics of imposed fertility control of free-living and captive wildlife will be discussed. The use of immunocastration and immunocontraception has been recognised as providing animal welfare benefits relative to alternative procedures (63, 66, 70, 80). In production animals, the animal welfare benefit is the provision of alternatives to the on-farm surgical removal of an animals gonads, usually undertaken without anaesthetic. In free-living wildlife, fertility control is recognised by animal welfare proponents as preferable to most lethal methods of controlling populations (63, 66).

Production animals

Ecological issues
It has been proposed that the widespread use of immunocontraception in wildlife may lead to the development of genetic resistance or heritable immune dysfunctions that reduce the ability of animals in the target populations to combat diseases (15, 16, 46, 57). However, most debate on applying immunocontraceptive products to wildlife relates to the relative environmental impacts of population control using sterility versus lethal methods and the theoretical risks of affecting non-target species in the case of viral-vectored immunocontraceptive agents (10). The actual impacts of behavioural modifications in wildlife and the risk to non-target species still need to be determined as few long-term data sets are available, although most authors concede that fertility control methods such as immunocontraception so far appear likely to provide significant environmental benefits over lethal control (9, 10, 11, 13, 38, 52, 80). Positive benefits of immunocontraception have been reported to include weight gain and increased longevity in populations of wild horses treated with PZP (38) and immunocontraception

In the pig industry, to avoid boar taint due to fat androstenone in the meat, male piglets are routinely castrated without anaesthetic during the first fews weeks of life. European Commission Directive 2001/93/EC still allows castration of piglets less than seven days of age without anaesthesia, even though research has shown that pain associated with castration is not affected by age (79). There have been campaigns by the Royal Society for the Prevention of Cruelty to Animals (RSPCA) in the United Kingdom (UK), and others, to ban the practice. Alternatives include no castration and early slaughter, surgical castration under general or local anaesthetic, immunocastration and sex-sorting of sperm for the selective production of female pigs (41). Immunocastration, using vaccination against GnRH (ImprovacTM CSL, Australia) has been shown to avoid the pain associated with surgical castration, suppress androstenone and improve the yield of lean meat in vaccinated pigs compared to those surgically castrated (36). Immunocontraception with another GnRH vaccine (VaxstrateTM. Arthur Webster, Australia) has been applied

Rev. sci. tech. Off. int. Epiz., 26 (2)

465

commercially as an alternative to surgical speying of heifers. This procedure is applied to stop them becoming pregnant whilst they are being fattened for slaughter under free-ranging conditions. Although originally promoted for the animal welfare benefits inherent in avoiding surgical speying without anaesthesia, the vaccine is no longer commercially available due to the additional cost associated with mustering of cattle for booster vaccinations, the cost of the vaccine itself and the lack of effect in stopping a proportion of heifers from cycling and becoming pregnant (12, 37).

Zoo animals
Population management in zoos is a particularly difficult issue as there is an imperative to allow for natural behaviour in captive animals, including reproduction, and to conduct successful breeding programmes while not producing surplus animals (83). Immunocontraception with PZP has been used succesfully in a range of zoo species. However, safety concerns preclude its use in pregnant animals and long-term effects have been described that include disruption to the reproductive endocrine system in some species. The time taken to return to fertility also varies between species (27).

of animals involved, the widespread use of non-target specific toxicants and the use of lethal disseminating disease agents has led to considerable debate on the ethics of controlling pest animals (15, 45, 63, 72, 80). A contentious ethical position for controlling these populations has been advocated that combines the principles of ecocentrism and animal welfare (22, 47). Ecocentrism holds that ecosystems have their own intrinsic values and moral standings which can be equal to or greater than individual species, including humans. Under this framework, the conservation value of the indigenous ecosystem (native plants and animals) requires the control of the non-indigenous populations and overrides the animal rights ethic of equal consideration of the interests of all sentient beings (72). However, it is considered that an animal welfare ethic should also be applied and control of non-indigenous sentient animals should be undertaken by the most effective and humane methods available, such as immunocontraception.

Conclusions
Immunocontraception and immunocastration are developing into viable management tools for fertility regulation in both captive and wild animals. In particular, fertility control by immunological means provides an attractive and ethically supported alternative to surgical sterilisation, as it is considerably less invasive and more humane. Nevertheless, apart from PZP vaccination in some zoos and promotion of GnRH vaccines as an alternative to castration in male piglets, immunocontraceptive vaccines are only available for experimental use. The lack of penetration of existing vaccines into many markets, such as companion animals, is due to a combination of factors, including uncertainties surrounding their long-term efficacy and safety, the requirement for repeated inoculation using potentially unacceptable adjuvants and relatively high production costs. However, if these issues can be resolved, the next generation of immunocontraceptive vaccines should be able to provide consistent, long-term infertility after a single application and provide realistic alternatives to chemical and surgical methods.

Free-living wildlife
There are a range of ethical issues associated with the differing strategies used to control free-living animals in most countries and three different ethical positions on wildlife population control have been taken (66). These are the Animal Rights position, i.e. an animals right to control its own life should not be interfered with in any way; the Environmental or Natural position, i.e. animal populations can only be controlled by predation or hunting and; the Animal Welfare position, i.e. populations should be controlled by non-lethal methods such as immunocontraception. Another ethical position has been taken in countries with large numbers of introduced, non-indigenous animals that are serious pests. These pests include rabbits (Oryctolagus cuniculus), foxes (Vulpes vulpes) and cats (Felis domesticus) in Australia and ferrets (Mustela putorius), stoats (Mustela erminea) and possums (Trichosurus vulpecula) in New Zealand. The large populations of non-indigenous species have severe negative impacts on native plants and animals. The control of these populations is undertaken for two reasons: their economic impact on agricultural production, and their damage to the environment. The large numbers

Acknowledgements
The authors wish to thank Tony Robinson for critically reviewing the manuscript.

466

Rev. sci. tech. Off. int. Epiz., 26 (2)

Les vaccins et le contrle immunologique de la fcondit chez les animaux


C.M. Hardy & A.L. Braid
Rsum Le contrle de la fcondit a pris un lan considrable, en tant quoutil de gestion des populations des animaux en captivit et sauvages et moyen de contenir les comportements agressifs et damliorer la qualit de la viande issue des animaux de rente. La vaccination visant contrler la fcondit (contraception et castration immunologiques) est une alternative aux mthodes bases sur la strilisation chirurgicale ou chimique ou sur le contrle ltal des populations. Deux types de vaccins exprimentaux ont t enregistrs pour la contraception immunologique des animaux, en vue dune utilisation sur le terrain. Ils contiennent soit des protines prleves de la zone pellucide (PZP) dovaires de truie, soit des conjugus de peptides synthtiques de lhormone stimulatrice de la gonadotrophine (GnRH). Ces vaccins devant tre administrs rgulirement, leur utilisation nest envisageable que pour des animaux maintenus en captivit ou pour des populations limites danimaux sauvages. Dautres possibilits de contraception immunologique sont ltude afin damliorer lefficacit de la mthode ou de permettre son utilisation sur des populations plus nombreuses danimaux sauvages. Certaines de ces mthodes, bases sur lutilisation de virus vivants gntiquement modifis pour empcher la conception, ont donn de bons rsultats au laboratoire. Les auteurs examinent les mrites et les risques respectifs des diffrentes mthodes de contraception immunologique disponibles ou en voie de dveloppement, ainsi que lacceptation sociale et la rglementation applicable en la matire. Mots-cls Contraception animale thique Lgislation Organisme modifi gntiquement Risque.

Vacunas para el control inmunolgico de la fertilidad en animales


C.M. Hardy & A.L. Braid
Resumen El control de la fertilidad ha cobrado un notable impulso como instrumento de gestin para regular las poblaciones de animales salvajes y en cautividad, y tambin para controlar la agresividad o mejorar la calidad de la carne en el ganado vacuno. Las vacunas anti-fertilidad (inmunoanticoncepcin e inmunocastracin) constituyen una alternativa suave a los mtodos basados en el sacrificio o en la esterilizacin quirrgica o qumica. Hoy en da estn registrados dos tipos de vacuna inmunoanticonceptiva para una utilizacin experimental sobre el terreno, constituidas en un caso por protenas de zona pelcida porcina, extradas a partir de ovarios de cerdo, y en el otro caso por un

Rev. sci. tech. Off. int. Epiz., 26 (2)

467

conjugado sinttico de pptidos de la hormona liberadora de gonadotropina (GnRH, por sus siglas en ingls). Estas vacunas requieren repetidas inyecciones y slo pueden usarse en el caso de poblaciones en cautividad o de un pequeo nmero de animales salvajes en libertad. Estn en marcha procesos para obtener vacunas alternativas, que mejoren la eficacia de las anteriores o puedan administrarse a grandes poblaciones de animales salvajes. En algunas de ellas se emplean virus vivos genticamente modificados para lograr la inmunoanticoncepcin, mtodo que se ha demostrado eficaz en condiciones de laboratorio. Los autores examinan las ventajas y riesgos, la aceptabilidad social y los reglamentos que rigen el uso de los mtodos de inmunoanticoncepcin animal ya arraigados o de reciente aparicin. Palabras clave Contracepcin animal tica Legislacin Organismos genticamente modificados Riesgo.

References
1. Abdennebi-Najar L. (2003). Interests and limits of immunocontraception. Gynecol. Obstet. Fertil., 31, 774-777. 2. Adiga P.R., Subramanian S., Rao J. & Kumar M. (1997). Prospects of riboflavin carrier protein (RCP) as an antifertility vaccine in male and female mammals. Hum. Reprod., 3, 325334. 3. Aitken R.J. (2002). Immunocontraceptive vaccines for human use. J. reprod. Immunol., 57, 273-287. 4. Angulo E. & Cooke B. (2002). First synthesize new viruses then regulate their release? The case of the wild rabbit. Mol. Ecol., 11, 2703-2709. 5. Asa C.S. (2005). Types of contraception. In Wildlife contraception: issues, methods, and applications (C.S. Asa & I.J. Porton, eds). The Johns Hopkins University Press, Baltimore, Maryland, 29-52. 6. Bagavant H., Sharp C., Kurth B. & Tung K.S. (2002). Induction and immunohistology of autoimmune ovarian disease in cynomolgus macaques (Macaca fascicularis). Am. J. Pathol., 160, 141-149. 7. Baird D.T., Collins J., Crosignani P.G., Glasier A., Jacobs H.S., La Vecchia C., Milsom I., Reid B., Templeton A., Vercellini P., Benagiano G., Frigerio G.C., Hoppe G., Liberatore S., Schmidt-Gollwitzer K., Skouby S.O. & Crosignani P.G. (2005). Noncontraceptive health benefits of combined oral contraception. Hum. Reprod., 11, 513-525. 8. Barber M.R. & Fayrer-Hosken R.A. (2000). Evaluation of somatic and reproductive immunotoxic effects of the porcine zona pellucida vaccination. J. experim. Zool., 286, 641-646. 9. Barfield J.P., Nieschlag E. & Cooper T.G. (2006). Fertility control in wildlife: humans as a model. Contraception, 73, 6-22. 10. Barlow N.D. (2000). The ecological challenge of immunocontraception: editors introduction. J. appl. Ecol., 37, 897-902. 11. Barr J.J.F Lurz P.W.W., Shirley M.D.F & Rushton S.P. (2002). ., . Evaluation of immunocontraception as a publicly acceptable form of vertebrate pest species control: the introduced grey squirrel in Britain as an example. Environ. Manag., 30, 342-351. 12. Bonneau M. & Enright W.J. (1995). Immunocastration in cattle and pigs. Livest. Prod. Sci., 42, 193-200. 13. Bradley M.P., Eade J., Penhale J. & Bird P. (1999). Vaccines for fertility regulation of wild and domestic species. J. Biotechnol., 73, 91-101. 14. Chen Y., Liu Z., Yang Y., Chen Y.Z. & Peng J.P. (2002). Infertility in mice induced by the rhesus monkey chorionic gonadotropin beta-subunit glycoprotein (rmCGbeta) using DNA immunization. Mol. cell. Biochem., 231, 89-96.

468

Rev. sci. tech. Off. int. Epiz., 26 (2)

15. Cooper D.W. & Herbert C.A. (2001). Genetics, biotechnology and population management of over-abundant mammalian wildlife in Australasia. Reprod. Fertil. Dev., 13, 451-458. 16. Cooper D.W. & Larsen E. (2006). Immunocontraception of mammalian wildlife: ecological and immunogenetic issues. Reproduction, 132, 821-828. 17. Cowan P.E., Ralston M.J., Heath D.D. & Grant W.N. (2006). Infection of naive, free-living brushtail possums (Trichosurus vulpecula) with the nematode parasite Parastrongyloides trichosuri and its subsequent spread. Int. J. Parasitol., 36, 287293. 18. De Vleeschouwer K., Leus K. & Van Elsacker L. (2000). An evaluation of the suitability of contraceptive methods in golden-headed lion tamarins (Leontopithecus chrysomelas), with emphasis on melengestrol acetate (MGA) implants: (i) effectiveness, reversibility and medical side-effects. Anim. Welf., 9, 251-271. 19. Delves P.J. (2004). How far from a hormone-based contraceptive vaccine? J. reprod. Immunol., 62, 69-78. 20. Delves P.J., Lund T. & Roitt I.M. (2002). Antifertility vaccines. Trends Immunol., 23, 213-219. 21. Dutton C.J. & Allchurch A.F (1998). A review of birth . control methods in mammals at the Jersey Wildlife Preservation Trust. Dodo, J. Jersey Wildl. Preserv. Trust, 34, 134-144. 22. Eggleston J.E., Rixecker S.S. & Hickling G.J. (2003). The role of ethics in the management of New Zealands wild mammals. New Zeal. J. Zool., 30, 361-376. 23. El Balaa R. & Marie M. (2006). Animal welfare considerations in small ruminant breeding specifications. J. agric. environ. Ethics, 19, 91-102. 24. Ferro V.A. (2002). Current advances in antifertility vaccines for fertility control and noncontraceptive applications. Expert Rev. Vaccines, 1, 443-452. 25. Ferro V.A. & Mordini E. (2004). Peptide vaccines in immunocontraception. Curr. Opin. molec. Therapeut, 6, 83-89. 26. Fitchen J., Beachy R.N. & Hein M.B. (1995). Plant virus expressing hybrid coat protein with added murine epitope elicits autoantibody response. Vaccine, 13, 1051-1057. 27. Frank K.M., Lyda R.O. & Kirkpatrick J.F. (2005). Immunocontraception of captive exotic species IV. Species differences in response to the porcine zona pellucida vaccine, timing of booster inoculations, and procedural failures. Zoo Biol., 24, 349-358. 28. Grant W.N., Skinner S.J.M., Newton-Howes J., Grant K., Shuttleworth G., Heath D.D. & Shoemaker C.B. (2006). Heritable transgenesis of Parastrongyloides trichosuri: a nematode parasite of mammals. Int. J. Parasitol., 36, 475-483.

29. Grimes D.A., Gallo M.F., Grigorieva V., Nanda K. & Schulz K.F (2005). Steroid hormones for contraception in . men: systematic review of randomized controlled trials. Contraception, 71, 89-94. 30. Gupta S.K., Srivastava N., Choudhury S., Rath A., Sivapurapu N., Gahlay G.K. & Batra D. (2004). Update on zona pellucida glycoproteins based contraceptive vaccine. J. reprod. Immunol., 62, 79-89. 31. Hardy C.M. (2007). Current status of virally vectored immunocontraception for biological control of mice. In Society of Reproduction and Fertility Supplement Number 63. Gamete biology: emerging frontiers in fertility and contraceptive development (S.K. Gupta, K. Koyama & J.F. Murray, eds). Nottingham University Press, United Kingdom, 495-506. 32. Hardy C.M., Hinds L.A., Kerr P.J., Lloyd M.L., Redwood A.J., Shellam G.R. & Strive T. (2006). Biological control of vertebrate pests using virally vectored immunocontraception. J. reprod. Immunol., 71, 102-111. 33. Hoskinson R.M., Rigby R.D., Mattner P.E., Huynh V.L., DOcchio M., Neish A., Trigg T.E., Moss B.A., Lindsey M.J., Coleman G.D. et al. (1990). Vaxstrate: an anti-reproductive vaccine for cattle. Aust. J. Biotechnol., 4, 166-170, 176. 34. Hunter R.A., Magner T. & Allingham P.G. (1999). Sustained growth promotion, carcass characteristics, and meat quality of steers treated with oestradiol-17 beta. Aust. J. agric. Res., 51, 133-138. 35. Jackson R.J., Maguire D.J., Hinds L.A. & Ramshaw I.A. (1998). Infertility in mice induced by a recombinant ectromelia virus expressing mouse zona pellucida glycoprotein 3. Biol. Reprod., 58, 152-159. 36. Jaros P., Burgi E., Stark K.D.C., Claus R., Hennessy D. & Thun R. (2005). Effect of active immunization against GnRH on androstenone concentration, growth performance and carcass quality in intact male pigs. Livest. Prod. Sci., 92, 31-38. 37. Jeffery M., Loxton I., Van der Mark S., James T., Shorthose R., Bell K. & DOcchio M. (1997). Liveweight gains, and carcass and meat characteristics of entire, surgically spayed or immunologically spayed beef heifers. Aust. J. experim. Agric., 37, 719-726. 38. Kirkpatrick J.F & Frank K.M. (2005). Contraception in . free-ranging wildlife. In Wildlife contraception: issues, methods, and applications (C.S. Asa & I.J. Porton, eds). The Johns Hopkins University Press, Baltimore, Maryland, 195221. 39. Kirkpatrick J.F & Jchle W. (2005). Regulatory issues. In . Wildlife contraception: issues, methods, and applications (C.S. Asa & I.J. Porton, eds). The John Hopkins University Press, Baltimore, Maryland, 17-25.

Rev. sci. tech. Off. int. Epiz., 26 (2)

469

40. Kiyma Z., Adams T.E., Hess B.W., Riley M.L., Murdoch W.J. & Moss G.E. (2000). Gonadal function, sexual behavior, feedlot performance, and carcass traits of ram lambs actively immunized against GnRH. J. Anim. Sci., 78, 2237-2243. 41. Klinc P. & Rath D. (2006). Application of flowcytometrically sexed spermatozoa in different farm animal species: a review. Arch. Tierzucht, 49, 41-54. 42. Kutzler M. & Wood A. (2006). Non-surgical methods of contraception and sterilization. Theriogenology, 66, 514-525. 43. Ladd A., Tsong Y.Y., Walfield A.M. & Thau R. (1994). Development of an antifertility vaccine for pets based on active immunization against luteinizing hormone-releasing hormone. Biol. Reprod., 51, 1076-1083. 44. Lloyd M.L., Shellam G.R., Papadimitriou J.M. & Lawson M.A. (2003). Immunocontraception is induced in BALB/c mice inoculated with murine cytomegalovirus expressing mouse zona pellucida 3. Biol. Reprod., 68, 2024-2032. 45. McCallum H. & Hocking B.A. (2005). Reflecting on ethical and legal issues in wildlife disease. Bioethics, 19, 336-347. 46. Magiafoglou A., Schiffer M., Hoffmann A.A. & McKechnie S.W. (2003). Immunocontraception for population control: will resistance evolve? Immunol. Cell Biol., 81, 152-159. 47. Marks C. (1999). Ethical issues in vertebrate pest management: can we balance the welfare of individuals in ecosystems? In The use of wildlife for research (D. Mellor & V. Monamy, eds). ANZCART, Glen Osmond, South Australia, 79-89. 48. Mepham T.B. & Forbes J.M. (1995). Ethical aspects of the use of immunomodulation in farm-animals. Livest. Prod. Sci., 42, 265-272. 49. Miller L.A., Johns B.E., Elias D.J. & Crane K.A. (1997). Comparative efficacy of two immunocontraceptive vaccines. Vaccine, 15, 1858-1862. 50. Miller L.A., Johns B.E., Elias D.J. & Killian G.J. (1999). Oral vaccination of white-tailed deer using a recombinant bacillus Calmette-Gurin vaccine expressing the Borrelia burgdorferi outer surface protein A: prospects for immunocontraception. Am. J. reprod. Immunol., 41, 279-285. 51. Miller L.A., Johns B.E. & Killian G.J. (2000). Immunocontraception of white-tailed deer with GnRH vaccine. Am. J. reprod. Immunol., 44, 266-274. 52. Miller L.A., Rhyan J.C. & Drew M. (2004). Contraception of bison by GnRH vaccine: a possible means of decreasing transmission of brucellosis in bison. J. Wildl. Dis., 40, 725-730. 53. Minteer B.A. & Collins J.P. (2005). Ecological ethics: building a new tool kit for ecologists and biodiversity managers. Conservation Biol., 19, 1803-1812.

54. Molenaar G.J., Lugard-Kok C., Meloen R.H., Oonk R.B., de Koning J. & Wensing C.J. (1993). Lesions in the hypothalamus after active immunisation against GnRH in the pig. J. Neuroimmunol., 48, 1-11. 55. Munson L. (2006). Contraception in felids. Theriogenology, 66, 126-134. 56. Munson L., Harrenstien L.A., Acton A.E., Graham P.A., Chassy L.M. & Kirkpatrick J.F (2005). Immunologic . responses and adverse reactions to Freunds-adjuvanted porcine zona pellucida immuno-contraceptives in domestic cats. Vaccine, 23, 5646-5654. 57. Munson L., Moresco A. & Calle P.P. (2005). Adverse effects of contraceptives. In Wildlife contraception: issues, methods, and applications (C.S. Asa & I.J. Porton, eds). The Johns Hopkins University Press, Baltimore, Maryland, 66-82. 58. Nass S.J. & Strauss J.F 3rd (2004). Strategies to facilitate ., the development of new contraceptives. Nat. Rev. Drug Discov., 3, 885-890. 59. Naz R.K. (2006). Effect of sperm DNA vaccine on fertility of female mice. Mol. Reprod. Dev., 73, 918-928. 60. Naz R.K., Gupta S.K., Gupta J.C., Vyas H.K. & Talwar G.P. (2005). Recent advances in contraceptive vaccine development. Hum. Reprod., 20, 3271-3283. 61. Nettles V.F (1997). Potential consequences and problems . with wildlife contraceptives. Reprod. Fertil. Dev., 9, 137-143. 62. Ni J., Ni Y., Wang X., Xu W., Wang Y. & Xiong S. (2004). Application of a gene vaccine targeting HER-2/Neu in immunocontraception. DNA Cell Biol., 23, 807-814. 63. Oogjes G. (1997). Ethical aspects and dilemmas of fertility control of unwanted wildlife: an animal welfarists perspective. Reprod. Fertil. Dev., 9, 163-167. 64. Paterson M., Wilson M.R., Morris K.D., van Duin M. & Aitken R.J. (1998). Evaluation of the contraceptive potential of recombinant human ZP3 and human ZP3 peptides in a primate model: their safety and efficacy. Am. J. reprod. Immunol., 40, 198-209. 65. Penfold L.M., Patton M.L. & Jchle W. (2005). Contraceptive agents in agression control. In Wildlife contraception: issues, methods, and applications (C.S. Asa & I.J. Porton, eds). The Johns Hopkins University Press, Baltimore, Maryland, 184-194. 66. Porton I.J. (2005). The ethics of wildlife contraception. In Wildlife contraception: issues, methods, and applications (C.S. Asa & I.J. Porton, eds). The Johns Hopkins University Press, Baltimore, Maryland, 3-16. 67. Redwood A.J., Messerle M., Harvey N.L., Hardy C.M., Koszinowski U.H., Lawson M.A. & Shellam G.R. (2005). Use of a murine cytomegalovirus K181-derived bacterial artificial chromosome as a vaccine vector for immunocontraception. J. Virol., 79, 2998-3008.

470

Rev. sci. tech. Off. int. Epiz., 26 (2)

68. Rhim S.H., Millar S.E., Robey F Luo A.M., Lou Y.H., Yule T., ., Allen P., Dean J. & Tung K.S. (1992). Autoimmune disease of the ovary induced by a ZP3 peptide from the mouse zona pellucida. J. clin. Invest., 89, 28-35. 69. Roitt I.M., Hutchings P.R., Dawe K.I., Sumar N., Bodman K.B. & Cooke A. (1992). The forces driving autoimmune disease. J. Autoimmun., 5, Suppl A, 11-26. 70. Rutberg A.T. (2005). Humane wildlife solutions: the role of immunocontraception. Humane Society Press, Washington, DC. 71. Sillence M.N. (2004). Technologies for the control of fat and lean deposition in livestock. Vet. J., 167, 242-257. 72. Singer P. (1997). Neither human nor natural: ethics and feral animals. Reprod. Fertil. Dev., 9, 157-162. 73. Singh M., Das S.K., Suri S., Singh O. & Talwar G.P. (1998). Regain of fertility and normality of progeny born during below protective threshold antibody titers in women immunized with the HSD-hCG vaccine. Am. J. reprod. Immunol., 39, 395-398. 74. Skinner S.M., Mills T., Kirchick H.J. & Dunbar B.S. (1984). Immunization with zona pellucida proteins results in abnormal ovarian follicular differentiation and inhibition of gonadotropin-induced steroid secretion. Endocrinology, 115, 2418-2432. 75. Srinivasan J., Tinge S., Wright R., Herr J.C. & Curtiss R., 3rd (1995). Oral immunization with attenuated salmonella expressing human sperm antigen induces antibodies in serum and the reproductive tract. Biol. Reprod., 53, 462-471. 76. Stolker A.A.M., Linders S., van Ginkel L.A. & Brinkman U.A.T. (2004). Application of the revised EU criteria for the confirmation of anabolic steroids in meat using GC-MS. Analyt. bioanalyt. Chem., 378, 1313-1321. 77. Talwar G.P. (1997). Fertility regulating and immunotherapeutic vaccines reaching human trials stage. Hum. Reprod., 3, 301-310. 78. Talwar G.P., Singh O.M., Gupta S.K., Hasnain S.E., Pal R., Majumbar S.S., Vrati S., Mukhopadhay A., Srinivasan J., Deshmukh U., Ganga S., Mandokhot A. & Gupta A. (1997). The HSD-hCG vaccine prevents pregnancy in women: feasibility study of a reversible safe contraceptive vaccine. Am. J. reprod. Immunol., 37, 153-160.

79. Taylor A.A., Weary D.M., Lessard M. & Braithwaite L. (2001). Behavioural responses of piglets to castration: the effect of piglet age. Appl. anim. Behav. Sci., 73, 35-43. 80. Tyndale-Biscoe C.H. (1994). Virus-vectored immunocontraception of feral mammals. Reprod. Fertil. Dev., 6, 281-287. 81. Upadhyay S.N., Thillaikoothan P., Bamezai A., Jayaraman S. & Talwar G.P. (1989). Role of adjuvants in inhibitory influence of immunization with porcine zona pellucida antigen (ZP-3) on ovarian folliculogenesis in bonnet monkeys: a morphological study. Biol. Reprod., 41, 665-673. 82. Volpe P., Izzo B., Russo M. & Iannetti L. (2001). Intrauterine device for contraception in dogs. Vet. Rec., 149, 77-79. 83. Webster J. (1995). Animal welfare: a cool eye towards Eden. Blackwell Science Ltd, Oxford. 84. Windsor P.A., Bush R., Links I. & Eppleston J. (2005). Injury caused by self-inoculation with a vaccine of a Freunds complete adjuvant nature (Gudair) used for control of ovine paratuberculosis. Aust. vet. J., 83, 216-220. 85. World Association of Zoos and Aquariums (WAZA) (2005). Chapter 9: Ethics and animal welfare. In Building a future for wildlife The world zoo and aquarium conservation strategy (P.J.S. Olney, ed.). WAZA Executive Office, Bern, Switzerland, 1-5. 86. Xu L., Peng J.P. & Shi S.Q. (2006). Evaluation of the contraceptive potential of brZPCp vaccine in BALB/c mice: their safety and efficacy. DNA Cell Biol., 25, 87-94. 87. Yoder C.A., Graham J.K., Miller L.A., Bynum K.S., Johnston J.J. & Goodall M.J. (2006). Effect of method of delivering nicarbazin to mallards on plasma 4,4-dinitrocarbanilide levels and reproduction. Poult. Sci., 85, 1442-1448. 88. Zhang X., Lou Y.H., Koopman M., Doggett T., Tung K.S. & Curtiss R., 3rd (1997). Antibody responses and infertility in mice following oral immunization with attenuated Salmonella typhimurium expressing recombinant murine ZP3 [published erratum appears in Biol. Reprod., 56, 1069 (1997)]. Biol. Reprod., 56, 33-41.

Rev. sci. tech. Off. int. Epiz., 2007, 26 (2), 471-477

Animal vaccination and the veterinary pharmaceutical industry


D. OBrien (1) & S. Zanker (2)
(1) International Federation for Animal Health (IFAH) Europe, Rue Defacqz, 1 B-1000 Brussels, Belgium (2) International Association for Soaps, Detergents and Maintenance Products (AISE), Avenue Herrmann Debroux 15A, 1160 Brussels, Belgium

Summary The market for veterinary vaccines is spread across species but it is limited in size and the development of vaccines is becoming more complex and expensive. Vaccines are amongst the most effective means of preventing disease in both animals and humans. In many cases, diseases have been eradicated or their impact on animal health and welfare greatly reduced. It is an ethical responsibility to ensure the availability of a wide range of vaccines even where the market needs to be financially supported, as in the case of less common animal species and those with less common conditions (commonly referred to by the acronym MUMS: Minor Use and Minor Species). Mass slaughter is becoming unacceptable to society and we must move to a vaccinate to live policy wherever possible. We need to use vaccines to avoid the high costs of disease and to enhance food safety. In developing vaccines, we need to minimise animal testing. In addition, we need to ensure that the public accept the use of vaccines in food-producing animals as a means of protecting the health and welfare of all animals. As we look to the future, vaccines will be vital to ensure our ability to provide more food to a growing global population. The European Technology Platform for Global Animal Health has a key role to play in identifying key research priorities. Keywords Disease Ethical approach Food safety Health Research Veterinary vaccine Welfare.

The global veterinary vaccine market


In 2004, the international veterinary vaccine market was worth US$3.1 billion (i.e. 21% of the total veterinary medicine market, which is valued at US$15 billion). Fortyone per cent of the market was based in the Americas, 37% in Europe and 22% across the rest of the globe. Table I provides a breakdown of the global veterinary vaccines market in terms of species.

Table I A breakdown of the monetary value of the global veterinary vaccines market in 2004
Species Livestock Poultry Other Value in US$ billions 1.50 0.68 0.92

Source: Wood Mackenzie Ltd, 2004 Global Veterinary Biologicals Market by Region and Species in 2004

472

Rev. sci. tech. Off. int. Epiz., 26 (2)

Developing a vaccine: the process


Put simply, the process of developing a vaccine involves identifying a disease target, isolating the disease organism, growing it in eggs or cells, treating it physically or chemically so that it does not cause disease (inactivated vaccines) or attenuating virulent characteristics of the strain (live vaccines) and then administering it to the animal. The result is an immune response that protects the animal against the disease. Modern genetics technology allows for more possibilities and options such as taking genes from the disease organism, inserting them into harmless viruses, growing the viruses and then administering the live virus to the animal to create the immune response. In addition, genes may be added to a known virus that is harmless, the virus may then be grown, treated in a manner that kills the virus and the resultant material may be used as a vaccine. Finally, a second disease may be identified, genes from the first disease may be added to the second disease, cultivated and the resultant treated material may be used to vaccinate against two diseases. In many cases, industry develops vaccines in cooperation with universities, institutes and other bodies, thereby combining its expertise with that of third parties. This is increasingly necessary as new scientific disciplines develop (such as in the area of genomics), as it is much more efficient to combine resources than for each party to have its own specialists.

cost of developing vaccines. This is in contrast to the value of food-producing animals, which is tending to decline over time. It is clear, therefore, that commercial vaccine production can only be undertaken for diseases that generate sales covering the cost of development, along with some profits. As a result, we have disease problems where the development of vaccines must be supported by public authorities, e.g. anthrax and botulism.

The benefits of animal vaccination


Vaccination is part and parcel of good husbandry and management. Vaccines are amongst the most effective means of preventing disease in both animals and humans. Preventing disease protects the health and welfare of the animals (i.e. less mortality and fewer animals that survive the disease but are negatively impacted for the rest of their life, e.g. with impaired lung function), helps to prevent the spread of disease to humans (e.g. salmonella vaccination ensures high quality food is produced from healthy animals) and helps to protect the environment, as fewer animals are required for food-producing purposes. Vaccines, coupled with diagnostic tests, eradication programmes and surveillance, help to eradicate diseases, e.g. foot and mouth disease (FMD), rabies and Aujeszkys disease in many EU countries. They also help to reduce the annual 17% loss of production associated with disease in animals. With globalisation, the potential for diseases to travel with animals, animal products and humans is greatly increased, as was seen during the 2003 outbreak of severe acute respiratory syndrome (SARS), thus the role of vaccines is becoming increasingly important. The European Technology Platform for Global Animal Health (ETPGAH) is actively involved in identifying priority diseases and research needs so that our ability to control diseases into the future will be increased. Coordination of research on a global basis via the ETPGAH has the potential to deliver significant benefits to society in the medium to long term. The advantage of being able to eradicate diseases, e.g. avian influenza, at source and so prevent them becoming global threats is clear and the benefits to society are huge.

Costs of developing a vaccine: benchmark data


Industry surveys indicate that the time taken to develop a vaccine varies across species. The process may take eight to ten years for brand new products. During this period, the development of the product is coupled with studies to demonstrate the safety, quality and efficacy of the product. The product must be formulated in a way that facilitates use and optimises efficacy, e.g. by including an adjuvant to stimulate a maximum immune response that lasts for a reasonably long period of time. The cost of this process also varies, but it is estimated that the average cost is US$50 million. As technology develops in complexity, regulatory requirements also become more complex, with both factors leading to an increase in the

Socio-ethical issues
The benefits of vaccination are clear, but there are several issues to deal with when developing vaccines for animals and each will be examined in this section.

Rev. sci. tech. Off. int. Epiz., 26 (2)

473

Vaccine availability
In the context of vaccines for less common animal species and those with less common conditions (often referred to with the acronym MUMS: Minor Use and Minor Species), the issue of availability is very real. Where there is a rare disease in a major species, such as FMD, anthrax or brucellosis, public funding must be made available if we wish to develop a vaccine as opposed to relying on antibiotic treatment of sick animals. It is ethically unacceptable to allow animals to become ill if we have tools, such as vaccines, to prevent illness. What price is society willing to pay to follow this ethical approach? In the case of minor species such as goats, fish, rabbits or deer, again, third party intervention is necessary if vaccines are to be developed. This could come in the form of public funding or the funding of the development of a vaccine, under contract, by groups of farmers who pool funds. The alternatives to vaccination are to treat animals that become ill or to stop producing some of these species for food purposes.

future cost of promotion to stimulate demand cost of lobbying government. Related sectors are impacted in various ways that may include loss of feed sales, decline in the value of finished product, scarcity of raw materials, loss of markets, etc. Indirect costs can include: government subvention (vaccination programmes, culling, supervision of quarantine areas, compensation for culling, scientific facilities, negotiation of the lifting of sales bans with other countries, etc.) loss of tourism revenue (the FMD outbreak in the UK resulted in many areas of the country being placed off limits to tourists) loss of overall employment, treatment of people who become ill. It is clear, therefore, that major disease outbreaks entail significant costs to society. Consequently, the idea of investing significantly in the prevention of disease makes very good economic sense. In this context, many issues need to be considered, including: a) the availability of vaccines to prevent a disease and the need to invest in better vaccines b) the availability of strategic reserves of vaccines and/or sera to respond to diseases that occur rarely (FMD) c) the ability of industry to respond to a surge in demand (as was the case during the recent outbreak of avian influenza) and the need to stimulate production capacity by public intervention such as the regular purchase of strategic reserves. In the context of avian influenza, the threat of a human pandemic is of great concern. The Spanish flu killed between 20 million and 40 million people in 1918. The Asian flu of 1957 killed between 1 million and 4 million people, as did the 1968 Hong Kong flu. The ability to use biosecurity, stamping out and vaccination to attempt to eliminate avian influenza and so possibly avoid a human pandemic is of major societal importance and once again highlights the value of vaccinating animals. Indeed, we need to consider vaccine use on a global scale to stop diseases at source and so prevent diseases becoming global problems.

Mass slaughter
The outbreak of FMD in the United Kingdom (UK) in 2001 (other areas of the globe are likely to be similarly impacted) represented a turning point in policy in the European Union (EU). It is no longer acceptable to the public to engage in the mass slaughter of animals that was witnessed during that outbreak. Instead of focusing exclusively on prevention and eradication, vaccination is now seen as a tool in the armoury to fight exotic diseases. This change in policy has become known as the vaccinate to live policy. Whilst a stamping out approach is still the most widely implemented strategy to eradicate disease in the first instance, vaccination is now regarded as an appropriate strategy if initial efforts to eradicate the disease via slaughtering are not succeeding.

Disease costs
The UK FMD outbreak is estimated to have cost 3,800 million (US$5,186 million) (1). The 2003 outbreak of avian influenza in the Netherlands cost at least US$612 million. Also in the Netherlands, the 1997-1998 outbreak of classical swine fever cost US$2.52 billion. The cost of disease is a complex issue. Direct costs that may arise for the producer include those that result from: loss of production culling of animals decline in overall consumption in the sector affecting overall pricing both during a disease outbreak and for some time afterwards

Food safety and quality: the value of vaccines


The control of salmonella in the UK, as depicted in Figure 1, indicates the value of vaccination to society in terms of public health. The elimination of FMD from Western Europe, as depicted in Figure 2, highlights the animal welfare value of vaccination. Similarly, from the

474

Rev. sci. tech. Off. int. Epiz., 26 (2)

viewpoint of protecting health as opposed to treating disease, the decline in the use of antibiotics in Norwegian salmon production after the introduction of appropriate vaccines, as depicted in Figure 3, further highlights the benefits of vaccination.

Fig. 3 Norwegian salmon production, consumption of pure antibiotics and the effect of vaccines Source: Norwegian Ministry of Fisheries and Coastal Affairs, 2005

*The Lion Quality Code of Practice, which was launched in 1998, includes independent auditing, improved traceability, packing station hygiene controls, a best-before date that is earlier than that required by law and compulsory vaccination against Salmonella enteritidis.

Clearly, vaccination has a tremendous benefit directly to society, as well as to animal health and welfare. The desirability of using vaccines to minimise the threat of other zoonoses to society is very clear: Escherichia coli, campylobacter, listeria, West Nile virus and Lyme disease are cases in point.

Fig. 1 The incidence of human salmonellosis in England and Wales (1981 to 2000) Source: Advisory Committee on the Microbiological Safety of Food, United Kingdom

Animal testing
In developing vaccines, animal testing is needed at various stages in order to assess safety and efficacy. Many of the testing requirements are contained in the European legislative framework, as detailed in the European Pharmacopoeia. Guidelines also exist clarifying when animal tests are required. It is the wish of everybody concerned to minimise the use of animals in testing vaccines. In order to move forward, we need alternatives that are accepted across the globe. It is of no value to introduce an alternative in one part of the globe if other regions demand the animal-based test. The company is still forced to use the animal test if it wishes to continue to supply its product in the region demanding the test. In this context, the International Cooperation on Harmonisation of Technical Requirements for the Registration of Veterinary Products (VICH) has a key role to play. VICH has the potential to harmonise regulatory requirements ensuring that the same test procedure is accepted globally this prevents duplication of testing. In addition, agreement may be reached where testing may be replaced by alternatives not involving animals. Progress through this forum has the potential to deliver global agreement on alternatives to animal testing.

Fig. 2 The elimination of foot and mouth disease from Western Europe Mass vaccination was introduced in Western Europe in the mid 1960s and was discontinued in 1992 Source: Intervet, Netherlands

Rev. sci. tech. Off. int. Epiz., 26 (2)

475

The initiative announced by the European Commission in November 2005 on alternatives to animal testing (European Partnership to Promote Alternative Approaches to Animal Testing [EPPAA]) is also important. The objective of the EPPAA is to replace, reduce or refine the use of animals in testing (this strategy is referred to as the 3 Rs initiative). It is important that we make every effort to replace animal testing, and to this end the Commission and industry from many sectors have combined to jointly work on alternatives. Over the years, progress has been made. For example, intra-cerebral safety testing in mice in relation to Aujeszkys disease vaccines has been eliminated.

Safety of food from vaccinated animals


A recently published EU study concerning the knowledge of consumers in relation to avian influenza is of interest (2). Fifty-three per cent of the public believe that it is dangerous to eat the meat of a chicken vaccinated against avian influenza. In the UK in 2001, the authorities did not use vaccination to try to stop the spread of FMD as various parties in the UK food chain indicated that they would not handle produce from vaccinated animals. When one examines these two pieces of information together, the need to communicate the safety of food from vaccinated animals becomes evident. In a context where the public are concerned about the safety of produce from vaccinated animals, the benefits of vaccination, particularly in the context of a major disease outbreak, may not be realisable. Most food animals receive a number of vaccinations during their life to protect them against various diseases and the public usually consume such produce without concern. It may be that they do not realise that animals are vaccinated routinely or they may be more cautious in the context of a disease outbreak that is widely reported in the media and that may or may not have human health implications. The challenge for both industry and government is to communicate information in a manner in which it will be accepted by society at large and will assure the public that produce from vaccinated animals is safe to eat.
Fig. 4 Predicted increase in the global consumption of livestock products Source: World Organisation for Animal Health

countries. For milk, the factors are two and five, respectively. For eggs, the factors are three and eight, respectively. Herd health management will be key in attempting to meet this challenge and vaccination will have a central role to play. As we move into the future, we anticipate, based on historical knowledge, that new diseases will emerge. This is all the more likely as farming intensifies in developing countries and as a result, intensive agriculture comes into contact with wild species. Our ability to be alert for such diseases and to have the scientific capacity and infrastructure to respond to such challenges is critical. The ETPGAH recognises this reality. Amongst its recommendations is the need to identify the threats to Europe from pathogens which are not considered important at present (i.e. horizon scanning) and conduct full risk assessment of potential threats from new and emerging diseases in particular those outside the EU boundaries (3). Whilst reference is made to Europe, the recommendation is valid in any part of the globe. The ETPGAH has also identified societal acceptance of technology as an issue that needs attention. The Strategic Research Agenda (SRA) from the ETPGAH proposes research into consumer perceptions and expectations of new technology and factors which affect consumer behaviour in relation to food safety. The SRA also proposes the development of risk communication strategies to communicate with the public in relation to new technology in the most effective manner. Part of this work should be aimed at communicating the fact that food from vaccinated animals is safe to eat.

Future food needs


Figure 4 indicates the scale of the challenge facing global agriculture if future food needs are to be satisfied. As previously mentioned, animal diseases are responsible for at least a 17% loss in production, and the elimination/reduction of these losses must be a priority. Meat consumption will increase by a factor of two in developed countries and by a factor of five in developing

476

Rev. sci. tech. Off. int. Epiz., 26 (2)

Conclusion
The development of vaccines is becoming more complex and expensive. The need to ensure the availability of vaccines requires public and producer cooperation. Vaccines deliver considerable benefits to both animals and society at large and have the potential to deliver even greater benefits in the context of the ETPGAH. Mass slaughter must be avoided if possible and animal health needs to be protected to avoid the huge costs associated with the outbreak of exotic diseases. The development of a wider range of effective vaccines has the potential to deliver benefits to society far in excess of the cost of development and this opportunity should not be missed. Food safety and quality benefits flow from vaccine use and we need to invest in alternatives to animal testing.

Future food needs at the global level highlight the important role and need for new and effective vaccines. The ETPGAH shows the way forward and this development needs to be coupled with effective communication with the public so that new vaccine technologies are accepted. From the viewpoint of society, vaccination delivers very considerable benefits. The human health benefits of reducing zoonoses are very substantial and need to be communicated in an effective manner. From the ethical viewpoint, we need to protect animal health and welfare by using vaccines to the greatest extent possible. In addition, we need to prove safety and efficacy whilst minimising the use of animals in testing.

La vaccination animale et lindustrie pharmaceutique vtrinaire


D. OBrien & S. Zanker
Rsum Le march des vaccins vtrinaires sintresse de nombreuses espces animales mais reste limit en volume et il devient de plus en plus difficile et onreux de dvelopper de nouveaux vaccins. Les vaccins sont lun des moyens les plus efficaces de prvenir la maladie chez les animaux comme chez lhomme. Dans bien des cas, la vaccination a permis dliminer des maladies ou den rduire limpact sur la sant et le bien-tre des animaux. La responsabilit thique impose de faire en sorte quun large ventail de vaccins soit disponible, y compris lorsque le march doit tre soutenu financirement comme cest le cas pour les espces animales plus rares ou les pathologies moins frquentes dsignes par lacronyme MUMS (Minor Use and Minor Species). Labattage sanitaire massif tant devenu intolrable pour la socit, nous devons voluer, dans la mesure du possible, vers des politiques de vaccination pour la vie . Les vaccins nous permettent dviter le fardeau du cot de la maladie tout en amliorant la scurit sanitaire des aliments. Le dveloppement des vaccins devra se faire en recourant le moins possible lexprimentation animale. En outre, il faudra faire en sorte que le public accepte la vaccination des animaux destins la consommation, en tant que mesure destine protger la sant et le bien-tre de tous les animaux. Dans une perspective davenir, grce aux vaccins nous serons en mesure dassurer la scurit alimentaire dune population mondiale de plus en plus nombreuse. La plateforme technologique sur la sant animale dans le monde lance par la Commission europenne a un rle dterminant jouer pour fixer les priorits de la recherche. Mots-cls Bien-tre animal Dmarche thique Maladie Recherche Sant Scurit sanitaire des aliments Vaccin vtrinaire.

Rev. sci. tech. Off. int. Epiz., 26 (2)

477

Vacunacin animal e industria farmacutica veterinaria


D. OBrien & S. Zanker
Resumen Si bien el mercado de la inmunizacin veterinaria comprende la lucha contra las infecciones que afectan a muchas especies animales, su talla es limitada y el desarrollo de vacunas cada vez ms complejo y oneroso. La vacunacin, que es uno de los instrumentos ms eficaces para prevenir enfermedades tanto en los animales, como en los seres humanos, ha permitido erradicar muchas enfermedades, o reducir la gravedad de sus consecuencias en la salud y el bienestar de los primeros. Desde el punto de vista tico, debe garantizarse la existencia de una gran variedad de vacunas, aunque para ello sea preciso prestar apoyo financiero al mercado, como en los casos de las especies animales poco comunes y las indicaciones poco frecuentes (denominadas habitualmente con la sigla MUMS, por las iniciales en ingls de Minor Use and Minor Species). En la sociedad contempornea, el rechazo del sacrificio masivo es cada vez mayor; por ello, siempre que resulte posible debe adoptarse una poltica de vacunacin que impida la circulacin de los agentes patgenos en los rebaos. La inmunizacin es necesaria para evitar los elevados costos de las enfermedades y garantizar la inocuidad de los alimentos. Tambin es preciso reducir al mnimo las pruebas en animales durante el desarrollo de vacunas. La vacunacin constituye un medio para proteger la salud y el bienestar de todos los animales; por consiguiente, debe darse a comprender al pblico la necesidad de administrarla a aquellos que se destinan al consumo. Las vacunas sern indispensables para producir las cantidades de alimentos que sern necesarias en el futuro a fin de alimentar a una poblacin mundial cada vez ms numerosa. La Plataforma Tecnolgica Europea para la Sanidad Animal Mundial desempear un papel clave a la hora de determinar los mbitos de investigacin prioritarios. Palabras clave Bienestar Enfermedad tica Inocuidad de los alimentos Investigacin Salud Vacuna veterinaria.

References
1. Anon. (2005). Cost benefit analysis of foot and mouth disease controls. A report for the Department for Environment, Food and Rural Affairs, United Kingdom. Risk Solutions. Available at: http://www.defra.gov.uk/animalh/ diseases/fmd/pdf/costben.pdf (accessed on 8 May 2007). 2. European Commission (2006). Special Eurobarometer 257. Avian Influenza. Directorate General for Health and Consumer Protection, Brussels. Available at: http://ec.europa. eu/public_opinion/archives/ebs/ebs_257_en.pdf (accessed on 7 May 2007). 3. European Technology Platform for Global Animal Health (ETPGAH) (2006). Prioritization of Animal Diseases. Stakeholder Meeting, 15 February, Brussels. Available at: http://www.ifahsec.org/2_CS_Disease%20prioritization.ppt (accessed on 7 May 2007).

Rev. sci. tech. Off. int. Epiz., 2007, 26 (2), 485-488

The opinion of the production sector on the role of vaccines in the control and eradication of livestock diseases in Argentina
L. Miguens
Sociedad Rural Argentina, Florida 460, (1005) Ciudad Autnoma de Buenos Aires, Argentina. E-mail: presidencia@sra.org.ar

Summary Research by a number of international organisations indicates that world demand for red meat protein is set to increase significantly in the coming years. However, faced with the risk of infectious animal diseases and zoonoses factors that could limit the growth of this production sector the fight against livestock diseases must continue, especially against those that affect food safety or pose a threat to human life. The use of vaccination to prevent infectious animal diseases is of key importance, not only because it helps to control and effectively eradicate infectious livestock diseases, but also because it makes it possible to introduce new technologies for intensive or semi-intensive production, to protect the environment, to care for animal welfare and to guarantee the safety of animal-derived foodstuffs. As part of their professional culture, livestock producers have come to fully appreciate the advantages of using vaccination to prevent disease rather than curative measures, which are more costly to implement and in some cases not very effective. The control of anthrax and rabies by means of effective vaccines was a factor in the widespread development of livestock in Argentina and other parts of Latin America. Recent results in the control and eradication of foot and mouth disease have made producers even more convinced of the merits of this technology. Animal disease prevention has proven to be highly conducive to the production of healthy foodstuffs. It is the responsibility of international organisations to draw up appropriate regulations to protect trade, supply safe and healthy products and prevent the application of unjustified non-tariff measures. Keywords Animal disease Argentina Control Eradication International trade Livestock producer Vaccination Vaccine.

Introduction
Cattle, which were introduced into America from Europe at the time of the Spanish conquest in the 15th Century, went on to breed easily in various regions of the Americas. Their numbers grew to unparalleled proportions in the River Plate region, which covers vast areas of Argentina,

Uruguay, Paraguay and southern Brazil. There was a huge natural increase in the livestock population in the Argentine grasslands, especially during the 16th and 17th centuries, so much so that by the early 18th Century the estimated population was similar to that of the present day (56 million head). Up until then no technological improvements had yet been introduced into livestock production, which was 100% pastoral (4).

486

Rev. sci. tech. Off. int. Epiz., 26 (2)

Livestock production was the main engine of the Argentine economy up until the mid-20th Century. Livestock producers were able to play the key role in this development thanks to new technology for genetic improvement, the introduction of fencing, improved animal feeding techniques, the development and application of agrochemicals and fertilisers, the production of modern farming machinery, and, finally, public sector initiatives to control livestock diseases, which received unstinting support from livestock organisations and producers. According to studies by specialised international organisations, half of the worlds population earns less than two dollars per person per day (although one third of this population, particularly in Asia and Latin America, is due to increase its income significantly over the next twenty years). What is more, 75% of the 1.2 billion or so people subsisting on less than one dollar per day live and work in rural areas of developing countries (1). Although world demand for red meat protein is set to increase significantly over the coming years, one of the main constraints on meeting this demand will be the occurrence of infectious animal diseases and in particular zoonoses (3). Production rates must therefore be increased and livestock diseases reduced or eradicated, especially those affecting food safety or posing a threat to human life. The prevention of infectious animal diseases by means of vaccination is paramount to safeguarding human life, not only because it helps to control and effectively eradicate infectious livestock diseases, but also because it makes it possible to introduce new technologies for intensive or semi-intensive production, to protect the environment, to meet animal welfare requirements and to guarantee the safety of animalderived foodstuffs.

which are organised and operate much like animal health groups (Grupos de Defensa Sanitaria) in other countries, work in a coordinated manner at regional and national level, facilitating the implementation of disease programmes.

The example of foot and mouth disease


The success achieved in controlling and eradicating foot and mouth disease in Argentina in the 1990s and the speedy control and eradication of subsequent events were attributable to three key factors: a) a realistic control and eradication plan b) an excellent quality vaccine (safe, potent and pure) c) the resolute and committed participation of production sectors. This convinced all stakeholders of the benefit of using vaccination to control foot and mouth disease. The first reports of foot and mouth disease in Argentina were documented in 1890. The disease was present in the country up until the late 1980s and was endemic throughout much of Argentina. Thereafter, the private sector, working together with the health authorities, began to take decisive action and to actively participate in strategic actions to control foot and mouth disease. Major health changes began to be observed in affected regions covered by the National Control and Eradication Programme, with a drastic reduction in prevalence in affected herds, showing that the programme was working well. Bi-annual administration of the foot and mouth disease vaccine with an oily adjuvant by the official Veterinary Services, paid for by livestock producers, extended the protection period and facilitated the effective surveillance of all farms and herds (around 100% vaccine coverage against foot and mouth virus serotypes A, O and C for more than eight years). Furthermore, the involvement of producers and producer organisations guaranteed the continuity of programme activities and was one of the key factors in arresting and ultimately eradicating the disease. As part of their professional culture, livestock producers have come to fully appreciate the advantages of using vaccination to prevent disease rather than curative measures, which are more costly to implement and in some cases not very effective. The control of anthrax and

The livestock sector and the use of vaccination


The livestock production sector in Argentina (and the River Plate region as a whole) has taken an active part in the process of technology introduction to improve production conditions, especially health conditions, and in the early 20th Century it began to use a number of tools enabling it to overcome some serious health obstacles. In the past fifty years, the production sector has grouped itself into local animal health committees (Comisiones Locales de Sanidad Animal) in every area of the country. The committees work jointly with official veterinary sector representatives on schemes for the early diagnosis of diseases, especially foot and mouth disease, and for monitoring vaccination campaigns against foot and mouth and other livestock diseases. These local committees,

Rev. sci. tech. Off. int. Epiz., 26 (2)

487

rabies by means of highly effective vaccines facilitated the widespread development of livestock in Argentina and the rest of Latin America. Recent results in the control and eradication of foot and mouth disease have made producers even more convinced of the merits of this technology. Throughout this process, livestock producers have always relied on the professional advice of veterinarians and of the official Veterinary Services. Indeed, this tripartite relationship is the key to successfully implementing any disease prevention action.

measures. The slaughter and destruction of thousands of healthy animals (2) and the subsequent environmental damage cause by the disposal of their remains (as in the case of cattle during the bovine spongiform encephalopathy epidemic in Europe) elicited a profoundly negative reaction among world public opinion. Fortunately, major changes have occurred in this area, such as the Dutch Farmers Union proposal to use vaccination to control the 2001 foot and mouth disease outbreak in the Netherlands. These episodes also revealed that animal welfare conditions had been severely undermined. However, livestock producers are the first link in the chain for proper implementation of animal welfare practices: the animals belong to them animals which they breed, select and care for and it is they who are ultimately responsible for what happens to their animals.

Modernisation of the sector


Nowadays, increasing world demand for protein poses a greater challenge to livestock producers. They know that their production must satisfy basic environmental conservation criteria, whilst world climate change determines the movement of their production to other latitudes, with different ecosystems, where there is a growing threat of emerging and re-emerging infectious animal diseases (for the most part zoonoses). All these factors, coupled with greater demand for food safety, have led producers to play a growing and committed role in the introduction of new production technology. Any economically sustainable development of livestock production, including access to international markets, is therefore inconceivable without the use of vaccines and of effective biosafety measures for the prevention, control and eradication of infectious livestock diseases. For this to happen, international animal health regulations must take into account scientific advances in the field and reflect the state of the art, bearing in mind that livestock producers are unconditional allies in animal health management and animal production food safety. However, these standards and recommendations must clearly reflect the optimum use of vaccination technologies in disease prevention and control, including their limitations, which in some cases may appear insurmountable in the current state of knowledge. One example is brucellosis control, where vaccination plans and the destruction of reactor animals are a necessary and economically valid prerequisite for definitively controlling and eradicating the disease, particularly among dairy herds. Clearly, the use of stamping out measures to control and eradicate livestock diseases poses ethical and environmental problems. The recent episodes in Europe (foot and mouth disease in the United Kingdom and the Netherlands), Asia and Africa (avian influenza) have demonstrated the serious limitations of stamping out

The producers standpoint


Argentinas livestock producers played an active and committed role in conjunction with the official veterinary sector in a successful ten-year programme for the control and eradication of foot and mouth disease, applying preventive vaccination to the entire cattle population in accordance with the international standards and recommendations of the World Organisation for Animal Health (OIE). In 2000, Argentina completed the eradication process and received international recognition of its status by the OIE, proving, together with other countries and regions in the Americas, that the route mapped out by the OIE was indeed feasible (5). Fortunately, a number of important measures continued to be taken after the disease was eradicated: the surveillance and warning system was maintained, vaccine was produced on an industrial scale and an antigen/vaccine bank was set up. All this enabled Argentina to respond rapidly when foot and mouth disease recurred in the region and to use emergency vaccination to bring the disease under control in less than one year, leading to the recovery of Argentinas foot and mouth disease status in 2002. All these measures were taken without eliciting a negative response from the public, without affecting the environment by the mass destruction of animals and with no economic impact on livestock producers, who were able to continue maintaining and increasing their herds in compliance with strict biosafety and animal welfare standards. Argentinas livestock producers are proud of this achievement and believe it demonstrates their efficiency, something which would not have been possible without the aid of a good preventive tool and an excellent vaccination programme. The investment in an antigen/vaccine bank has turned out to be amply justified

488

Rev. sci. tech. Off. int. Epiz., 26 (2)

economically, in view of the success in recovering markets and production value. Other examples in other parts of the world concerning other livestock diseases have yielded the same results, to the great satisfaction of livestock producers in those countries.

Based on their experience of controlling various epizootics, livestock producers will therefore continue to work actively with the official Veterinary Services and private veterinarians to control and eradicate livestock diseases. The need to guarantee healthy foodstuffs, in compliance with new standards on animal production food safety, is a challenge for disease prevention programmes. Nowadays livestock producers have two basic technologies at their disposal: biosafety and preventive vaccination. Biosafety, which can be implemented efficiently in integrated production systems, is very expensive to set up and maintain, whereas preventive vaccination, which can be implemented in all production systems, tends to be cheaper. The recommendations of international organisations must be followed in order to ensure the proper and efficient implementation of the two systems and so facilitate market access for safe and healthy products. Clearly, these standards, which are based on leading-edge technology, do not introduce tariff measures. The current vaccination strategy, which consists of vaccinating the entire herd with an immunogen that is extremely pure as regards non-structural proteins, serves as a guarantee for hazard-free trade in animals and animal products from free countries practising vaccination to free countries without vaccination. Only if this type of strategy is used will vaccination be able to play its role in the prevention, control and eradication of livestock diseases.

Conclusion
In summary, it is livestock producers opinion that, over the years, the use of vaccines has proven to be an excellent technological tool in preventing livestock diseases. However, producers acknowledge that vaccines and their use should be subject to a series of basic requirements: a) vaccines must comply with international regulations (OIE), which are considered to be minimum standards b) vaccines must be safe and should not jeopardise food safety in any way. No discrimination must be made between products from vaccinated infection-free animals and the same products from unvaccinated infection-free animals c) vaccines used preventively should guarantee a long immunity period, preferably for life. To achieve the desired outcomes, basic research on the subject will need to be promoted and encouraged d) vaccines should be stable and effective under environmental extremes of cold and heat e) vaccines should be reasonably priced to enable their use even in countries/regions with scarce economic resources. Meeting growing world demand for foodstuffs, particularly from the poorest countries, is acknowledged to be one of the responsibilities of livestock producers, together with other sectors. In economic terms, producers know how to produce more and better products (healthier foodstuffs).

Acknowledgements
The author is grateful for the comments of Doctors Federico Gonzalez Grey, Carlos van Gelderen and Alejandro Schudel, who revised this article.

References
1. Daz Bonilla E. & Gulati A. (2003). Developing countries and the WTO negotiations, trade policies and food security. The International Food Policy Research Institute, Washington, DC, United States of America. 2. Follet B. (ed.) (2002). Infectious disease in livestock. The Royal Society Inquiry into Infectious Diseases in Livestock, chaired by Sir Brian Follet FRS. Perfect Page, London, United Kingdom. 3. Morgan N. & Prakash A. (2006). International livestock markets and the impact of animal diseases. In Animal production food safety challenges in global markets (S.A. Slorach, ed.). Rev. sci. tech. Off. int. Epiz., 25 (2), 517-528. 4. Ras N. (2001). El origen de la riqueza en una frontera ganadera. Academia Nacional de Agronoma y Veterinaria, Buenos Aires, Argentina. 5. Secretara de Agricultura, Ganadera y Pesca (1990). Plan de control y erradicacin de la fiebre aftosa en Argentina Bases para el control de la fiebre aftosa. Secretara de Agricultura, Ganadera y Pesca de la Repblica Argentina, Buenos Aires.

Rev. sci. tech. Off. int. Epiz., 2007, 26 (2), 489-494

Conclusions
Future trends in veterinary vaccinology
As stated in the introduction to these two issues of the Scientic and Technical Review (the Review) of the World Organisation for Animal Health (OIE), vaccination is without a doubt the best means of preventing and, where possible, eliminating animal diseases. This is particularly true for viral infections, if only because we do not have large-spectrum antivirals appropriate for use in veterinary medicine. Vaccination can thus be considered an investment in animal health insurance. Nonetheless, there are many unmet needs in animal health, and infections for which there are as yet no vaccines.

Obstacles to the development of veterinary vaccines


Certain obstacles to the development of vaccines have already been cited in the introduction to these two issues of the OIE Review; these difculties, and others, are discussed in further detail here.

Biological and technical obstacles


The rst set of difculties is biological and technical in nature. They account, for example, for the scarcity of antiparasitic vaccines on the market. Parasites coexist with their hosts, and in the course of their evolution have devised many strategies to maintain this coexistence. This does not only apply to parasites, but also to viruses and bacteria. Viruses have co-evolved with their hosts, and have also developed many strategies to survive, in relative state of equilibrium, in a population or with a host. For example, they have acquired, often at the expense of their host, many molecules or mechanisms that enable them to withstand the various assaults of the immune response. This explains in part why, despite years of research, there is as yet no vaccine available for African swine fever. Moreover, many viral and bacterial infections, for example foot and mouth disease and avian inuenza, are caused by pathogens that have several serotypes and which constantly evolve. A recent example of the problems brought about by serotypical variation is provided by the appearance of bluetongue in Northern Europe due to serotype 8 (one amongst the 24 known serotypes). Existing vaccines proved useless, as they did not contain the corresponding antigens. However, for vector-borne diseases such as bluetongue, vaccination seems to be the only solution. It is very difcult to attack the vector, especially through methods that have no negative environmental impact. Another difculty inherent in veterinary vaccinology is the wide variety of target species, and the number of different infections in each one. Vaccines, as opposed to therapeutic molecules (antibiotics, anthelmintics), are for the most part extremely specic; there are only a few multispecies vaccines containing the same antigenic valence (tetanus, rabies ).

490

Rev. sci. tech. Off. int. Epiz., 26 (2)

Another problem for animal vaccination is that the target species include wildlife, which are not always easy to access for the purpose of vaccination. For example it is not possible to vaccinate vampire bats (Desmodus rotundus) in Latin America against rabies because of the difculty in gaining access to these animals. Other wild species, however, such as foxes (Vulpes vulpes) (rabies) or wild boars (Sus scrofa) (classical swine fever) are relatively easy to gain access to and can be vaccinated by means of vaccinal baits. The appearance of new emerging diseases, for which, obviously, no vaccine exists at the moment of emergence, presents another challenge for the development of animal vaccines. One example of this is the Nipah virus infection that occurred in Malaysia: as this was a lethal zoonosis, the only solution was the systematic slaughter of infected pigs and the destruction or burial of their carcasses. However, in the United States of America, the emergence of West Nile virus infection, which affects humans and a variety of animals species, but principally horses, led to the development and marketing of eight different equine vaccines in an exceptionally short space of time. Similarly, the re-emergence of transboundary diseases in countries hitherto free of them is another threat, the risk of which is heightened by growth in the ve Ts (Transport, Travel, Tourism, Trade, Terrorism). These diseases generally originate in developing or transition countries that lack the means to protect themselves; for the international community, assisting the ght against these diseases where they occur, particularly through vaccination, means protecting the entire planet. These diseases often have a wildlife reservoir that is not easy to access, such as the African buffalo (Syncerus caffer) for the SAT (South African Territories) strains of FMD.

Economic obstacles
Other obstacles to vaccination are of an economic nature. The most important one is the low return on investment for private companies that develop veterinary vaccines for livestock, especially for diseases specic to developing countries, which unfortunately is where the needs are the greatest. However, such problems are not conned to developing or transition countries, as certain target species get little attention even in developed countries if they are considered minor species. In Europe, for example, this is the case for milking sheep, goats, rabbits, sh other than salmonids and fowl other than chicken. In addition, these minor species are distributed unevenly over the different zones of Europe, which does not facilitate the implementation of European-wide procedures for marketing veterinary vaccines. There is also the problem of rare diseases both in the so-called major species (e.g. the European form of malignant catarrhal fever in ruminants) and the most catastrophic scenario in minor species (e.g. tularaemia in hares). This has led to the MUMS concept (Minor Use, Minor Species). Developing vaccines for these types of diseases requires an equitable public-private partnership.

Legal and regulatory obstacles


The last set of obstacles is legal and regulatory in nature. Many animal health regulations favour hygienic prophylaxis and rule out preventive vaccination, especially in the developed countries in which the major epizootics affecting domestic herds (foot and

Rev. sci. tech. Off. int. Epiz., 26 (2)

491

mouth disease, classical swine fever) have been eliminated. Fortunately, there is a trend toward the implementation of health policies that include vaccination (vaccination to live), thanks in particular to the availability of new-generation vaccines which have a companion diagnostic test and which contain a serological marker (DIVA technology: Differentiating Infected and Vaccinated Animals). This availability has moved international organisations such as the OIE to modify vaccination regulations. Another regulatory obstacle to the development of veterinary vaccines for livestock is the existence of very stringent and inexible regulations governing the registration and marketing of veterinary drugs (including vaccines). Such legislation does not promote exibility in the choice of vaccinal strains for vaccines capable of preventing infections caused by pathogens with multiple serotypes, or in their adaptation to the epidemiological conditions in the eld. In addition, these very heavy regulations (which, however, present the considerable advantage of guaranteeing quality and efcacy against the pathogen involved, as well as the safety of commercial products) have recently been supplemented by additional regulations as a result of environmental impact studies for veterinary drugs (certain anthelmintics for example). In this respect, vaccines present a clear advantage over therapeutic molecules in that they do not leave residues, and generally have no direct impact on non-target species (including arthropods). Attenuated vaccines, however, require special attention, as they may become dispersed amongst the target population or infect other, non-target species in their vicinity, either in an agro-system or in an eco-system if the target species is a wildlife species.

Other obstacles
Public apprehension over vaccination or over certain products such as genetically modied organisms may constitute a nal obstacle. A specic case of this occurred during the recent episode of FMD in the United Kingdom when it was found that consumers would have been reluctant to consume products from vaccinated animals. This mistrust was one of the (minor) reasons behind the decision not to resort to vaccination in attempting to control the disease in 2001. The current availability of DIVA-type vaccines against FMD, used in combination with serological tests to detect antibodies against non-structural proteins (NSP) is, however, an additional argument in favour of vaccination.

Veterinary vaccination: a complex issue


Veterinary vaccination is a particularly complex subject, due to the many factors that are involved, such as the disparity of targets. As was said earlier, one of the main problems encountered is the number of target species and the number of pathogens involved, as well as their antigenic variation and the need to vaccinate several wildlife species that cannot be easily accessed. The problem is compounded by the fact that the populations concerned are not uniform: rstly, they are distributed across diverse geographical areas and secondly, within each species the economic use to which animals are destined may differ. Although companion animals and horses constitute relatively homogenous groups, this is not the case with livestock, whose breeding conditions and intended use may vary enormously.

492

Rev. sci. tech. Off. int. Epiz., 26 (2)

Different species present biological differences, in particular in terms of their immune response modes and, more specically, with regard to the transfer of maternal immunity from mother to progeny. Whereas in primates this transfer is almost entirely transplacental, and occurs during gestation, this is not the case with dogs and cats, in which a small portion of passive immunity is transmitted during gestation, but most of which is transferred after birth. Since these species are multiparous, there is a disparity in the quantity of immunoglobins ingested by the different puppies or kittens of a given litter. In addition, there are quantitative and qualitative differences between the immune statuses of different mothers, and a mother can only transmit what she herself possesses. In perissodactyles and artiodactyles, transmission occurs only after birth through the colostrum. In birds, this transmission occurs via egg vitellus. In addition to these fundamental biological differences that will inuence vaccination protocols, the intended use of the individuals within each species may vary enormously. Thus, the lives of broilers are very different from those of laying hens; the same can be said for the lives of young meat cattle and dairy cattle. Similarly, the use of breeders (bulls used for natural or articial insemination) differs from that of production animals. We could provide many more examples. These differences bring about variations not only in the vaccination protocols to be recommended, but also in the vaccines desired characteristics, in particular efcacy and duration of protection. Laying hens live much longer than broilers, and dairy cattle live much longer than cattle raised for slaughter. Moreover, within a given species there are clear differences between breeds, e.g. the small dog breeds live longer than the large ones (one of the major genes responsible for size in dogs has just been located). Owing to all these differences, the desired length of vaccinal protection will vary. Dogs, cats and horses must be protected throughout their biological life, which is long; broilers only require protection for the few weeks of their short lives; laying hens should be protected for a year. Duration of protection for wildlife species requires special attention, given population renewal rates and life expectancy. For example, a survey has shown that the average life expectancy for foxes (Vulpes vulpes) in continental Europe was no longer than three years. A vaccine against rabies in foxes therefore need not confer longer protection. Another decisive factor in veterinary vaccinology is the role played by the epidemiological and pathogenic characteristics of an infection. These characteristics will have a great inuence on vaccination protocols. For example, vaccinating female cattle against the pestivirus responsible for bovine viral diarrhoea/mucosal disease, in order to control the infection, will aim at protecting the fetus during gestation, which will have a major impact on the vaccination protocol.

New hope: future trends in veterinary vaccinology


Research and selective breeding
There has been much recent interest in unmet animal health needs on the part of several international bodies, in particular the European Technology Platform for Global Animal Health (ETPGAH) or, more recently, a joint European Union/United States Workshop on Advances in Immunology and Vaccine Discovery. Their meetings have concluded that veterinary medical

Rev. sci. tech. Off. int. Epiz., 26 (2)

493

research in immunology in domestic or wildlife target species should be intensied. To this end, a list of international priority diseases has been drawn up. In particular, immunological research into innate immunity in different species and means of stimulating it should be pursued further; such research is facilitated by the fact that we now have access to complete genome sequences for certain major species (chicken, cattle, pigs, dogs ) and to methods for comparing them with those of other species (man, mice). More concrete (vaccinological) research into adjuvants capable of stimulating innate immunity is also a priority.

With regard to the major epizootic or enzootic diseases that we wish to eradicate, the development of DIVA-type vaccines has provided us with a form of soft disease control. This is one of the most signicant advances in animal health in recent years (this type of vaccine has been used against Aujeszkys disease and infectious bovine rhinotracheitis among others).

Another important eld for research in veterinary vaccinology is methods of vaccine administration; this applies in particular to vaccines for wildlife or feral domestic animals. These are receiving increasing attention since wildlife and feral domestic animals act as a reservoir for identied diseases, but also for non-identied potential infections. Any intervention in wildlife must take account of the need to protect biodiversity; indeed, certain vaccinal interventions aim solely at the preservation of an endangered species (conservation medicine).

The use of new biotechnologies and knowledge derived from the study of the genomics of target species and their pathogens will undoubtedly help solve some of the problems encountered in animal health. For example, there is a growing trend to orient livestock selection not only toward production, but also toward animal health objectives by selecting animals resistant to certain diseases. However, this approach appears somewhat limited, as it is difcult to imagine a return to the production of more rustic disease-resistant animals without losing the economic advantages of previous selections. The example of resistance to Mareks disease shows how difcult it is to reconcile disease-resistance and productivity. Owing to the economic impact of this disease on industrial poultry breeding, geneticists selected lines that were resistant to the Mareks disease virus, but in which certain characteristics that were positive from a production point of view were unfortunately lost. As soon as an effective vaccine was made available at an affordable price, breeders turned to vaccination, since they could thus conserve the economic advantages of the previous selection.

Another approach could lie in the selection of animals that respond well to vaccination. An analysis of the immune response in different dog breeds to rabies vaccination, in the framework of the pet scheme for the introduction of properly vaccinated animals into the United Kingdom, showed considerable variation between breeds. Animal response to vaccination is thus subject to great variability, a factor which could be used in the selection, with or without markers, of good vaccination responders. It should, however, be noted that there is a risk of encountering the same sort of unexpected results as when selecting for resistance to specic diseases.

494

Rev. sci. tech. Off. int. Epiz., 26 (2)

Sustainable solutions: flexible regulations and public/private partnership


Finally, it is to be hoped that regulations governing registration and marketing of vaccines for livestock or wildlife will become more exible, so that veterinary vaccines can meet the epidemilogical requirements of eld conditions, and that animal disease control recommendations will take account of scientic and technological progress as well as of the new vaccinal solutions available. In developing or transition countries, equitable cooperation between the public and private sectors is advisable, in order to make available high-quality, safe, effective and affordable products which can meet those countries immense needs with regard to infectious tropical or parasitic diseases. Whichever solutions are adopted, they must include provision for upholding animal welfare and protecting public health and the environment, and not neglect the need to maintain biodiversity and livestock sustainability over the long term. Although further avenues for development are beginning to emerge, such as vaccines for therapeutic rather than preventive purposes and anti-tumoral vaccination, it is too early to draw any conclusions regarding this research.
Professor P.-P. Pastoret Head of the Publications Department World Organisation for Animal Health (OIE) 12 rue de Prony 75017 Paris France Email: pp.pastoret@oie.int Dr A.A. Schudel Vice-President of the Scientic Commission World Organisation for Animal Health (OIE) Urraca 1366, Carilo (7167) Partido de Pinamar Provincia de Buenos Aires Argentina Email: alejandro.schudel@gmail.com Dr M. Lombard Consultant in Biology 22 rue Crillon 69006 Lyons France Email: lombard.family@wanadoo.fr

Rev. sci. tech. Off. int. Epiz., 2007, 26 (2), 495-501

Conclusions
Tendances futures de la vaccinologie vtrinaire
Comme dj mentionn dans lintroduction de ces deux numros de la Revue scientique et technique de lOrganisation mondiale de la sant animale (OIE), la vaccination est sans conteste la meilleure mesure pour prvenir les maladies infectieuses animales et ventuellement les liminer. Ceci est particulirement vrai pour les infections dorigine virale, notamment parce que nous ne possdons pas encore de molcules antivirales large spectre utilisables en mdecine vtrinaire. La vaccination peut donc tre considre comme un investissement visant garantir la sant animale. Il existe nanmoins de nombreux besoins en sant animale qui ne sont pas encore satisfaits et des infections contre lesquelles il nexiste pas encore de vaccins.

Les obstacles au dveloppement de vaccins vtrinaires


Les obstacles au dveloppement de vaccins seront examins en dtail ci-aprs ; certains dentre eux ont dj t mentionns dans lintroduction de ces deux numros thmatiques de la Revue.

Obstacles biologiques et techniques


Les premires difcults sont dordre biologique et technique. Ceci permet, par exemple, dexpliquer pourquoi il existe peu de vaccins antiparasitaires sur le march. Les parasites cohabitent en effet avec leur hte et ont dvelopp au cours de lvolution de nombreuses stratgies qui autorisent cette cohabitation. Ceci nest pas seulement vrai pour les parasites, mais vaut galement pour les virus ou les bactries. Les virus ont co-volu avec leur hte et ont galement dvelopp de nombreuses stratgies pour survivre dans une population ou chez leur hte dans un certain tat dquilibre. Ils ont notamment acquis, souvent aux dpens de leur hte, de nombreuses molcules ou des mcanismes qui leur permettent de circonvenir les diffrents bras arms de la rponse immune. Cest ce qui explique, en partie, labsence de vaccin disponible contre la peste porcine africaine, malgr de nombreuses annes de recherche. Dautre part, nombre dinfections virales et bactriennes sont provoques par des agents pathognes possdant plusieurs srotypes (comme la vre aphteuse et linuenza aviaire), et qui voluent constamment. Un exemple rcent des difcults engendres par cette variation srotypique est celui de la vre catarrhale ovine apparue dans le Nord de lEurope, qui tait due au srotype 8 (un parmi les 24 srotypes connus). Les vaccins existants taient inutilisables car ils ne contenaient pas les antignes correspondants. Pourtant, pour des maladies vectorielles comme la vre catarrhale ovine, la vaccination

496

Rev. sci. tech. Off. int. Epiz., 26 (2)

semble tre la seule solution, car il est particulirement difcile de sattaquer au vecteur, surtout si lon souhaite recourir des mthodes qui naient pas un impact ngatif sur lenvironnement. Une autre difcult inhrente la vaccinologie vtrinaire est la grande diversit des espces cibles et des infections affectant chacune dentre elles. Les vaccins ont la particularit, contrairement aux molcules thrapeutiques (antibiotiques, anthelminthiques) dtre le plus souvent troitement spciques ; il nexiste que peu de vaccins multi-espces comportant la mme valence antignique (ttanos, rage). Enn, le fait que les cibles comptent nombre despces sauvages, difciles daccs par la vaccination, pose encore un autre problme. Par exemple, il parat difcile de vacciner contre la rage les chauves-souris vampires (Desmodus rotundus) en Amrique latine. En revanche, dautres espces sauvages comme le renard (Vulpes vulpes) (dans le cas de la rage) ou le sanglier (Sus scrofa) (dans le cas de la peste porcine classique) sont dun accs relativement ais et peuvent tre vaccines facilement grce la mthode des appts vaccinaux. Lapparition de nouvelles maladies mergentes pour lesquelles aucun vaccin nexiste au moment de leur apparition reprsente un d supplmentaire pour le dveloppement des vaccins vtrinaires. Linfection par le virus Nipah apparue en Malaisie en est un exemple : comme il sagissait dune anthropozoonose mortelle, la seule solution a t labattage systmatique des porcs infects et la destruction ou lenfouissement de leurs cadavres. En revanche, aux tats-Unis dAmerique, lmergence de linfection par le virus de la vre du Nil occidental, qui affecte lhomme et nombre despces animales, mais particulirement les chevaux, a conduit au dveloppement et la mise sur le march de huit vaccins quins diffrents dans un laps de temps exceptionnellement court. De mme, la rmergence de maladies transfrontalires dans certains pays auparavant indemnes constitue une menace, dont le risque est augment par leffet accru des cinq T (Transport, Travel, Tourism, Trade, Terrorism : les transports, les dplacements, le tourisme, le commerce et le terrorisme). Ces maladies proviennent gnralement de pays en dveloppement ou en transition qui nont pas les moyens de sen dfendre ; pour la communaut internationale, aider combattre ces maladies l o elles svissent, notamment par la vaccination, cest protger lensemble de la plante. Ces maladies possdent souvent un rservoir sauvage difcilement accessible, comme le bufe africain (Syncerus caffer) pour les souches SAT (South African Territories) de la vre aphteuse.

Obstacles conomiques
Dautres obstacles la vaccination sont de nature conomique : le premier dentre eux est le faible retour dinvestissement que peuvent esprer les laboratoires privs impliqus dans le dveloppement de vaccins vtrinaires destins aux animaux de rente, particulirement pour des maladies spciques des pays en dveloppement, l o malheureusement les besoins se font le plus sentir. Ces problmes, toutefois, ne se limitent pas aux pays en dveloppement ou en transition car, mme dans les pays dvelopps, certaines espces cibles font lobjet de peu

Rev. sci. tech. Off. int. Epiz., 26 (2)

497

dattention lorsquelles sont juges mineures ; par exemple, en Europe, sont qualis despces mineures les moutons producteurs de lait, les chvres, les lapins, les poissons autres que les salmonids et les oiseaux autres que les poulets. cela sajoute le fait que ces espces mineures sont diversement rparties dans les diffrentes zones de lespace europen, ce qui ne facilite pas la mise en uvre de procdures europennes pour la mise sur le march des vaccins vtrinaires. Sajoute encore lexistence de maladies rares la fois dans les espces considres comme majeures (par exemple la forme europenne du coryza gangrneux chez les ruminants) et, scnario le plus catastrophique, dans les espces dites mineures (par exemple la tularmie du livre). Ceci a conduit au concept dsign par lacronyme MUMS (Minor Use, Minor Species [utilisations mineures et espces mineures]). Le dveloppement de vaccins pour ces types de maladies passe ncessairement par un partenariat public-priv quitable.

Obstacles juridiques et rglementaires


Les derniers obstacles sont de nature rglementaire et juridique. Nombre de rglementations applicables la sant animale privilgient la prophylaxie sanitaire et excluent la vaccination prventive, particulirement dans les pays dvelopps qui ont limin les grandes pizooties du cheptel domestique (vre aphteuse, peste porcine classique). Heureusement, un changement se dessine en faveur de la mise en uvre de politiques incluant la vaccination ( vaccination-to-live , cest--dire visant au maintien en vie des animaux), notamment grce la disponibilit de vaccins de nouvelle gnration, associs un test de diagnostic compagnon et pourvus dun marqueur srologique (technologie DIVA permettant de diffrencier les animaux infects des animaux vaccins). Cette possibilit a permis aux instances internationales telles que lOIE de modier les normes applicables la vaccination.

Un autre frein rglementaire au dveloppement de vaccins vtrinaires pour les animaux de rente est lexistence de rglementations trs strictes et rigides pour lenregistrement des mdicaments vtrinaires (y compris les vaccins) et leur mise sur le march. Cette lgislation ne favorise pas la exibilit dans le choix des souches vaccinales pour la composition de vaccins appropris la prvention des infections provoques par des agents pathognes aux multiples srotypes, et leur adaptation aux conditions pidmiologiques du terrain. cette rglementation trs contraignante, mais qui a cependant le grand mrite de garantir la qualit et lefcacit vis--vis de lagent pathogne vis ainsi que la scurit des produits commercialiss, se sont plus rcemment ajoutes de nouvelles exigences, rendues ncessaires par ltude de limpact des mdicaments vtrinaires sur lenvironnement (cest notamment le cas de certains anthelminthiques).

cet gard, les vaccins prsentent un net avantage par rapport aux molcules thrapeutiques car ils ne provoquent pas la formation de rsidus et nont gnralement pas dimpact direct sur les espces non cibles (arthropodes inclus). Une attention particulire doit cependant tre apporte aux vaccins attnus qui peuvent se dissminer dans la population cible ou infecter dautres espces non cibles qui leur sont proches, que ce soit dans un agro-systme ou dans un cosystme sil sagit despces sauvages.

498

Rev. sci. tech. Off. int. Epiz., 26 (2)

Autres obstacles
Un dernier obstacle peut tre constitu par la mance du public lgard de la vaccination ou de certains produits comme les organismes gntiquement modis. Un cas particulier a t la rticence du public consommer des produits issus danimaux qui auraient t vaccins contre la vre aphteuse lors du rcent pisode de cette maladie au Royaume-Uni. Cette mance a t lune des raisons (mineure) qui ont motiv la dcision de ne pas utiliser la vaccination pour tenter de juguler lpizootie en 2001. La disponibilit actuelle de vaccins de type DIVA contre la vre aphteuse, qui sutilisent avec des tests srologiques de dtection des anticorps dirigs contre les protines non structurelles est dsormais un argument supplmentaire en faveur de la vaccination.

La vaccination vtrinaire : une thmatique complexe


La vaccination vtrinaire est une question particulirement complique du fait des nombreux facteurs qui interviennent, par exemple la disparit des cibles. Ainsi quil a t mentionn auparavant, lun des principaux problmes rencontrs est le nombre despces cibles et le nombre de pathognes impliqus, ainsi que leur variation antignique et le fait de devoir vacciner certaines espces sauvages difcilement accessibles. Le problme se complique du fait que les populations animales ne sont pas uniformes : dune part, leur rpartition gographique est htrogne et, dautre part, une mme espce peut faire lobjet de plusieurs types de production diffrents. Si les animaux de compagnie et les chevaux constituent des groupes relativement homognes, il nen va pas de mme pour les animaux de rente dont lutilisation conomique et les conditions dlevage sont extrmement diverses. Les espces diffrent biologiquement entre elles, notamment dans leur mode de rponse immune et, plus spciquement, dans le transfert de limmunit maternelle de la mre sa descendance. Alors que chez les primates ce transfert se fait presque entirement durant la gestation par voie transplacentaire, il nen va pas de mme chez les chats et les chiens, chez qui une faible partie de limmunit passive est transmise durant la gestation, la part la plus importante tant transmise aprs la naissance. Ces espces tant multipares, ceci cre une disparit dans la quantit dimmunoglobines ingres par les diffrents chiots ou chatons de la mme porte ; cela sajoutent les diffrences quantitatives et qualitatives de ltat immun des mres : or une mre ne peut transmettre que ce quelle possde elle-mme. Chez les prissodactyles et les artiodactyles, la transmission sopre exclusivement aprs la mise-bas par le biais du colostrum. Chez les oiseaux, cette transmission sopre par le vitellus de luf. En plus de ces diffrences biologiques fondamentales qui vont inuencer les protocoles de vaccination, lusage auquel sont destins les individus dune mme espce peut varier considrablement. Ainsi, les conditions de vie des poulets de chair diffrent de celles des poules pondeuses ; celles des veaux de boucherie diffrent de celles des vaches laitires. De mme, les animaux reproducteurs (taureaux utiliss pour linsmination naturelle ou articielle) ne sont pas exploits de la mme manire que les animaux producteurs. On

Rev. sci. tech. Off. int. Epiz., 26 (2)

499

pourrait multiplier les exemples. Ces diffrences entranent des variations non seulement dans les protocoles de vaccination recommands, mais galement dans les caractristiques souhaitables des vaccins, notamment en termes defcacit et de dure de protection. Les poules pondeuses vivent bien plus longtemps que les poulets de chair, il en va de mme des vaches laitires par rapport aux bovins de boucherie. De plus, au sein dune mme espce il existe de nettes diffrences entre les races ; ainsi les petites races canines vivent plus longtemps que les grandes (lun des principaux gnes responsables de la taille chez le chien vient dtre localis). Du fait de toutes ces diffrences, la dure de protection attendue dun vaccin est elle-mme variable. Les chiens, les chats, les chevaux doivent tre protgs durant toute leur vie biologique, qui est longue. Les poulets de chair ne doivent tre protgs que pendant les quelques semaines que dure leur vie ; les poules pondeuses doivent tre protges durant une anne entire. Une attention particulire doit tre porte sur la dure de protection des espces sauvages, en tenant compte du renouvellement de la population et de son esprance de vie. Une enqute a, par exemple, montr que la dure moyenne de vie des renards (Vulpes vulpes) en Europe continentale nexcdait pas trois ans. Par consquent, la protection confre par un vaccin antirabique destin au renard peut ne pas dpasser cette priode. Un autre facteur dterminant en vaccinologie vtrinaire est le rle jou par les caractristiques pidmiologiques et pathogniques dune infection, qui vont inuencer de manire dterminante les protocoles de vaccination. Par exemple, la vaccination des vaches pour contrler linfection due au Pestivirus responsable de la diarrhe virale bovine/maladie des muqueuses, aura pour but de protger le ftus durant la gestation, ce qui aura un impact majeur sur le protocole de vaccination.

Un nouvel espoir : les tendances futures de la vaccinologie vtrinaire


La recherche et llevage slectif
Plusieurs instances internationales se sont rcemment penches sur les besoins non satisfaits en sant animale ; cest en particulier le cas de la Plateforme technologique europenne sur la sant animale dans le monde (ETPGAH), ou plus rcemment de lAtelier conjoint tats-Unis dAmrique/Union europenne sur les avances en matire dimmunologie et les dcouvertes en vaccinologie. Il ressort de ces rencontres que la recherche en mdecine vtrinaire consacre limmunologie des espces domestiques ou sauvages doit tre intensie. Une liste de maladies prioritaires au niveau mondial a galement t tablie. Il convient notamment dapprofondir la recherche sur limmunit inne des diffrentes espces et sur les moyens de la stimuler ; cette approche est actuellement facilite par le fait que les squences gnomiques compltes de certaines espces majeures (poulet, bovins, porc, chien) sont dsormais connues et que des approches comparatives relatives au gnome dautres espces (homme, souris) sont disponibles. La recherche dadjuvants capables de stimuler limmunit inne gure galement parmi les priorits des applications concrtes de la vaccinologie.

500

Rev. sci. tech. Off. int. Epiz., 26 (2)

Pour ce qui est des grandes pizooties ou des maladies enzootiques dont on souhaite llimination, le dveloppement de vaccins de type DIVA a apport une solution qui autorise une mthode douce de contrle. Il sagit l dune des avances les plus remarquables en sant animale ces dernires annes (ce type de vaccin a t utilis avec succs contre la maladie dAujeszky et la rhinotrachite infectieuse bovine, parmi dautres). Un autre grand chantier de la vaccinologie vtrinaire est celui du mode dadministration des vaccins ; ceci vaut en particulier pour les vaccins destins la faune sauvage ou aux espces domestiques errantes. On y porte de plus en plus dattention car ces animaux jouent souvent un rle de rservoir pour des infections connues, mais aussi pour des infections potentielles non encore identies. Toute intervention sur la faune sauvage doit prendre en compte le souci du maintien de la biodiversit, certaines interventions vaccinales ayant dailleurs pour seul but la conservation dune espce menace (conservation medicine). Lutilisation des nouvelles biotechnologies et des connaissances drives de la gnomique des espces cibles et de leurs pathognes aidera certainement rsoudre certains problmes rencontrs en sant animale. Par exemple, une tendance actuelle consiste orienter la slection des animaux de rente non plus seulement vers des objectifs de production, mais aussi vers des objectifs sanitaires en slectionnant des animaux rsistants vis--vis de certaines maladies. Toutefois, cette approche prsente certaines limites, car il parat difcile de revenir une production danimaux plus rustiques et rsistants aux maladies sans perdre les acquis conomiques des slections antrieures. Lexemple de la rsistance la maladie de Marek est l pour nous rappeler la difcult concilier la rsistance aux maladies avec les impratifs de productivit. Du fait de limpact conomique de cette maladie sur llevage industriel des volailles, des gnticiens avaient slectionn des lignes rsistantes au virus de la maladie de Marek, en perdant malheureusement certains traits de production intressants. Ds quun vaccin efcace fut disponible un prix abordable, les leveurs ont privilgi la vaccination, car elle permettait de conserver les acquis conomiques de la slection antrieure. Une autre voie dapproche serait la slection danimaux bons rpondeurs la vaccination. Une analyse de la rponse immune des diffrentes races de chien la vaccination antirabique dans le cadre du programme dintroduction danimaux dment vaccins au Royaume-Uni (pet scheme) a rvl une variation considrable entre les races lgard de cette vaccination. Il existe donc une variabilit importante dans la rponse des animaux la vaccination, qui pourrait tre mise prot dans la slection, assiste ou non de marqueurs, danimaux bons rpondeurs. Il faut cependant remarquer que lon risque de rencontrer le mme type dalas que celui rencontr lors de la slection de traits de rsistance vis--vis de maladies spciques.

Des solutions durables : une rglementation plus souple et des partenariats public/priv
Enn, il faut souhaiter que la rglementation en matire denregistrement et dautorisation de mise sur le march des vaccins destins aux animaux de rente ou sauvages sassouplisse, an de permettre aux vaccins vtrinaires de satisfaire aux

Rev. sci. tech. Off. int. Epiz., 26 (2)

501

besoins pidmiologiques du terrain, et que les prescriptions zoosanitaires pour le contrle des maladies tiennent compte des avances scientiques et technologiques ainsi que des nouvelles solutions vaccinales disponibles. Pour ce qui est des pays en dveloppement ou en transition, il faut esprer quune coopration quitable entre le secteur priv ou public stablisse an de rendre disponibles des produits de qualit, efcaces et srs, conomiquement abordables, qui permettent de satisfaire les immenses besoins de ces pays confronts aux maladies tropicales infectieuses ou parasitaires. Quelles que soient les solutions apportes, elles doivent prendre en compte la bientraitance animale, la protection de la sant publique, la protection de lenvironnement et le ncessaire maintien de la biodiversit et de la durabilit de llevage long terme. Dautres secteurs de dveloppement commencent tre identis : il sagit des vaccins vise thrapeutique plutt que prventive et de la vaccination antitumorale ; il est cependant trop tt pour en dresser un quelconque bilan aujourdhui.
Professeur P.-P. Pastoret Chef du Service des publications Organisation mondiale de la sant animale (OIE) 12 rue de Prony 75017 Paris France e-mail : pp.pastoret@oie.int Dr A.A. Schudel Vice-Prsident de la Commission scientique de lOrganisation mondiale de la sant animale (OIE) Urraca 1366 Carilo (7167) Partido de Pinamar Provincia de Buenos Aires Argentine e-mail : alejandro.schudel@gmail.com Dr M. Lombard Consultant en biologie 22 rue Crillon 69006 Lyon France e-mail : lombard.family@wanadoo.fr

Rev. sci. tech. Off. int. Epiz., 2007, 26 (2), 503-509

Conclusiones
Tendencias de la vacunologa veterinaria en el futuro
Como queda dicho en la introduccin a estos dos nmeros de la Revista cientca y tcnica de la Organizacin Mundial de Sanidad Animal (OIE), la vacunacin es, incontestablemente, la medida ms ecaz para prevenir y eventualmente eliminar las enfermedades infecciosas de los animales, en especial las infecciones de origen vrico, sobre todo porque an no disponemos de molculas antivirales de amplio espectro que puedan utilizarse en medicina veterinaria. Desde este punto de vista, cabe equiparar la vacunacin a invertir en un seguro de enfermedad animal. Sin embargo, hay numerosas necesidades todava desatendidas en materia de sanidad animal, as como infecciones contra las que no existe ninguna vacuna.

Los obstculos a la obtencin de vacunas veterinarias


Algunos de los obstculos que dicultan la obtencin de vacunas vienen ya mencionados en la introduccin a estos dos nmeros temticos de la Revista de la OIE. Estos obstculos y otros sern discutidos en mayor detalle a continuacin.

Obstculos biolgicos y tcnicos


Las primeras dicultades son de orden biolgico y tcnico, y permiten explicar, por ejemplo, por qu hay tan pocas vacunas antiparasitarias disponibles en el mercado. Los parsitos, en efecto, conviven con el hospedador, y en el curso de la evolucin han ido desarrollando numerosas estrategias para adaptarse a esa situacin. Ello se aplica no slo a los parsitos, sino tambin a los virus y bacterias. Los virus han coevolucionado con sus hospedadores, y generado asimismo muchas estrategias para sobrevivir en un cierto estado de equilibrio dentro de una poblacin o un organismo hospedador. Sobre todo han adquirido, a menudo a costa del hospedador, numerosas molculas o mecanismos que les permiten eludir los diversos brazos armados de la respuesta inmunitaria. Esto explica, en parte, que tras muchos aos de investigacin, an no dispongamos de una vacuna contra la peste porcina africana. Por otro lado, muchas infecciones vricas y bacterianas son causadas por agentes patgenos que poseen mltiples serotipos, sujetos adems a constante evolucin, como es el caso de la ebre aftosa o la inuenza aviar. La reciente aparicin en el Norte de Europa de la ebre catarral ovina causada por el serotipo 8 (uno de los 24 que se conocen) brinda buen ejemplo de las dicultades que puede acarrear semejante variabilidad serotpica. Las vacunas existentes eran inutilizables porque carecan de los correspondientes antgenos. Y sin embargo, en el caso de enfermedades vectoriales como la ebre catarral ovina, la nica solucin parece radicar en las vacunaciones, pues resulta especialmente difcil atacar al vector, sobre todo con mtodos que no daen el medio ambiente.

504

Rev. sci. tech. Off. int. Epiz., 26 (2)

Otra de las dicultades inherentes a la vacunologa veterinaria estriba en la gran variedad de especies destinatarias y las numerosas infecciones distintas que pueden afectar a cada una de ellas. Ahora bien, las vacunas, a diferencia de las molculas teraputicas (antibiticos, antihelmnticos, etc.), tienen la particularidad de ser casi siempre muy especcas; hay pocas vacunas multiespeccas que presenten la misma valencia antignica (ttanos, rabia). Otro problema relativo a la vacunacin de los animales es que entre las especies destinatarias hay animales salvajes a los que es difcil llegar para administrarles una vacuna. Buen ejemplo en este sentido es la dicultad de vacunar contra la rabia a los murcilagos vampiro (Desmodus rotundus) de Amrica Latina. Hay otras especies salvajes, en cambio, como el zorro (Vulpes vulpes) (rabia) o el jabal (Sus scrofa) (peste porcina clsica), que son de un acceso relativamente fcil y se les puede administrar vacunas empleando cebos vacunales. La aparicin de nuevas enfermedades emergentes, contra las cuales, naturalmente, no hay vacuna alguna en el momento en el que aparecen constituye un desafo adicional para el desarrollo de vacunas veterinarias. Tal fue el caso de la infeccin por el virus Nipah surgida en Malasia: como adems se trataba de una antropozoonosis mortal, la nica solucin posible fue el sacricio sistemtico de los cerdos infectados y la destruccin o inhumacin de los cadveres. Sin embargo, en los Estados Unidos de Amrica, la emergencia de la infeccin del virus de la ebre del Nilo Occidental, que afecta a los hombres y a varias especies animales, principalmente a los equinos, ha llevado a elaborar y comercializar ocho vacunas equinas distintas en un lapso de tiempo excepcionalmente corto. Una amenaza similar proviene de la reaparicin de enfermedades transfronterizas en determinados pases hasta entonces indemnes, cuyo nivel de riesgo aumenta a medida que se intensican los factores conocidos como los cinco T (Transport, Travel, Tourism, Trade, Terrorism: transporte, viaje, turismo, comercio, terrorismo). Estas enfermedades proceden generalmente de pases en desarrollo o en transicin que no tienen medios para defenderse de ellas. Para la comunidad internacional, ayudar a combatirlas all donde atacan, sobre todo mediante la vacunacin, equivale a ayudar al planeta entero. Adems, suelen poseer un reservorio salvaje al que es difcil acceder, como ocurre con el bfalo africano (Syncerus caffer), reservorio de las cepas SAT (South African Territories) de la ebre aftosa.

Obstculos econmicos
Otros obstculos a la vacunacin son de ndole econmica, y el principal es la escasa rentabilidad que las empresas privadas del sector pueden esperar obtener de la elaboracin de vacunas veterinarias para los animales de granja, sobre todo en el caso de enfermedades especcas de los pases en desarrollo, que por desgracia son justamente los ms necesitados en la materia. Estos problemas, sin embargo, no afectan slo a los pases en desarrollo o en transicin, ya que incluso en los pases desarrollados hay ciertas especies que reciben poca atencin y son consideradas menores. En Europa, por ejemplo, se calican de tales la oveja productora de leche, la cabra, el conejo, todos los peces salvo los salmnidos y

Rev. sci. tech. Off. int. Epiz., 26 (2)

505

todas las aves menos el pollo. Por si fuera poco, esas especies estn repartidas de forma heterognea en las distintas zonas del espacio europeo, cosa que no ayuda a instituir mecanismos de dimensin europea para comercializar vacunas veterinarias. La situacin se agrava adems por la existencia de enfermedades raras a la vez en especies consideradas mayores (por ejemplo la forma europea de ebre catarral maligna en rumiantes) y en especies llamadas menores (la peor combinacin posible) (por ejemplo, la tularemia en la liebre). De ah que se haya acuado el concepto de especie MUMS (Minor Use, Minor Species: usos menores y especies menores). El desarrollo de vacunas para enfermedades de este tipo pasa obligatoriamente por una alianza equilibrada entre los sectores pblico y privado.

Obstculos jurdicos y normativos


Los ltimos obstculos son de carcter normativo y jurdico. Muchos reglamentos zoosanitarios privilegian la prolaxis higinica con exclusin de la vacunacin preventiva, sobre todo en los pases desarrollados que ya han logrado eliminar las grandes epizootias de los animales domsticos (ebre aftosa o peste porcina clsica). Afortunadamente, empieza a perlarse un cambio de rumbo en favor de la aplicacin de polticas sanitarias que incorporen la vacunacin (vaccination to live), gracias sobre todo a la existencia de vacunas de nueva generacin que tienen una prueba de diagnstico complementaria y que estn provistas de un marcador serolgico (tecnologa DIVA: diferenciar a los animales infectados de los animales vacunados), lo que ha permitido a instancias internacionales como la OIE modicar las normas relativas a la vacunacin. Otro factor jurdico que diculta la obtencin de vacunas veterinarias destinadas a los animales de granja es la existencia de reglamentos muy estrictos y rgidos para registrar y comercializar medicamentos veterinarios (incluidas las vacunas); esta legislacin no favorece la exibilidad a la hora de elegir cepas que sirvan para crear vacunas adecuadas para prevenir infecciones debidas a patgenos con mltiples serotipos y para adaptar esas vacunas a las condiciones epidemiolgicas reinantes sobre el terreno. Esta aparatosa reglamentacin, que sin embargo presenta la gran ventaja de garantizar la calidad, la ecacia para el patgeno en cuestin y la seguridad de los productos comercializados, ha sido reforzada ltimamente por normas adicionales, fruto de los estudios de impacto ambiental de los medicamentos (ciertos antihelmnticos, por ejemplo). En este terreno las vacunas presentan una neta ventaja con respecto a las molculas teraputicas, ya que no inducen la formacin de residuos ni suelen tener efectos directos sobre las especies no destinatarias (comprendidos los artrpodos). Pese a ello, conviene prestar especial atencin a las vacunas atenuadas que pueden extenderse dentro de la poblacin destinataria o infectar a otras especies anes, ya sea en un agrosistema o en un ecosistema (cuando se trata de vacunar a una especie salvaje).

Otros obstculos
Un ltimo obstculo puede provenir de los recelos que alimenta el gran pblico respecto a la vacunacin o a ciertos productos como los organismos genticamente modicados. Un caso particular, en este sentido, fue el de la reticencia a consumir productos

506

Rev. sci. tech. Off. int. Epiz., 26 (2)

procedentes de animales que hubieran sido vacunados contra la ebre aftosa durante el episodio infeccioso surgido hace pocos aos en el Reino Unido. Esa desconanza fue uno de los factores (de importancia menor) que en 2001 motivaron la decisin de no utilizar las vacunaciones para tratar de atajar la epizootia. Ahora bien, la actual existencia de vacunas de tipo DIVA contra la ebre aftosa, que se usan con pruebas serolgicas para detectar anticuerpos contra protenas no estructurales (NSP), constituye un argumento suplementario en favor de la vacunacin.

La vacunacin veterinaria: un tema complicado


Los numerosos factores que entran en juego, por ejemplo la disparidad de especies destinatarias, hacen de la vacunacin veterinaria un asunto especialmente complejo. Como queda dicho, uno de los principales problemas reside en la intervencin de un gran nmero de especies y de patgenos, en la variabilidad antignica de stos y en la necesidad de vacunar a ciertas especies salvajes a las que es difcil acceder. El asunto se complica an ms por la falta de uniformidad entre las poblaciones: por un lado su distribucin geogrca es heterognea, y por el otro las distintas poblaciones de una especie pueden destinarse a usos econmicos muy diversos. Los animales de compaa y los caballos forman grupos relativamente homogneos, a diferencia de los animales de granja, cuyas condiciones de cra y utilizacin pueden ser muy variables. Las especies son distintas biolgicamente, en especial en su modo de respuesta inmunitaria, y ms especcamente en el modo de transferencia de la inmunidad materna a la progenie. En los primates el proceso tiene lugar casi totalmente por va placentaria durante la gestacin. En los gatos y perros, en cambio, se transmite por dicha va slo una pequea parte de la inmunidad pasiva, mientras que la parte ms importante se adquiere despus del nacimiento. As, al tratarse de especies multparas, los cachorros de una misma camada habrn ingerido una cantidad de inmunoglobulinas distinta en cada caso. A ello se aaden diferencias cuantitativas y cualitativas en el estado inmunitario de la madre, pues sta slo puede transmitir lo que ella misma posee. En los perisodctilos y artiodctilos, la transmisin se realiza exclusivamente despus del parto a travs del calostro. En las aves tiene lugar a travs del vitelo del huevo. Adems de estas diferencias biolgicas fundamentales, que van a inuir en gran medida en los protocolos de vacunacin, dentro de cada especie tambin pueden variar sobremanera los usos a los que vayan a destinarse los distintos individuos. As, por ejemplo, la vida de los pollos de engorde es completamente distinta a la de las gallinas ponedoras, y la de un joven bovino de una raza de produccin crnica tiene poco que ver con la de una vaca lechera. Los ejemplares reproductores (toros en inseminacin natural o articial) son destinados a un uso muy distinto al de los animales productores. Cabra ofrecer an multitud de ejemplos en este sentido. Estas diferencias entraan variaciones no slo en los protocolos de vacunacin recomendados, sino tambin en las caractersticas que deben revestir las vacunas, sobre todo en cuanto a su ecacia y a la duracin de la proteccin que coneren. Una gallina ponedora vive mucho ms tiempo que un pollo de engorde, una vaca lechera mucho ms que sus congneres destinados

Rev. sci. tech. Off. int. Epiz., 26 (2)

507

directamente a la carnicera. Adems, dentro de una misma especie hay marcadas diferencias entre las razas: los perros de raza pequea, por ejemplo, viven mucho ms tiempo que los de raza grande (se acaba de localizar uno de los principales genes que determinan el tamao de un perro). En funcin de todas estas diferencias variar la duracin prevista de la proteccin que ofrezca una vacuna. Los perros, gatos y caballos requieren proteccin durante toda su vida biolgica, que suele ser larga. En el caso de los pollos de engorde, en cambio, slo hay que protegerlos las pocas semanas que dura su vida, mientras que las gallinas ponedoras deben ser protegidas durante un ao. Especial atencin conviene prestar al intervalo de proteccin en las especies salvajes, teniendo en cuenta la renovacin de la poblacin y la esperanza de vida de los individuos. Una investigacin demostr, por ejemplo, que en Europa continental los zorros (Vulpes vulpes) no viven en promedio ms de tres aos. Por ello una vacuna antirrbica destinada al zorro no debe necesariamente conferir proteccin durante un lapso de tiempo superior. Otro factor clave para la vacunologa veterinaria son las caractersticas epidemiolgicas y patognicas de una infeccin, que van a inuir decisivamente en los protocolos de vacunacin. Por ejemplo: la vacunacin de hembras de bovino contra el Pestivirus responsable de la diarrea viral bovina (o enfermedad de las mucosas), utilizada para controlar la infeccin, tiene por nalidad proteger al feto durante la gestacin, cosa que determina el protocolo de vacunacin.

Una nueva esperanza: tendencias de la vacunologa veterinaria en el futuro


Investigacin y cra selectiva
En los ltimos aos ha habido varias instancias internacionales que han trabajado sobre las lagunas existentes en el terreno de la sanidad animal. Entre ellas destacan la Plataforma Tecnolgica Europea para la Sanidad Animal Mundial (ETPGAH) o, ms recientemente, un grupo de trabajo mixto de los Estados Unidos de Amrica y la Unin Europea que se ocupa de los avances en materia de inmunologa y del descubrimiento de vacunas. De las reuniones de esos foros de estudio se desprende que es indispensable intensicar la investigacin sobre la inmunologa de las especies de las que se ocupa la medicina veterinaria, tanto domsticas como salvajes. Se ha elaborado asimismo una lista de enfermedades prioritarias a escala mundial. Desde el punto de vista de la inmunologa, es preciso proseguir las investigaciones sobre inmunidad innata en las distintas especies y sobre posibles formas de estimularla, trabajo que hoy en da se ve facilitado por el conocimiento de la secuencia genmica completa de algunas de las especies ms importantes (pollo, bovinos, cerdo, perro, etc.) y por los mtodos comparativos en relacin con la de otras especies (hombre, ratn). Ms concretamente (en vacunologa), la bsqueda de adyuvantes capaces de estimular la inmunidad innata gura tambin entre las prioridades. Por lo que respecta a las grandes epizootias o las enfermedades enzoticas que aspiramos a eliminar, la obtencin de vacunas de tipo DIVA ha aportado una solucin que permite controlarlas con mtodos poco agresivos. Se trata de uno de los avances ms

508

Rev. sci. tech. Off. int. Epiz., 26 (2)

notables de los ltimos aos en el terreno de la sanidad animal (este tipo de vacunas han sido utilizadas exitosamente contra la enfermedad de Aujeszky y la rinotraquetis infecciosa bovina, entre otras enfermedades). Otra de las grandes lneas de trabajo de la vacunologa veterinaria gira en torno al modo de administracin de las vacunas, sobre todo en el caso de las vacunas destinadas a la fauna salvaje o a ejemplares cimarrones de especies domsticas. Este tema viene mereciendo una atencin creciente, ya que estos animales cumplen a menudo una funcin de reservorio de infecciones, tanto conocidas como an no descritas pero potencialmente peligrosas. Toda actuacin que afecte a la fauna salvaje debe integrar la voluntad de preservar la diversidad biolgica. Tanto es as que ciertas intervenciones vacunales tienen por objetivo nico la proteccin de una especie amenazada (conservation medicine). El uso de las nuevas tecnologas y del conocimiento obtenido con la genmica de las especies destinatarias y sus patgenos ayudar sin duda a resolver algunos de los problemas que tiene planteados la sanidad animal. Por ejemplo, una tendencia actual que se viene aanzando es la de orientar la seleccin de los animales de granja hacia el cumplimiento no slo de objetivos de produccin, como era el caso hasta ahora, sino tambin de objetivos zoosanitarios mediante la seleccin de animales resistentes a ciertas enfermedades. Sin embargo, esta lnea de trabajo parece un tanto limitada, pues cuesta imaginar un retorno a la produccin de animales ms rsticos y resistentes a enfermedades sin sacricar por el camino las ventajas econmicas logradas con anteriores selecciones. El ejemplo de la enfermedad de Marek es un buen recordatorio de la dicultad de conciliar la resistencia a las enfermedades con la productividad. En vista de las grandes prdidas econmicas que la enfermedad de Marek iniga a la industria avcola, una serie de genetistas haba seleccionado linajes resistentes al virus responsable de esa dolencia, pero ello, lamentablemente, supuso la prdida de ciertos rasgos de produccin interesantes. En cuanto se obtuvo una vacuna ecaz y de precio asequible para los productores, stos empezaron a privilegiar la vacunacin, que les permita conservar la rentabilidad econmica obtenida con la seleccin anterior. Otro posible ngulo de trabajo consistira en seleccionar animales que respondieran bien a la vacunacin. El anlisis de la respuesta inmunitaria que distintas razas de perro ofrecieron a la vacunacin antirrbica, como parte del pet scheme para introducir animales correctamente vacunados en el Reino Unido, puso de relieve sobre todo una considerable variacin entre razas. As pues, hay un nivel importante de variabilidad en la respuesta de los animales a una vacuna, factor que podra utilizarse para seleccionar, con o sin ayuda de marcadores, animales que ofrecieran una buena respuesta a la vacunacin. Conviene sealar sin embargo que quiz tropecemos con el mismo tipo de imponderables que los surgidos en los procesos de seleccin con respecto a enfermedades especcas.

Soluciones sostenibles: reglamentaciones ms flexibles y asociaciones entre sectores pblico y privado


Es deseable, por ltimo, que la reglamentacin en materia de registro y comercializacin de vacunas para animales salvajes o de granja presente mayor exibilidad, de manera que las vacunas puedan responder a las necesidades epidemiolgicas que surjan sobre

Rev. sci. tech. Off. int. Epiz., 26 (2)

509

el terreno, y que las prescripciones zoosanitarias para el control de enfermedades tengan en cuenta los adelantos cientcos y tecnolgicos, as como la aparicin de nuevas soluciones basadas en el uso de vacunas. Por lo que respecta a los pases en desarrollo o en transicin, cabe esperar que en ellos empiecen a establecerse mecanismos equitativos de colaboracin entre los sectores pblico y privado para comercializar productos de calidad, ecaces, seguros y de precio asequible, que permitan satisfacer las inmensas necesidades de dichos pases en materia de enfermedades tropicales infecciosas o parasitarias. Sean cuales sean las soluciones que se ofrezcan, stas debern integrar debidamente planteamientos como el bienestar de los animales, la proteccin de la salud pblica o la proteccin del medio ambiente, sin olvidar el indispensable mantenimiento de la diversidad biolgica a largo plazo y la consecucin de una ganadera duradera. Otros mbitos de trabajo empiezan a perlarse, entre ellos el de las vacunas de uso teraputico en lugar de preventivo y el de la vacunacin antitumoral. An es demasiado pronto, sin embargo, para hacer algn tipo de balance al respecto.
Profesor P.-P. Pastoret Jefe del Servicio de Publicaciones Organizacin Mundial de Sanidad Animal (OIE) 12 rue de Prony 75017 Pars Francia e-mail: pp.pastoret@oie.int Dr. A.A. Schudel Vicepresidente de la Comisin Cientca de la Organizacin Mundial de Sanidad Animal (OIE) Urraca 1366 Carilo (7167) Partido de Pinamar Provincia de Buenos Aires Argentina e-mail: alejandro.schudel@gmail.com Dr. M. Lombard Consultor en biologa 22 rue Crillon 69006 Lyon Francia e-mail: lombard.family@wanadoo.fr

Rev. sci. tech. Off. int. Epiz., 2007, 26 (2), 511-517

Vaccination guidelines: a bridge between official requirements and the daily use of vaccines
E. Thiry (1) & M.C. Horzinek (2)
(1) Virology and Viral Diseases, Department of Infectious and Parasitic Diseases, Faculty of Veterinary Medicine, University of Lige, B-4000 Lige, Belgium. Email: Etienne.thiry@ulg.ac.be (2) Veterinary Research Consult, Haydnlaan 15, 3723KE Bilthoven, the Netherlands

Summary Vaccination guidelines are non-compulsory recommendations which assist the veterinary practitioner to use vaccines efficiently. They complement the official information contained in the shortened form of the summary of product characteristics that is included in the package insert of the product. The aim of this article is to clarify the role of guidelines and examine how they can improve the use of vaccines in practical conditions. The development of vaccination guidelines is explained. Several issues are discussed: primary vaccination schedule; interference with maternally derived antibodies; duration of immunity; vaccination and ageing. Three guidelines dealing with the vaccination of cats against upper respiratory tract disease are compared, as an example. In conclusion, vaccination guidelines are essential tools to assist veterinarians in good vaccination practices. They fill the gap that exists between the official recommendations included in the regulations and the licensing dossiers and the daily use of the vaccines. Keywords Canine Feline Guidelines Vaccine Virus.

Introduction
Vaccination guidelines are non-compulsory recommendations which assist the veterinary practitioner to use vaccines efficiently. They complement the official information contained in the summary of product characteristics (SPC) that can be found in the package insert (an SPC is part of the documentation required for an authorised medicinal product) (21). However, although the vaccination schedule that appears in the package insert has been reviewed and approved by the licensing authorities, the practical use of the vaccine in the field can deviate from this official schedule. Furthermore, new scientific data can modify the vaccination approach, but the respective changes in the regulations can take several years before coming into force. In such cases, guidelines can serve as a bridge between official and unofficial recommendations for vaccine use. The aim of this article is to clarify the role of guidelines and the ways in which they can ensure that vaccines are used effectively in practical conditions. The situation in the

European Union (EU) will be taken as an example. The focus on pet animal vaccination is appropriate because it is less regulated by official policy and sanitary measures and more influenced by the behaviour of veterinarians and pet owners. The authors also undertake a comparison of several guidelines independently issued on the same subject. The role of vaccine producers and national and international authorities in the development and production of vaccines has been described and discussed by Jones et al. elsewhere in this issue of the Scientific and Technical Review (15). Therefore, this paper intends to explain the increasing role of expert committees in publishing vaccination guidelines in order to help the veterinarian in his/her daily practice.

Why are guidelines useful?


It is important to produce guidelines because the time between the initial development of a vaccine and its use in

512

Rev. sci. tech. Off. int. Epiz., 26 (2)

the field can be very long: several different players become involved and scientific knowledge develops, so the initial recommendations for use may no longer be the most appropriate. In addition, a vaccine is usually used as part of a complex vaccination programme and also in very diverse field situations, not all of which can be documented in the package insert of the product. There are two main sets of factors which can affect the use of vaccines; the first concerns the process of developing and marketing a vaccine and the second concerns the variety of different bodies that make recommendations about their production and use. The first is as follows: a) academic research provides scientific data about the identification of infectious agents, their incidence and the fulfilment of Kochs postulates. Pathogenesis and immunity are also studied and all this scientific information is used to stimulate applied research on the development of vaccines (6); b) the pharmaceutical company designs a vaccine and carries out experimental and field studies in order to meet the requirements of the regulatory authorities for quality, safety and efficacy of the vaccine in a given species, a given age and for a given vaccination schedule (6); c) the pharmaceutical company submits a dossier to obtain a marketing authorisation in the EU, which must comply with several regulations: Directives of the European Commission, monographs of the European Pharmacopoeia and other international and national rules (8, 9, 26); d) the veterinary immunological product will follow a centralised or a decentralised procedure for granting a marketing authorisation in the EU (3); e) when the marketing authorisation is granted, the SPC of the vaccine is available in the product information. It gives the main properties of the compound and the approved vaccination schedule as it will appear in the package insert. It is based on the results of experiments performed in experimental stations and field trials carried out by the company (21); f) the product is released on the market and the company informs veterinary practitioners through its marketing communication; this communication may deviate from the officially approved recommendations for vaccine use (21); g) veterinary practitioners may use the vaccination in ways that are noticeably different from the recommendations of the package insert, particulary if it is incorporated in a larger scheme involving other vaccines. The second set of factors relates to the official and nonofficial bodies that provide information and/or make recommendations about the production and use of vaccines. It can be described as follows: a) faculties and schools of veterinary medicine as well as other scientific institutes provide scientific knowledge;

b) pharmaceutical companies develop vaccines using their own scientific and technological data and provide their own technical information about the product; c) regulatory authorities publish rules and guidelines and are associated in the writing of regulations regarding the production of vaccines in order to guarantee the quality of the new veterinary immunological medicinal products (21). Examples of the main regulations that must be taken into account when preparing and using a new vaccine are the European Pharmacopoeia, European Commission Directives on medicinal products, and, in the United States of America, the Code of Federal Regulations (18). Given the time that it takes to develop legislation, these regulations are sometimes not based on the most recent scientific knowledge; d) various kinds of veterinary associations produce guidelines for the proper use of vaccines in field conditions (12); e) animal owners and breeder associations can also influence the use of vaccines by veterinary practitioners (12). The different sources of information regarding the use of vaccines explain why a discrepancy can be observed between the recommendations of the vaccine package insert and its use in the real conditions of veterinary practice. It is of primary importance that the vaccination schedules followed by the veterinary practitioners are the most efficacious ones even if this means that they do not strictly follow the recommendations of the package inserts.

How are guidelines developed?


The identified discrepancy between the official requirements for vaccine use and the practical use of vaccines in the field has been well recognised and whether this is acceptable or not should no longer be a matter of discussion. Therefore, there is a need to make a link between the authorised characteristics of the vaccine as described in the SPC and the package insert, the scientific knowledge available in published literature, and the requirements of veterinary practice. To this end, several committees have been created on the initiative of professional associations or experts in the field. These committees can be sponsored by pharmaceutical companies but, if they are, they must maintain full scientific independence from the sponsors. Their objective is to produce guidelines for the proper use of vaccines in the field and therefore to propose a coherent vaccination scheme which does not depend on the individual characteristics of licensed products.

Rev. sci. tech. Off. int. Epiz., 26 (2)

513

The authority of these committees is based on the experience of their members, who must be recognised experts in the field of infectious diseases, vaccination and internal medicine. Several committees are already at work, for example: the Feline Vaccine Advisory Panel of the American Association of Feline Practitioners (20); the European Advisory Board on Cat Diseases (7) and the German Federal Association of Veterinary Practitioners (2). Guidelines are issued after a long reviewing procedure which takes into consideration information from multiple sources such as: scientific literature, including scientific articles, conference proceedings and clinical reports providing results of controlled trials with a strong statistical significance vaccine package inserts, based on the marketing authorisation dossier and controlled experiments carried out by the company to fulfil the official requirements; the results of these trials are strictly limited to the product under testing information on the current vaccination practices of veterinarians (in order to take into account the experience gained in field conditions) information from evidence-based veterinary medicine, which combines the above-mentioned sources of different levels of evidence with a pragmatic approach to reach a decision (16). These committees write general and specific recommendations for good vaccination practices and remain independent of any brand or any particular commercial product. They can skip the slow process of the development of regulations and can therefore make recommendations on adapting vaccination programmes on a yearly basis according to the latest scientific knowledge. Scientific literature may provide new data on already licensed vaccines or very similar products, and these new data may differ from those contained in the marketing authorisation dossier. These committees can modify the vaccination approach and make new recommendations which are not strictly supported by either the vaccine marketing authorisation dossier or the official regulations. For example, they can provide advice on the vaccine valences to be included in a vaccination programme (the so-called core vaccines are recommended for all companion animals; non-core vaccines should be administered in specific risk conditions [20, 23]) and make recommendations on which infectious agents should be vaccinated against depending on the age, the condition and the environment of the animal (24). These committees can influence practitioners use of vaccines because they are very close to their needs.

Therefore, they are expected to become more and more important in the future. They could recommend alternative methods of using the vaccine that are not described in the SPC. This point should be taken into consideration by the official drug agencies and, in general, by all regulationsetting institutions. Regulations evolve quite slowly compared to the rapidly expanding needs of veterinary medicine and the evolution of scientific knowledge. It is likely that these committees could not only influence the vaccination habits of veterinary practitioners but also the official institutions like national and international drug agencies.

What issues do guidelines address?


Onset of immunity
Primary vaccination schedule
The SPC gives a minimum age for vaccination, which can, in practice, vary depending on several factors: the age at which the puppy or the kitten is shown to the practitioner for the first time; the behaviour and the environment of the animal; and the infectious agents that the vaccination is intended to combat. For instance, vaccination against canine parvovirus (CPV) can start early, at five to six weeks of age. The efficacy of the vaccination at this very young age is influenced by the level of maternally derived antibodies (MDA). Furthermore, the interval between injections cannot always be strictly applied due to practical constraints, and especially if the pet animal is not shown to the practitioner at the appropriate age. Certain combinations of vaccines will also mean that some deviation from the recommendations of the vaccine SPC are necessary. Guidelines are needed, therefore, to advise the veterinarian about the best decision to take in all the various circumstances encountered in practice.

Interference with maternally derived antibodies


At a very young age, MDA can significantly interfere with the efficacy of vaccination. This is a general statement since no individual testing is performed on a regular basis. In the future, diagnostic kits to measure the level of MDA interference will become available for some vaccines and are already in use, for canine distemper virus (CDV) and CPV (25). At a later age, duration of MDA depends on the level of immunity of the bitch or the queen, on the amount of colostrum uptake by the puppies or the kittens (differences between offspring of the same litter) and on the infectious agent.

514

Rev. sci. tech. Off. int. Epiz., 26 (2)

For example, vaccine package inserts propose a double vaccination schedule in cats at 9 and 12 weeks of age for both feline herpesvirus 1 (FeHV-1) and feline calicivirus (FCV). Knowing that MDA have a prolonged duration, especially for FCV, guidelines can recommend that a third round of vaccination be performed at 16 weeks of age (7, 20).

Combination of vaccines
Vaccine SPCs do not give any recommendation about the simultaneous or the sequential use of vaccines sold by different companies. The compatibility between vaccines is therefore a safety concern which is usually not addressed by the package inserts of the products and which needs specific guidelines. Both safety and efficacy must be considered since some vaccine valences may have interfering effects that decrease the efficacy of other vaccine components. In addition, it is the role of guidelines to provide the practitioner with coherent vaccination programmes (including the all important valences) and to propose modifications to the vaccine schedule to take account of various different factors such as specific epidemiological conditions or the type of animal being vaccinated, e.g. immunodepressed animals, animals in shelters, animals in breeding colonies, etc. (7, 24). Therefore, guidelines aim to provide comprehensive advice on vaccination programmes for both core and non-core vaccines, justifying their use under specific conditions.

Duration of immunity
In European vaccine SPCs, intervals between vaccinations are strictly regulated by analysing the results of protection experiments at the end of the claimed duration of immunity (8, 9).

First booster vaccination


The recommendation for the time of the first booster vaccination is based on the results of the duration of immunity experimental studies. It is most often one year after the primary vaccination. However, experts can suggest that cats and dogs be vaccinated earlier, six months after the primary vaccination (7). This statement is based on the fact that several animals are not properly vaccinated with the primary vaccination because of interference by MDA. This intermediate vaccination is designed to prevent dogs remaining unprotected over a one-year period. However, such modification remains compatible with the vaccine SPC because revaccination is carried out within the demonstrated duration of immunity.

Vaccination and ageing


Ageing is known to increase susceptibility to infectious diseases and decrease the efficacy of vaccinations, mainly due to a partial loss of the antibody response and a reduction of T helper lymphocyte activity (10, 19). A few data are available in domestic animals (10, 13). However, even the average age when ageing could modify the immune response is not known for companion animals and can only be extrapolated from the measurement of some physiological modifications. For example, while many cats begin to show clinically significant changes between the ages of 7 and 10, most of them start to be affected at 12 years of age (1). Several recommendations have already been made in the literature (22). For example, old dogs have high anti-rabies antibody titres before revaccination, meaning that a good vaccination schedule applied in the young and adult period can efficiently protect aged animals (13). More data should be made available by other studies in order to better substantiate the vaccination of aged animals. Indeed this matter is considered neither by regulations nor by pharmaceutical companies and is not properly included in vaccination guidelines due to this lack of knowledge (20).

Intervals between vaccinations


The current trend is to increase the interval between booster vaccinations. It is supported by several scientific publications (4, 5), but drug companies are required to carry out their own experiments and publish their own results before any modification can be made to the SPC, and as this is expensive and not in their interest they do not provide the necessary data and SPCs remain unchanged. Performing the first booster vaccination after one year is highly recommended (14). Taking CDV and canine adenovirus 2 (CAV-2) as examples, the protection conferred by attenuated vaccines is long-lasting and most likely longer than the duration of the immunity officially reported in the vaccine package inserts (14). One challenge study supports claims that there is a three-year duration of immunity after vaccination at 7 and 11 weeks of age (11). CDV and CAV-2 antibodies persist at least four years after vaccination (17). Other studies suggest that there may be an even longer duration of immunity against these viruses (23). This development is now apparent in the most recent versions of vaccine guidelines, where three-year intervals between rounds of vaccination are usually proposed for core vaccines (20).

Comparison of three guidelines


The three sets of guidelines compared here were all produced to guide the veterinary practitioner in the vaccination of cats. They were produced by the American

Rev. sci. tech. Off. int. Epiz., 26 (2)

515

Table I Comparison of the main features of guidelines on the vaccination of cats against the two viruses involved in upper respiratory tract disease (feline herpesvirus 1 and feline calicivirus) (2, 7, 20)
Criteria Primary vaccination schedule American Association of Feline Practitioners Administer two doses, 3 to 4 weeks apart European Advisory Board on Cat Diseases Two vaccinations, at an interval of 2 to 4 weeks Special recommendations for primary vaccination Begin as early as 6 weeks of age, then every 3 to 4 weeks until 16 weeks of age Vaccination is usually started at around 9 weeks of age; the second vaccination is administered 2 to 4 weeks later, with the third given around 12 weeks of age In high-risk situations, a third vaccination at 16 weeks should be considered Booster vaccination schedule duration of immunity A single dose is given one year following the last dose of the initial series Booster vaccinations every three years At annual intervals In cases of cats in low-risk situations, three-yearly intervals would be recommended. An informed decision should be made on the basis of a risk benefit analysis Special recommendations In unusual circumstances, if a cat is going to be placed in a known high-risk situation, an additional booster vaccination shortly before such risk is encountered may be considered Annual boosters are particularly important to cats that may be exposed to high-risk situations, e.g. boarding catteries Booster vaccinations every two years Vaccination at 15 months of age is included in the basic vaccination schedule German Federal Association of Veterinary Practitioners Two vaccinations, at an interval of 3 to 4 weeks Three vaccinations at 8, 12 and 16 weeks of age

Association of Feline Practitioners (20), the European Advisory Board on Cat Diseases (7) and the German Federal Association of Veterinary Practitioners (2), respectively (Table I). Only certain sections of these guidelines have been compared, namely, the sections that deal with vaccination programmes to protect cats against upper respiratory tract disease caused by FeHV-1 and FCV. The differences which can be observed between the three guidelines can be explained by at least two factors. First, the disease approach is different depending on the country or the region: the cultural aspects (e.g. with regard to the importance of animal welfare), the level of medical care of pet animals and the available vaccines. Secondly, there are so far no definite rules to control infectious diseases and the expert opinion, as reflected in the guidelines, is always based on a consensus between experts. This consensus opinion may differ depending on the composition of the expert panel.

that exists between the official recommendations issuing from the regulations and the licensing dossiers, and the daily use of vaccines. Vaccines are developed and produced with the aim of protecting animals against important, often lethal, infectious diseases. Therefore, good vaccination guidelines can improve the quality of the vaccine-induced protection in the field, provided they take into account both the latest scientific data and knowledge of the practice conditions.

Acknowledgements
We would like to offer our sincere thanks to our colleagues at the European Advisory Board on Cat Diseases, with whom we had many helpful discussions. We extend our thanks to Herv Poulet and Jean-Christophe Thibault (Mrial, Lyons), Anglique Zicola, Marie-Lys Van de Weerdt and Dominique Quatpers (Virology, Faculty of Veterinary Medicine, University of Lige) for stimulating discussions.

Conclusions
Vaccination guidelines are essential tools to help veterinarians use vaccination effectively. They fill the gap

516

Rev. sci. tech. Off. int. Epiz., 26 (2)

Les lignes directrices de la vaccination : un pont entre les besoins officiels et lutilisation quotidienne des vaccins
E. Thiry & M.C. Horzinek
Rsum Les lignes directrices de vaccination sont des recommandations non obligatoires qui aident le vtrinaire praticien utiliser les vaccins de manire efficace. Elles compltent linformation officielle prsente dans la notice dutilisation du produit et qui peut tre retrouve dans le rsum des caractristiques du produit. Lobjectif de cet article est de clarifier le rle des lignes directrices et leurs bnfices dans le cadre dune bonne utilisation des vaccins dans les conditions de la pratique. Le concept des lignes directrices de vaccination est dabord expliqu. Plusieurs points dintrt sont dvelopps : programme de primo-vaccination, interfrence par les anticorps dorigine maternelle, dure dimmunit, vaccination et vieillissement. Trois lignes directrices traitant de la vaccination du chat contre le coryza flin sont compares, titre dexemple. En conclusion, les lignes directrices sont des outils essentiels pour aider les vtrinaires dans de bonnes pratiques de vaccination. Elles forment un lien entre les recommandations officielles manant des rglements et des dossiers denregistrement et lusage quotidien des vaccins. Mots-cls Canin Flin Ligne directrice Vaccin Virus.

Las directrices de vacunacin como nexo entre los requisitos oficiales y el uso cotidiano de las vacunas
E. Thiry & M.C. Horzinek
Resumen Las directrices de vacunacin son recomendaciones de aplicacin optativa, que ayudan al veterinario a utilizar las vacunas de manera eficaz. Representan un complemento de la informacin oficial que figura en el resumen de caractersticas que acompaa al producto empaquetado. Tras aclarar la funcin de las directrices y explicar el modo en que pueden servir para mejorar el uso de las vacunas en la prctica, los autores exponen el proceso que lleva a su elaboracin. Entre los temas tratados figuran el calendario de vacunaciones primarias, la interferencia con anticuerpos de origen materno, la duracin de la inmunidad y la relacin entre vacunacin y envejecimiento. A modo de ejemplo comparan tres directrices de vacunacin de gatos contra la infeccin del tracto respiratorio superior. Las directrices de vacunacin, en suma, son una herramienta bsica para ayudar al veterinario a proceder correctamente al administrar una vacuna, pues vienen a tender un puente entre el uso cotidiano de las vacunas y las recomendaciones oficiales contenidas en los reglamentos o los expedientes de solicitud de licencia. Palabras clave Canino Directrices Felino Vacuna Virus.

Rev. sci. tech. Off. int. Epiz., 26 (2)

517

References
1. American Association of Feline Practitioners (AAFP) (2005). American Association of Feline Practitioners/Academy of Feline Medicine panel report on feline senior care. J. feline Med. Surg., 7, 3-32. 2. Anon. (2006). Deutsche Impfempfehlungen fr die Kleintierpraxis [German recommendations for vaccination in small animal practice]. Bundesverband praktizierender Tierrzte [German Federal Association of Veterinary Practitioners]. 3. Brunko P. (1997). Procedures and technical requirements in the European Union. In Veterinary vaccinology (P.-P. Pastoret, J. Blancou, P. Vannier & C. Verschueren, eds). Elsevier, Amsterdam, 674-679. 4. Coyne M.J., Burr J.H.H., Yule T.D., Harding M.J., Tresnan D.B. & McGavin D. (2001). Duration of immunity in dogs after vaccination or naturally acquired infection. Vet. Rec., 149, 509-515. 5. Coyne M.J., Burr J.H.H., Yule T.D., Harding M.J., Tresnan D.B. & McGavin D. (2001). Duration of immunity in cats after vaccination or naturally acquired infection. Vet. Rec., 149, 545-548. 6. Desmettre P. & Martinod S. (1997). Research and development. In Veterinary vaccinology (P.-P. Pastoret, J. Blancou, P. Vannier & C. Verschueren, eds). Elsevier, Amsterdam, 175-194. 7. European Advisory Board on Cat Diseases (2007). Guidelines on feline infectious diseases. Available at: www.abcd-vets.org/guidelines/ (accessed on 17 May 2007). 8. European Communities (EC) (2001). Directive 2001/82/EC of the European Parliament and of the Council of 6 November 2001 on the Community code relating to veterinary medicinal products. Off. J. Eur. Communities, L 311, 28/11/2001, 0001-0066. 9. European Communities (EC) (2004). Directive 2004/28/EC of the European Parliament and of the Council of 31 March 2004 amending Directive 2001/82/EC on the Community code relating to veterinary medicinal products. Off. J. Eur. Communities, L 136, 30/04/2004, 0058-0084. 10. Fermaglich D.H. & Horohov D.W. (2002). The effect of aging on immune responses. Vet. Clin. North Am. Equine Pract., 18 (3), 621-630. 11. Gore T.C., Lakshmanan N., Duncan K.L., Coyne M.J., Lum M.A. & Sterner F (2005). Three-year duration of .J. immunity in dogs following vaccination against canine adenovirus type-1, canine parvovirus, and canine distemper virus. Vet. Ther., 6, 5-14. 12. Hill R.E. (1997). Role of the consumer in the use and regulation of veterinary biological products. In Veterinary Vaccinology (P.-P. Pastoret, J. Blancou, P. Vannier & C. Verschueren, eds). Elsevier, Amsterdam, 744-762. 13. HogenEsch H., Thompson S., Dunham A., Ceddia M. & Hayek M. (2004). Effect of age on immune parameters and the immune response of dogs to vaccines: a cross-sectional study. Vet. Immunol. Immunopathol., 97, 77-85. 14. Horzinek M.C. (2006). Vaccine use and disease prevalence in dogs and cats. Vet. Microbiol., 117, 2-8. 15. Jones P.G.H., Cowan G., Gravendyck M., Nagata T., Robinson S. & Waits M. (2007). Regulatory requirements for vaccine authorisation. In Animal vaccination. Part 2: scientific, economic, regulatory and socio-ethical issues (P.-P. Pastoret, M. Lombard & A.A. Schudel, eds). Rev. sci. tech. Off. int. Epiz., 26 (2), 379-393. 16. Kastelic J.P. (2006). Critical evaluation of scientific articles and other sources of information: an introduction to evidence-based veterinary medicine. Theriogenology, 66, 534-542. 17. Mouzin D.E., Lorenzen M.J., Haworth J.D. & King V.L. (2004). Duration of serologic response to five antigens in dogs. J. Am. vet. med. Assoc., 224, 55-60. 18. Pastoret P.-P. & Falize F (1999). Viral veterinary vaccines. . Dev. biol. Stand., 101, 73-78. 19. Plowden J., Renshaw-Hoelscher M., Engleman C., Katz J. & Sambhara S. (2004). Innate immunity in aging: impact on macrophage function. Aging Cell, 3, 161-167. 20. Richards J.R., Elston T.H., Gaskell R.M., Hartmann K., Hurley K.F Lappin M.R., Levy J.L., Rodan I., Scherk M., ., Schultz R.D. & Sparkes A.W. (2006). The 2006 American Association of Feline Practitioners Feline Vaccine Advisory Panel Report. J. Am. vet. med. Assoc., 229, 1405-1441. 21. Roberts B. & Sanders A. (1997). Registration and marketing. In Veterinary vaccinology (P.-P. Pastoret, J. Blancou, P. Vannier & C. Verschueren, eds). Elsevier, Amsterdam, 243-254. 22. Schultz R.D. (1982). Theoretical and practical aspects of an immunization program for dogs and cats. J. Am. vet. med. Assoc., 181, 1142-1149. 23. Schultz R.D. (2006). Duration of immunity for canine and feline vaccines: a review. Vet. Microbiol., 117, 75-79 . 24. Thiry E. (2006). Clinical virology of the dog and cat. Editions du Point Vtrinaire, Paris. 25. Twark L. & Dodds W.J. (2000). Clinical use of serum parvovirus and distemper virus antibody titers for determining revaccination strategies in healthy dogs. J. Am. vet. med. Assoc., 217, 1021-1024. 26. World Organisation for Animal Health (OIE) (2004). Manual of Diagnostic Tests and Vaccines for Terrestrial Animals (mammals, birds and bees), 5th Ed. OIE, Paris.

Rev. sci. tech. Off. int. Epiz., 2007, 26 (2), 519-521

Recommendations of the OIE International Conference on the Control of Infectious Animal Diseases by Vaccination, Buenos Aires, Argentina, 13 to 16 April 2004
A.A. Schudel (1) & M. Lombard (2)
(1) Urraca 1366, Carilo (7167), Partido de Pinamar, Provincia de Buenos Aires, Buenos Aires, Argentina. Email: incoalex@hotmail.com (2) Consultant in Biologicals, 22, rue Crillon, 69006, Lyons, France

Introduction
In recent years, epidemics of emerging and re-emerging infectious animal diseases and zoonoses have often been controlled by implementing a policy of mass slaughter. However, this approach to disease control poses considerable ethical, technical, ecological and economic problems and there is a need for alternative control strategies. Therefore, in 2004, the World Organisation for Animal Health (OIE), the International Association for Biologicals (IABS), and the National Animal Health Service in Argentina (Servicio Nacional de Sanidad Animal) organised an international conference on the control of infectious animal diseases by vaccination. The conference was held in Buenos Aires, Argentina, from 13 to 16 April and was attended by over 300 scientists from some 50 countries.

Considering that:
1. Preventing the spread of animal disease through international trade of animals and animal products is one of the primary missions of the OIE. This is accomplished by establishing and updating international standards and guidelines that prevent the spread of pathogens while avoiding unjustified sanitary barriers. 2. The OIE standards for terrestrial animals are contained in the Terrestrial Animal Health Code (the Terrestrial Code) and the Manual of Diagnostic Tests and Vaccines for Terrestrial Animals (the Terrestrial Manual). 3. The collection, analysis and dissemination of veterinary scientific information is also one of the main missions of the OIE. 4. The standards developed by the OIE are recognised as international standards for animal health and zoonoses by the Agreement on the Application of Sanitary and Phytosanitary Measures (SPS) of the World Trade Organization (WTO). 5. Infectious animal diseases and zoonoses represent a major constraint to the maintenance and development of livestock and present a major threat to public health, to

Recommendations
The recommendations of the Conference attendees (1) support the position that veterinary vaccinology can help to build a better world, particularly given the quality of the veterinary biologics available today.

520

Rev. sci. tech. Off. int. Epiz., 26 (2)

the livelihood of farmers (especially in developing countries) and to national economies. 6. Over the past few years, the world has witnessed the emergence and re-emergence of several infectious animal diseases that have had a major impact on animal and human health. These have severely affected the economy in both developed and developing countries. 7. New scientific and technological knowledge about the prevention of many of these infectious diseases could contribute to the development of safer and more efficacious vaccines and diagnostic tests. 8. For ethical, ecological and economic reasons, it is no longer acceptable to control and eradicate disease outbreaks mainly by applying a policy of mass slaughter. 9. Vaccines help to improve animal health, public health, animal welfare, and agricultural sustainability, thus protecting the environment, maintaining biodiversity, and protecting consumers of animal products. 10. The OIE, being the international reference organisation for animal health and zoonoses has, wherever possible, incorporated into its standards, the best state of the art scientific knowledge on the use of appropriate diagnostic tests, disease prevention and control by vaccination. 11. Vaccination is without doubt one of the most useful measures which can be used to prevent animal diseases, and since its inception veterinary science has been strongly linked with the development of vaccinology. 12. Vaccination has proved that it is capable of preventing, controlling and eradicating disease, as exemplified by the control of smallpox, rinderpest and rabies. 13. Recent scientific advances in the diagnostic field, in particular the ability to differentiate vaccinated animals from those that have undergone pathogen replication as a result of natural challenge, have been recently incorporated into the Terrestrial Manual. Their implications have either already been reflected or are currently being discussed by the OIE in order to amend those sections of the Terrestrial Code which deal with disease control and recovery of disease-free status after an occurrence of a disease. 14. This International Conference is based on the valuable experience gained in the control and elimination of foot and mouth disease and other significant animal diseases and zoonoses through the use of vaccination. 15. The Conference is an opportunity for the exchange of the latest scientific information at the global level that will,

at the same time, assist in the evaluation and improvement of the current OIE standards and guidelines for better control of animal infectious diseases. 16. For this event, the OIE has acted in collaboration with the IABS, which has a long and valuable tradition in the dissemination of the most relevant scientific information on human and animal health.

Conference attendees of the OIE International Conference on the Control of Infectious Animal Diseases by Vaccination recommend the following:
1. Current approaches to animal disease prevention, control, and eradication by vaccination should be reviewed, wherever possible, according to the latest scientific information and incorporated into the OIE standards, recommendations and guidelines in order to facilitate both animal disease control and trade in animals and animal products. 2. Whenever feasible, the OIE should formulate vaccination policies as an alternative to the mass slaughtering of animals. 3. Greater emphasis should also be placed on the use of vaccination for the control of food-borne and other zoonotic diseases in animals in order to safeguard public health. This may include wildlife reservoirs of pathogens. 4. The OIE should develop and incorporate into its standards, recommendations and guidelines all relevant new information on diagnostic tests and the effective prevention, control and subsequent eradication of infectious animal diseases by vaccination. 5. The OIE should ask Member Countries to produce and use vaccines manufactured, tested and approved according to OIE standards and guidelines in order to improve their safety and potency. The same principles should apply to diagnostic tests. 6. The OIE should encourage Member Countries to strengthen the capacity of their antigen and vaccine banks to control emerging or re-emerging infectious diseases and zoonoses. 7. The OIE should recommend the development of more flexible marketing authorisation regulations in order to be able to adapt vaccines to the epidemiological situation in the field when facing pathogens with multiple serotypes, as exemplified by vaccines against human influenza (provided good epidemiological tools are in place).

Rev. sci. tech. Off. int. Epiz., 26 (2)

521

8. The OIE should support all research efforts in veterinary vaccinology and encourage funding agencies to put research into new veterinary biological products on their agendas and make it a priority. Public research is still needed to fill the gap where the private sector does not invest in new products due to the lack of foreseen investment return. 9. The OIE should provide, on official request from Member Countries, international standards and general information on the availability of antigen and vaccine banks. 10. The OIE should encourage other international and regional organisations to adopt a similar approach in the

control and eradication of other infectious animal diseases by vaccination. 11. The OIE and the IABS should disseminate all information concerning the International Conference to OIE Member Countries, international and regional organisations, and other stakeholders. 12. The OIE should refer the scientific information generated and discussed at this International Conference, including these recommendations, to the OIE Regional Commissions and relevant Specialist Commissions before submission for endorsement by the OIE International Committee.

References
1. Schudel A.A. & Lombard M. (eds) (2004). Control of infectious animal diseases by vaccination. Proc. OIE Conference, Buenos Aires, Argentina, 13-16 April 2004. Dev. Biol. (Basel), 119.

Rev. sci. tech. Off. int. Epiz., 2007, 26 (2), 523-525

Vaccines and OIE listed diseases


A.A. Schudel
Urraca 1366, Carilo (7167), Partido de Pinamar, Provincia de Buenos Aires, Buenos Aires, Argentina. Email: incoalex@hotmail.com

OIE activities
The activities of the World Organisation for Animal Health (OIE) in relation to the prevention and control of infectious animal diseases are primarily focused on the following areas: collection, analysis and dissemination of veterinary scientific information in a timely and transparent manner promotion of technical assistance and the strengthening of international cooperation for the enhancement of the control and eradication of animal diseases worldwide improvement of the legal framework and capacity building of official Veterinary Services, especially in developing countries promotion of safe trade in animals and animal products by eliminating the unjustified barriers that are sometimes put in place as a result of animal diseases, while minimising the entry of pathogens into importing countries. These activities are principally carried out through the development and application of standards, recommendations and guidelines. The OIE is regarded by the Sanitary and Phytosanitary Agreement of the World Trade Organization (WTO) as the only international organisation responsible for setting standards on animal diseases, including zoonoses.

reviews the request or problem and may seek expert opinion from other experts or Commissions or may decide to refer it to an ad hoc group of specialists for consideration and advice. The final advice or suggestion is reviewed by the Specialist Commission, which then proposes a draft text for an appropriate standard to be developed. This draft text is then circulated to all OIE Member Countries, which are given sixty days to comment on the proposal. The comments are considered by the Commission, which may decide to withdraw the text altogether or to make certain amendments to accommodate the comments received. The revised version is then submitted to the International Committee during the General Session in May for discussions and subsequent adoption. Once adopted, it becomes an OIE standard. The OIE standards are contained in the Terrestrial Animal Health Code (the Terrestrial Code), the Aquatic Animal Health Code (the Aquatic Code), the Manual of Diagnostic Tests and Vaccines for Terrestrial Animals (the Terrestrial Manual) and the Manual of Diagnostic Tests for Aquatic Animals (the Aquatic Manual) (1, 2, 3, 4).

Terrestrial Animal Health Code/ Aquatic Animal Health Code


One of the main purposes of the Codes is to reduce the risk of the spread of infectious animal diseases or agents through the international trade of animals and animal products while at the same time facilitating safe trade. Through its Early Warning Disease Information System, the OIE promptly contacts its Member Countries via emails, faxes or other means of communication to inform them (in the official languages of the OIE) of the occurrences of major animal diseases in the world. This allows countries at risk, i.e. neighbouring countries or those importing animals or animal products from the infected country to take appropriate measures to prevent the entry of pathogens into their territory. The Codes contain some generic chapters dealing with general issues (e.g. obligations and ethics in international

OIE standards
The procedure adopted by the OIE to create or update a standard is complex, but fully transparent. The request to create or update a standard can come from an OIE Delegate, a Specialist Commission, the OIE International Committee or any other authority following the outbreak of a disease or any significant epidemiological event. On receipt of the request, the OIE Central Bureau forwards it to the relevant Specialist Commission. The Commission

524

Rev. sci. tech. Off. int. Epiz., 26 (2)

trade, import risk analysis, principles of epidemiosurveillance with respect to specific diseases) and other chapters are devoted to specific diseases. Each disease chapter spells out the requirements for a country or zone to be considered free from the disease and lists the commodities that can be safely traded irrespective of the disease status. It also describes the health measures to be followed by importing countries when importing live animals and specific animal products from countries or zones where the disease exists. Importing countries are strongly urged to follow the recommendations prescribed by the OIE. However, it is the sovereign right of an importing country to apply stricter controls or a higher level of protection. In such cases, and in accordance with WTO rules, the importing country has to carry out a science-based risk assessment to demonstrate clearly why it wants to apply a higher level of protection. The OIE standards provide Member Countries with the appropriate guidance to apply an import risk analysis. As is clearly indicated in the OIE standards, the mere presence of a disease in an exporting country does not provide full justification for a total ban on animals or animal products from that country. Instead, a decision on whether or not to accept imports from infected countries should be based on the results of an import risk analysis. This is how, under certain conditions, countries with bovine spongiform encephalopathy (BSE) and foot and mouth disease (FMD) can continue to export meat and meat products. The OIE also has an in-house dispute settlement system to resolve technical or scientific conflicts that may arise among Member Countries. This mediation system is science-based and is quick, effective and less costly than the complex system in place at the WTO.

agreement between importing and exporting countries. The aims of the Terrestrial Manual are to facilitate the international trade of animals and animal products and also to contribute to the improvement of animal health services and the control of animal diseases by internationally recognised measures. It spells out the requirements for the manufacture and quality control of animal vaccines, including vaccines for 48 of the OIE listed diseases. The guidance on the requirements for vaccine manufacture lays emphasis on: a) b) c) Seed management: characteristics of the seed method of choice validation as a vaccine. Method of manufacture: in-process control batch control sterility safety potency duration of immunity stability preservatives precautions (hazards). Tests on final products: safety potency purity.

Manual of Diagnostic Tests and Vaccines


The OIE Terrestrial Manual is a companion volume of the Terrestrial Code. It also contains some generic chapters dealing with important issues such as: sampling methods quality management of veterinary laboratories principles of validation of diagnostic tests tests for sterility and freedom from contaminants human safety in veterinary vaccine manufacture biotechnology in the diagnosis of infectious diseases the role of official bodies in the international regulation of veterinary biologicals. Other chapters are devoted to specific diseases. These chapters include the prescribed tests for international trade and alternative tests that can be used under a bilateral

The Terrestrial Manual also describes the various diagnostic tests that are recognised not only to prove the presence of disease but also the tests that can be used to demonstrate freedom from infection or absence of virus circulation. In this respect, the diagnostic tests that have recently been developed for the diagnosis and control of certain diseases such as FMD, classical swine fever, Aujeszkys disease and avian influenza have received considerable attention from the OIE. The Terrestrial Manual recognises the application of marker vaccines and the accompanying diagnostic tests that serve to differentiate vaccinated from infected animals (DIVA tests). These tests have in a certain way revolutionised disease control in that in certain conditions they help avoid the mass killing of animals, which poses ethical, ecological and economic problems. They have also immensely contributed to international trade, because infected countries can use vaccination as an additional tool to control important animal diseases and continue with trade, provided of course that vaccinated animals are clearly identified and only healthy animals are vaccinated. These tests are also accepted by the OIE for the recovery of country disease status. In the case of FMD, non-structural

Rev. sci. tech. Off. int. Epiz., 26 (2)

525

protein (NSP) tests are now validated as screening tests and can be used to demonstrate absence of virus circulation. As regards Aujeszkys disease, the absence of gE antibodies in vaccinated pigs is accepted by the OIE as evidence of absence of virus circulation. Similarly, the OIE Ad hoc Group of experts on avian influenza has proposed that vaccination be used as an additional tool in the control of the disease, provided that DIVA tests are applied to demonstrate the absence of circulating virus. This should also enable trade of poultry and poultry products to continue from countries or zones which have experienced the disease.

will not compete with the existing vaccine banks but will complement them by including vaccines against diseases which are of particular relevance to developing countries. It is also the intention of the OIE, together with its world network of Reference Laboratories and international experts, to pursue research on DIVA tests to enable these tests to cover as many animal diseases as possible. To date there are no vaccines recommended in the Manual of Diagnostic Tests for Aquatic Animals.

Future developments
The OIE continues to work towards creating vaccine and antigen banks which will be useful to Member Countries on a national, regional and international basis. These banks

References
1. World Organisation for Animal Health (OIE) (2004). Manual of Diagnostic Tests and Vaccines for Terrestrial Animals, 5th Ed. OIE, Paris. 2. World Organisation for Animal Health (OIE) (2006). Aquatic Animal Health Code, 9th Ed. OIE, Paris. 3. World Organisation for Animal Health (OIE) (2006). Manual of Diagnostic Tests for Aquatic Animals, 5th Ed. OIE, Paris. 4. World Organisation for Animal Health (OIE) (2006). Terrestrial Animal Health Code, 15th Ed. OIE, Paris.

Vous aimerez peut-être aussi