Вы находитесь на странице: 1из 435

British Pharl11.

acopoeia
(Veterinary) 2016
British Pharmacopoeia (Veterinary)
2016

The British Pharmacopoeia Commission has caused this British


Pharmacopoeia 01eterinary) 2016 to be prepared under regulation
317(3)(b) of the Human Medicines Regulations 2012 and, in accordance
with regulation 317 (4), the Ministers have arranged for it to be published.
It has been notified in draft to the European Commission in accordance
with Directive 98/34/EEC.

The monographs of the Eighth Edition of the European Pharmacopoeia


(2013), as amended by Supplements 8.1 to 8.5, published by the Council
of Europe are reproduced either in this edition of the British
Pharmacopoeia 01eterinary) or in the associated edition of the British
Pharmacopoeia.

see General Notices

Effective date: 1 January 2016

see Notices

London: The Stationery Office


In respect of Great Britain:

THE DEPARTMENT OF HEALTH

In respect of N orthern Ireland:

THE DEPARTMENT OF HEALTH, SOCIAL SERVICES AND


PUBLIC SAFETY

© Crown Copyright 2015


Published by The Stationery Office on behalf of the Medicines and
Healthcare products Regulatory Agency (MHRA) except that:

European Pharmacopoeia monographs are reproduced with the permission


of the Council of Europe and are not Crown Copyright. These are
identified in the publication by a chaplet of stars.

This publication is a 'value added' producto If you wish to re-use the


Crown Copyright material from this publication, applications must be made
in writing, clearly stating the material requested for re-use, and the purpose
for which it is required. Applications should be sent to: Dr S Atkinson,
MHRA, 5th Floor, 151 Buckingham Palace Road, London SW1W 9SZ.

First Published 2015

ISBN 978 011 3230 006

British Pharmacopoeia Commission Office:


MHRA
151 Buckingham Palace Road
London SW1 W 9SZ
Telephone: +44 (0)20 3080 6561
E-mail: bpcom@mhra.gsi.gov.uk
Web site: http://www.pharmacopoeia.com

Laboratory:
British Pharmacopoeia Commission Laboratory
Queen' s Road
Teddington
Middlesex TW11 OLY
Telephone: +44 (0)20 8943 8960
E-mail: bpcrs@mhra.gsi.gov.uk
Web site: http://www.pharmacopoeia.com
Contents

NOTICES

PREFACE

BRITISH PHARMACOPOEIA COMMISSION

EXPERT ADVISORY GROUPS, PANELS OF EXPERTS AND


WORIZING PARTIES

CODE OF PRACTICE

MEMBERSHIP

BP Commission, Expert Advisory Groups, Panels of Experts, Working


Parties

STAFF

British Pharmacopoeia, BP Laboratory, Publisher


INTRODUCTION

Additions, Omissions, Technical Changes

GENERAL NOTICES
MONOGRAPHS

Medicinal and Pharmaceutical Substances

Formulated Preparations: General Monographs

Formulated Preparations: Specific Monographs

Immunological Products

Surgical Materials

INFRARED REFERENCE SPECTRA


APPENDICES

SUPPLEMENTARY CHAPTERS
INDEX

Vet-v
Notices

Monographs of the European Pharmacopoeia are distinguished by a chaplet


of stars against the title. The term European Pharmacopoeia, used without
qualification, means the eighth edition of the European Pharmacopoeia
comprising, unless otherwise stated, the main volume, published in 2013, as
amended by any subsequent supplements and revisions.

Patents In this Pharmacopoeia certain drugs and preparations have been included
notwithstanding the existence of actual or potential patent rights. In so far
as such substances are protected by Letters Patent their inclusion in this
Pharmacopoeia neither conveys, nor implies, licence to manufacture.

Effective dates N ew and revised monographs of national origin enter into force on
1 January 2016. The monographs are brought into effect under regulation
320(2) of the Human Medicines Regulations 2012.

Monographs of the European Pharmacopoeia have previously been


published by the European Directorate for the Quality of Medicines &
HealthCare in accordance with the Convention on the Elaboration of a
European Pharmacopoeia and have been brought into effect under
European Directives 200l/82/EC, 200l/83/EC and 2003/63/EC, as
amended, on medicines for human and veterinary use.

Vet-vi
Preface

The British Pharmacopoeia Commission has caused to be prepared under


regulation 317(3)(b) of the Human Medicines Regulations 2012 a
compendium, namely this British Pharmacopoeia (Veterinary) 2016, and, in
accordance with regulation 317(4), the Ministers have arranged for it to be
published. It is a companion volume to the British Pharmacopoeia 2016.

The British Pharmacopoeia (Veterinary) 2016 contributes significantly to


the quality control of materials used in the practice of veterinary medicine.
It contains publidy available, legally enforceable standards that provide an
authoritative statement of the quality that a product, material or artide is
expected to meet at any time during its period of use. The Pharmacopoeial
standards are designed to complement and assist the licensing and
inspection processes and are part of the overall system for safeguarding
animal and human health in the VK.

The British Pharmacopoeia Commission wishes to record its appreciation of


the services of all those who have contributed to the preparation of this
important work.

Vet-vii
British Phartnacopoeia
C01n111.ission

The British Pharmacopoeia Commission is appointed, on behalf of the


Secretary of State for Health, by the Department of Health's Appointments
Team who are responsible for appointments to all of the Advisory Bodies
appointed under the Human Medicines Regulations 2012.

Under the terms of the Human Medicines Regulations 2012, the duties of
the British Pharmacopoeia Commission are as follows:

(a) the preparation and publication of any new edition of the British
Pharmacopoeia [regulations 317(1) and 317(4)];

(b) the preparation and publication of any compendium containing


information relating to substances and articles which are or may be
used in the practice of veterinary medicine or veterinary surgery
[regulations 317(3)(b) and 317(4)];

(c) the preparation and publication of a list of names to be used as the


headings to monographs in the British Pharmacopoeia [regulations
318(1) and 318(2)];

(d) the preparation of any amendments to the aboye publications


[regulation 317(5)(a)].

Members of the British Pharmacopoeia Commission are appointed for a


renewable term of 4 years and, under the requirements laid down by the
Office of the Commissioner for Public Appointments, can serve for a
maximum of 10 years.

In order to ensure that the British Pharmacopoeia Commission fulfils its


duties under the Human Medicines Regulations 2012, the members also
have the following duties:

(1) to frame clear and unequivocal technical advice in order to


discharge the Commission's responsibilities both for the British
Pharmacopoeia, the British Pharmacopoeia (Veterinary) and British
Approved Names and as the national pharmacopoeial authority with
respect to the European Pharmacopoeia;

(2) to develop clear policies for the preparation and publication of the
British Pharmacopoeia and its related publications;

(3) to serve on one or more Expert Advisory Groups or Panels of


Experts of the BP Commission, usually in the position of Chair or
Vice-Chair;

(4) to approve new and revised text for inclusion in n~w editions of the
British Pharmacopoeia and British Pharmacopoeia (Veterinary);

(5) to approve new and revised names for inclusion in new editions of
British Approved Names and its annual supplements.

Vet-viii
In addition to the duties listed aboye, the Chair of the British
Pharmacopoeia Commission has the following additional duties:

(1) To chair all scheduled and unscheduled meetings;

(2) To carry out members appraisals in accordance with Department of


Health policies and timelines;

(3) To participate in the process to appoint/re-appoint members of the


British Pharmacopoeia Commission.

Vet-ix
Expert Advisory Groups, Panels
of Experts and W orking Parties

Members of Expert Advisory Groups, Panels of Experts and Working


Parties are appointed by the British Pharmacopoeia Commission.

The duties of the members are as follows:

(a) to collaborate in the preparation and revision of Monographs,


Appendices and Supplementary Chapters for inclusion in the British
Pharmacopoeia and British Pharmacopoeia 0leterinary);

(b) to collaborate in the preparation and revision of Monographs,


Methods and General Chapters of the European Pharmacopoeia;

(c) to review reports from the British Pharmacopoeia Laboratory in


terms of technical content and, where possible, provide independent
experimental data to assist in decision making;

(d) to collaborate in the preparation and revision of the list of names to


be used as titles for monographs of the British Pharmacopoeia and
British Pharmacopoeia (Veterinary).

Members of Expert Advisory Groups, Panels of Experts and W orking


Parties are usually appointed for a renewable term of 4 years.

Vet-x
Code of Practice

Members of the British Pharmacopoeia Commission and its supporting


Expert Advisory Groups, Panels of Experts and Working Parties are
required to comply with aCode of Practice on Declaration of Interests in
the Pharmaceutical Industry.

British Pharmacopoeia Commission

Chairs and members of the British Pharmacopoeia Commission are


required to make a full declaration of interests on appointment and
annually thereafter. They must also inform the BP Secretariat promptly of
any changes to these interests during the year. These interests are published
in the Medicines Advisory Bodies Annual Reports.

Relevant interests must be declared at meetings and are recorded in the


Minutes.

Expert Advisory Groups, Panels of Experts and Working Parties

Chairs and members are required to make a full declaration of interests on


appointment and to update the Secretariat if these interests change during
their term of office. A record is kept of those experts who have declared
specific interests, but these are not published.

Relevant interests must be declared at meetings and are recorded in the


Minutes.

Vet-xi
Mel11.bership of the British
Pharrnacopoeia COl11.tnission

The list below includes those members who served during the period 2014
to 2015.

Chair Professor K.evin M G Taylor BPharm PhD FRPharmS


Professor of Clinical Phannaceutics) UCL School of Pharmacy

Vice-Chair Professor Alastair Davidson BSe PhD FRPharmS


Visiting Professor of Pharmaceutical Sciences) University of Strathclyde

Professor Donald Cairns BSe PhD MRPharmS CSei CChem FRSC


Head: School of Pharmacy and Lije Sciences) Robert Gordon University)
Aberdeen

Mr Barry Capon CBE MA DL (Lay representative)


Former Non-executive Director) Noifolk and Suffolk NHS Foundation Trust

Dr Graham D Cook BPharm PhD MRPharmS


Senior Director) Process I(nowledge/Quality by Design) Pfizer

Mr Andrew Coulson BVetMed MSe MRCVS


Member of the Royal College of Veterinary Surgeons)· former Superintending
Inspector) Science & Research Group) The Home Office

Mr Christopher Goddard BSe DIS CSei EurChem CChem FRSC


Quality Control Technical Manager) Reciphan1'l Limited

Dr K.eith Helliwell BPharm PhD


Senior Technical Adviser) William Ransom & Son PLC

Dr Rodney L Horder BPharm PhD MRPharmS


Fon1'ler Divisional Vice President) Buropean Quality and Regulatory Strategy)
Abbott

Dr Gerard Lee BPharm PhD FRPharmS MRSC CChem


Former Group Manager) British Pharmacopoeia and Laboratory Services
(MHRA))· former Secretary & Scientific Director of the British Pharmacopoeia
Commission

Dr Brian R Matthews BPharm PhD FRPharmS FTOPRA MRI


Consultant on pharmaceutical and medical device regulatory affairs)· former Senior
Director) BC Registration) Alcon Laboratories

Professor John Miller MSe PhD MRSC CChem


Visiting Professor) Strathclyde Institute of Phan1'lacy and Biomedical Sciences)·
former Head of the BDQM Laboratory

Dr Ronald Torano BSe PhD MRSCCChem


Pharmacopoeial Intelligence and Advisory Specialist)· GlaxoSmithI<1ine

Vet-xii
Dr Lineoln Tsang BPharm LLB PhD FRSC FIBiol FRSA FRPharmS
Solicitor
Lije Sciences LawyerJ' PartnerJ Arnold & Porter LLP

Mrs Josephine Turnbull LLB (Lay representative)


Former Chair of Tees J Esk and Wear Valley NHS Foundation Trust

Dr Paul Varley BSe PhD


Vice President of Biopharmaceutical DevelopmentJ Medimmune Limited

Professor Elizabeth Williamson BPharm PhD MRPharmS


Former Professor of PharmacYJ University of Reading

Secretary and Scientific Dr Samantha Atkinson BSe MSe PhD MRSC


Director Visiting Fellow J University of Reading

Vet-xiii
Metnbership of Expert Advisory
Groups, Panels of Experts and
W orking Parties

The Commission appointed the foIlowing Expert Advisory Groups, Panel s


of Experts and Working Parties to advise it in earrying out its duties.
Membership has ehanged from time to tÍlne; the lists below include aIl who
have served during the period 2014 to 2015.

EXPERT ADVISORY GROUPS


ABS: Antibiotics R L Horder (Chair), G Cook (Vice-Chair), P EIlis, E Flahive, A Gibson,
V Jaitely, A Livingstone, W Mann, J Miller, N Thomas, B White,
1 R Williams
BIO: Biological and L Tsang (Chair), P Varley (Vice-Chair), L Bisset*, A F Bristow*, C Burns,
Biotechnological D H Calam, IZ Chidwiek*, A Cook*, J Cook*, L Findlay*, S Gill,
Products E Griffiths, C Jones*, A lZippen*, B Patel, A M Pickett*, T Pronee,
L Randon, 1 Rees*, S Sehepelmann*, D Sesardie, P Sheppard,
P Stiekings*, W J Tarbit, A H Thomas, R Thorpe, M Wadhwa*
(Corresponding members A Onadipe, J N A Tettey)
HCM: Herbal and E WiIliamson (Chair)) L A Anderson (Vice-Chair), P Anderson, A Bligh,
Complementary T Chapman, A Charvill, S Gibbons, IZ Helliwell, P Hylands, C Leon,
Medicines R Middleton, A C Moffat, B Moore, M Pires, M Rowan, K Strohfeldt-
Venables, J Sumal*, P Viner, C Welham, C Wright, K Zhao
(Corresponding lnembers SS Handa, A Krauss, Z-T Wang)
MC1: Medicinal A G Davidson (Chair), D Cairns (Vice-Chair), M Ahmed, J C Berridge,
Chemicals M Broughton, A J Caws, P Fleming, A James, V Loh, W J Lough,
D Malpas
MC2: Medicinal G Cook (Chair), C T Goddard (Vice-Chair), M Cole, J Cowie, D Edwards,
Chemicals A Gibson, J Lim, J Miller, P Murray, J Qiu, A Ruggiero, M Turgoose,
NWynne
(Corresponding members M Brits, W Sherwin)
MC3: Medicinal V Fenton-May (Chair), E Williamson (Vice-Chair), M Almond, S Arkle,
Chemicals J Beaeh, J Beaman, C T Goddard, P Hampshire, W K L Pugh,
B Raekstraw, R Torano, M Tubby, 1 R Williams
NOM: Nomenclature J K Aronson (Chair), L Tsang (Vice-Chair), M Ahmed, D Mehta,
G P Moss, R Thorpe
(Corresponding meJ1'zbers R G Baloeeo Mattavelli, E M Cortés Montejano,
J S Robertson)
PCY: Pharmacy R L Horder (Chair), B R Matthews (Vice-Chair), M Ahmed*, M Aulton,
E Baker, J Beaeh, N Broad, G Davison, G Eecleston, D Elder, B GraneIl-
Villen, J Lim*, R Lowe, J MaeDonald, J F MeGuire, T Púrewal,
L Randon, IZ Taylor
(Corresponding wlelnber J Churehill)
* Denotes a specialist member.

Vet-xiv
ULM: Unlicensed M G Lee (Chair), V Fenton-May (Vice-Chair), G Bennett, S Branch,
Medicines D Caulfield, A Charvill, W Goddard, N Hussain, S Jones, M A Oldcorne,
N J Precious, J Rothwell, M Santillo, J Smith, A Sully, P Weir

PANELS OF EXPERTS
BLP: Blood K Chidwick, A R Hubbard, J More, P Varley
Products

CX: Excipients B R Matthews (Chair), C Mroz (Vice-Chair), E Anno, C Cable,


R Cawthorne, W Cook, D Deutsch, N Hussain, M 1 Robertson, K Slevin

IGC: Inorganic and C T Goddard (Chair), M Almond, S Atherton, S Boland, A C Cartwright,


General Chemicals D Caulfield, P Henrys, G Lay, D Malpas, C Mroz, D Riches

MIC: Microbiology V Fenton-May (Chair), S Denyer, D P Hargreaves, B R Matthews,


P Newby

RAD: Radioactive J Ballinger, J Brain, D Graham, S R Hesslewood, G Inwards, P Maltby,


Materials R D Pickett, R Smith, S Waters

VET: Veterinary E Williamson (Chair), A Coulson (Vice-Chair), A Cairns, S Cockbill,


Medicines D Evans, E Flahive, P Lees, B Ward

VIP: Veterinary A M Brady, R Banks, IZ Redhead, J Salt, P W Wells, R Woodland


Immunological
Products

WORKING PARTIES
AQbD: Analytical G Cook (Chair), S Brown, S Ellison, M Hanna-Brown, S Jones,
Quality by Design D Makohon, P N ethercote, E Razzano
(Corresponding member K Barnett)

DNA: Identification K Helliwell (Chair), J Hawkins, E Mee, A Slater, E Williamson


Techniques

MCS: Microscopy E Williamson (Chair), R Arroo, R Fleck, K Helliwell, IZ MacLellan Gibson

Vet-xv
Current British Pharrnacopoeia
Staff

Secretariat M Vallender (Editor-in-Chief)

S Young (Head of Analytical Science)

M Barrett, H Corns, P Crowley, A Evans, J Francomb, A Gibb, P Holland,


G Li-Ship, R A Pask-Hughes, C Pitt, J Pound, F J Swanson, M Whaley

NIBSC-based StatI C Howard, C Lockie-Williams

Administrative B F Delahunty, W Jeffries, J Paine

1509001
F527268

Vet-xvi
Current British Phartnacopoeia
Laboratory Staff

IZ Courtney (Laboratory Manager)


A Ciesluk, D Colmer, J Elliot, C Galdino, P Gallagher, S Humphries,
R Mannan, C Marcolan, A Murphy, M Nanasi, A Panchal, E Sanderson,
N Vadukal, A Vasilaki, M Wallis, S Wilson

/509001
FS 27613

Vet-xvii
Current Staff of the Publisher of
the British Phartnacopoeia

J Hook (MD Public Sector EMEA)

A Prince (Client Services Director)

N Billington (Client Services Manager)

A Hughes (Account Manager)

e Hackett (Proy'ect Manager)


P Allard, A Dampier, M Grant, A Hood, S Page, M Parka, N Pope,
M Rainbird, A Ray, P Relfe, e Spary, J Stoker, 1 Webb, K Williams

1509007
F522428

Vet-xviii
2016 Introduction Vet-xix

Introduction

Triennial Review of the British Pharmacopoeia Commission

The Department of Health conducted a Triennial Review of the British


Pharmacopoeia Commission (BPC) to provide assurance to the Department
and the public that the functions of the Commission are required and that
it is operating effectively. The review of the BPC was undertaken in two
stages, the first examined the functions and form of the BPC and the
second examined the efficiency, governance and performance of the BPC.
The recommendation from the review of form and function is that "the
BP Commission should continue to deliver its existing functions as an
Advisory Non-Departmental Public Body". This model facilitates the
BP Commission to be in a position close to the Medicines and Healthcare
products Regulatory Agency whilst being independent of it and facilitates
harnessing potential information, communication, laboratory and expertise
synergles.
The evidence gathered from the review of efficiency, governance and
performance suggested that "the BP commission operates efficiently, is
mostly compliant with the principIes of good corporate governance and is
considered to be a leading pharmacopoeia. In particular, stakeholders
highlighted the BP Commission's innovative work, the dedication and
expertise of members and the Secretariat, industry engagement and
transparency" .
The Triennial Review report, published in March 2015, makes a number
of minor recommendations which will be taken forward by the BPC
Secretariat. The full report of the Review can be found on the website
WWW.gov. uk/governmenti consul ta tions/british-pharmacopoeia-commission-
triennial-review.

British Pharmacopoeia (Veterinary) 2016

The British Pharmacopoeia (Veterinary) 2016 supersedes the British


Pharmacopoeia (Veterinary) 2015. The British Pharmacopoeia Commission
has caused this edition to be prepared under regulation 317 (3) (b) of the
Human Medicines Regulations 2012 and, in accordance with regulation
317 (4), the Ministers have arranged for it to be published. This empowers
the British Pharmacopoeia Commission to prepare a compendium
containing information relating to substances, combinations of substances
and articles (whether veterinary medicinal products or not) which are or
may be used in the practice of veterinary medicine or veterinary surgery.
Under the terms of the Human Medicines Regulations 2012 it is an offence
to sell or supply a medicinal product in thé United Kingdom that is the
subject of a monograph in the Pharmacopoeia if that product does not
comply with the standards specified in the monograph.

The British Pharmacopoeia (Veterinary) 2016 is published as a companion


volume to the British Pharmacopoeia 2016 and thus contains only those
monographs for substances and preparations used exclusively or
predominantly in veterinary medicine within the United I<ingdom, together
Vet-xx Introduction 2016

with such additional texts as are necessary to support them. It therefore


follows that any reference to a monograph, appendix or reagent not
contained within this edition is to be construed as a reference to the said
monograph, appendix or reagent contained within the British
Pharmacopoeia 2016.

This edition, together with the British Pharmacopoeia 2016, contains all the
monographs reproduced from the 8th Edition of the European
Pharmacopoeia as amended by Supplements 8.1 to S.S. Users of the British
Pharmacopoeia and British Pharmacopoeia (Veterinary) therefore benefit by
finding within these two compendia all current pharmacopoeial standards
for veterinary medicines used within the United Kingdom.

Effective Date The effective date for this edition is 1 January 2016.

National monographs omitted from this or earlier editions of the British


Pharmacopoeia remain effective in accordance with Regulation 252(2)(c) of
the Human Medicines Regulations 2012.

Implementation dates regarding European Pharmacopoeia publications are


provided in Supplementary Chapter IV B: Dates of Implementation.
European Pharmacopoeia monographs are identified by a chaplet of stars
alongside the title.

Additions A list of monographs included for the first time in the British
Pharmacopoeia (Veterinary) 2016 is given at the end of this Introduction.
It includes 2 new monographs reproduced from the Sth Edition of the
European Pharmacopoeia as amended by Supplements 8.1 to 8.5.

Omissions One monograph, that for Fenthion, has been omitted from the British
Pharmacopoeia (Veterinary) 2016.

Infrared Reference As with the previous edition, the reference spectra are placed in alphabetical
Spectra order within this edition.

European Co-operation Agreement


Pharmacopoeia
As a consequence of the Co-operation Agreement with the European
Directorate for the Quality of Medicines & Healthcare (EDQM) of the
Council of Europe, the British Pharmacopoeia Commission is pleased to
note the integration of European Pharmacopoeia texts for the British
Pharmacopoeia 2015 in-year online updates and for this edition of the
British Pharmacopoeia.

All monographs of the Sth Edition of the European Pharmacopoeia, as


amended by Supplements 8.1 to S.5, which are used in veterinary practice
but not normally in human medicine in the U nited Kingdom, are
reproduced in this edition of the British Pharmacopoeia (Veterinary). Each
of these monographs is signified by a chaplet of stars alongside its title.
Additionally, reference to the European Pharmacopoeia monograph number
is included immediately below the title in italics in the form 'Ph. Eur.
111onograph xxxx'. Where the title in the British Pharmacopoeia (Veterinary)
is different from that in the European Pharmacopoeia, an approved
synonym has been created (see Appendix XXI B (Vet)) and the European
Pharmacopoeia title is included before the monograph number. The entire
2016 Introduction Vet-xxi

European Pharmacopoeia text is delineated by two horizontallines bearing


the symbol 'Ph. Eur.'.

The European Pharmacopoeia texts have been reproduced in their entirety


but, where deemed appropriate, additional statements of relevance to UIZ
usage have been added (e.g. action and use statement, a list of British
Pharmacopoeia (Veterinary) preparations). It should be noted, however,
that in the event of doubt of interpretation in any text of the European
Pharmacopoeia, the text published in English under the direction of the
Council of Europe should be consulted.

Correspondence between the general methods of the European


Pharmacopoeia and the appendices of the British Pharmacopoeia
(Veterinary) is indicated in each appendix. A list is also provided at the
beginning of the appendices section. This provides a full listing of the
European Pharmacopoeia method texts with their British Pharmacopoeia
and British Pharmacopoeia (Veterinary) equivalents.

Pharmacopoeial Pharmacopoeial requirements for articles used in veterinary medicine are


Requirements established on the same basis as those used in human medicine. A proper
understanding of the basis upon which these requirements are established is
essential for their application and advice is provided within the General
Notices of the British Pharmacopoeia (Veterinary) and the Supplementary
Chapters of the British Pharmacopoeia. It should be noted that no
requirement of the Pharmacopoeia can be taken in isolation. A valid
interpretation of any particular requirement depends upon it being read in
the context of (i) the monograph as a whoIe, (ii) the specified method of
analysis, (iii) the relevant General Notices and (iv) where appropriate, the
relevant General Monograph(s).

Where a preparation that is the subject of a monograph in the British


Pharmacopoeia is supplied for use in veterinary medicine, the standards of
the British Pharmacopoeia apply, unless otherwise justified and authorised.
Attention is drawn to the Notice permitting the designation British
Pharmacopoeia (Veterinary) [BP (Vet)] to be used in place of the
designation British Pharmacopoeia [BP] where a preparation complying
with the British Pharmacopoeia is supplied for use in veterinary medicine
with the approval of the competent authority.

Expert Advisory Groups; Panels of Experts; Working Parties

The British Pharmacopoeia Commission has reviewed the membership of


all of its Expert Advisory Groups, PaneIs of Experts and Working Parties.
The tenure of appointed members runs for a period of 4 years from
1 January 2015 and is renewable. Criteria for the appointment of experts
are available on the consolidated website (www.pharmacopoeia.com).

Panels of Experts The British Pharmacopoeia Commission has changed


the status of the former Working Party on Excipients to a Panel of Experts
based on its current worldoad.

Working Parties The British Pharmacopoeia Commission has established


3 new Working Parties as follows. Two of the Working Parties, DNA:
Identification Techniques and MCS: Microscopy, relate to the programme
of work on herbal analysis. The third Working Party, AQbD: Analytical
Vet-xxii Introduction 2016

Quality by Design, has been established to support the work of the joint
Medicines and Healthcare products Regulatory Agency (MHRA)-BP
Analytical Quality by Design project. The joint project examines the
application of Quality by Design concepts to analytical methods and
includes representation from the MHRA Licensing Division, the Good
Manufacturing Practice Inspectorate and industry.

eode of Practice Members of the British Pharmacopoeia Commission and its supporting
Expert Advisory Groups, Panel s of Experts and Working Parties are
required to comply with aCode of Practice on Declaration of Interests in
the pharmaceutical industry. Details of the Code are published on the
website (www.pharmacopoeia.com).

Websites British Pharmacopoeia Websites

The British Pharmacopoeia websites, www.pharmacopoeia.co.uk and


www.pharmacopoeia.com. have been consolidated and the new website,
https://www.pharmacopoeia.com. is now available, containing information
relating to the British Pharmacopoeia and allowing subscribers to access the
British Pharmacopoeia 2016 and British Pharmacopoeia (Veterinary) 2016
online and British Approved Names publications.

The consolidated website provides new and improved functionality together


with greater access to improved BP-supporting resources. These resources
include freely available example chromatograms, herbal micrographs and
omitted surgical material monographs for all users. Draft new and revised
texts of the BP will continue to be placed on the website in an improved
and more accessible way.

Subscribers to the BP online will find that these resources will also be
linked with relevant texts and directly accessible from the BP online
contento

Access to previous editions of the BP will be available as a BP archive


product for purchase by new and existing BP online subscribers. The
content of the archive will start from the BP 2014 onwards and will grow
year-on-year as superseded editions are added to the archive.

Improvements and new features have also been made to the BPCRS
catalogue and ordering facility. U sers will be able to receive notifications on
the status of out-of-stock items and view order histories. BPCRS products
will also be linked with relevant BP monographs and subscribers to the BP
online will be able to purchase these directly from the BP online contento
BPCRS customers will continue to be able to make purchases through
invoice or credit card orders.

An email subscription feature will allow users to keep abreast with BP news
and BPCRS updates.

In line with a policy of continuous improvement, users of the newly


consolidated www.pharmacopoeia.com website are invited to provide the
Secretariat with feedback on their experience.
2016 Introduction Vet-xxiii

European Pharmacopoeia Websites

https://extranet.edqm.eu/publications/recherches sw.shtml For those texts


reproduced from the European Pharmacopoeia, the EDQM website
provides access to a database (the Knowledge database) containing
information of various sorts related to monographs and intended to
facilitate their proper use. Information is provided on chromatographic
columns used in monograph development, suppliers of reagents and
equipment that may be difficult to find for sorne users, the status of
monographs (in development, adopted, published, under revision), revisions
of the monographs on a historical basis, beginning from the 5th Edition of
the European Pharmacopoeia as well as other useful information.

https://pharmeuropa.edqm.eu/home The European Pharmacopoeia Forum,


Pharmeuropa, is published quarterly as an aid for the elaboration of
monographs and as a vehiele for information on pharmacopoeial and related
matters. Pharmeuropa is available as a free on-line publication.

Forward Look Electronic Updates The British Pharmacopoeia 2016 online updates will
be published on the website, www.pharmacopoeia.com. to enable users to
keep up to date with monographs published in the European
Pharmacopoeia. These updates will be integrated annually with the
publication of the main edition of the British Pharmacopoeia.

Monograph Development The British Pharmacopoeia Commission will


continue to collaborate with stakeholders to develop BP monographs for
veterinary medicines, to ensure that the BP (Veterinary) continues to
provide authoritative quality standards for veterinary preparations.

Acknowledgements The British Pharmacopoeia Commission is greatly indebted to the members


of its Expert Advisory Groups, Panels of Experts, in particular, the Panel of
Experts on Veterinary Medicines and W orking Parties for their dedicated
enthusiasm and assistance in the preparation of this edition. The
Commission is especially indebted to the advice, commitment and
leadership of Professor Peter Lees, former Vice-Chair of the Veterinary
Medicines Panel.

Close co-operation has continued with many organisations at home and


overseas. These inelude the Veterinary Medicines Directorate, the
Medicines and Healthcare products Regulatory Agency, the National Office
of Animal Health, the Association of the British Pharmaceutical Industry,
the European Pharmacopoeia Commission and the European Directorate
for the Quality of Medicines & HealthCare, the Therapeutic Goods
Administration (Australia), the Health Products and Food Branch of Health
Canada, the United States Pharmacopeia, the Quality Assurance and
Safety: Medicines Department of the World Health Organization (WHO)
and the Health Sciences Authority of Singapore.

The British Pharmacopoeia Commission wishes to thank the European


Directorate for the Quality of Medicines & HealthCare for their support
and assistance in the reproduction of the European Pharmacopoeia texts
and monographs. The British Pharmacopoeia Coínmission acknowledges
the importance of the work of the European Pharmacopoeia (Ph. Eur.)
Commission and its Groups of Experts and Working Parties. The British
Pharmacopoeia Commission is also grateful for the generous contribution
Vet-xxiv Introduction 2016

by the UK experts to the work of the Groups of Experts and W orking


Parties of the European Pharrnacopoeia Cornrnission.

The British Pharrnacopoeia is grateful for the contribution of Ms Harjit


Jagpal for adrninistrative and events rnanagernent.
The British Pharrnacopoeia Cornrnission also acknowledges and appreciates
the advice of the publishing tearn at The Stationery Office, in particular,
Ms Nichola Billington, Mr Colin Hackett, Mr Paul Allard, Mr Paul Relfe
and Mr Ian Webb, in the production of this edition.

The British Pharrnacopoeia Cornrnission is grateful for the cornrnitrnent and


contribution of the website tearn at The Stationery Office, in particular,
Mr Terry Blake, Mr Andrew Hood and Mr Vinod Sathyarnoorthy, in the
consolidation of the two BP websites.

Additions The following rnonographs of the British Pharrnacopoeia (Veterinary) 2016


were not included in the British Pharrnacopoeia (Veterinary) 2015.

Medicinal and Pharmaceutical Substances


Sulfadirnethoxine Sodiurn*
Triclabendazole*

Omissions The following rnonographs of the British Pharrnacopoeia (Veterinary) 2015


are not included in the British Pharrnacopoeia (Veterinary) 2016.

Medicinal and Pharmaceutical Substances


Fenthion

Technical Changes The following rnonographs in the British Pharrnacopoeia (Veterinary)


2016 have been technically arnended since the publication of the British
Pharrnacopoeia (Veterinary) 2015, or have had a significant editorial
change. This list does not include revised rnonographs of the European
Pharrnacopoeia. An indication of the nature of the change or the section of
the rnonograph that has been changed is given in italic type in the right
hand colurnn.

Medicinal and Pharmaceutical Substances

Piperonyl Butoxide Definitian; Identificatian; Related substances;


Assay; Impurities

* denotes a monograph of the European Pharmacopoeia


Monographs

Medicinal and Phartnaceutical


Substances
2016 General Monographs Vet-37

The manufacture of active substances must take place under


MEDICINAL AND PHARMACEUTICAL conditions of good manufacturing practice.
SUBSTANCES The provisions of general chapter 5.10 apply to the control of
impurities in substances for phannaceutical use.
Whether or not it is specifically stated in the individual
monograph that the substance for phannaceutical use:
Substances -- is a recombinant protein or another substance obtained as
a direct gene product based on genetic modification,
tor Pharmaceutical Use where applicable, the substance also complies with the
(Ph. Eur. monograph 2034) requirements of the general monograph Products of
PhElf _____________________________________________ recombinant DNA technology (0784);
- is obtained from animals susceptible to transmissible
DEFINITION spongiform encephalopathies other than by experimental
Substances for phannaceutical use are any organic or challenge, where applicable, the substance also complies
inorganic substances that are used as active substances or with the requirements of the general monograph Products
excipients for the production of medicinal products for with nsk of transmitting agents of animal spongifonn
human or veterinary use. They may be obtained from natural encephalopathies (1483);
sources or produced by exu'action from raw materials, - is a substance derived from a fennentation process,
fennentation or synthesis. whether or not the micro-organisms involved are modified
This general monograph does not apply to herbal drugs, by traditional procedures or recombinant DNA (rDNA)
herbal drugs for homoeopathic preparations, herbal drug technology, where applicable, the substance also complies
preparations, extracts, or mother tinctures for homoeopathic with the requirements of the general monograph Products
preparations, which are the subject of separate general of fennentation (1468).
monographs (Herbal drugs (1433), Herbal drugs for If solvents are used during production, they are of suitable
homoeopathic preparations (2045), Herbal drug quality. In addition, their toxicity and their residuallevel are
preparations (1434), Extracts (0765), Mother tinctures for taken into consideration (5.4). If water is used during
homoeopathic preparations (2029)). It do es not apply to raw production, it is of suitable quality.
materials for homoeopathic preparations, except where there If substances are produced or processed to yield a certain
is an individual monograph for the substance in the non- form or grade, that specific fonn or grade of the substance
homoeopathic part of the Phannacopoeia. complies with the requirements of the monograph. Certain
Where a substance for phannaceutical use not described in functionality-related tests may be described to control
an individual monograph of the Phannacopoeia is used in a properties that may influence the suitability of the substance
medicinal product prepared for the special needs of and subsequently the properties of dosage forms prepared
individual patients, the need for compliance with the present from it.
general monograph is decided in the light of a risk Powdered substances May be processed to obtain a certain
assessment that takes account of the available quality of the degree of fineness (2.9.35).
substance and its intended use.
Compacted substances Are processed to increase the particle
Where medicinal products are manufactured using size or to obtain partic1es of a specific fonn and/or to obtain
substances for phannaceutical use of human or animal origin, a substance with a higher bulk density.
the requirements of chapter 5.1.7. Viral safety apply.
Coated active substances Consist of partic1es of the active
Substances for phannaceutical use may be used as such or as substance coated with one or more suitable excipients.
starting materials for subsequent formulation to prepare
Granulated active substances Are partic1es of a specified size
medicinal products. Depending on the fonnulation, certain
and/or form produced from the active substance by
substances may be used either as active substances or as
granulation directly or with one or more suitable excipients.
excipients. Solid substances may be compacted, coated,
granulated, powdered to a certain fineness, or processed in If substances are processed with excipients, these excipients
other ways. A monograph is applicable to a substance comply with the requirements of the relevant monograph or,
processed with an excipient only where such processing is where no such monograph exists, the approved specification.
mentioned in the definition section of the monograph. Where active substances have been processed with excipients
Substance for pharmaceutical use of special grade Unless to produce, for exampk, coated or granulated substances, the
otherwise indicated or restricted in the individual processing is carried out under conditions of good
monographs, a substance for phannaceutical use is intended manufacturing practice and the processed substances are
for human and veterinary use, and is of appropriate quality regarded as intermediates in the manufacture of a medicinal
for the manufacture of all dosage forms in which it can be producto
used. CHARACTERS
Polym01phism Individual monographs do not usually specify The statements under the heading Characters
ctystalline or amorphous forms, unless bioavailability is (e.g. statements abóut the solubility or a decomposition
affected. All forms of a substance for phannaceutical use point) are not to be interpreted in a strict sense and are not
comply with the requirements of the monograph, unless requirements. They are given for infonnation.
otherwise indicated. Where a substance may show polymorphism, this may be
PRODUCTION stated under Characters in 9rder to draw this to the attention
Substances for phannaceutical use are manufactured by of the user who may have to take this characteristic into
procedures that are designed to ensure a consistent quality consideration during formulation of a preparation.
and comply with the requirements of the individual
monograph or approved specification.
Vet-38 General Monographs 2016

IDENTIFICATION The requirements aboye do not apply to biological and


Where under Identification an individual monograph biotechnological products, oligonuc1eotides,
contains subdivisions entitled 'First identification' and radiopharmaceuticals, products of fermentation and semi-
'Second identification', the test or tests that constitute the synthetic products derived therefrom, to crude products of
'First identification' may be used in all circumstances. animal or plant origin or herbal products.
The test or tests that constitute the 'Second identification' For active substances in a new application for a medicinal
may be used in pharmacies provided it can be demonstrated product for human use, the requirements of the guideline on
that the substance or preparation is fully traceable to a batch the limits of genotoxic impurities and the corresponding
certified to comply with aH the other requirements of the questions and answers documents published on the website
monograph. of the European Medicines Agency (or similar evaluation
Certain monographs give two or more sets of tests for the principIes for non-European Union member states) must be
purpose of the first identification, which are equivalent and followed.
may be used independently. One or more of these sets Residual solvents
usually contain a cross-reference to a test prescribed in the are limited according to the principIes defined in chapter 5.4,
Tests section of the monograph. It may be used to simpli:fy using general method 2.4.24 or another suitable method.
the work of the analyst carrying out the identification and the Where a quantitative determination of a residual solvent is
prescribed tests. For example, one identification set cross- carried out and a test for loss on drying is not carried out,
refers to a test for enantiomeric purity while the other set the content of residual solvent is taken into account for
gives a test for specific optical rotation: the intended purpose calculation of the assay content of the substance, the specific
of the two is the same, that is, verification that the correct optical rotation and the specific absorbance.
enantiomer is presento
Microbiological quality
TESTS Individual monographs give acceptance criteria for
Polymorphism (5.9) microbiological quality wherever such control is necessary.
If the nature of a crystaHine or amorphous form imposes Table 5.1.4.-2. -Acceptance criteriajor nzicrobiological qualit:y oj
restrictions on its use in preparations, the nature of the non-sterile substances jor pharmaceutical use in chapter 5.1.4.
specific crystalline or amorphous form is identified, its Microbiological qualit:y oj non-sterile pharmaceutical preparations
morphology is adequately controlled and its identity is stated and substances jor pharmaceutical use gives recornmendations
on the label. on microbiological quality that are of general relevance for
Related substances substances subject to microbial contamination. Depending on
Unless otherwise prescribed or justified and authorised, the nature of the substance and its intended use, different
organic impurities in active substances are to be reported, acceptance criteria may be justified.
identified wherever possible, and qualified as indicated in Sterility (2.6.1)
Table 2034.-1 or in Table 2034.-2 for peptides obtained by If intended for use in the manufacture of sterile dosage forms
chemical synthesis. without a further appropriate sterilisation procedure, or if
offered as sterile grade, the substance for pharmaceutical use
Table 2034.-1. - Reporting, identification and qualification of complies with the test for sterility.
organic impurities in active substances Bacterial endotoxins (2.6.14)
Use Maximum Report- Identification Qualification If offered as bacterial endotoxin-free grade, tlle substance for
daily ing threshold threshold pharmaceutical use complies with the test for bacterial
dose threshold
endotoxins. The limit and test method (if not gelation
Human ::; 2 g/day > 0.05 per > 0.10 per > 0.15 per
use or cent cent or a cent or a method A) are stated in the individual monograph. The limit
human daily intake daily intake is calculated in accordance with the recommendations in
and of> 1.0 mg of> 1.0 mg general chapter 5.1.10. Guidelines jor using the test jor bactelial
veterinary (whichever is (whichever is
use the lower) the lower) endotoxins, unless a lower limit is justified from results from
Human > 2 g/day > 0.03 per > 0.05 per cent > 0.05 per production batches or is required by the competent authority.
use or cent cent Where a test for bacterial endotoxins is prescribed, a test for
human
and pyrogens is not required.
veterinary
use
Pyrogens (2.6.8)
Veterinary Not > 0.10 per > 0.20 per cent > 0.50 per If the test for pyrogens is justified rather than the test for
use only applicable cent cent bacterial endotoxins and if a pyrogen-free grade is offered,
the substance for pharmaceutical use complies with the test
for pyrogens. The limit and test method are stated in the .
Table 2034.-2. - Reportíng, identification and qualification of individual monograph or approved by the competent
organic impurities in peptides obtained by chemical synthesis authority. Based on appropriate test validation for bacterial
endotoxins and pyrogens, the test for bacterial endotoxins
Reporting Identification Qualification
threshold
may replace the test for pyrogens.
threshold threshold
> 0.1 per cent > 0.5 per cent > 1.0 per cent Additional properties
Control of additional properties (e.g. physical characteristics,
functionality-relat.ed characteristics) may be necessary for
Specific thresholds may be applied for impurities known to individual manufacturing processes or formulations. Grades
be unusually potent or to produce toxic or unexpected (such as sterile, endotoxin-free, pyrogen-free) may be
pharmacological effects. produced with a view to manufacture of preparations for
If the individual monograph does not provide suitable control parenteral administration or other dosage forms and
for a new impurity, a suitable test for control must be
developed and inc1uded in the specification for the substance.
2016 Alfaxalone Vet-39

appropriate requirements may be specified in an individual IDENTIFICATION


monograph. A. Dissolve 20 mg in 2 mL of water, add 3 mL of 2M sodiu111
ASSAY hydroxide, extract with 5 mL of cyclohexane and evaporate to
dryness under reduced pressure. The il1frared absorption
Unless justified and authorised, contents of substances for
spectrum of the residue, Appendix U A, is concordant with
pharmaceutical use are determined. Suitable methods are
the reference spectrum of acepromazine (RSV 01).
used.
B. Complies with the test for identijication of phenothiazines,
LABELLING Appendix IU A, applying to the plate 1 ~lL of each solution
In general, labelling is subject to supranational and national and using aceprol11azine maleate BPCRS for the preparation of
regulation and to intemational agreements. The statements solution (2).
under the heading Labelling therefore are not comprehensive
C. Dissolve 0.2 g in a mixture of 3 mL of water and 2 mL of
and, moreover, for the purposes of the Pharmacopoeia only
5M sodium hydroxide and shake with three 3-mL quantities of
those statements that are necessary to demonstrate
ether. Add to the aqueous solution 2 mL of bromine solution,
compliance or non-compliance with the monograph are
warm in a water bath for 10 minutes, heat to boiling, cool
mandatory. Any other labelling statements are included as
and add 0.25 mL to a solution of 10 mg of resorcinol in 3 mL
recommendations. When the term 'label' is used in the
of sulfuric acid. A bluish black colour develops on heating for
Pharmacopoeia, the labelling statements may appear on the
15 minutes in a water bath.
container, the package, a leafiet accompanying the package or
a certificate of analysis accompanying the article, as decided TESTS
by the competent authority. Acidity
Where appropriate, the label states that the substance is: pH of a 1.0% w/v solution, 4.0 to 4.5, Appendix V L.
-- intended for a specific use; Melting point
-- of a distinct crystalline form; 136° to 139°, Appendix V A.
-- of a specific degree of fineness; Related substances
-- compacted; Complies with the test for related substances in phenothiazines,
-- coated; Appendix IU A, but using a mixture of 75 volumes of
-- granulated; n-hexane, 17 volumes of butan-2-one and 8 volumes of
-- sterile; diethylamine as the mobile phase.
-- free from bacterial endotoxins;
-- free from pyrogens; Loss on drying
0

-- containing gliding agents. When dried to constant weight at 105 , loses not more than
1.0% of its weight. Use 1 g.
Where applicable, the label states:
-- the degree of hydration; Sulfated ash
-- the name and concentration of any excipient. Not more than 0.2%, Appendix IX A.
____________________________________________ PhEm ASSAY
Dissolve 0.4 g in 50 mL of acetic anhydride and carry out
Method I for non-aqueous titration, Appendix VIU A, using
clystal violet solution as indicator. Each mL of 0.1 M perchloric
acid VS is equivalent to 44.25 mg of C19H22N20S,C4H404'
Acepromazine Maleate

Alfaxalone

442.5 3598-37-6

Action and use


Dopamine receptor antagonist; neuroleptic. H ~
Preparations HO H
Acepromazine Injection
Acepromazine Tablets 332.5 23930-19-0
DEFINITION Action and use
Acepromazine Maleate is 2-acetyl-10- Intravenous generalanaesthetic.
(3-dimethylaminopropyl) phenothiazine hydrogen maleate.
It contains not les s than 98.5% and not more than 101.0% DEFINITION
of C19H22N20S,C4H404, calculated with reference to the Alfaxalone is 3a-hydroxy-5a-pregnane-11, 20-dione.
dried substance. It contains not les s than 95.0% and not more than 103.0%
CHARACTERISTICS of C21H3203' calculated with reference to the dried
A yellow, crystalline powder. substance.
Soluble in water; freely soluble in chloroform; soluble in CHARACTERISTICS
ethanol (96%); slightly soluble in ether. A white to creamy white powder.
Vet-40 Amitraz 2016

Practically insoluble in water; freely soluble in chloroform; Calculate the content of C21H3203 in the substance being
soluble in ethanol (96%); practically insoluble in petroleu11l examined using the declared content of C21H3203 in
spirit (boiling range) 60° to 80°). alfaxalone BPCRS; peak areas 01' peak heights may be used
IDENTIFICATION irrespective of the symmetry factor.
A. The infrared absol'ption spectnl11z, Appendix n A, is
concordant with the reference spectnl11z of alfaxalone (RSV 03).
B. Complies with the test for identification of steroids,
Appendix In A, using impregnating solvent JI and Amitraz
mobile phase D.
C. In the Assay, the chromatogram obtained with solution
(2) shows a peak having the same retention time as the peak
due to alfaxalone BPCRS in the chromatogram obtained with
solution (1).
TESTS
Light absorption
Absorbance of a 0.20% w/v solution in ethanol (96%) at 293.4 33089-61-1
235 nm, not more than 0.20, calculated with reference to the
dried substance, Appendix n B. Action and use
Topical parasiticide; acaricide.
Related substances
Carry out the method for thin-layer chromatography, Preparation
Appendix nI A, using silica gel G as the coating substance Amitraz Dip Concentrate (Liquid)
and a mixture of equal volumes of ethyl acetate and toluene as
the mobile phase. Apply separately to the plate 1O ~lL of each DEFINITION
of three solutions of the substance being examined in a Amitraz is N-methylbis (2,4-xylyliminomethyl) amine.
mixture of equal volumes of chlorofo1"m and methanol It contains not les s than 97.0% and not more than 101.0%
containing (1) 5.0% w/v, (2) 0.15% w/vand (3) 0.050% w/v. of C19H23N3, calculated with reference to the anhydrous
After removal of the plate, dry it in a current of air until the substance.
solvent has evaporated, spray with a saturated solution of CHARACTERISTICS
cerál11z (nj sulfate in sulfmic acid (50%) and heat at 110° for A white to buff powder.
1 hour. Any secondaly spot in the chromatogram obtained Practically insoluble in water; decomposes slowly in ethanol
with solution (1) is not more intense than the spot in the (96%); freely soluble in acetone.
chromatogram obtained with solution (2) (3%) and not more
than one such spot is more intense than the spot in the IDENTIFICATION
chromatogram obtained with solution (3) (1 %). The infrared abs01ption spectrwn, Appendix n A, is concordant
with the reference spectru77Z of amitraz (RSV 04) .
Loss on drying
When dried to constant weight at 105°, loses not more than TESTS
1.0% of its weight. Use 1 g. Related substances
Sulfated ash Carry out the method for gas chromatography,
Not more than 0.1 %, Appendix IX A. Appendix In B, using the following solutions.
0.010% w/v of 2)4-dimethylaniline, 0.20% w/v offor111-2')4'-
ASSAY
xylidide BPCRS and 0.20% w/v of N)N'-bis(2) 4-
Carry out the method for liquid chromatography,
xylyl)fol"mamidine BPCRS in methyl aceta te (solution A)
Appendix nI D, using the following solutions. For solution
(1) dilute 25 mL of a solution in propan-2-01 R1 containing Disperse 30 mg of N-methyl-N' -(2)4-xylyl)fo1711amidine
0.2% w/v of alfaxalone BPCRS, 0.01 % w/v of alfadolone hydl"ochlO1ide BPCRS in 5 mL of methyl aceta te, add about
acetate BPCRS and 0.03% w/v of betamethasone BPCRS 32 mg of tliethylamine, mix with the aid of ultrasound for
(internal standard) to 100 mL with carbon dioxide-free water. 2 minutes, filter, wash the filter with a small amount of
For solution (2) dilute 25 mL of a solution in propan-2-01 R1 methyl acetate and add sufficient methyl aceta te to the
containing 0.2% w/v of the substance being examined to combined filtrate and washings to produce 25 mL (solution
100 mL with caJ'bon dioxide-free water. For solution (3) dilute B) (about 0.1 % w/v of N-methyl-N' -(2)4-xylyl)fo1711amidine).
25 mL of a solution in propan-2-01 R1 containing 0.2% w/v of (1) 5.0% w/v solution of the substance being examined in
the substance being examined and 0.03% w/v of the internal methyl acetate.
standard to 100 mL with carbon dioxide-free water. (2) A mixture of equal volumes of solution A and solution B.
The chromatographic procedure may be carried out using CHROMATOGRAPHIC CONDITIONS
(a) a stainless steel column (10 cm x 5 mm) packed with (a) Use a fused silica capiZZaJy column (10 m x 0.53 mm)
octadecylsilyl silica gel for chromatography (5 ~lm) (Spherisorb bonded with a film (5 ~lm) of poly
ODS 1 is suitable) and maintained at 60°, (b) as the mobile [methyl(95)phenyl(5)}siloxane (Chrompack CP-SIL 8 CB is
phase with a flow rate of 1 mL per minute a mixture of suitable) .
propan-2-ol R1 and caJ'bon dioxide-free water adjusted so that
the resolution factor between the peaks due to alfadolone (b ) Use helium as the carrier gas at 12 mL per minute.
acetate (retention time about 5 minutes) and alfaxalone (c) Use gradient conditions at an initi~t1 temperature of 125°,
(retention time about 6 minutes) is more than 1.0 (a mixture maintained at 125° for 5 minutes, increasing linearly to 270°
of 25 volumes of propan-2-01 R1 and 75 volumes of carbon at arate of 5° per minute and maintained at 270° for
dioxide-free water is usually suitable) and (c) a detection 15 minutes.
wavelength of 205 nm. (d) Use an inlet temperature of 230°.
2016 Amprolium Hydrochloride Vet-41

(e) Use a fiame ionisation detector at a temperature of 300°. IMPURlTIES


(f) Inject 1 ~lL of each of solutions (1) and (2).
In the chromatogram obtained with solution (2) the peaks ~NH2
following the solvent peak, in order of emergence, are due to
2,4-dimethylaniline, form-2' ,4'-xylidide, N-methyl-N'-(2,4- Me Avl Me
xylyl)formamidine and N,N '-bis(2,4-xylyl)formamidine.
LIMITS A. 2,4-dimethylaniline (2,4-xylidine),
In the chromatogram obtained with solution (1):
~NHCHO
the area of any peak corresponding to 2,4-dimethylaniline,
form-2',4'-xylidide, N-methyl-N'-(2,4-xylyl)formamidine and
N,N'-bis(2,4-xylyl)formamidine is not greater than the area Me Avl Me
of the corresponding peak in the chromatogram obtained
with solution (2) (0.1 %, 2%, 1% and 2% respectively); B. form-2',4'-xylidide,
the area of any other secondary peak is not greater than the
area of the peak due to 2,4-dimethylaniline in the ~N~NHMe
chromatogram obtained with solution (2) (0.1 %).
Water Me Avl Me
Not more than 0.1 % w/w, Appendix IX C, Method lA.
U se 5 g and a mixture of equal volumes of chloroform and C. N-methyl-N'-(2,4-xylyl)formamidine,
2-chloroethanol in place of anhydrous methanol.
Sulfated ash
Not more than 0.2%, Appendix IX A.
ASSAY
Carry out the method for gas chromatography,
Appendix nI B, using the following solutions.
Prepare a 2 % v/v solution of squalane (internal standard) in D. N,N'-bis(2,4-xylyl)formamidine.
methyl acetate (solution C).
(1) 0.15 g of the substance being examined in sufficient
methyl acetate to produce 30 mL.
(2) 0.15 g of the substance being examined in 10 mL of Amprolium Hydrochloride
solution C and add sufficient methyl acetate to produce
30 mL.
(3) 1.50% w/v solution of amitraz BPCRS in solution C and
dilute 1 volume of this solution to 3 volumes with methyl CI- ,HCI
acetate.
CHROMATOGRAPHIC CONDITIONS
(a) Use a fused silica capillary column (15 m x 0.53 mm) 315.3 137-88-2
coated with a 1.5 ~lm film of methyl silicone gum
(Chrompack CP-Sil 5 CB is suitable). Action and use
(b) Use helium as the carrier gas at 12 mL per minute. Antiprotozoal; prevention and treatment of coccidiosis
(veterinary) .
(c) Use isothermal conditions maintained at 220°.
(d) Use an inlet temperature of 230°. DEFINITION
(e) Use a fiame ionisation detector at a temperature of 300°. Amprolium Hydrochloride is 1-(4-amino-2-propylpyrimidin-
(f) Inject 1 ~L of each solution. 5-ylmethyl)-2-methylpyridinium chloride hydrochloride.
It contains not les s than 97.5% and not more than 101.0%
SYSTEM SUIT ABILITY
of C14H19ClN4,HCl, calculated with reference to the dried
The assay is not valid unless, in the chromatogram obtained substance.
with solution (3), the resolution factor between the peaks
corresponding to squalane and amitraz is at least 3.0. CHARACTERISTICS
A white or almost white powder; odourless or almost
LIMITS
odourless.
Calculate the content of C19H23N3 from the chromatograms Freely soluble in water; slightly soluble in ethanol (96%); very
obtained using the declared content of C19H23N3 in slightly soluble in ether; practically insoluble in chloroform.
amitraz BPCRS.
IDENTIFICATION
STORAGE
A. The infrared absorption spectrum, Appendix n A, is
Amitraz should be kept in a well-closed container, which may concordant with the reference spectrum of amprolium
contain paraformaldehyde, packed in separate sachets as a hydrochloride (RSV 07).
stabiliser.
B. The light absorption, Appendix n B, in the range 230 to
350 nm of a 0.002% w/v solution in O.lM hydrochloric acid
exhibits two maxima, at 246 nm and 262 nm. The
absorbances at the maxima are about 0.84 and about 0.80,
respectively.
Vet-42 Aprarnycin Sulfate 2016

C. To 1 mg add 5 mL of naphthalenediol reagent solution; CHARACTERISTICS


a deep violet colour is produced. A light brown powder or granular material; hygroscopic.
D. Yields the reactions characteristic of chlorides, Freely soluble in 'Water; practically insoluble in acetone, in
Appendix VI. ethanol (96%), in ether and in methanol.
TESTS IDENTIFICATION
Picoline A. Carry out the method for thin-Iayer chro111atography,
Dissolve 1.5 g in 30 mL of 'Water in a distillation flask, add Appendix In A, using the following solutions in 'Water.
20 mL of a saturated solution of potassium carbonate (1) 0.1 % w/v ofthe substance being examined.
sesquihydrate, connect the flask to a coarse-frined aerator
(2) 0.07% w/v of apramycin BPCRS.
extending to the bonom of a 100-mL graduated cylinder
containing 50 mL of 0.05M hydrochloric acid, and pass air, (3) 0.07% w/v of each of apramycin BPCRS and
which has previously been passed through sulfuric acid and tobramycin BPCRS.
glass wool, through the system for 60 minutes. To 5 mL of CHROMATOGRAPHIC CONDITIONS
the hydrochloric acid solution add sufficient (a) Use a silica gel F Z54 precoated plate (Merck silica gel 60
0.05M hydrochloric acid to produce 200 mL. The absorbance of F Z54 plates are suitable).
the resulting solution at 262 nm is not greater than 0.52, (b) Use the mobile phase as described below.
Appendix n B.
(c) Apply 5 /lL of each solution.
Loss on drying
0 (d) Develop the plate to 10 cm.
When dried to constant weight at 100 at a pressure not
exceeding 0.7 kPa, loses not more than 1.0% of its weight. (e) After removal of the plate, allow it to dry in a cunent of
Use 1 g. warm air, spray with a mixture of equal volumes of a
46% w/v solution of sulfLl1ic acid and a 0.2% w/v solution of
Sulfated ash 0
naphthalene-1 J 3-diol in ethanol (96%) and heat at 150 for
Not more than 0.1 %, Appendix IX A.
5 to 10 minutes.
ASSAY MOBILE PHASE
Carry out Method 1 for non-aqueous titration,
20 volumes of chlorofo1711, 40 volumes of 13.5M ammonia and
Appendix VIII A, using 0.3 g and 1-naphtholbenzein solution
60 volumes of methanol, equilibrated for 1 hour before use.
as indicator. Each mL of O.lM perchloric acid VS is equivalent
to 15.77 mg of C14H19CIN4,HCl. SYSTEM SUITABILITY
The test is not valid unless the chromatogram obtained with
solution (3) shows two clear1y separated principal spots.
CONFIRMATION

Aprarnycin Sulfate The principal spot in the chromatogram obtained with


Apramycin Sulphate solution (1) corresponds in colour and position to that in the
chromatogram obtained with solution (2).
B. In the test for Related substances, the retention time of

~
HO o the principal peak in the chromatogram obtained with
solution (1) corresponds to that of the principal peak in the
H2N
chromatogramobtained with solution (2).
HO OH ~NHMe
OH
C. Yields reaction A characteristic of sulfates, Appendix VI.
O O O HO OH

R =H
R
NH 2 O
J;;j NH 2
NH2
TESTS
Sulfate
26.0 to 33.0% of S04, calculated with reference to the
anhydrous substance, when determined by the following
method. Dissolve 0.25 g in 100 mL of 'Water, adjust to pH 11
with 13.5M al1unonia and add 10 mL of O.lM barium chlO1ide
Vs. Titrate with O.lM disodiu111 edetate VS using 0.5 rng of
784.8 41194-16-5
phthalein purple as indicator; add 50 rnL of ethanol (96%)
Action and use when the colour of the solution begins to change and
Aminoglycoside antibacterial. continue the titration until the violet-blue colour disappears.
Each rnL of 0.1 M baJiu111 chloride VS is equivalent to
Preparations
9.606 rng of S04'
Apramycin Veterinary Oral Powder
Caerulomycin and dipyridyI derivatives
Apramycin Premix
Dissolve 0.5 g in 'Water in a 100 rnL graduated flask, add
DEFINITION 10 mL of methanol and dilute to 100 mL with 'Water. Place
Apramycin Sulfate is the sulfate of 4-0- 5 mL in a 25 mL graduated flásk and add 5 rnL of an
[(2R,3RAaS,6R, 7S,8R,8aR)-3-amino-6-(4-amino-4-deoxy-o:- acetate buffer prepared by dissolving 8.3 g of anhydrous
D-glucopyranosyloxy)-8-hydroxy-7 - sodiu111 acetate in 25 rnL of 'Water, adding 12 rnL of glacial
methylaminoperhydropyrano[3,2-b]pyran-2-yl]-2- acetic acid and diluting to 100 rnL with 'Water. Mix, add 1 rnL
deoxystreptamine. It is produced by the growth of certain of a 10% w/v solution of hydroxylamine hydrochloride in 'Water
strains of Strept0111yces tenebrarius or obtained by any other and mix again. Add 5 mL of a 1% w/v-solution of am1110niu111
means. The potency is not less than 450 Units per mg, iron(JI) sulfate and dilute to 25 rnL with 'Water. Measure the
calculated with reference to the anhydrous substance. absorbance of the resulting solution at 520 nm,
Appendix n B, using in the reference cell a solution obtained
2016 Apramycin Sulfate Vet-43

by carrying out the same procedure without the substance peak in the chromatogram obtained with solution (3)
being examined. The absorbance is not greater than that (7%, 5%, 2% and 2% respectively);
obtained by repeating the test using 5 mg of 2)2' -dipyridyl the area of any other seconda1y peak is not greater than
dissolved in 10 mL of nzethanol and diluted to 100 mL with 0.4 times the area of the principal peak in the chromatogram
'Water and beginning at the words 'Place 5 mL in a 25 mL obtained with solution (3) (2%);
graduated fiask ... ' (1 %). the sum of the areas of all the seconda1y peaks is not greater
Related substances than 3 times the area of the principal peak in the
Carry out the method for liquid chromatography, chromatogram obtained with solution (3) (15%).
Appendix III D, using the following solutions in 'Water. Disregard any peak with an area les s than the area of the
(1) 0.50% w/v of the substance being examined. peak in the chromatogram obtained with solution (4) (0.1 %).
(2) 0.35% w/v of apramycin BPCRS. Sulfated ash
(3) Dilute 1 volume of solution (1) to 20 volumes. Not more than 1.0%, Appendix IX A, Method n. Use 1 g.
(4) Dilute 1 volume of solution (3) to 50 volumes. Water
CHROMATOGRAPHIC CONDITIONS Not more than 14.0% w/w, Appendix IX C. Use 0.2 g and
20 mL of a mixture containing 1 volume of methanol and
(a) Use a column (25 cm x 4 mm) packed with fast cation-
2 volumes of fonnamide as the solvento The solvent mixture
exchange polymeric beads (13 ~lm) with sulfonic acid
must be prepared at least 12 hours before use and should be
functional groups (Dionex Fast Cation-1 R is suitable) and a
stored in an airtight container.
stainless steel post-column reaction coil (380 cm x 0.4 mm)
with internal baffies. Use in the reaction coil ninhydrin reagent ASSAY
1 at a fiow rate approximately the same as that for the mobile Cany out the microbiological assay of antibiotics,
phase. Appendix XIV A, Method B. The precision of the assay is
(b) U se gradient elution and the mobile phase described such that the fiduciallimits of error are not les s than 95%
below. and not more than 105 % of the estimated potency.
(c) Use a fiow rate of 0.8 mL per minute. IMPURITIES
0
(d) Use a column temperature of 130 Maintain the post-

column reaction coil at the same temperature.


(e) Use a detection wavelength of 568 nm. OMe
(f) Inject 20 ~lL of each solution.
MOBILE PRASE
lvIobile phase A A solution containing 1.961 % w/v of sodiunz
citrate, 0.08% v/v of liquefied phenol and 0.5% v/v of H N
I
thiodiglycol, adjusted to pH 4.25 using hydrochloric acid. OH
lvIobile phase B A solution containing 4.09% w/v of sodium
chloride and 3.922% w/v of sodium citrate with 0.08% v/v of A. caerulomycin,
liquefied phenol, adjusted to pH 7.4 with hydrochloric acid.
Equilibrate the column using a mixture containing 75% of
HO

HO~O\
mobile phase A and 25% of mobile phase E. After each
injection elute for 3 minutes using the same mixture and OH
then cany out a linear gradient elution for 6 minutes to
100 % of mobile phase B. Elute for a further 21 minutes ~~NH2
using 100% of mobile phase E, then step-wise re-equilibrate NH2 O NH
2
to a mixture of 75% of mobile phase A and 25% of mobile
phase E and elute for at least 10 minutes.
B. lividamine,
SYSTEM SUITABILITY
The test is not valid unless, in the chromatogram obtained
with solution (2), the resolution factor between the peaks due OH
to compound A and 3-hydroxyapramycin, identified using HO~NH2
the reference chromatogram supplied with apramycin BPCRS, HO~
is at least 0.8. NH 2
LIMITS
Multiply the areas of all the seconda1y peaks by 0.5. [NOTE: C. 2-deoxystreptamine,
This is to ensure that the impurities are calculated relative to D. 3-hydroxyapramycin; R = OH,
Apramycin Sulfate (which contains about 50% w/w of
E. 'compound A',
apramycin).]
F. 'compound E'.
In the chromatogram obtained with solution (1):
the areas of any peaks corresponding to 3-hydroxyapramycin,
lividamine/2-deoxystreptamine (combined), compound A and
compound E (identified using the reference chromatogram
supplied with apramycin BPCRS) are not greater than 1.4,
1.0, 0.4 and 0.4 times respectively the are a of the principal
Vet-44 Azaperone 2016

Mobile phase:
Azaperone -- mobile phase A: dissolve 1.4 g of anhydrous sodiu11l sulfate R
(Azaperone for Vetel'inmy Use) in 900 mL of 'Water R, add 16.0 mL of 0.01 1\11 sulfuric
Ph Bur l1zonograph 1708) acid and dilute to 1000 mL with 'Water R;
-- mobile phase B: methanol R;

Time Mobile phase A Mobile phase B


(min) (per cent V/V) (per cent V/V)
0- 15 95 -7 20 5 -7 80

15 - 20 20 80

327.4 1649-18-9 Flo'W rate 1.5 mUmin.


Detection Spectrophotometer at 230 nm.
Action and use
Jnjection 1O ~lL.
Dopamine receptor antagonist; neuroleptic (veterinary).
Relative retention With reference to azaperone (retention
Preparation
time = about 9 min): impurity A = about 0.9;
Azaperone Injection
impurity B = about 1.1; impurity C = about 1.15.
PhE~ _____________________________________________ System suitability: reference solution (a):
-- resolution: minimum 8.0 between the peaks due to
DEFINITION
azaperone and to benperidol.
1-(4-Fluorophenyl)-4- [4-(pyridin-2-yl)piperazin-1-yl]butan-1-
Li111its:
one.
-- impurity A: not more than the area of the principal peak
Content in the chromatogram obtained with reference solution (b)
99.0 per cent to 101.0 per cent (dried substance). (0.25 per cent);
CHARACTERS -- ul1specified impurities: for each impurity, not more than
Appearance 0.8 times the area of the principal peak in the
White or almost white powder. chromatogram obtained with reference solution (b)
(0.20 per cent);
Solubility
- - SU1J1 of i112purities B and C: not more than 3 times the area
Practically insoluble in water, freely soluble in acetone and in
of the principal peak in the chromatogram obtained with
methylene chloride, soluble in ethanol (96 per cent).
reference solution (b) (0.75 per cent);
It shows polymorphism (5.9). -- total: not more than 4 times the area of the principal peak
IDENTIFICATION in the chromatogram obtained with reference solution (b)
Infrared absorption spectrophotometry (2.2.24). (1.0 per cent);
Preparatíon Discs. -- disregard limit: 0.2 times the area of the principal peak in
the chromatogram obtained with reference solution (b)
C0112parison azaperone CRS.
(0.05 per cent).
If the spectra obtained show differences, dissolve the
Loss on drying (2.2.32)
substance to be examined and the reference substance
Maximum 0.5 per cent, determined on 1.000 g by drying
separately in acetone R, evaporate to dryness and record new
in vacuo at 60 oC for 4 h.
spectra using the residues.
Sulfated ash (2.4.14)
TESTS
Maximum 0.1 per cent, detennined on 1.0 g.
Appearance of solution
The solution is clear (2.2.1) and not more intensely coloured ASSAY
than reference solution Y6 (2.2.2) Method JI). Dissolve 0.130 g in 70 mL of a mixture of 1 volume of
Dissolve 1.0 g in 25 mL of a 14 gIL solution of tartaric anhydrous acetic acid R and 7 volumes of methyl ethyl ketone R.
acid R. Titrate with 0.1 M perchlorz'c acid, using 0.2 mL of
naphtholbenzein solution R as indicator.
Related substances
1 mL of 0.1 M perchloric acid is equivalent to 16.37 mg of
Liquid chromatography (2.2.29).
C19H22FN30.
Test solution Dissolve 0.100 g of the substance to be
examined in methanol R and dilute to 10.0 mL with the same STORAGE
solvento Protected from light.
Reference solution (a) Dissolve 5.0 mg of azaperone CRS and IMPURITIES
6.0 mg of benperidol CRS in methanol R and dilute to Specified il11purities A, B, C
200.0 mL with the same solvento
Reference solution (b) Dilute 1.0 mL of the test solution to
100.0 mL with methanol R. Dilute 5.0 mL ofthe solution to
20.0 mL with methanol R.
Column:
-- size: l = 0.10 m, 0 = 4.6 mm;
-- stationmy phase: base-deactivated octadecylsi1yl silica gel for
chromatography R (3 ~lm);
-- temperature: 25 oC.
2016 Calcium Copperedetate Vet-45

B. Ignite 0.2 g, dissolve the residue in 3 mL of


2M hydrochloric acid, neutralise the solution with 5M a11111l011ia
and add 1 mL of 6M acetic acid and 2 mL of dilute potassium
iodide solutiol1. A white precipitate is produced and iodine is
liberated, colouring the supernatant liquid brown.
C. Dissolve 0.5 g in 10 mL of water, acidify with
~ 2M hydrochloric acid, add 25 mL of a 10% v/v solution of
~R 111acaptoacetic acid and :filter. Make the :filtrate alkaline with
5M a11111Zonia and add 5 mL of a 2.5% w/v solution of
F o
a11Z111onium oxalate. A white precipitate is produced which is
A. 1-(2-fluorophenyl)-4- [4-(pyridin-2-yl)piperazin-1-yl]butan- soluble in hydrochloric acid but only sparingly soluble in
l-one, 6M acetic acid.
TESTS
RIII Lead
Not more than 25 ppm of Pb when determined by the
~R following method. Dissolve 1.25 g in 10 mL of hydrochloric
o acid, dilute to 25 mL with water and determine by atomic
absorption spectrophotometly, Appendix II D, measuring at
B. 4-[4-(pyridin-2-yl)piperazin-1-yl]-1-[4-[ 4-(pyridin-2- 283.3 nm and using alead hollow-cathode lamp as the
yl)piperazin-1-yl]phenyl]butan-1-one, radiation source and lead standard solution (lOO ppm Pb),
diluted if necessary with water, to prepare the standard
RIíI . solutions.

~OHo
Zinc
N ot more than 200 ppm of Zn when determined by the
following method. Dissolve 1.0 g in 20 mL of hydrochloric
acid, dilute to 200 mL with water and determine by atomic
C. 4-hydroxy-1-[4-[4-(pyridin-2-yl)piperazin-1-
absOlption spectrophotometly, Appendix II D, measuring at
yl] phenyl]butan-1-one.
213.9 nm and using a zinc hollow-cathode lamp as the
_____________________________________________ ~E~

radiation source and zinc standard solution (5 mg/111L Zn)


diluted if necessary with water, to prepare the standard
solutions.
Loss on drying
Calcium Copperedetate When dried to constant weight at 105°, loses not more than
2.0% of its weight. Use 1 g.
ASSAY
For copper
Ignite 4 g at 600 0 to 700°, cool and heat the residue with
12 mL of a mixture of equal volumes of hydrochloric acid and
wata on a water bath for 15 minutes. Add 10 mL of water,
filter and dilute the filtrate to 100 mL with water (solution
A); reserve a portion for the Assay for calcium. To 25 mL of
solution A add 25 mL of water and 10 mL of bromine
solution, boil to remove the bromine, cool and add dilute
sodiu11Z carbonate solution until a faint permanent precipitate is
Action and use produced. Add 3 g of potassium iodide and 5 mL of 6M acetic
Used in the treatment of copper deficiency. acid and titrate the liberated iodine with O.1M sodium
Preparation thiosulfate VS, using starch 111ucilage as indicator, until only a
Calcium Copperedetate Injection faint blue colour remains; add 2 g of potassium thiocyanate
and continue the titration until the blue colour disappears.
DEFINITION Each mL of O.lM sodium thiosulfate VS is equivalent to
Calcium Copperedetate is the dihydrate of calcium 6.354 mg of Cu.
[ethylenediaminetetra-acetato(4- )-N,NI ,0,0 1] copper(n). For calcium
It contains not less than 9.1 % and not more than 9.7 % of To 5 mL of solution A add 10 mL of water and 10 mL of a
calcium, Ca, and not les s than 14.4% and not more than 10% v/v solution of 111ercaptoacetic acid, allow to stand until
15.3% of copper, Cu, both calculated with reference to the the precipitate has coagulated, dilute to 100 mL with water,
dried substance. add 5 mL of 5M sodium hydroxide and titrate with
CHARACTERISTICS 0.05M disodiu111 edetate VS, using methyl thY11101 blue mixture as
A blue, crystalline powder. indicator, until the solution becomes a full purple colour,
adding ,the titrant slowly as the end point is approached.
Freely soluble in water, the solution gradually precipitating
Each mL of 0.05M disodiu11Z edetate VS is equivalent to
the tetrahydrate; practically insoluble in ethanol (96%).
2.004 mg of Ca.
IDENTIFICATION
A. Dissolve 0.2 g in 5 mL of water and add 1 mL of
6M acetic acid and 2 mL of dilute potassiu111 iodide solutiol1.
The solution remains clear and deep blue.
Vet-46 Carprofen 2016

Detection Spectrophotometer at 235 nm.


Carprofen
Jnjection 20 ~lL.
(Cal'pmfen for Veterinary Use) Run time 4 times the retention time of carprofen.
Ph Eur monograph 2201)
Retention time Carprofen = about 10 mino
System suitability: reference solution (a):
H\eH3

~
- resolution: minimum 1.5 between the peaks due to
N I ~ eOzH impurity C and carprofen.
Limits:
f _ '\ .# and enantiomer
- unspecijied i11lpurities: for each impurity, not more than
el twice the area of the principal peak in the chromatogram
obtained with reference solution (b) (0.20 per cent);
- total: not more than 5 times the area of the principal peak
273.7 53716-49-7
in the chromatogram obtained with reference solution (b)
Action and use (0.5 per cent);
Cyc1o-oxygenase inhibitor; analgesic; anti-inflarnmatory. - disregard limit: the area of the principal peak in the
chromatogram obtained with reference solution (b)
~Ew ___________________________________________ (0.1 per cent).
DEFINITION Heavy metals (2.4.8)
(2RS)- 2-( 6-Chloro-9 H-carbazol-2-yl) propanoic acid. Maximum 20 ppm.
Content Dissolve 1.0 g in ethanol (96 per cent) R and dilute to 20 mL
98.5 per cent to 101.5 per cent (dried substance). with the same solvento 12 mL of the solution complies with
test B. Prepare the reference solution using lead standard
CHARACTERS solution (1 ppm Pb) R.
Appearance
Loss on drying (2.2.32)
White or almost white, crystalline powder.
Maximum 0.5 per cent, determined on 1.000 g by drying in
Solubility an oven at 105 oC for 2 h.
Practically insoluble in water, freely soluble in acetone,
Sulfated ash (2.4.14)
soluble in methanol, slightly soluble in 2-propanol.
Maximum 0.1 per cent, determined on 1.0 g.
It shows polymorphism (5.9).
ASSAY
IDENTIFICATION Dissolve 0.200 g in 50 mL of ethanol (96 per cent) R.
Infrared absorption spectrophotometry (2.2.24). Add 1.0 mL of 0.1 M hydrochloric acid. Titrate with 0.1 NI
Campa riso n carpmfen CRS. sodium hydroxide, determining the end-point
If the spectra obtained in the solid state show differences, potentiometrically (2.2.20). Read the volume added between
dissolve the substance to be examined and the reference the 2 points of inflexion.
substance separately in acetone R, evaporate to dryness and 1 mL of 0.1 NI sodiu11l hydroxide is equivalent to 27.37 mg of
record new spectra using the residues. ClsH12ClN02'
TESTS STORAGE
Appearance of solution Protected from light.
The solution is c1ear (2.2.1) and not more intensely coloured
IMPURITIES
than reference solution BY3 (2.2.2) Method JI).
Other detectable i711purities (the following substances would, if
Dissolve 1.0 g in methanol R and dilute to 25 mL with the present at a sufficient level, be detected by one or other of
same solvento the tests in the monograph. They are limited by the general
Related substances acceptance criterion for other/unspecified impurities and/or
Liquid chromatography (2.2.29). Cany out the test protected by the general monograph Substances for pharmaceutical use
from light. (2034). It is therefore not necessary to identify these
Test solution Dissolve 50 mg of the substance to be examined impurities for demonstration of compliance. See also 5.10.
in the mobile phase and dilute to 100.0 mL with the mobile Control of impurities in substances for pharmaceutical use): A) B)
phase. C) D) E) F) G) H.
Reference solution (a) Dissolve 2.5 mg of carprofen for system

.-<~«:::H
suitability CRS (containing impurity C) in the mobile phase
and dilute to 10.0 mL with the mobile phase.
Reference solution (b) Dilute 1.0 mL of the test solution to
100.0 mL with the mobile phase. Dilute 1.0 mL ofthis
solution to 10.0 mL with the mobile phase.
Column:
- size: 1 = 0.25 m, 0 = 4.6 mm;
r
el

A. 2-( 6-chloro-QH-carbazol-2-yl)-2-methylpropanedioic acid,


- stationaJY phase: end-capped polar-embedded octadecylsilyl
amorphous organosilica polymer R (5 ~lm).
Mobile phase Mix 30 volumes of a 1.36 gIL solution of
potassiu111 dihydrogen phosphate R adjusted to pH 3.0 with
phosphonc acid R and 70 volumes of methanol R2.
Flow rate 1.3 mUmin.
2016 Cefalonium Vet-47

Cefalonium

B. (2RS)-2-(9H-carbazol-2-yl)propanoic acid,

494.5 5575-21-3 (anhydrous)

Action and use


Cephalosporin antibacterial.
Preparation
c. (1 RS) -1-( 6-chloro-9 H-carbazol-2-yl) ethanol,
Cefalonium Intramammary Infusion (Dry Cow)

DEFINITION
Cefalonium is 3-( 4-carbamoyl-1-pyridiniomethyl)-7-
[(2-thienyl)acetamido] -3-cephem-4-carboxylate dihydrate.
It contains not less than 95.0% and not more than 103.5%
of C2oHlSN40SS2, calculated with reference to the
anhydrous substance.

D. 1-( 6-chloro-9H-carbazol-2-yl)ethanone, CHARACTERISTICS


A white or almost white crystalline powder.
Very slight1y soluble in water and in methanol; soluble in
dimethyl sulfoxide; insoluble in dichloromethane, in ethanol
(96%) and in ether. It dissolves in dilute acids and in alkaline
solutions.
IDENTIFICATION
A. The infrared abs01ption spectrum, Appendix II A, is
E. 3-chloro-9H-carbazole, concordant with the reference spectrum of cefalonium
(RSV09).
B. The light abs01ption, Appendix II B, in the range 220 to
350 nm of a 0.002% w/v solution in water exhibits two
maxima, at 235 nm and at 262 nm. The absorbance at
235 nm is about 0.76 and at 262 nm is about 0.62.
TESTS
el Specific optical rotation
Dissolve 0.25 g with the aid of gentle heat in sufficient
dimethyl sulfoxide to produce 50 mL. Allow the solution to
F. diethyl 2-(6-chloro-9H-carbazol-2-yl)-2-
stand for 30 minutes before measurement of the optical
methylpropanedioate,
rotation. The specific optical rotation of the resulting solution is
-50 to -56, ca1culated with reference to the anhydrous
substance, Appendix V F.
Related substances
Carry out the method for thin-layer chromatography,
Appendix lIT A, using the following solutions in 8.3M acetic
acid.
(1) 2.5% w/v of the substance being examined.
G. ethyl (2RS)-2-( 6-chloro-9H-carbazol-2-yl)propanoate, (2) 0.05% w/v of the substance being examined.
(3) 0.025% w/v of the substance being examined.
(4) 0.005% w/v of the substance being examined.
(5) 0.05% w/v of each of cefalotin sodium EPCRS and
isonicotinamide.
CHROMATOGRAPHIC CONDITIONS
(a) Use as the coating silica gel F254 .
H. 6-chloro-2-ethyl-9H-carbazole. (b) Use the mobile phase as described below.
___________________________________________ PhE~ (c) Apply 4 ¡..tL of each solútion.
(d) Develop the plate to 12 cm.
(e) After removal of the plate, allow it to dry in air and
examine under ultraviolet light (254 11m).
Vet-48 Clazuril 2016

MOBILE PHASE
10 volumes of glacial acetic acid, 10 volumes of 1M sodium
acetate and 30 volumes of propan-2-0l.
SYSTEM SUITABILITY
y CONH 2
The test is not valid unless the chromatogram obtained with
solution (5) shows two clearly separated spots. D. isonicotinamide.
LIMITS
Any secondary spot in the chroma1Ogram obtained with
solution (1) is not more intense than the spot in the
chromatogram obtained with solution (2) (2%), not more
than one such spot is more intense than the spot in the
Clazuril
chromatogram obtained with solution (3) (1 %) and not more (Clazw'il for Veterinmy Use~ Ph Bur 1110nograph 1714)
than three such spots are more intense than the spot in the
chromatogram obtained with solution (4) (0.2% each).

'
H eN el

dO
Sulfated ash
Not more than 0.2%, Appendix IX A.
~ :P",
~ o
)l and enantiomer
Water el" ~ N NH
6.5 to 8.5% w/w, Appendix IX C. Use 0.5 g. ~0 o
ASSAY
Measure the absorbance of a 0.002% w/v solution at the 373.2 101831-36-1
maximum at 262 nm, Appendix II B. Calculate the content
of C2oHlSN40SS2 from the absorbance obtained using a Action and use
0.002% w/v solution of cefalonium BPCRS and from the Treatment of coccidiosis; antiprotozoal (veterinary).
declared content of C2oHlsN40SS2 in cefalonium BPCRS.
PhE~ ___________________________________________
STORAGE
Cefalonium should be protected from light and stored at a DEFINITION
temperature not exceeding 30°. (2RS)- [2-Chloro-4- (3 ,5-dioxo-4,5-dihydro-1, 2, 4-triazin-
Cefalonium intended for use in the manufacture of either a 2 (3H)-yl)phenyl] (4-chlorophenyl)ace1Onitrile.
parenteral dosage form or an intramammmy infusion without a Content
further appropriate sterilisation procedure complies with the 99.0 per cent to 101.0 per cent (dried substance).
following additional requirement. CHARACTERS
Sterility Appearance
Complies with the test for sterility, Appendix XVI A. White or light yellow powder.
IMPURlTIES Solubility
Practically insoluble in water, freely soluble in
COOH dimethylformamide, slightly soluble in ethanol (96 per cent)

~O~~~OAC
and in methylene chloride.
n IDENTIFICATION
~r-~N\\\~Sj A. Melting point (2.2.14): 199 oC to 203 oC.
S H H H B. Infrared absorption spectrophotometry (2.2.24).
Comparison clazurzl CRS.
A. cefalotin,
TESTS
Related substances
COOH Liquid chromatography (2.2.29).

n ~O~~~OH Solvent mixture tetrahydrofuran R, water R (50:50 V/V).

~r-~N\\\~Sj Test solution Dissolve 20.0 mg of the substance 10 be


examined in the solvent mixture and dilute to 20.0 mL with
S H H H
the solvent mixture.
Reference solution (a) Dissolve 5 mg of clazuril for system
B. 3-hydroxymethyl-7 ~-(2-thienylacetamido)-3-cephem-4- suitability CRS (containing impurities A, B, C, D, E, F, G, H
carboxylic acid, and 1) in the solvent mixture and dilute to 5.0 mL with the
solvent mixture.

~ o~;:t-J
Reference solution (b) Dilute 1.Ó mL of the test solution to
100.0 mL with the solvent mixture. Dilute 2.0 mL of this
n solution to 10.0 mL with the solvent mixture.

"~~N"'~SJ
S H H H
Column:
-- size: 1 = 0.10 m, 0 = 4.6 mm;
-- stationary phase: octadecylsilyl silica gel for chromatography R
(3 [lm);
C. 3-hydroxymethyl-7 ~-(2-thienylacetamido)- 3-cephem-4- -- temperature: 35 oc.
carboxylic acid lactone,
2016 Clazuril Vet-49

Mobile phase: determining the end-point potentiometrically (2.2.20). Carry


- nzobile phase A: mix 100 volumes of a 7.7 giL solution of out a blank titration.
am1110nium acetate R adjusted to pH 6.2 with a 1 mL of 0.1 M sodium hydroxide is equivalent to 37.32 mg of
10 per cent V/V solution of anhydrous fotmic acid R, C17HlQC12N4Ü2'
150 volumes of acetonittile R and 750 volumes of water R;
- 1110bile phase B: mix 50 volumes of water R, 100 volumes STORAGE
of a 7.7 giL solution of a111111oniu111 acetate R adjusted to Protected from light.
pH 6.2 with a 10 per cent V/V solution of anhydrous IMPURITIES
formic acid R and 850 volumes of acetonitrile R; Specified i111purities A, B, C, D, E, F, G, H, 1

Time Mobile phase A Mobile phase B


(min) (per cent V/V) (per cent V/V)
0-20 100 -7 O 0-7 100
and enantiomer
20 - 25 O 100 CI

Flow rate 1. O mUmin.


Detection Spectrophotometer at 230 nm. A. (2RS)- [2-chloro-4-(3, 5-dioxo-4,5-dihydro-1 ,2,4-triazin-
2 (3H)-yl)phenyl] (4-chlorophenyl) acetic acid,
Injection 5 ~lL.
Identificatíon of impwities U se the chromatogram supplied
with clazuril for system suitability CRS and the chromatogram
obtained with reference solution (a) to identi:fy the peaks due
to impurities A, B, C, D, E, F, G, H and 1. and enantiomer
Relative retention With reference to clazuril (retention
time = about 16 min): impurity A = about 0.6;
impurity B = about 0.78; impurity C = about 0.80;
impurity D = about 0.86; impurity E = about 0.9;
impurity F = about 0.95; impurity G = about 0.98; B. 2- [3-chloro-4- [(RS)-( 4-chlorophenyl) cyanomethyl]phenyl]-
impurity H = about 1.1; impurity 1 = about 1.2.
3,5-dioxo-2,3,4,5-tetrahydro-1,2,4-triazine-6-carboxamide,
System suitability: reference solution (a):
- peak-to-valley ratio: minimum 1.5, where Hp = height
above the baseline of the peak due to impurity G o
and H v = height above the baseline of the lowest point of
the curve separating this peak from the peak due to and enantiomer
clazuril, CI
- the chromatogram obtained is similar to the
chromatogram supplied with claZ'wil for system
suitability CRS.
C. (2RS)-2-[2-chloro-4-(3,5-dioxo-4,5-dihydro-1,2,4-triazin-
Limits:
2 (3H)-yl)phenyl] -2-(4-chlorophenyl) acetamide,
- cortection factors: for the calculation of contents, multiply
the peak areas of the following impurities by the
corresponding correction factor: impurity G = 1.4;
impurity H = 0.8;
- impuritíes AJ B J CJ DJ EJ F J GJ H J I: for each impurity,
and enantiomer
not more than the area of the principal peak in the
chromatogram obtained with reference solution (b)
(0.2 per cent);
- unspecified i111purities: for each impurity, not more than the
area of the principal peak in the chromatogram obtained
with reference solution (b) (0.20 per cent);
- total: not more than 3 times the area of the principal peak D. 2-[3-chloro-4-[(RS)-
in the chromatogram obtained with reference solution (b) (4-chlorophenyl) cyanomethyl]phenyl] -N,N-dimethyl-3,5-
(0.6 per cent); dioxo-2,3,4,5-tetrahydro-1 ,2, 4-triazine-6-carboxamide,
- disregard limit: 0.25 times the area of the principal peak in
the chromatogram obtained with reference solution (b)
(0.05 per cent); disregard the peaks due to the solvents.
Loss on drying (2.2.32)
and enantiomer
Maximum 0.5 per cent, determined on 1.000 g by drying in
an oven at 105 oC for 4 h.
Sulfated ash (2.4.14)
Maximum 0.1 per cent, determined on 1.0 g.
ASSAY E. methyl 2-[3-chloro-4-[(RS)-
Dissolve about 0.260 g in 35 mL of tetrahydrofuran R and (4-chlorophenyl) cyanomethyl] phenyl] -3,5-dioxo-2,3,4,5-
add 35 mL of water R. Titrate with 0.1 M sodiu111 hydroxide, tetrahydro-1,2,4-triazine-6-carboxylate,
Vet-50 Cloprostenol Sodium 2016

DEFINITION
Cloprostenol Sodium is (± )-(52)-7 -(1R,3R,5S)-2-[ (lE,3R)-
4-(3-chlorophenoxy)-3-hydroxybut-1-enyl] -3,5-
and enantiomer
dihydroxycyclopentylhept-5-enoate. It contains not less than
97.5% and not more than 102.5% of C22H2sClNa06,
calculated with l'eference to the anhydrous substance.
CAUTION Cloprostenol Sodiumis extremely potent and
extraol'dinaiy care should be taken in any procedul'e in zuhich it is
F. ethyl 2-[3-chloro-4-[(RS)- used.
(4-chlorophenyl) cyanomethyl] phenyl] -3 ,5-dioxo-2,3 ,4,5-
CHARACTERISTICS
tetrahydro-1,2,4-triazine-6-carboxylate,
A white or almost white, amorphous powder; hygroscopic.
Freely soluble in water, in ethanol (96%) and in methanol;
pl'actically insoluble in acetone.
IDENTIFICATION
A. The infrared abs01ption spectrunz, Appendix II A, is
concordant with the reference spectrum of cloprostenol sodium
(RSV 11).
G. 2-[3-chloro-4-(4-chlorobenzoyl)phenyl]-1,2,4- E. Yields reaction A characteristic of sodium salts,
triazine3,5(2H,4H)-dione, Appendix VI.
TESTS
o~~ H eN el Related substances
Carry out the method for liquid chromatography,
HNyN
o Appendix III D, using the following solutions in absolute
o N)(NH
ethanol.

~~ o (1) 2.0% w/v of the substance being examined.


(2) 0.050% w/v of the substance being examined.
CHROMATOGRAPHIC CONDITIONS
el
(a) Use a stainless steel column (25 cm x 4.6 mm) packed
H. [2-chloro-4-(3,5-dioxo-4,5-dihydro-1,2,4-triazin-2(3H)- with silica gel for chromatography (5 ~lm) (Partisil is suitable).
yl)phenyl] [4- [[2-chloro-4-(3,5-dioxo-4,5-dihydro-1,2,4- (b) U se isocratic elution and the mobile phase described
triazin-2 (3H)- below.
yl)phenyl] cyanomethyl]phenyl] (4-chlorophenyl)acetonitl'ile, (c) Use a fiow rate of 1.8 mL per minute.
(d) Use an ambient column temperature.

N~
and enantiomer
(e) Use a detection wavelength of 220 nm.
(f) Inject 5 ~lL of each solution.
el U NH
I
NH
I 2
(g) Allow the chromatography to proceed fol' twice the
retention time of the peak due to Cloprostenol.
N~
o
MOBILE PHASE

1. (2)-2- [[3-chloro-4- [(RS)-( 4-chlorophenyl)cyanomethyl] 1 volume of glacial acetic acid, 70 volumes of absolute ethanol
phenyl] diazanylidene] acetamide. and 930 volumes of hexane.
___________________________________________ ~E~ LIMITS
In the chromatogram obtained with solution (1):
the sum of the al'eas of any secondaiy peaks is not greatel' than
the area of the principal peak in the chromatogram obtained
with solution (2) (2.5%).
Cloprostenol Sodium
Water
Not more than 3.0% w/w, Appendix IX C. Use 50 mg
OH
dissolved in 1 mL of absolute ethanol.
H~=H\\\.... ~ ~
_ :'" ~- ~ -COONa
ASSAY
Carry out the method for liquid chromatography,
Appendix lIT D, using the following solutions in absolute
H ~ H % 0yyCI
OH H OH V ethanol.
(1) 0.08% w/v of the substance being examined.
(2) 0.08% w/v of cloprostenol sodium BPCRS.
446.9 55028-72-3 CHROMATOGRAPHIC CONDITIONS
(a) Use a stainless steel column (25 crri x 4.6 mm) packed
Action and use
with silica gel for chromatography (5 ~lm) (Partisil is suitable).
Prostaglandin (PGF2rJ analogue.
(b) Use isocratic elution and the mobile phase described
Preparation
below.
Cloprostenol Injection
2016 Closantel Sodium Dihydrate Vet-51

(c) Use a fiow rate of 1.8 mL per minute. PhE~ ___________________________________________

(d) Use an ambient column temperature. DEFINITION


(e) Use a detection wavelength of 220 nm. N- [S-Chloro-4- [(RS)-( 4-chlorophenyl)cyanomethyl] -2-
(f) Inject S ~LL of each solution. methylphenyl] -2-hydroxy-3,S-diiodobenzamide sodium salt
dihydrate.
MOBILE PHASE
1 volume of glacial acetic acid, 100 volumes of absolute ethanol Content
and 900 volumes of hexane. 98.S per cent to 101.S per cent (anhydrous substance).

DETERMINATION OF CONTENT CHARACTERS


Appearance
Ca1culate the content of C22H2sClNa06 from the
Yellow powder, slightly hygroscopic.
chromatograms obtained and using the declared content of
C22H2sClNa06 in clop1'Ostenol sodium BPCRS. Solubility
Very slightly soluble in water, freely soluble in ethanol
STORAGE (96 per cent), soluble in methanol.
Cloprostenol Sodium should be protected from light and
It shows polymorphism (5.9).
moisture.
IDENTIFICATION
IMPURITIES
A. Infrared absorption spectrophotometry (2.2.24).
Preparation Discs without recrystallisation.
OH
H ~ H Comparison closantel sodiu111 dihydrate CRS.

~
= \\\\\\~COONa B. Dissolve 0.1 g in 2 mL of ethanol (96 per cent) R.
The solution gives reaction (a) of sodium (2.3.1).
::: o;::?
~ -
H OHH
$

H$ OH
0yyCI
V TESTS
Appearance of solution
The solution is clear (2.2.1) and not more intensely coloured
than reference solution GY4 (2.2.2) Method JI).
A. (± )-(SZ )-7 -(1R,3R,SS)-2-[ (lB,3S)-4-(3-chlorophenoxy)- Dissolve O.SO g in ethanol (96 per cent) R and dilute to
3-hydroxybut-l-enyl] -3 ,S-dihydroxycyclopentyIhept-S-enoate SO mL with the same solvento
(epime¡))
Related substances
Liquid chromatography (2.2.29). Prepare the solutions
OH im11Zediately befare use and protect f1'Om light.
H H
\\\\\\~COONa
Test solution Dissolve 0.100 g of the substance to be
examined in methanol R and dilute to 10.0 mL with the same
solvento
~ 0yyCI Reference solution (a) Dissolve 10 mg of closantel for system
H
H OH V suitability CRS (containing impurities A to D in methanol R
and dilute to 1.0 mL with the same solvento
Reference solution (b) Dilute 1.0 mL of the test solution to
B. (±)-(SE )-7-(lR,3R,SS)-2-[(lB,3 R)-4- 100.0 mL with methanol R. Dilute S.O mL ofthis solution to
(3-chlorophenoxy)-3-hydroxybut-l-enyl] -3,S- 2S.0 mL with methanol R.
dihydroxycyclopentyIhept-S-enoate (trans-is01nel). Colu111n:
- size: 1 = 0.10 m, 0 = 4.6 mm,
- stationa¡y phase: base-deactivated octadecylsilyl silica gel for
ch1'Omatography R (3 ~Lm),
*** - temperature: 3S oc.
Closantel Sodium Dihydrate
*** *** Mobile phase:
(Closantel Sodium Dihydrate for Veterina¡y Use) *** - 1110bile phase A: to 100 mL of a 7.7 giL solution of
Ph Bur monograph 1716) ammonium acetate R previously adjusted to pH 4.3 with
acetic acid R, add SO mL of acetonitrile R and 8S0 mL of
H ,eN 'Water R;
oH3e~ - mobile phase B: to 100 mL of a 7.7 gIL solution of
ammonium aceta te R previously adjusted to pH 4.3 with
I«N~eIUel
I H
acetic acid R, add SO mL of 'Water R and 8S0 mL of
acetonitrile R;
~ ONa

I and enantiomer
Time Mobile phase A Mobile phase B
(min) (per cent Vil') (per cent Vil')
61438-64-0 0-2 50 50

Action and use 2 - 22 50 --7 20 50 --7 80


Antihelminthic. 22 - 27 20 80
Vet-52 Closantel Sodium Dihydrate 2016

PlO'lO rate 1.5 mUmin.


Detection Spectrophotometer at 240 nm.
111jection 1O ~lL.
Relative retention With reference to closantel (retention
time = about 16 min): impurity A = about 0.07; B. (2R~-(4-amino-2-chloro-5-
impurity B = about 0.48; impurity C = about 0.62; methylphenyl) (4-chlorophenyl)ethanenitrile,
impurity D = about 0.65; impurity E = about 0.82;
impurity F = about 0.89; impurity G = about 0.93; R1 R2

oH3e~
impurity H = about 1.13; impurity 1 = about 1.16;
impurity J = about 1.55.
Syste11l suitability: reference solution (a):
-- resolution: baseline separation between the peaks due to
I«N~el~el
I H
impurity G and closantel, ~ OH

-- the chromatogram obtained is similar to the R3 and enantiomer


chromatogram supplied with closantel for system
suitability CRS. C. R1 = H, R2 = COzH, R3 = 1: (2R~-[2-chloro-4-
Limits: [(2-hydroxy-3,5-diiodobenzoyl)amino]-5-
-- correction factors: for the calculation of contents, multiply methylphenyl] (4-chlorophenyl)acetic acid,
the peak areas of the following impurities by the D. R1 = H, R2 = CONHz, R3 = 1: N-[4-[(1R~-2-amino-
corresponding correction factor: impurity A = 1.5; 1-(4-chlorophenyl)-2-oxoethyl] -5-chloro-2-methylphenyl] -2-
impurity B = 1.3; hydroxy-3,5-diiodobenzamide,
-- impurity G: not more than 2.5 times the area of the
E. R1 = H, R2 = CN, R3 = Cl: 3-chloro-N-[5-chloro-4-
principal peak in the chromatogram obtained with
[(R~-( 4-chlorophenyl)cyanomethyl] -2-methylphenyl] -2-
reference solution (b) (0.5 per cent);
hydroxy-5-iodobenzamide,
-- impurities F, H, 1: for each impurity, not more than
1.5 times the area of the principal peak in the F. R1 + R2 = 0, R3 = 1: N-[5-chloro-4-(4-chlorobenzoyl)-
chromatogram obtained with reference solution (b) 2-methylphenyl] -2-hydroxy-3 ,5-diiodo benzamide,
(0.3 per cent); G. R1 = H, R2 = C(=NH)OCH3 , R3 = 1: methyl (2R~-2-
-- impurities A, B, C, D, E, J: for each impurity, not more [2-chloro-4- [(2-hydroxy-3, 5-diiodobenzoyl)amino]-5-
than the area of the principal peak in the chromatogram methylphenyl] -2-(4-chl orophenyl) acetimidate,
obtained with reference solution (b) (0.2 per cent); H. R1 = H, R2 = CO-OCH3 , R3 = 1: med1yl (2R~-[2-
-- any other impurity: for each impurity, not more than the chloro-4- [(2-hydroxy-3,5-diiodobenzoyl) amino] -5-
area of the principal peak in the chromatogram obtained methylphenyl] (4-chlorophenyl)acetate,
with reference solution (b) (0.2 per cent); I. R1 = R3 = H, R2 = CN: N-[5-chloro-4-[(R~-
-- total: not more than 7.5 times the area of the principal (4-chlorophenyl) cyanomed1yl] -2-med1 ylphenyl] -2-hydroxy -5-
peak in the clu'omatogram obtained with reference iodobenzamide,
solution (b) (1.5 per cent);

»
-- disregard limit: 0.25 times the area of the principal peak in I
the chromatogram obtained with reference solution (b)
(0.05 per cent).
Water (2.5.12) o 1#
eN I
4.8 per cent to 5.8 per cent, determined on 0.250 g. el NH
~ ~
Use a mixture of 1 volume of dimethylformamide R and
4 volumes of 111ethanol R as the solvento
ASSAY
Dissolve 0.500 g in 50 mL of a mixture of 1 volume of
el #

f , * ~

eN
eH 3

anhydrous acetic acid R and 7 volumes of methyl ethyl ketone R.


Titrate with 0.1 M perchloric acid, determining the end-point el
potentiometrically (2.2.20).
1 mL of 0.1 M perchlO1ic acid is equivalent to 68.5 mg J.N-[5-chloro-4-[[4-[[2-chloro-4-[(2-hydroxy-3,5-
of CzzH13ClzIzNzNaOz. diiodobenzoyl) amino] -5-methylphenyl] cyanomethyl] phenyl]
STORAGE (4-chlorophenyl)cyanomethyl] -2-methylphenyl] -2-hydroxy-
In an airtight container, protected from light. 3,5-diiodobenzamide.
___________________________________________ ~E~

IMPURITIES
Specijied impurities: A, B, C, D, E, F, G, H, 1, J.

A. 2-hydroxy-3,5-diiodobenzoic acid,
2016 Cobalt Oxide Vet-53

immediately with 0.02M sodiu111 thiosulfate VS, using stareh


Cloxacillin Benzathine mueilage, added towards the end of the titration, as indicator.
The difference between the titrations represents the volurne
of O.OlM iodine VS equivalent to the total penicillins presento
p I Me H H

of!
H Calculate the content of C16HzoNz,(C19HlSC1N30SS)Z from
~ N ~ ~ S the difference obtained by canying out the assay

~
~ - Me

OoJ=t-fMe
I
CI H COOH
simultaneously using cloxacillin benzathine BPCRS and from
the declared content of C16HzoNz,(C19HlSC1N30SS)Z in
cloxaeillin benzathine BPCRS.
For benzathine
To 1 g add 30 mL of a saturated solution of sodium ehloride
32222-55-2 and 10 mL of 5M sodium hydroxide, shake well, and extract
with four 50 mL quantities of ether. Wash the combined
Action and use extracts with three 10 mL quantities of water, extract the
Penicillin antibacterial. combined washings with 25 mL of ether and add the extract
Preparations to the main ether solution. Evaporate the ether solution to
Cloxacillin Benzathine Intramammary Infusion (Dry Cow) low bulk, add 2 mL of absolute ethanol and evaporate to
Ampicillin Trihydrate and Cloxacillin Benzathine dlyness. To the residue add 50 mL of anhydrous aeetie acid
IntramammalY Infusion (Dly Cow) and titrate with O.lM perehlorie acid VS, using 0.1 mL of
l-naphtholbenzein solution as indicator. Repeat the operation
DEFINITION without the substance being examined. The difference
Cloxacillin Benzathine is N,N' -dibenzylethylenediammonium between the titrations represents the amount of perchloric
bis [( 6R)-6-(3-0-chlorophenyl-5-methylisoxazole-4- acid required to neutralise the liberated base. Each mL of
carboxamido)penicillanate]. It contains not less than 92.0% O.lM perehlorie aeid VS is equivalent to 12.02 mg of
of C16HzoNz,(C19HlSCIN30SS)Z and not less than 20.0% C16HzoNz·
and not more than 22.0% of benzathine, C16HzoNz, each STORAGE
calculated with reference to the anhydrous substance. Cloxacillin Benzathine should be kept in an airtight
CHARACTERISTICS container. If the material is sterile, the container should be
A white 01' almost white powder. sterile, tamper-evident and sealed so as to exclude micro-
Slightly soluble in water; freely soluble in 17lethanol; slightly organisms.
soluble in ethanol (96%) and in propan-2-01. LABELLING
IDENTIFICATION The label states, where applicable, that the material is sterile.
A. The infrared absO/ptioll spectrum, Appendix II A, is Cloxacillin Benzathine intended for use in the manufacturer of
concordant with the refere71ee ~pectrul1l of cloxacillin either a parenteral dosage for711 01' an imranzammar.y infusion
benzathine (RSV 12). without a furrher appropriate sterilisation proeedure eomplies 'luith
B. Shake 0.1 g witll 1 mL of 1M sodiWll hydroxide for the following additional requirement.
2 minutes, add 2 mL of ether, shake for 1 minute and allow Sterility
to separate. Evaporate 1 mL of the ether layer to dryness, Complies with the test for sterility, Appendix XVI A.
dissolve the residue in 2 mL of glacial aeetie acid and add
1 mL of dilute potassium diehromate solution. A golden yellow
precipitate is produced.
C. Shake 50 mg with 10 mL of water and filter. To 5 mL of
the filtrate add a few drops of silver nitrate solution.
Cobalt Oxide
No precipitate is produced. Heat 50 mg with 2 mL of 240.8 1307-96-9
aleoholie potassium hydroxide solution on a water bath for
15 minutes, add 15 mg of aetlvated eharcoal, shake and filter. Action and use
Acidify the filtrate with 2M nitrie acid. The solution yields Used in the prevention of cobalt deficiency in ruminants.
reaction A characteristic of ehlorides, Appendix VI.
DEFINITION
TESTS Cobalt Oxide consists of cobalt(n,m) oxide (tricobalt
Water tetraoxide) with a small proportion of cobalt(m) oxide
Not more than 5.0% w/w, Appendix IX C. Use 0.5 g. (dicobalt trioxide). It contains not les s than 70.0% and not
ASSAY more than 75.0% of Co, calculated with reference to the
0
substance ignited at about 600 •
For cloxacillin benzathine
To 60 mg add 40 mL of methanol, shake to dissolve, add CHARACTERISTICS
25 mL of 1M sodium hydroxide and allow to stand for A blackpowder.
30 minutes. Add 27.5 mL of 1M hydroehlorie acid and Practically insoluble in water. It dissolves in mineral acids and
sufficient water to produce 100 mL, mix, transfer 20 mL of in solutions of tlle alkali hydroxides.
the solution to a stoppered flask, add 30 mL of O.OlM iodine
VS, close the flask with a wet stopper and allow to stand for IDENTIFICATION
15 minutes protected from light. Titrate the excess of iodine A. Dissolve 50 mg, with warming, in 5 mL of hydroehlorie
with 0.02M sodiu11Z thiosulfate VS, using stareh mueilage, added acid and add 10 mL of water. To 2 mL of the solution add
towards the end of the titration, as indicator. Add a further 1 mL of 5M sodium hydroxide. A blue precipitate which
12 mg of the substance being examined to 10 mL of water, becomes pink on warming is produced. Reserve the
swirl to dIsperse, add 30 mL of O.OlM iodine VS and titrate remainder of the solution for use in test B.
Vet-54 Decoquinate 2016

B. Neutralise 10 mL of the solution reserved in test A with Related substances


5M sodium hydroxide and add 0.5 mL of 6M acetic acid and Carry out the method for thin-layer chromatography,
10 mL of a 10% w/v solution of potassium nitlite. A yellow Appendix In A, using the following solutions.
crystalline precipitate is produced. (1) 1.0% w/v of the substance being examined in chlo1'Oform,
Loss on ignition prepared with the aid of heat.
When ignited at about 600°, loses not more than 1.0% of its (2) 0.0050% w/v of diethyl 4-decyloxy-3-
weight. Use 1 g. ethoxyanilinomethylenemalonate BPCRS in chlo1'Ofo1'111.
ASSAY (3) 0.010% w/v of the substance being examined in
Dissolve 0.1 g in 20 mL of hyd1'OchlO1ic acid, by repeated chloroform.
evaporation if necessary. Add 300 mL of 'Water, 4 g of CHROMATOGRAPHIC CONDITIONS
hyd1'Oxylamine hyd1'Ochloride and 25 mL of 13.5M aml1zonia. (a) Use as the coating silica gel F254 (Merck silica gel 60 F 2S4
Warm to 80° and titrate with 0.05M disodium edetate VS, plates are suitable).
using methyl thymol blue mixture as indicator, until the colour
changes from blue to purple. Each mL of 0.05M disodium (b) Use the mobile phase as described below.
edetate VS is equivalent to 2.946 mg of Co. (c) Apply 10 ¡.tI of each solution.
(d) Develop the plate to 15 cm.
(e) After removal of the plate, dry in air and examine under
ultraviolet light (254 nm).
Decoquinate MOBILE PHASE
5 volumes of anhydrous fo1'mic acid, 10 volumes of absolute
ethanol and 85 volumes of chloroform.
LIMITS
Any secondary spot corresponding to diethyl 4-decyloxy-3-
ethoxyanilinomethylenemalonate in the chromatogram
obtained with salution (1) is not more intense than the spat
in the chromatogram obtained with solution (2) (0.5%) and
417.6 18507-89-6 any other secondaly spot is nat more intense than the spot in
the chromatogram obtained with solution (3) (1 %).
Action and use
Loss on drying
Antiprotozoal (veterinary).
When dried to constant weight at 105°, loses not more than
Preparation 0.5% of its weight. Use 1 g.
Decoquinate Premix
Sulphated ash
DEFINITION Not more than 0.1 %, Appendix IX A.
Decoquinate is ethyl 6-decyloxy-7 -ethoxy-4- ASSAY
hydroxyquinoline-3-carboxylate. It contains not less than Dissolve 1 g in a mixture of 50 mI of chloroform and 50 mI of
99.0% and not more than 101.0% of C24H3SNOs, calculated anhydrous acetic acid and carry out Method I for 11on-aqueous
with reference to the dried substance. titration, Appendix VIII A, using C1ystal violet solution as
CHARACTERISTICS indicator. Each mI of O.lM perchl017c acid VS is equivalent to
A cream to buff-coloured, microcrystalline powder; odourless 41.76 mg of C24H3SNOs.
or almost odourless.
Insoluble in 'Wate/"; very slightly soluble in chlo1'Ofon11 and in
ether; practically insoluble in ethanol (96%).
IDENTIFICATION Deltameth ri n
A. The infrared abs01ption spectrum, Appendix n A, is
concordant with the reference spectrwl1 of decoquinate Me

~rH H:p~I ÚI
(RSV 14).
B. The light abs01ption, Appendix n B, in the range 230 to Me
350 nm of the solution used in the test for Light absorption Sr O, ~ O ~
exhibits a well-defined maximum only at 265 nm. :::
O H eN
TESTS
Light absorption 505.2 52918-63-5
Dissolve 40 mg in 10 mI of hot chlo1'Oform and, keeping the
solution warm, dilute slowly with 70 mI of absolute ethanol. Action and use
Cool and dilute to 100 mI with absolute ethanol. Irnmediately Insecticide (veterinary).
dilute 10 mI to 100 mI with absolute ethanol. To 10 mI of the Preparation
solution add 10 mI of O.lM hyd1'OchlO1ic acid and dilute to Deltamethrin Pour:-on
100 mI with absolute ethanol. The absorbance of the resulting
solution at the maximum at 265 nm is 0.38 to 0.42, DEFINITION
calculated with reference to the dried substance, Deltamethrin is (S)-a-cyano-3-phenoxybenzyl-( 1R,3R)-3-
Appendix n B. (2,2-dibromovinyl)-2,2-dimethylcyclopropane carboxylate.
It contains not less than 97.0% and nat more than 101.0%
of C22H19BrzN03'
2016 Deltamethrin Vet-55

CHARACTERISTICS CHROMATOGRAPHIC CONDITIONS


A white to buff-coloured, crystalline powder. (a) Use a silica gel F'254 precoated plate (Merck silica gel 60
Insoluble in 'Water; soluble in ethanol (96%) and in acetone. F'254 plates are suitable).
IDENTIFICATION (b) Use the mobile phase as described below.
A. The injrared abs01ption spectru11l, Appendix II A, is (c) Apply 10 /11 of each solution.
concordant with the reference spectrum of deltamethrin (d) Develop the plate to 15 cm.
(RSV 47). (e) After removal of the plate, dry in air and examine under
B. In the test for Related substances, the principal spot in the ultraviolet light (254 nm).
chromatogram obtained with solution (2) corresponds to that MOBILE PHASE
in the chromatogram obtained with solution (5).
20 volumes of di-isopropyl ether and 80 volumes of hexane.
TESTS
LIMITS
Specific optical rotation
Any secondaly spot in the chromatogram obtained with
In a 4% w/v solution in toluene, +55.5 to +61.5,
solution (1) is not more intense than the spot in the
Appendix V F.
chromatogram obtained with solution (3) (1 %) and not more
Becisthemic acid chloride than two such spots are more intense than the spot in the
Not more than 0.2% when detelmined by the following chromatogram obtained with solution (4) (0.5%).
method. Dissolve 2 g in 100 mI of methanol with moderate
heating, if necessary, and coo1. Titrate with 0.02M potassium ASSAY
hydroxide VS using a solution containing 0.8% w/v of dimethyl Carry out the method for liquid chromatography,
yello'W and 0.08% w/v of methylene blue in methanol as Appendix III D, using the following solutions in the mobile
indicator to a green end point. Each mI of 0.02M potassium phase.
hydroxide VS is equivalent to 6.329 mg of becisthemic acid (1) 0.1 % w/v of the substance being examined.
chloride, CsHgBr2ClO. (2) 0.1 % w/v of deltamethl1n BPCRS.
Becisthemic acid and becisthemic anhydride (3) 0.1 % w/v of deltameth1in imp'w1ty standard BPCRS.
Not more than 1% in total when determined by the following
CHROMATOGRAPHIC CONDITIONS
methods.
(a) Use a stainless steel column (25 cm x 4.6 mm) packed
Becisthemic acid with silica gel for chromatography (5 ~l1n) (Zorbax Sil is
Dissolve 2 g in 100 mI of ethanol (96%) with moderate suitable) .
heating. Cool in an ice-bath and immediately titrate with
(b) U se isocratic elution and the mobile phase described
0.02M sodium hydroxide VS using a 1 % w/v solution of
below.
l-naphtholbenzein in ethanol (96%) solution as indicator to a
green end point. Correct the volume of titrant for any (c) U se a flow rate of 1.3 mI per minute.
contribution due to the becisthemic acid chloride content (d) U se an ambient column temperature.
using the following expression: (e) Use a detection wavelength of 278 nm.
(f) Inject 20 ~tl of each solution.
MOBILE PHASE
where V titration volume obtained in the becisthemic
acid chloride test, 0.04 volume of propan-2-ol, 2 volumes of acetonitrzle,
PI weight of sample used in the becisthemic 10 volumes of dichloromethane and 100 volumes of hexane.
chloride test, SYSTEM SUIT ABILITY
P'2 weight of sample used in this test. The test is not valid unless, in the chromatogram obtained
Each mI of 0.02M sodium hydroxide VS is equivalent to with solution (3), a peak due to (R)-deltamethrin appears
5.959 mg of becisthemic acid, CsHlQBr'202. immediately before the principal peak, as indicated in the
Becisthemic anhydride reference chromatogram supplied with deltamethl1n
To 1.0 g add 10 mI of O.OlM aniline in cyclohexane and 10 mI imp'w1ty standard BPCRS.
of glacial acetic acid. Stopper the flask and allow to stand at DETERMINATION OF CONTENT
room temperature for 1 hour. Titrate with O.OlM perchloric Calculate the content of C'22HI9Br2N03 using the dec1ared
acid VS using clystal violet solution as indicator. Repeat the content of C22HI9Br2N03 in delta111ethrin BPCRS.
procedure omitting the substance being examined. Correct
the volume of titrant for any contribution due to twice the IMPURITIES
becisthemic acid chloride content calculated using the above The impurities limited by the requirements of this
fOlmula. Each mI of O.OlM perchloric acid VS is equivalent to monograph inc1ude:
5.779 mg ofbecisthemic anhydride, CI6HISBr403. - Becisthemic acid,
- Becisthemic anhydride,
Related substances - Becisthemic acid chloride.
Carry out the method for thin-layer chromatography,
Appendix III A, using the following solutions in toluene.
(1) 2.0% w/v of the substance being examined.
(2) 0.5% w/v of the substance being examined.
(3) 0.020% w/v of the substance being examined.
(4) 0.010% w/v ofthe substance being examined.
(5) 0.5% w/v of deltamethrin BPCRS.
Vet-56 Dembrexine Hydrochloride Monohydrate 2016

Dembrexine Hydrochloride *** Time Mobile phase A Mobile phase B

** ** (min) (per cent VIV) (per cent VIV)


Monohydrate ***** 0-7 75 25

(De111brexine Hydrochlonde Monohydrate for 7 - 15 75 ~ 50 25 ~ 50


Vetennary Use, Ph Bur 1110nogmph 2169) 15 - 20 50 50

20 - 25 50 ~ 75 50 ~ 25

25 - 30 75 25
, Hel, H2 0

Flow rate 1.0 mLlmin.


Detection Specu-ophotometer at 250 nm.
Injection 10 ¡..tL.
C13HlSBr2ClN02,H20 433.6 52702-51-9
Relative retention With reference to dembrexine (retention
PhE~ ___________________________________________
time = about 6 min): impurity A = about 2.3;
DEFINITION impurity B = about 1.3.
tmns-4-[(3,5-Dibromo-2-hydroxybenzyl)amino]cyc1ohexanol System suitability: reference solution (b):
hydrochIoride monohydrate. - resolution: minimum 2 between the peaks due to
Content dembrexine and impurity E.
98.0 per cent to 101.0 per cent (anhydrous substance). Li111its:
- impunties A, B: for each impurity, not more than the area
CHARACTERS of the principal peak in the chromatogram obtained with
Appearance reference solution (a) (0.2 per cent);
White or almost white, crystalline powder. - unspecified i111pUtities: for each impurity, not more than the
Solubility area of the principal peak in the chromatogram obtained
Slight1y soluble in water, freely soluble in methanol, slight1y with reference solution (a) (0.2 per cent);
soluble in anhydrous ethano!. - total: not more than 2.5 times the area of the principal
IDENTIFICATION peak in the chromatogram obtained with reference
solution (a) (0.5 per cent);
A. Infrared absorption spectrophotometry (2.2.24).
- disregard limit: 0.5 times the area of the principal peak in
C0111panson de111brexine hydrochlotide monohydmte CRS. the chromatogram obtained with reference solution (a)
B. It gives reaction (a) of chIorides (2.3.1). (0.1 per cent); disregard any peak due to the blank.
TESTS Water (2.5.12)
Related substances 3.5 per cent to 5.0 per cent, detelmined on 0.500 g.
Liquid chromatography (2.2.29). Prepare the solutions Sulfated ash (2.4.14)
immediately before use. Maximum 0.1 per cent, determined on 1.0 g.
Test solution Dissolve 25.0 mg of the substance to be
ASSAY
examined in 111ethanol R and dilute to 10.0 mL with the same
Dissolve 0.350 g in 40 mL of methanol R. Add 40 mL of
solvento
acetone R and 1 mL of 0.1 M hydrochloric acid. Carry out a
Reference solution (a) Dilute 1.0 mL of the test solution to potentiometric titration (2.2.20) using 0.1 M sodium
50.0 mL with methanol R. Dilute 1.0 mL of this solution to hydroxide. Read the volume added between the 2 points of
10. O mL with methanol R. infiexion.
Reference solution (b) Dissolve 2.5 mg of t1"ibromophenol R 1 mL of 0.1 M sodium hydroxide is equivalent to 41.56 mg
(impurity E) in methanol R and dilute to 50.0 mL with the of C13HlSBr2CIN02'
same solvento To 1.0 mL of this solution add 1.0 mL of the
test solution and dilute to 10.0 mL with methanol R. IMPURITIES
Blank solution Methanol R. Specified i111punties A, B.
Column: Other detectable i111purities (the following substances would, if
- size: l = 0.15 m, 0 = 4.0 mm; present at a sufficient level, be detected by one or other of
- stationary phase: end-capped octadecylsilyl silica gel for the tests in the monograph. They are limited by the general
chromatography R (5 ¡..tm); acceptance criterion for other/unspecified impurities and/or
- te111pemture: 40 oc. by the general monograph Substances for phar111aceutical use
(2034). It is therefore not necessary to identify these
Mobile phase:
impurities for demonstration of compliance. See also 5.10.
- mobile phase A: dissolve 1.0 g of potassium dihydrogen
Control of i111punties in substances for pharmaceutical use): C, D,
phosphate R in 900 mL of water R, adjust to pH 7.4 with
E.
0.5 M potassium hydroxide and dilute to 1000 mL with
water R; mix 80 volumes of this solution with 20 volumes H~ __ H

HO"~NÁ/OH
of methanol R;
- 1110bile phase B: methanol R, acetonitnle R (20:80 V/V);

Sr
)J Sr

A. tmns-4-[(3,5-dibromo-2-
hydroxybenzylidene )amino] cyc1ohexanol,
2016 Detomidine Hydrochloride Vet-57

Test solution Dissolve 25 mg of the substance to be examined


in 20 mL of the mobile phase and dilute to 50.0 mL with
the mobile phase.
Reference solution (a) Dilute 0.20 mL of the test solution to
100.0 mL with the mobile phase.
B. cis-4- [(3, 5-dibromo-2-hydroxybenzyl) amino] cyclohexanol, Reference solution (b) Dissolve 1 mg of detomidine
i111purity B CRS in the mobile phase and dilute to 100.0 mL
R1 with the mobile phase. Dilute 1.0 mL of the solution to
~OH 10.0 mL with reference solution (a).
Column:
R2 M R3 -- size: 1 = 0.15 m, 0 = 4.6 mm;
-- stationary phase: end-capped octylsilyl silica gel for
C. R1 = CHO, R2 = R3 = Br: 3,5-dibromo-2- chromatography R (5 [lm).
hydroxybenzaldehyde, Mobile phase acetonitrile Rl, 2.64 giL solution of am1110nium
D. R1 = CHO, R2 = R3 = H: 2-hydroxybenzaldehyde phosphate R (35:65 V/V).
(salicylaldehyde) , Flow rate 1 mUmin.
E. R1 = R2 = R3 = Br: 2,4,6-tribromophenol. Detection Spectrophotometer at 220 nm.
___________________________________________ ~E~
Injection 20 [lL.
Run time 4 times the retention time of detomidine.
Jdentification of impurities Use the chromatogram obtained
with reference solution (b) to identify the peak due to
Detomidine Hydrochloride *** impurity B.
*** *** Relative retention With reference to detomidine (retention
(Det0111idine Hydrochlo1"ide for Veterinary Use, *** time = about 6 min): impurity B = about 2.1.
Ph. Eur. 1110nograph 1414) System suitability: reference solution (b):
-- resolution: minimum 5.0 between the peaks due to
detomidine and impurity B.
, Hel Calculation of percentage contents:
-- for each impurity, use the concentration of detomidine in
reference solution (a).
Limits:
222.7 90038-01-0
-- unspecified impurities: for each impurity, maximum
Action and use 0.20 per cent;
Alphaz-adrenoceptor agonist. -- total: maximum 0.5 per cent;
-- reporting threshold: 0.10 per cent.
PhE~ ___________________________________________
Water (2.5.12)
DEFINITION Maximum 2.0 per cent, determined on 0.250 g.
4- [(2,3-Dimethylphenyl)methyl]-lH-imidazole hydrochloride. Sulfated ash (2.4.14)
Content Maximum 0.1 per cent, determined on 1.0 g.
99.0 per cent to 101.0 per cent (anhydrous substance). ASSAY
CHARACTERS Dissolve 0.170 g in 50 mL of ethanol (96 per cent) R.
Appearance Add 5.0 mL of 0.01 M hydrochloric acid. Carry out a
White or almost white, hygroscopic, crystalline powder. potentiometric titration (2.2.20), using 0.1 M sodiu112
Solubility hydroxide. Read the volume added between the 2 points of
Soluble in water, freely soluble in ethanol (96 per cent), very inflexion.
slightly soluble in methylene chloride. 1 mL of 0.1 M sodium hydroxide is equivalent to 22.27 mg of
C12HlSClN2'
IDENTIFICATION
A. Infrared absorption spectrophotometry (2.2.24). STORAGE
C0111parison deto111idine hydrochloride CRS. In an airtight container.
If the spectra obtained show differences, dry the substance to IMPURITIES
be examined and the reference substance separately in an Other detectable impurities (the following substances would, if
oven at 105 oC and record new spectra. present at a sufficient level, be detected by one or other of
B. It gives reaction (a) of chlorides (2.3.1). the tests in the monograph. They are limited by the general
acceptance criterion for other/unspecified impurities and/or
TESTS by the general monograph Substances for pharmaceutical use
Appearance of solution (2034). It is therefore not necessary to identify these
The solution is clear (2.2.1) and colourless (2.2.2, impurities for demonstration of compliance. See also 5.10.
Method JI). Control of i112purities in substances for pharmaceutical use): A, B,
Dissolve 0.25 g in water R and dilute to 25 mL with the C.
same solvento
Related substances
Liquid chromatography (2.2.29).
Vet-58 Diclazuril 2016

(j" rI N:~
Reference solution (a) Dissolve 5 mg of diclazuril for system
HCYY
3
i {/ and enantiomer suitability CRS in dimethylformamide R and dilute to 5.0 mL
with the same solvento
CH 3 H OH
Reference solution (b) Dilute 1.0 mL of the test solution to
A (RS)-(2,3-dimethylphenyl) (1 H-imidazol-4-yl)methanol, 100.0 mL with dimethylformamide R. Dilute 5.0 mL ofthe
solution to 20.0 mL with di111ethylforma111ide R.
Column:
-- size: 1 = 0.10 m, 0 = 4.6 mm,
-- stationa1Y phase: base-deactivated octadecylsilyl silica gel for
chromatography R (3 ~lm),
-- te111perature: 35 oc.
Mobile phase:
B. (RS)-(1-benzyl-1H-imidazol-5-yl) (2,3- -- mobile phase A: mix 10 volumes of a 6.3 gIL solution of
dimethylphenyl)methanol, am1110nium fonnate R adjusted to pH 4.0 with anhydrous
formic acid R, 15 volumes of acetonitrile R and 75 volumes
of water R,
-- J110bile phase B: mix 10 volumes of a 6.3 gIL solution of
ammoniu111 formate R adjusted to pH 4.0 with anhydrous
fonnic acid R, 85 volumes of acetonitnle R and 5 volumes
of water R,
C.4-[(2,3-dimethylcyclohexyl)methyl]-lH-imidazole.
___________________________________________ ~Ew
Time Mobile phase A Mobile phase B
(min) (per cent V/V) (per cent v/V)
0-20 100 -7 O 0-7100

20 - 25 O 100
Diclazuril ***
*** ***
(Diclazuril for Veterina1Y Use) *** Flow rate 1.0 mUmin.
Ph Eur monograph 1718) Detection Spectrophotometer at 230 nm.
Injection 5 ~lL.
System suitability: reference solution (a):
-- peak-to-valley ratio: minimum of 1.5, where H p = height
and enantiomer
above the baseline of the peak due to impurity D and
H v = height above the baseline of the lowest point of the
curve separating this peak from the peak due to diclazuril.
Limits:
407.6 101831-37-2 -- correction factors: for the calculation of contents, multiply
the peak areas of the following impurities by the
Action and use corresponding correction factor: impurity D = 1.9;
Antiprotozoal (veterinary); coccidiosis. impurity H = 1.4,
PhEw ___________________________________________ -- impurity D: not more than 0.4 times the area of the
principal peak in the chromatogram obtained with
DEFINITION reference solution (b) (0.1 per cent),
(RS)-( 4-Chlorophenyl) [2,6-dichloro-4-(3 ,5-dioxo-4,5- -- any other impurity: not more than the area of the principal
dihydro-1 ,2,4-triazin-2 (3H)-yl)phenyl] acetonitrile. peak in the chromatogram obtained with reference
Content solution (b) (0.25 per cent),
99.0 per cent to 101.0 per cent (dried substance). -- total: not more than 4 times the area of the principal peak
in the chromatogram obtained with reference solution (b)
CHARACTERS
(1.0 per cent),
Appearance -- disregard limit: 0.2 times the area of the principal peak in
White or light yellow powder. the chromatogram obtained with reference solution (b)
Solubility (0.05 per cent).
Practically insoluble in water, sparingly soluble in Loss on drying (2.2.32)
dimethylformamide, practically insoluble in alcohol and Maximum 0.5 per cent, determined on 1.000 g by drying in
methylene chloride. an oven at 105 oC for 4 h.
IDENTIFICATION Sulfated ash (2.4.14)
Infrared absorption spectrophotometry (2.2.24). Maximum 0.1 per cent, determined on 1.0 g.
Companson Ph. Eur. reference spectrum of diclazunl.
ASSAY
TESTS Dissolve 0.150 g' in 75 mL of dimethylformamide R. Carry out
Related substances a potentiometric titration (2.2.20), usil1g 0.1 M
Liquid chromatography (2.2.29). tetrabutylammonium hydroxide. Read the volume added at the
Test solution Dissolve 20.0 mg of the substance to be second inflexion point. Carry out a blank titration.
examined in dimethylformamide R and dilute to 20.0 mL with 1 mL of 0.1 M tetrabutylam1110nium hydroxide is equivalent to
the same solvento 20.38 mg of C 17H 9 CbN4 0 2 .
2016 Difloxacin Hydrochloride Trihydrate Vet-59

STORAGE
Difloxacin Hydrochloride ****
Protected from light. ** *
IMPURITIES
Trihydrate *****
Specified impurities: A, B, e, D, E, F, G, H, 1. (Difloxacin Hydrochloride Tn}¡,ydrate for Veterinary
Use, Ph Bur 1110nograph 2239)

~o
R~ el~N)lNH andenantiomer

~0o
R'

A. R = el, R' = e0 2 H: 2-[3,5-dichloro-4-[(RS)-


(4-chlorophenyl)cyanomethyl]phenyl]-3,5-dioxo-2,3,4,5-
tetrahydro-1,2,4-triazine-6-carboxylic acid,
490.0 91296-86-5
B. R = OH, R' = H: (RS)-[2,6-dichloro-4-(3,5-dioxo-4,5-
dihydro-1,2,4-triazin-2(3H)- Action and use
yl)phenyl] (4-hydroxyphenyl) acetonitrile, Fluroquinolone antibacterial.
C. R = el, R' = eONH2 : 2-[3,5-dichloro-4-[(RS)- ~Ew _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ ___
(4-chlorophenyl) cyanomethyl] phenyl] -3,5-dioxo-2,3,4,5-
tet:rahydro-1,2 ,4-triazine-6-carboxamide, DEFINITION
G. R = el, R' = eO-0-[eH 2h-eH 3 : butyl 2-[3,5-dichloro- 6-Fluoro-1-(4-fluorophenyl)-7 -(4-methylpiperazin1-yl)-4-oxo-
4- [(RS)-( 4-chlorophenyl) cyanomethyl] phenyl] -3,5-dioxo- 1,4-dihydroquinoline-3-carboxylic acid hydrochloride
2,3,4,5-tetrahydro-1,2,4-triazine-6-carboxylate, trihydrate.
Content
X el 99.0 per cent to 101.0 per cent (anhydrous substance).

~o CHARACTERS
el~ el~N)lNH Appearance
White 01' light yellow, crystalline powder.
~0 o Solubility
Slightly soluble in water and in methanol, very slightly
D. X = O: 2-[3,5-dichloro-4-(4-chlorobenzoyl)phenyl]-1,2,4- soluble in methylene chloride.
triazine-3,5(2H,4H)-dione, It shows polymorphism (5.9).
F. X = H 2 : 2-[3,5-dichloro-4-(4-chlorobenzyl)phenyl]-1,2,4- IDENTIFICATION
triazine-3,5(2H,4H)-dione, A. Infrared absorption spectrophotomet:ry (2.2.24).
Comparison dzfioxacin hydrochloride CRS.
If the spectra obtained in the solid state show differences,
dissolve the substance to be examined and the reference
substance separately in methanol R, evaporate to dryness and
record new spectra using the residues.
B. Suspend 30 mg in 2 mL of 'Water R, acidify with dilute
E. R = NH 2 : (RS)-( 4-amino-2,6-
nitric acid R and filter. The clear filtrate gives reaction (a) of
dichlorophenyl) (4-chlorophenyl) acetonitrile,
chlorides (2.3.1).
H. R = H: (RS)-(4-chlorophenyl)(2,6-
C. Water (see Tests).
dichlorophenyl) acetonitrile,
TESTS
Related substances
Liquid chromatography (2.2.29).
Solvent mixture acetonitrile R, 'Water R (50:50 V/V).
Solution A Dissolve 2.72 g of potassiu111 dihydrogen phosphate R
in 900 mL of 'Water R and adjust to pH 2.5 with phosphonc
acid R; dilute to 1000 mL with 'Water R.
Test solution Dissolve 30.0 mg of the substance to be
examined in 50.0 mL of the solvent mixture and dilute to
100.0 mL with mobile phase A.
1. N,2-bis[3,5-dichloro-4- Reference solution (a) Dissolve 6.0 mg of difloxacin
[(4-chlorophenyl)cyanomethyl]phenyl]-3,5-dioxo-2,3,4,5- impurity G CRS in acetonitn7e R and dilute to 20.0 mL with
tetrahydro-1,2,4-triazine-6-carboxamide. the same solvento
- _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ PhEur Reference solution (b) Mix 0.5 mL of reference solution (a),
1.0 mL of the test solution and 50 mL of the solvent mixture
and dilute to 100.0 mL with mobile phase A.
Vet-60 Difioxacin Hydrochloride Trihydrate 2016

Referenee solution (e) Dissolve 3 mg of sarafioxacin Sulfated ash (2.4.14)


hydroehloride R (impurity B) in 100.0 mL of solution A. Maximum 0.1 per eent, determined on 1.0 g in a platinum
Dilute 1.0 mL of the solution to 50.0 mL with the test erueible.
solution.
ASSAY
Column: Dissolve 0.150 g in 5 mL of anhydrous formie acid R and add
- size: 1 = 0.15 m, 0 = 4.6 mm; 50 mL of aeetie anhydrz'de R. Titrate with 0.1 M perehlorie
- stationary phase: end-capped oetadeeylsilyl siliea gel for aeid, detetmining the end-point potentiometrieally (2.2.20).
ehronzatography R (5 ~lm). Read the volume added at the 2nd point of infiexion.
Mobile phase: 1 mL of 0.1 M perehlorie acid is equivalent to 21.79 mg of
- mobile pltase A: aeetonitrile R, tetrahydrofuran R, solution A
C21H20ClF2N303'
(5:5:90 V/V/V);
- mobile phase B: aeetonitrz'le R, solution A, tetrahydrofuran R IMPURITIES
(5:35:60 V/V/V); Speeified impurities G
Other detectable impurities (the following substanees would, if
Time Mobile phase A Mobile phase B present at a suffieient level, be deteeted by one or other of
(min) (per cent Vil') (per cent Vil') the tests in the monograph. They are limited by the general
0-15 100 O aeeeptanee eriterion for otherlunspeeified impurities and/or
by the general monograph Substanees for pharmaeeutieal use
15 - 50 100 -7 O 0-7100
(2034). It is therefore not neeessary to identify these
50 - 60 O 100 impurities for demonstration of eomplianee. See also 5.10.
Control of impurities in substanees for pha17naeeutieal use): A, B,
C,D,E,F.
Flow rate 1. O mUmin.
Detection Speetrophotometer at 325 nm.
Injection 30 ~L of the test solution and referenee solutions (b)
and (e).
Identifieation of impurities Use the ehromatogram obtained
with referenee solution (e) to identify the peak due to
impurity B; use the ehromatogram obtained with referenee
solution (b) to identify the peak due to impurity G.
Relative retention With referenee to difioxaein (retention
time = about 10 min): impurity B = about 1.2;
impurity G = about 4.0.
System suitability: referenee solution (e):
- peak-to-valley ratio: minimum 2.0, where Hp = height
above the baseline of the peak due to impurity B and
H v = height above the baseline of the lowest point of the A. 6-fiuoro-7 -(4-methylpiperazin-1-yl)-1- [4-
curve separating this peak from the peak due to (4-methylpiperazin-1-yl)phenyl] -4-oxo-1 ,4-dihydroquinoline-
difioxaein. 3-earboxylie aeid,
Li711its:
- impurity G: not more than the area of the eorresponding
peak in the ehromatogram obtained with referenee
solution (b) (0.5 per eent);
- unspeeified impurities: for eaeh impurity, not more than
0.2 times the area of the peak due to difioxaein in the
ehromatogram obtained with referenee solution (b)
(0.20 per eent);
- total: maximum 1.0 per eent;
- disregard limit: 0.1 times the area of the peak due to
difioxaein in the ehromatogram obtained with referenee B. 6-fiuoro-1-( 4-fiuorophenyl)-4-oxo- 7-piperazin-1-yl-1,4-
solution (b) (0.10 per eent). dihydroquinoline-3-earboxylie aeid (sarafioxaein),
Heavy metals (2.4.8)
Maximum 20 ppm.
Solvent mixture Dissolve 30 g of propylene glyeol R in 30 mL
of methanol R, add 4 g of arginine R and dilute to 100 mL
with water R.
0.25 g eomplies with test H. Prepare the referenee solution
using 0.5 mL of lead standard solution (10 ppm Pb) R.
The substanee precipita tes after addition of buffer solution
pH 3.5 R. Dilute to 20 mL with methanol R; the substanee
re-dissolves eompletely.
Water (2.5.12) C. 1-(4-ehlorophenyl)-6-fiuoro-7 -(4-methylpiperazin-1-yl)-4-
8.0 per eent to 12.0 per eent, determined on 0.100 g, using a oxo-1 ,4-dihydroquinoline-3-earboxylie aeid,
mixture of 20 volumes of formamide R and 25 volumes of
711ethanol R as solvento
2016 Dihydrostreptornycin Sulfate Vet-61

Dihydrostreptomycin Sulfate ***


** **
Dihydrostreptomycin Sulphate *****
(Dihydrostreptomycin Sulfate for Veterina1Y Use,
Ph Bur monograph 0485)

D. 6-chloro-1-(4-fiuorophenyl)-7 -( 4-methylpiperazin-1-yl)-4-
oxo-1 ,4-dihydroquinoline-3-carboxylic acid,

Compound R Molec. Formula M,

E. 7-chloro-1-(4-fiuorophenyl)-6-(4-methylpiperazin-1-yl)-4- dihydrostreptomycin
CH 20H C42HssN14036S3 1461
sulphate
oxo-1 ,4-dihydroquinoline-3-carboxylic acid,
streptomycin
CHO C42Hs4N14036S3 1457
sulphate

5490-27-7

Action and use


Aminoglycoside antibacterial.
~E~ ___________________________________________

DEFINITION
lVlain compound Bis[N,N"'-[(1R,2R,3S,4R,5R,6S)-4-[[5-
deoxy-2-0- [2-deoxy-2-(methylamino )-Ci-L-glucopyranosyl] -3-
F. 6-fiuoro-N, 1-bis (4-fiuorophenyl) -7-( 4-methylpiperazin-1-
C-(hydroxymethyl) -Ci- L-lyxofuranosyl] oxy] -2,5,6-
yl)-4-oxo-1 ,4-dihydroquinoline-3-carboxamide,
trihydroxycyclohexane-1 ,3-diyl] diguanidine] trisulfate.
Sulfate of a substance obtained by catalytic hydrogenation of
streptomycin or by any other means.
Semi-synthetic product derived from a fermentation producto
Stabilisers may be added.
Content
-- sum of the percentage contents of dihydrostreptomycin sulfate
and streptomycin sulfate: 95.0 per cent to 102.0 per cent
(dried substance);
-- streptomycin sulfate: maximum 2.0 per cent (dried
G. 7-chloro-6-fiuoro-1-(4-fiuorophenyl)-4-oxo-1,4- substance) .
dihydroquinoline-3-carboxylic acid. PRODUCTION
___________________________________________ PhE~
The method of manufacture is validated to demonstrate that
the product, if tested, would comply with the following test.
Abnormal toxicity (2.6.9)
Inject into each mouse 1 mg dissolved in 0.5 mL of water for
injections R.
CHARACTERS
Appearance
White or almost white, hygroscopic powder.
Solubility
Freely soluble in water, practically insoluble in acetone, in
ethanol (96 per cent) and in methanol.
IDENTIFICATION
Fint identification A, E
Second identification B, C, D, E
Vet-62 Dihydrostreptomycin Sulfate 2016

A. Examine the chromatograms obtained in the assay. Test solution Dissolve 50.0 mg of the substance to be
Results The principal peak in the cmomatogram obtained examined in water R and dilute to 10.0 mL with the same
with the test solution is similar in retention time and size to solvento
the principal peak in the chromatogram obtained with Referenee solution (a) Dissolve the contents of a vial of
reference solution (a). dihydrostreptomycin sulfate CRS (containing impurities A, B
B. Thin-Iayer cmomatography (2.2.27). and e) in 5.0 mL of water R.
Test solution Dissolve 10 mg of the substance to be examined Referenee solution (b) Dilute 1.0 mL of the test solution to
in water R and dilute to 10 mL with the same solvento 100.0 mL with water R.
Referenee solution (a) Dissolve the contents of a vial of Referenee solution (e) Dilute 5.0 mL of reference solution (b)
dihydrostreptomycin sulfate CRS in 5.0 mL of water R. to 50.0 mL with water R.
Referenee solution (b) Dilute 1.0 mL of reference solution (a) Referenee solution (d) Dissolve 10 mg of streptomycin
to 5.0 mL with water R. sulfate CRS in water R and dilute to 20 rnL with the same
Referenee solution (e) Dissolve 10 mg of kanamycin solvento Mix 0.1 mL of this solution with 1.0 mL of
monosulfate CRS and 10 mg of neomycin sulfate CRS in reference solution (a).
water R, add 2.0 mL of reference solution (a), mix Referenee solution (e) Dilute 1.0 mL of reference solution (a)
thoroughly and dilute to 10 mL with water R. to 100.0 mL with water R.
Plate TLC siliea gel plate R. Column:
- size: l = 0.25 m, 0 = 4.6 mm;
Mobile phase 70 gIL solution of potassium dihydrogen
- stationaJJ' phase: octadeeylsilyl siliea gel for ehromatography R
phosphate R.
(5 ~lm);
Applieatioll 1O ~lL. - temperature: 45 oc.
Development Over 2/3 of the plateo Mobile phase Solution in water R containing 4.6 gIL of
Drying In a current of warm airo anhydrous sodium sulfate R, 1.5 gIL of sodium
Deteetion Spray with a mixture of equal volumes of a 2 giL oetanesulfonate R, 120 mUL of aeetonitlile R1 and 50 mUL of
solution of 1,3-dihydroxynaphthalene R in ethanol a 27.2 giL solution of potassium dihydrogen phosphate R
(96 per eent) R and a 460 gIL solution of sulfurie acid R; heat adjusted to pH 3.0 with a 22.5 giL solution of phosphorie
at 150 oC for 5-10 mino acid R.
System suitability: reference solution (c): Flow rate 1.0 mUmin.
- the cmomatogram shows 3 clear1y separated spots. Detection Spectrophotometer at 205 nm.
Results The principal spot in the cmomatogram obtained with Injection 2 O ~lL.
the test solution is similar in position, colour and size to the Run time 1.5 times the retention time of
principal spot in the chromatogram obtained with reference dihydrostreptomycin.
solution (b).
Identification of impurities Use the chromatogram supplied
e. Dissolve 0.1 g in 2 mL of water R and add 1 mL of (J.- with dihydrostreptomycin sulfate CRS and the chromatogram
naphthol solution R and 2 mL of a mixture of equal volumes obtained with reference solution (a) to identify the peaks due
of strong sodium hypoehlorite solution R and water R. A red to streptomycin and impurities A, B and e.
colour develops.
Relative retention With reference to dihydrostreptomycin
D. Dissolve 10 mg in 5 mL of water R and add 1 mL of 1 M (retention time = about 57 min): impurity A = about 0.2;
hydroehlorie acid. Heat in a water-bath for 2 mino Add 2 mL impurity B = about 0.8; streptomycin = about 0.9;
of a 5 giL solution of (J.-naphthol R in 1 M sodium hydroxide
impurity e = about 0.95.
and heat in a water-bath for 1 mino A violet-pink colour is
System suitability:
produced.
- peak-to-valley ratio (a): minimum 1.1, where H p = height
E. It gives reaction (a) of sulfates (2.3.1). aboye the baseline of the peak due to streptomycin and
TESTS H v = height aboye the baseline of the lowest point of the
Solution S curve separating this peak from the peak due to
Dissolve 2.5 g in earbon dioxide-free water R and dilute to impurity e in the cmomatogram obtained with reference
10 mL with the same solvento solution (d);
- peak-to-valley ratio (b): minimum 5, where H p = height
Appearance of solution
aboye the baseline of the peak due to impurity e and
Solution S is not more intensely coloured than intensity 5 of
H v = height aboye the baseline of the lowest point of the
the range of reference solutions of the most appropriate
curve separating this peak from the peak due to
colour (2.2.2, Method 11). A110w to stand protected from light
dihydrostreptomycin in the cmomatogram obtained with
at about 20 oC for 24 h; solution S is not more opalescent
reference solution (d);
than reference suspension II (2.2.1).
- the cmomatogram obtained with reference solution (a) is
pH (2.2.3) similar to the cmomatogI.~am supplied with
5.0 to 7.0 for solution S. dihydrostreptomycin sulfate CRS.
Specific optical rotation (2.2. 7) Limits:
-83.0 to -91.0 (dried substance). - eorrection factor: for the calculation of content, multiply the
Dissolve 0.200 g in water R and dilute to 10.0 mL with the peak area of impurity A by 0.5;
same solvento - impurity C: not more than twice the area of the principal
Related substances peak in the cmomatogram obtained with reference
Liquid cmomatography (2.2.29). solution (b) (2.0 per cent);
2016 Dimpylate Vet-63

- impurities A, B: for each impurity, not more than the area


of the principal peak in the chromatogram obtained with
reference solution (b) (1.0 per cent);
- any other impwity: for each impurity, not more than the
area of the principal peak in the chromatogram obtained
with reference solution (b) (1.0 per cent);
- total: not more than 5 times the area of the principal peak
in the chromatogram obtained with reference solution (b)
(5.0 per cent);
- disregard limit: the area of the principal peak in the
chromatogram obtained with reference solution (c)
(0.1 per cent); disregard the peak due to streptomycin.
Heavy metals (2.4.8)
20 ppm.
1.0 g complies with test C. Prepare the reference solution B. N,N"'- [( 1S,2R,3R,4S,5R, 6R)-2,4,5-trihydroxy-6- [[~-D­
using 2 mL of lead standard solution (lO ppm Pb) R. mannopyranosyl-(1-+4)-2-deoxy-2-(methylamino)-cx-L-
glucopyranosyl-(1-+ 2)-5-deoxy-3-C-(hydroxymethyl)-cx-L-
Loss on drying (2.2.32)
lyxofuranosyl] oxy] cyclohexane-1 ,3-diyl] diguanidine
Maximum 5.0 per cent, determined on 1.000 g by drying
(dihydrostreptomycin B),
under high vacuum at 60 oC for 4 h.
C. unknown structure,
Sulfated ash (2.4.14)
Maximum 1.0 per cent, determined on 1.0 g.
Bacterial endotoxins (2.6.14)
Less than 0.50 IU/mg, if intended for use in the manufacture
of parenteral preparations without a further appropriate
procedure for removal of bacterial endotoxins.
ASSAY
Liquid chromatography (2.2.29) as described in the test for
related substances with the following modification.
Injection Test solution and reference solutions (a) and (e).
Calculate the percentage content of streptomycin sulfate
using the chromatogram obtained with reference solution (e)
and the declared content of dihydrostreptomycin sulfate CRS.
Calculate the percentage content of dihydrostreptomycin
sulfate using the chromatogram obtained with reference
D. N,N"'-[(lR,2R,3S,4R,5R,6S)-4-[[3,5-dideoxy-2-0-[2-
solution (a) and the declared content of dihydrostreptomycin
deoxy-2-(methylamino )-cx-L-glucopyranosyl] -3-
sulfate CRS.
(hydroxymethyl)-cx-L-arabinofuranosyl] oxy] -2,5,6-
STORAGE trihydroxycyclohexane-1 ,3-diyl] diguanidine
In an airtight container, protected from light. If the substance (deoxydihydrostreptomycin) .
is sterile, store in a sterile, airtight, tamper-proof container. ___________________________________________ PhE~

IMPURITIES
Specijied impurities A, B, C
Other detectable impwities (the following substances would, if
present at a sufficient level, be detected by one 01' other of Dimpylate
the tests in the monograph. They are limited by the general
acceptance criterion for other/unspecified impurities and/or
by the general monograph Substances for pharmaceutical use Me

1~ ~
(2034). It is therefore not necessary to identify these
impurities for demonstration of compliance. See also 5.10.
Control of impwities in substances for pha17naceutical use): D. Pr' N O-P-OEt
I
OEt
NH

HO HN)lNH 2
304.4 333-41-5
HO--~OH Action arid use
Insecticide.
HO HN NH 2
y DEFINITION
NH
Dimpylate is O,O-diethyl O~(2-isopropyl-6-methylpyrimidin-
A. N,N"'-[(lR,2s,3S,4R,5r,6S)-2,4,5,6- 4-yl)phosphorothioate. It contains not less than 95.0% and
tetrahydroxycyclohexane-1 ,3-diyl] diguanidine (streptidine), not more than 101.0% of C12H21N2Ü3PS, calculated with
reference to the anhydrous substance.
It may contain suitable stabilisers such as antioxidants.
Vet-64 Dimpylate 2016

CHARACTERISTICS 3-ethyl-2-isopropyl-6-methyl-4-oxo-3 ,4-dihydropyrimidine is


A clear, yellowish brown, slightly viscous liquido not greater than 2.5 times the area of the peak due to
Practically insoluble in water; miscible with ethanol (96%), dimpylate in the chromatogram obtained with solution (3)
with ether and with most orgaruc solvents. (1 % of each) , the area of any peak corresponding to O,O,S-
triethyl phosphorothioate is not greater than 1.25 times the
IDENTIFICATION area of the peak due to dimpylate in the chromatogram
A. The infrared abs01ption spectrum, Appendix n A, is obtained with solution (3) (0.5%), the are a of any peak
concordant with the reference spectrum of dimpylate (RSV 49). corresponding to tetraethyl thionopyrophosphate is not
B. In the Assay, the chromatogram obtained with solution greater than 0.02 times the area of the peak due to dimpylate
(1) shows a peak with the same retention time as the peak in the chromatogram obtained with solution (3) (0.02%,
due to dimpylate in the chromatogram obtained with assuming a response factor of 0.4), the area of any peak
solution (3). corresponding to tetraethyI dithionopyrophosphate is not
TESTS greater than 0.25 times the area of the peak due to dimpylate
in the chromatogram obtained with solution (3) (0.2%,
Toluene
assuming a response factor of 0.5), the area of any peak
Not more than 1% v/v when determined by the test for
corresponding to O,O-diethyl 0-(2-isopropyl-6-
residual solvents, Appendix VIII L.
methylpyrimidin-4yl)phosphate is not greater than 0.75 times
Related substances the area of the peak due to dimpylate in the chromatogram
Carry out the method for gas chromatography, obtained with solution (3) (0.3%) and the area of any other
Appendix In B, using the following solutions. Solution (1) secondary peak is not greater than 0.5 times the area of the
contains 0.5% w/v of the substance being examined in peak due to dimpylate in the chromatogram obtained with
dichloromethane. Solution (2) contains 0.5% w/v of the solution (3) (0.2%). Disregard any peak corresponding to
substance being examined and 0.025% w/v of diethyl toluene.
phthalate (internal standard) in dichloromethane. Solution (3)
contains 0.0020% w/v of the substance being examined and
Water
Not more than 0.1 % w/w, Appendix IX C. Use 2 g.
0.025% w/v of diethyl phthalate in dichloromethane. Solution
(4) contains 0.5% w/v of dimpylate for chromatography BPCRS ASSAY
and 0.025% w/v of diethyl phthalate in dichloromethane. Carry out the method for gas chromatography,
Solution (5) contains 0.012% v/v of toluene in Appendix In B, using solutions in 4-methylpentan-2-one
dichloromethane. containing (1) 0.2% w/v of the substance being examined,
The chromatographic procedure may be carried out using (2) 0.2% w/v of the substance being examined and
(a) a fused silica capillary column (15 m x 0.32 mm) coated 0.15% w/v of diethyl phthalate (internal standard) and (3)
with a 1 ~lm film of dimethyl silicone gum (SE-54 is suitable) 0.2% w/v of dimpylate BPCRS and 0.15% w/v of diethyl
fitted with a precolumn (0.2 m x 0.53 mm) coated with a phthalate.
1 ~lm film of dimethyl silicone gum, the temperature The chromatographic procedure may be carried out using
programme described below with the inlet port at room (a) a fused silica capillary column (15 m x 0.53 mm) coated
with a 1.5 ¡.tm film of dimethyl silicone gum (SE-54 from J &
0
temperature and the detector at 280 and (b) hydrogen as the
can·ier gas at a fiow rate of 40 mL per minute and nitrogen as W Scientific is suitable) at a temperature of 110° increasing
the make up gas with a fiow rate of 50 mL per minute. linearly at arate of 6° per minute to 215° with the inlet port
at 250° and the detector at 250 0 and (b) helium as the carrier
gas at a fiow rate of 20 mL per minute with nitrogen as the
Time (Minutes) Temperature Comment
make up gas with a fiow rate of 10 mL per minute.
0-1 35° isothermal The assay is not valid unless, in the chromatogram obtained
35°---+180° linear increase
with solution (3), the resolution factor between the peaks due
1 - 15.5
0
10 /minute to dimpylate and the internal standard is at least 2.
Calculate the content of C12H21N2Ü3PS from the
15.5 - 23.5 180° isothermal
chromatograms obtained and using the declared content of
23.5 - 30.5 180°---+250° linear increase C12H21N2Ü3PS in dimpylate BPCRS.
10 0 /minute IMPURITIES
30.5 -40.5 250° isothermal The impurities limited by the requirements of this
monograph include:

In the chromatogram obtained with solution (3) the retention


time of the internal standard is about 18.5 minutes and of
dimpylate, about 23 minutes. The test is not valid unless the
chromatogram obtained with solution (4) closely resembles
that supplied with dimpylate for chromatography BPCRS. A. Diethyl disulfide,
In the chromatogram obtained with solution (2) identify any
peaks corresponding to 4-ethoxy-2-isopropyl-6- S
11
methylpyrimidine, O,O,S-triethyl phosphorothioate, 3-ethyl- CI-P-OEt
1
2-isopropyl-6-methyl-4-oxo-3,4-dihydropyrimidine, tetraethyl OEt
thionopyrophosphate, tetraethyl dithionopyrophosphate and
O,O-diethyl 0-(2-isopropyl-6-methylpyrimidin-4-
B. 0,0-Diethyl chlorophosphorothioate,
yl)phosphate from the chromatogram supplied with dimpylate
for chromatography BPCRS. The area of any peak
corresponding to 4-ethoxy-2-isopropyl-6-methylpyrimidine or
2016 Diprenorphine Hydrochloride Vet-65

Me Me
N~
pri)lNÁOEt Pr l
1\ ~
N S-P-OEt
1
OEt
c. 4-Ethoxy-2-isopropyI-6-methylpyrimidine,
K. 0,0-DiethyI S-(2-isopropyI-6-methylpyrimidin-4-
S yI)phosphorothiophate,
11
EtO-P-OEt
1 Me
OEt

D. O, O, 0-TriethyI phosphorothioate,
Pr l
1\ ~
N O-P-SEt
1

OEt
S
11
EtS-P-OEt
1 L. O,S- DiethyI 0-(2-isopropyI-6-methylpyrimidin-4-
OEt yI)phosphorothiophate,

E. 0, O,S-TriethyI phosphorothioate,

Me
N~
pri)lN~O 1

Et

F. 3-EthyI-2-isopropyI-6-methyI-4-oxo-3,4- M. Bis (2-isopropyI-6-methylpyrimidin-4-yl) sulfide,


dihydropyrimidine,

Me
N~
pri)lNÁOH

G. 4-Hydroxy-2-isopropyI-6-methylpyrimidine,

s
11
O
11
EtO-P- O-P-OEt N. O-ethyI O,O-(2-isopropyI-6-methylpyrimidin-4-
1 1
OEt OEt yl)phosphorothiophate.

H. TetraethyI thionopyrophosphate,

s s Diprenorphine Hydrochloride
11 11
EtO - P - 0- P - OEt
1 1
OEt OEt

I. T etraethyI dithionopyrophosphate,

Me

Pr l
1\ ~
N O-P-OEt
1

OEt
H
Me Me

462.1 16808-86-9

J.0,0-DiethyI 0-(2-isopropyI-6-methylpyrimidin-4- Action and use


yI)phosphate, Opioid receptor antagonist.
Preparation
Diprenorphine Injection
Vet-66 Enilconazole 2016

DEFINITION
Diprenorphine Hydrochloride is N-cyclopropylmethyl-7,8-
Enilconazole
dihydro-7 a-(1-hydroxy-1-methylethyl)-6-0-methyl-6a, 14a- (Bnilconazole for VeterinaJY Use,
ethanonOllliorphine hydrochloride. It contains not less than Ph Bur 11Zonograph 1720)
98.5% and not more than 101.0% of C26H3sN04,HCI,
ca1culated with reference to the dried substance. CI

CHARACTERISTICS
A white or almost white, crystalline powder. CI~H ,
Sparingly soluble in water; slightly soluble in ethanol (96%);
very slightly soluble in chlorofomz; practically insoluble in
H2C~O~ and enantiomer

ether. N

IDENTIFICATION
(17 N
A. The infrared abs01ption spectrum, Appendix n A, is
concordant with the reference spectrum of diprenorphine 297.2 35554-44-0
hydrochloride (RSV 18).
B. The light abs01ption, Appendix n B, in the range 230 to Action and use
350 nm of a 0.02% w/v solution in O.lM hydrochloric acid Antifungal.
exhibits a maximum only at 287 nm. The absorbance at the ~E~ ___________________________________________
maximum is about 0.70.
C. The light absorption, Appendix n B, in the range 230 to DEFINITION
350 nm of a 0.02% w/v solution in O.lM sodium hydroxide 1-[(2RS)-2-(2,4-Dichlorophenyl)-2-(prop-2-enyloxy)ethyl]-
exhibits a maximum only at 301 nm. The absorbance at the 1H-imidazole.
maximum is about 1.1. Content
D. Yields reaction A characteristic of chlorides, Appendix VI. 98.5 per cent to 101.5 per cent (dried substance).
TESTS CHARACTERS
Acidity Appearance
pH of a 2.0% w/v solution, 4.5 to 6.0, Appendix V L. Clear, yellowish, oily liquid or solid mass.
Specific optical rotation Solubility
In a solution prepared by dissolving 0.5 g in sufficient Very slightly soluble in water, freely soluble in ethanol
methanol to produce 25 mL, -97.0 to -107.0, ca1culated with (96 per cent), in methanol and in toluene.
reference to the dried substance, Appendix V F. IDENTIFICATION
Related substances Infrared absorption spectrophotometry (2.2.24).
Can'Y out the method for thin-Iayer chromatography, Comparison enilconazole CRS.
Appendix nI A, using silica gel G as the coating substance
and a mixture of 1 volume of water, 5 volumes of methanol, TESTS
30 volumes of butan-2-one, 80 volumes of acetone and Optical rotation (2.2.7)
100 volumes of cyclohexane as the mobile phase. Apply -0.10° to + 0.10°.
separately to the plate 20 ~lL of each of two solutions of the Dissolve 0.1 g in methanol R and dilute to 10 mL with the
substance being examined in methanol containing (1) same solvento
2.0% w/v and (2) 0.020% w/v. Add at each point of Related substances
application 1O ~lL of a mixture of 4 volumes of methanol and Gas chromatography (2.2.28). Prepare the solutions i11Zmediately
1 volume of 13.5M ammonia. After removal of the plate, before use and protect from light.
allow it to dry in air and spray with a 1% w/v solution of
Test solution Dissolve 0.100 g of the substance to be
iodine in methanol. Any secondaJY spot in the chromatogram
examined in toluene R and dilute to 100.0 mL with the same
obtained with solution (1) is not more intense than the spot
solvento
in the chromatogram obtained with solution (2) (1 %).
Reference solution (a) Dissolve 10. O mg of enilconazole CRS
Loss on drying
and 10.0 mg of enilconazole impurity B CRS in toluene R and
When dried to constant weight at 105°, loses not more than
dilute to 100.0 mL with the same solvento
2.0% of its weight. Use 1 g.
Reference solution (b) Dilute 5.0 mL of the test solution to
Sulfated ash 100.0 mL with toluene R. Dilute 1.0 mL ofthis solution to
Not more tl1an 0.1 %, Appendix IX A. 10.0 mL with toluene R.
ASSAY Column:
Carry out Method I for non-aqueous titration, - material: fused silica;
Appendix VIII A, using 0.5 g, adding 7 mL of mercU1y(JI) - size: l = 25 m, 0 = 0.32 mm;
acetate solution and using c¡ystal violet solution as indicator. - stationalY phase: chemically bonded
Each mL of O.lM perchloric acid VS is equivalent to 46.21 mg poly (dimethyl) (diphenyl) siloxane R (film thickness
C26H3SN04,HCl. 0.52 ~lm).
STORAGE Carné gas hélium for chromatography R.
Diprenorphine Hydrochloride should be protected from light. Plow rate 1.3 mUmin.
Split ratio 1:38.
2016 Enrofioxacin Vet-67

el
Te17lperature:

Time Temperature CI~


H, and enantiomer
(min) (OC)
Column 0-6.4 100 ~ 260 H2e~o~
6.4 - 14 260 N
R2/ 'R1
Injection port 250

Detector 300
A. R1 = R2 = H: (2RS)-2-(2,4-dichlorophenyl)-2-(prop-2-
enyloxy) ethanamine,
Detection Flame ionisation_ B. R1 = H, R2 = CHT CH=CH2: N-[(2RS)-2-(2,4-
Injection 2 ~lL. dichlorophenyl) -2-(prop-2-enyloxy) ethyl] prop-2-en-1-amine,
Identijication of impurities Use the chromatogram obtained C. R1 = CHO, R2 = H: N-[(2RS)-2-(2,4-dichlorophenyl)-2-
with reference solution (a) to identify the peak due to (prop-2-enyloxy)ethyl] formamide,
impurity E. D. R1 = CHO, R2 = CHT CH=CH2: N-[(2RS)-2-(2,4-
Relative retention With reference to enilconazole (retention dichlorophenyl) -2-(prop-2-enyloxy) ethyl] -N-(prop-2-
time = about 10 min): impurity A = aboutO.6; enyl)formamide,
impurity B = about 0.7; impurity C = about 0.8;
impurity D = about 0.9; impurity E = about 1.03;
impurity F = about 1.1.
System suitability: reference solution (a):
-- resolution: minimum 2.5 between the peaks due to and enantiomer
enilconazole and impurity E.
Limits:
-- impunties A, B, C, D, B, F: for each impurity, not more
than twice the area of the principal peak in the
chromatogram obtained with reference solution (b) E. (lRS)-1-(2,4-dichlorophenyl)-2-(-lH-imidazol-1-
(1.0 per cent), and not more than 1 such peak has an yl)ethanol,
area greater than the area of the principal peak in the
chromatogram obtained with reference solution (b) el el
(0.5 per cent);
unspecijied impurities: for each impurity, not more than
0.4 times the area of the principal peak in the
chromatogram obtained with reference solution (b)
(0.20 per cent);
H2e~o~
HO and enantiomer

N
-- total: not more than 4 times the area of the principal peak
in the chromatogram obtained with reference solution (b) () N
(2.0 per cent);
-- disregard limit: 0.1 times the area of the principal peak in F. 1-[(2RS)-2-(3,4-dichlorophenyl)-2-(prop-2-enyloxy)ethyl]-
the chromatogram obtained with reference solution (b)
1H-imidazole.
(0.05 per cent).
___________________________________________ PhE~

Loss on drying (2.2.32)


Maximum 0.5 per cent, determined on 1.000 g by drying
in vacuo at 40 oC for 4 h.
Sulfated ash (2.4.14)
Maximum 0.1 per cent, determined on 1.0 g. Enrofloxacin
ASSAY (Bnrofioxacin for Veterinary Use,
Dissolve 0.230 g in 50 mL of a mixture of 1 volume of Ph Bur 1110nograph 2229)
anhydrous acetic acid R and 7 volumes of methyl ethyl ketone R.
Titrate with 0.1 M perchloric acid using 0.2 mL of
naphtholbenzein solution R as indicator.
1 mL of 0.1 M perchloric acid is equivalent to 29.72 mg of
C14H14Cl2N20.
STORAGE
In an airtight container, protected from light.
IMPURITIES 359.4 93106-60-6
Specijied impurities A, B, C, D, E, F
Actipn and use
Fluoroquinolone antibacterial (veterinary).
PhE~ ___________________________________________

DEFINITION
1-Cyclopropyl-7 -( 4-ethylpiperazin-1-yl)-6-fluoro-4-oxo-1,4-
dihydroquinoline-3-carboxylic acid.
Vet-68 Enrofioxacin 2016

Content Flo'W rate 1.5 mUmin.


98.5 per cent to 101.5 per cent (dried substance). Detection Spectrophotometer at 270 nm.
CHARACTERS Jnjection 1O ~lL.
Appearance Run time 3 times the retention time of enrofioxacin.
Pale yellowish or light yellow, crystalline powder. Identification of i11lpurities U se the chromatogram supplied
Solubility with enrofioxacin for syste11l suitability CRS and the
Practically insoluble in water, freely soluble in methylene chromatogram obtained with reference solution (a) to
chloride, slightly soluble in methanol. identify the peaks due to impurities B and C.
IDENTIFICATION Relative retention With reference to emofioxacin (retention
Infrared absorption spectrophotometry (2.2.24). time = about 16 min): impurity C = about 0.6;
impurity B = about 0.8.
Comparison enrofioxacin CRS.
System suitability: reference solution (a):
TEST - resolution: minimum 2.0 between the peaks due to
Appearance of solution impurity B and enrofioxacin.
The solution is not more opalescent than reference Limits:
suspension II (2.2.1) and not more intensely coloured than - impurity B: not more than 2.5 times the area of the
reference solution GY4 (2.2.2, Method JI). principal peak in the chromatogram obtained with
To 1.0 g of the substance to be examined add about 0.25 g reference solution (b) (0.5 per cent);
of potassium hydroxide R and 7 mL of 'Water R. Sonicate to - impurity C: not more than the area of the principal peak
dissolve and dilute to 10.0 mL with 'Water R. in the chromatogram obtained with reference solution (b)
Impurity A (0.2 per cent);
Thin-layer chromatography(2.2.27). Prepare the solutions - unspecified impu1"Zúes: for each impurity, not more than the
immediately before use. area of the principal peak in the chromatogram obtained
Solvent mixture methanol R, methylene chloride R (50:50 V/V). with reference solution (b) (0.20 per cent);
- total: not more than 5 times the area of the principal peak
Test solution Dissolve 0.100 g of the substance to be
in the chromatogram obtained Witll reference solution (b)
examined in the solvent mixture and dilute to 5. O mL with
(1.0 per cent);
the solvent mixture.
- disregard limit: 0.5 times the area of the principal peak in
Reference solutioll Dissolve 5.0 mg of ciprofioxacin the chromatogram obtained with reference solution (b)
impurity A CRS (enrofioxacin impurity A) in the solvent (0.1 per cent).
mixture and dilute to 50.0 mL with the solvent mixture.
Heavy metals (2.4.8)
Dilute 4.0 mL ofthis solution to 10.0 mL with the solvent
Mu,ximum 20 ppm.
mixture.
Dissolve 1.5 g in a mixture of 5 mL of 2 ÑI acetic acid and
Plate TLC silica gel FZ54 plate R (2-10 ~lln).
10 mL of 'Water R. Filter. 12 mL of the filtrate after adding
Mobile phase butanol R, 'Water R, anhydrous acetic acid R, ethyl 2 mL of 'Water R (instead of buffer solution) complies with
acetate R (15:15:20:50 V/V/V/V). test E. Prepare the reference solution using 12 mL of lead
Application 1O ~lL. standard solution (2 ppm Pb) R.
Development Over 3/4 of the plateo Loss on drying (2.2.32)
D¡ying In air. Maximum 1.0 per cent, determined on 2.000 g by drying
Detection Examine in ultraviolet light at 254 nm. under high vacuum at 120 oC for 6 h.
Results: Sulfated ash (2.4.14)
- impunty A: any spot due to impurity A is not more Maximum 0.1 per cent, determined on 1.0 g.
intense than the spot in the chromatogram obtained with ASSAY
the reference solution (0.2 per cent).
Dissolve 0.250 g in 100 mL of anhydrous acetic acid R and
Related substances titrate with 0.1 M perchlO1'ic acid detennining the end-point
Liquid chromatography (2.2.29). potentiometrically (2.2.20).
Test solution Dissolve 50 mg of the substance to be examined 1 mL of 0.1 M perchloric acid is equivalent to 35.94 mg of
in the mobile phase and dilute to 50.0 mL with the mobile C19HzzFN303·
phase.
STORAGE
Reference solution (a) Dissolve 10 mg of enrofioxacin for system
Protected from light.
suitability CRS (containing impurities B and C) and dilute to
10 mL with the mobile phase. IMPURITIES
Reference solution (b) Dilute 1.0 mL of the test solution to Specified impurities A, B, C
50.0 mL with the mobile phase. Dilute 1.0 mL of this Other detectable impurities (the following substances would, if
solution to 10.0 mL with the mobile phase. present at a sufficient level, be detected by one or other of
Column: the tests in the monograph. They are limited by the general
- size: l = 0.15 m, 0 = 4.6 mm; acceptance criterion for other/unspecified impurities and/or
- stationa¡y phase: base-deactivated end-capped octadecylsilyl by the general monograph Substances for pha17naceutical use
silica gel for chro11latography R (5 ~lm); (2034). It is therefore not necessary to identify these
- temperature: 40 oC. impurities for demonstration of compliánce. See also 5.10.
Control of impurities in substances for pha17naceutical use): E, F,
Mobile phase Mix 15 volumes of methanol R and 85 volumes
of a 2.9 giL solution of phosphonc acid R, previously adjusted G.
to pH 2.3 with triethylamine R.
2016 Etamiphylline Camsilate Vet-69

Etamiphylline Camsilate

Me Me

J:;~03H
A. 7-chloro-1-cyclopropyl-6-fiuoro-4-oxo-1 ,4-
dihydroquinoline-3-carboxylic acid,

511.6 19326-29-5

Action and use


N on-selective phosphodiesterase inhibitor (xanthine);
treatment of reversible airways obstruction.
Preparations
Etamiphylline Injection
B. ciprofioxacin,
Etamiphylline Tablets

DEFINITION
Etamiphylline Camsilate is
7-(2-diethylaminoethyl) theophylline camphorsulfonate.
It contains not les s than 98.0% and not more than 102.0%
of C13H21Ns02,ClOH1604S, calculated with reference to the
dried substance, when determined by both methods
described under the Assay.
C. 1-cyclopropyl-7 -( 4-ethylpiperazin-1-yl)-4-oxo-1,4-
dihydroquinoline-3-carboxylic acid, CHARACTERISTICS
A white 01' almost white powder.
Very soluble in 'water; soluble in chlorofo17nand in ethanol
(96%); very slightly soluble in ether.
IDENTIFICATION
A. The light absmption, Appendix n B, in the range 230 to
350 nm of a 0.004% w/v solution exhibits a maximum only
at 274 nm. The absorbance at 274 nm is about 0.70.
B. The injrared abs01ptioll spectrum, Appendix n A, is
E. 6-chloro-1-cyclopropyl-7 -( 4-ethylpiperazin-1-yl)-4-oxo- concordant with the reference spectrwll of etamiphylline
1,4-dihydroquinoline-3-carboxylic acid, camsilate (RSV 51).
C. Fuse 0.1 g with a pellet of sodium hydroxide, dissolve in
water and neutralise with hydrochloric acid. The resulting
solution yields reaction A characteristic of sulfates,
Appendix VI.
TESTS
Melting point
202 0 to 206°, Appendix V A.
F. 1-cyclopropyl-7 -(4-ethylpiperazin-1-yl)-6-fiuoroquinolin- Acidity
4(1H)-one, pH of a 10% w/v solution, 3.9 to 5.4, Appendix V L.
Free etamiphyIline
Not more than 2.0% w/w when determined by Method I for
non-aqueous titration, Appendix VIII A, using 2 g dissolved in
75 mL of acetic anhyd17de and detelmining the end point
potentiometrically. Each mL of O.lM perchloric acid VS is
equivalent to 27.93 mg of free etamiphylline.
Related substances
Carry out the method for thin-layer chro111atography,
G. 7- [(2-aminoethyl) amino] -1-cyclopropyl-6-fiuoro-4-oxo- Appendix In A, using silica gel HF254 as the coating
1,4-dihydroquinoline-3-carboxylic acid. substance and a mixture of 80 volumes of chloroform,
___________________________________________ ~E~
20 volumes of ethanol (96%) and 1 volume of
13.5M a1111110nia as the mobile phase. Apply separately to the
plate .1 O ~lL of each of two solutions of the substance being
examined in water containmg (1) 4.0% w/v and (2)
0.0080% w/v. After removal of the plate, allow it to dry in air
and examine under ultraviolet light (254 nm). Any secondary
spot in the chromatogram obtained with solution (1) is not
Vet-70 Ethopabate 2016

more intense than the spot in the chromatogram obtained The chromatographic procedure may be canied out using
with solution (2) (0.2%). (a) a stainless steel column (30 cm x 3.9 mm) packed with
Loss on drying particles of silica the surface of which has been modified by
When dried to constant weight at 105°, loses not more than chemically-bonded phenyl groups (10 ~lm) (Waters
~lBondapak phenyl is suitable), (b) 30 volumes of acetonitrile,
0.5% of its weight. Use 1 g.
150 volumes of methanol and 450 volumes of 0.15M sodium
Sulfated ash hexanesulfonate, adjusted to pH 2.5 with 011hophosphoric acid,
Not more than 0.2%, Appendix IX A. as the mobile phase with a fiow rate of 1 mL per minute and
ASSAY (c) a detection wavelength of 268 nm. Maintain the
0
For camphorsulfonic acid temperature of the column at 45 •

Dissolve 1 g in 25 mL of nzethanol, previously neutralised The test is not valid unless, in the chromatogram obtained
with 1M sodiunz hydroxide VS, and titrate with O.lM sodium with solution (3), the resolution factor between the peaks due
hydroxide VS using thynzol blue solution as indicator. Each mL to ethopabate and methyl 4-acetamido-2-hydroxybenzoate is
of O.lM sodium hydroxide VS is equivalent to 51.16 mg of at least 1.2.
C13H21Ns02,ClOH1604S, In the chromatogram obtained with solution (1) the area of
For etamiphylline any peak eluting before the peak corresponding to methyl
Dissolve 0.15 g in 20 mL of 2M hydrochloric acid, add 12 mL 4-acetamido-2-hydroxybenzoate (identified by the peak in the
of a 5% w/v solution of silicotungstic acid and allow to stand chromatogram obtained with solution (3)) is not greater than
for 5 hours. Filter, wash the residue with 2M hydrochl017C acid 0.1 times the area of the principal peak in the chromatogram
until the filtrate yields no precipitate with a 1 % w/v solution obtained with solution (2) (0.1 %); the area of any other
of quinine hydrochloride and dry at 110°. Each g of residue is seconda¡y peak is not greater than the are a of the principal
equivalent to 0.2830 g of C13H21Ns02,ClOH1604S, peak in the chromatogram obtained with solution (2) (1 %)
and the sum of the areas of all the seconda¡y peaks is not
greater than twice the area of the principal peak in the
chromatogram obtained with solution (2) (2%).
Loss on drying
Ethopabate When dried at 60° at a pressure of 2 kPa for 2 hours, loses
not more than 1.0% of its weight. Use 1 g.
ÚCOOMe Sulfated ash

AcHN ~ I OEt
Not more than 0.1 %, Appendix IX A.
ASSAY
Carry out the method for liquid chromatography,
237.3 59-06-3 Appendix In D, using the following solutions in a mixture of
equal volumes of methanol and water. Solution (1) contains
Action and use 0.002% w/v of the substance being examined. Solution (2)
Antiprotozoal. contains 0.002% w/v of ethopabate BPCRS. Solution (3)
contains 0.002% w/v of ethopabate BPCRS and 0.010% w/v
DEFINITION of methyl 4-acetamido-2-hydroxybenzoate BPCRS.
Ethopabate is methyl 4-acetamido-2-ethoxybenzoate.
The chromatographic conditions described under Related
It contains not less than 96.0% and not more than 101.0%
substances may be used.
of C12HlSN04, calculated with reference to the dried
substance. The test is not valid unless, in the chromatogram obtained
with solution (3), the resolution factor between the peaks due
CHARACTERISTICS to ethopabate and methyl 4-acetamido-2-hydroxybenzoate is
0
A white or pinkish white powder. It melts at about 148 •
at least 1.2.
Very slightly soluble in water; soluble in chlorofor711 and in Calculate the content of C12HlSN04 in the substance being
methanol; sparingly soluble in ethanol (96%); slightly soluble examined using the declared content of C12HlSN04 in
in ether. ethopabate BPCRS.
IDENTIFICATION IMPURITIES
A. The infrared absorption spectru711, Appendix n A, is
concordant with the reference spectru711 of ethopabate
COOH
(RSV 20).
~OH
y
B. In the Assay, the retention time of the principal peak in
the chromatogram obtained with solution (1) is the same as
that of the principal peak in the chromatogram obtained with
solution (2).
TESTS
Related substances A. 4-aminosalicylic acid,
Carry out the method for liquid chronzatography,
Appendix In D, using the following solutions in a mixture of
equal volumes of methanol and water. Solution (1) contains
0.040% w/v of the substance being examined. For solution
(2) dilute 1 volume of solution (1) to 100 volumes. Solution
(3) contains 0.040% w/v of ethopabate BPCRS and
0.010% w/v of methyl 4-acetamido-2-hydroxybenzoate BPCRS.
2016 Etorphine Hydrochloride Vet-71

COOMe used. Any spillage on the skin should be washed off at once. In the
~OEt case of accidental injection 01' abs01ption through broken skin 01'

y NH 2
111UCOUS membl'aneJ a l'eversing agent should be injected
im11lediately.
CHARACTERISTICS
A white or almost white, microcrystalline powder.
B. methyl 2-ethoxy-4-aminobenzoate, Sparingly soluble in water and in ethanol (96%); very slightly
soluble in chloroforl11; practically insoluble in ether.
COOMe IDENTIFICATION
~OH A. The light absorption, Appendix n B, in the range 230 to

y 350 nm of a 0.02% w/v solution in O.lM hydrochloric acid


exhibits a maximum on1y at 289 nm. The absorbance at the
maximum is about 0.68.
B. The light abs01ption, Appendix n B, in the range 230 to
350 nm of a 0.02% w/v solution in O.lM sodiu111 hydroxide
C. methyl 2-hydroxy-4-aminobenzoate, exhibits a maximum on1y at 302 nm. The absorbance at the
maximum is about 1.2.
COOMe C. Carry out the method for thin-layer chl'omatography,
~OH Appendix In A, in subdued light using silica gel GF254 as the

y NHAc
coating substance and a mixture of 10 volumes of
diethylamine, 20 volumes of ethyl acetate and 70 volumes of
toluene as the mobile phase. Apply separately to each half of
the plate 1 [lL of each of two solutions in methanol containing
(1) 2.0% w/v of the substance being examined and (2)
D. methyl 4-acetamido-2-hydroxybenzoate, 1.8% w/v of dipl'en01phine BPCRS. Add at each point of
application 1O ~lL of a mixture of 4 volumes of 111ethanol and
COOEt 1 volume of 13.5M ammonia. After removal of tlle plate,
~OEt allow it to dry in air and examine under ultraviolet light

y NHAc
(254 nm). Spray one half of the plate with a mixture of
5 volumes of chloroplatinic acid solution, 35 volumes of dilute
potassium iodide solution and 60 volumes of acetone. Spray the
other half of the plate with a mixture of 1 volume of iron(m)
chloride solwion Rl and 1 volume of dilute potassium
E. ethyl 4-acetamido-2-ethoxybenzoate. hexacyanoferrate(m) solution. The principal spot in tlle
chromatogram obtained with solution (1) has an Rf value of
about 1.15 relative to that of the spot in the chromatogram
obtained with solution (2), absorbs ultraviolet light and yie1ds
a reddish violet colour with the iodoplatinate spray reagent
Etorphine Hydrochloride and a blue colour with the iron-hexacyanoferrate spray
reagent.
D. Yields reaction A characteristic of chlorides, Appendix VI.
TESTS
Acidity
,HCI pH of a 2.0% w/v solution, 4.0 to 5.5, Appendix V L.
Specific optical rotation
In a solution prepared by dissolving 0.5 g in sufficient
methanol to produce 25 mL, -122 to -132, calculated with
reference to the dried substance, Appendix V F.
Related substances
448.0 13764-49-3 Carry out in subdued light the method for thin-layer
chromatography, Appendix nI A, using silica gel G as the
Action and use coating substance and a mixture of 10 volumes of
Opioid receptor agonist; analgesic. diethylamine, 20 volumes of ethyl acetate and 70 volumes of
Preparation toluene as the mobile phase. Apply separately to the plate
Etorphine and Acepromazine Injection 20 [lL of each of two solutions of the substance being
examined in methanól containing (1) 2.0% w/v and (2)
DEFINITION 0.020% w/v. Add at each point of application 10 ~lL of a
Etorphine Hydrochloride is (6R, 7R, 14R)-7 ,8-dihydro-7- mixture of 4 volumes of methanol and 1 volume of
[( 1R) -1-hydroxy-1-methylbutyl] -6-0-methyl-6, 14- 13.5M 'ammonia. After removal of the plate, allow it to dry in
ethenomorphine hydrochloride. It contains not less than air and spray with a 1 % w/v _solution of iodine in methanol.
98.0% and not more than of 101.0% of C2sH33N04,HCI, Any secondary spot in the chromatogram obtained with
calculated with reference to the dried substance. solution (1) is not more intense than the spot in the
CAUTION Etorphine Hydrochloride is extremely potent and chromatogram obtained with solution (2) (1 %).
extraordinaJY care should be taken in any procedure in which it is
Vet-72 Febantel 2016

Loss on drying Test solution (b) Dilute 5.0 mL of test solution (a) to
When dried to constant weight at 105°, loses not more than 100.0 mL with the solvent mixture.
4.0% of its weight. Use 1 g. Referenee solution (a) Dilute 1.0 mL of test solution (a) to
Sulfated ash 100.0 mL with the solvent mixture. Dilute 1.0 mL of this
Not more than 0.1 %, Appendix IX A. solution to 10.0 mL with the solvent mixture.
ASSAY Referenee solution (b) Dissolve 50.0 mg offebantel CRS in the
Carry out Method I for non-aqueous titration, solvent mixture and dilute to 10.0 mL with the solvent
Appendix VIII A, using 0.5 g, adding 7 mL of mereulJl(IJ) mixture. Dilute 5.0 mL of this solution to 50.0 mL with the
aeetate solution and using erystal violet solution as indicator. solvent mixture.
Each mL of O.lM perehlorie acid VS is equivalent to 44.80 mg Referenee solution (e) Dissolve 5 mg of febantel for system
of CzsH33N04,HCl. suitability CRS (containing impurities A, B and C) in 1. O mL
of the solvent mixture.
STORAGE
Colunzn:
Etorphine Hydrochloride should be protected from light.
-- size: l = 0.15 m, 0 = 4.0 mm;
-- stationary phase: spherical end-eapped oetadeeylsilyl siliea gel
for ehromatography R1 (5 ~lm).
Mobile phase Dissolve 6.8 g of potassium dihydrogen
Febantel ***
** **
phosphate R in 1000 mL of water for ehromatography R.
Mix 350 mL of aeetonitlile R with 650 mL of this solution.
(Febantel for Veterinary Use) ***** Flow rate 1.0 mUmin.
Ph Eur monograph 2176)
Detection Spectrophotometer at 280 nm.
Injeetion 1O ~lL of test solution (a) and reference solutions (a)
and (c).
Run time 1.5 times the retention time of febantel.
Elution order Impurity A, impurity B, impurity C, febantel.
Retention time Febantel = about 32 mino
Syste111 suitabllity: reference solution (c):
-- resolution: minimum 3.0 between the peaks due to
impurities A and B and minimum 4.0 between the peaks
58306-30-2
due to impurities B and C.
Action and use Limits:
Antihelminthic. -- i111pwities A) B) C: for each impurity, not more than the
area of the principal peak in the chromatogram obtained
___________________________________________
PhE~
with reference solution (a) (0.1 per cent);
DEFINITION -- unspeeified impwities: for each impurity, not more than
Dimethyl N,N'-[[[2-[(methoxyacetyl)amino]-4- twice the area of the principal peak in the chromatogram
(phenylsulfanyl)phenyl]imino] methylene] dicarbamate. obtained with reference solution (a) (0.20 per cent);
-- total: not more than 5 times the area of the principal peak
Content in the chromatogram obtained with reference solution (a)
97.5 per cent to 102.0 per cent (dried substance). (0.5 per cent);
CHARACTERS -- disregard limit: 0.5 times the area of the principal peak in
Appearance the chromatogram obtained with reference solution (a)
White or almost white, crystalline powder. (0.05 per cent).
Solubility Heavy metals (2.4.8)
Practically insoluble in water, soluble in acetone, slightly Maximum 20 ppm.
soluble in anhydrous ethanol. 1.0 g complies with test F. Prepare the reference solution
It shows polymorphism (5.9). using 2 mL of lead standard solution (lO ppm Pb) R.
IDENTIFICATION Loss on drying (2.2.32)
Infrared absorption spectrophotometry (2.2.24). Maximum 0.5 per cent, determined on 1.000 g by drying in
an oven at 105 oC for 2 h.
Comparison febantel CRS.
Sulfated ash (2.4.14)
If the spectra obtained in the solid state show differences,
Maximum 0.1 per cent, determined on 1.0 g.
dissolve the substance to be examined and the reference
substance separately in aeetone R, evaporate to dryness and ASSAY
record new spectra using the residues. Liquid chromatography (2.2.29) as described in the test for
TESTS related substances with the following modification.
Related substances Injeetion Test solution (b) and reference solution (b).
Liquid chromatography (2.2.29). Calculate the percentage content of CzoHzzN406S from the
Solvent mixture aeetonitrile R, tetrahydrofuran R (50:50 V/V). dec1ared content of febantel CRS.
Test solution (a) Dissolve 0.100 g of the substance to be IMPURITIES
examined in the solvent mixture and dilute to 10. O mL with Speeified impurities A, B, C
the solvent mixture.
2016 Fenbendazole Vet-73

solution to 200.0 mL with 111ethanol R. Dilute 5.0 mL of this


solution to 10.0 mL with hydroehlorie 111ethanol R.
Rejerenee solution (b) Dissolve 10.0 mg ofjenbendazole
impurity A CRS in 100.0 mL of methanol R. Dilute 1.0 mL of
the solution to 10.0 mL with hydroehlorie methanol R.
Rejerenee solution (e) Dissolve 10.0 mg ofjenbendazole
A. methyl [[2- [(methoxyacetyl) amino] -4- impU11ty B CRS in 100.0 mL of methanol R. Dilute 1.0 mL of
(phenylsulfanyl)phenyl] carbamimidoyl] carbamate, the solution to 10.0 mL with hydroehlorie methanol R.
Rejerenee solution (d) Dissolve 10.0 mg ofjenbendazole CRS
and 10.0 mg of mebendazole CRS in 100.0 mL of methanol R.
Dilute 1.0 mL ofthe solution to 10.0 mL with hydroehlorie
methanol R.
Column:
B. R = CH z-OCH3 : 2-(methoxyrnethyl)-5-(phenylsulfanyl)-
- size: l = 0.25 m, 0 = 4.6 mm;
1H-benzimidazole,
- stationaJY phase: base-deaetivated end-eapped oetadeeylsilyl
C. R = NH-CO-OCH3 : methyl [5-(phenylsulfanyl)-lH- siliea geljor ehr0111atography R (5 ~lm).
benzimidazol-2-yl]carbamate (fenbendazole).
Mobile phase:
___________________________________________ PhE~

- mobile phase A: anhydrous aeetie acid R, methanol R,


water R (1:30:70 V/V/V);
- mobile phase B: anhydrous aeetie aeid R, water R,
methanol R (1 :30:70 V/V/V);
Fenbendazole ***
*** *** Time Mobile phase A Mobile phase B
(Fenbendazole jor Veterinary UseJ *** (min) (per cent V/V) (per cent V/V)
Ph Eur monograph 1208) 0- 10 100 -7 O O -7 100

10 - 40 O 100

Flow rate 1.0 mUmin.


Deteetion Spectrophotometer at 280 nm.
299.4 43210-67-9 Injeetion 1O ~L.
Identifieation oj impUI'I'ties Use the chromatogram obtained
Action and use with reference solution (b) to identify the peak due to
Antihelminthic. impurity A; use the chromatogram obtained with reference
Preparations solution (c) to identify the peak due to impurity B; use the
Fenbendazole Granules chromatogram obtained with reference solution (d) to
Fenbendazole Veterinary Oral Paste identify the peak due to mebendazole.
Fenbendazole Veterinary Oral Suspension Relative retention With reference to fenbendazole (retention
time = about 18 min): impurity A = about 0.2;
~E~ ___________________________________________ impurity B = about 0.6; mebendazole = about 0.8.
DEFINITION System suitability Reference solution (d):
Methyl [5-(phenylsulfanyl)-lH-benzimidazol-2-yl]carbamate. - resolution: minimum 1.5 between the peaks due to
mebendazole and fenbendazole.
Content
Limits:
98.0 per cent to 101.0 per cent (dried substance).
- impurity A: not more than 2.5 times the are a of the
CHARACTERS corresponding peak in the chromatogram obtained with
Appearance reference solution (b) (0.5 per cent);
White or almost white powder. - i111PUrity B: not more than 2.5 times the area of the
Solubility corresponding peak in the chromatogram obtained with
Practically insoluble in water, sparingly soluble in reference solution (c) (0.5 per cent);
dimethylformamide, very slightly soluble in methanol. - unspeeified impurities: for each impurity, not more than
twice the area of the principal peak in the chromatogram
IDENTIFICATION obtained with reference solution (a) (0.5 per cent);
Infrared absorption spectrophotometry (2.2.24). - total: maximum 1.0 per cent;
Comparison jenbendazole CRS. - disregard limit: 0.4 times the area of the principal peak in
TESTS the chromatogram obtained with reference solution (a)
Related substances (0.10 per cent).
Liquid chromatography (2.2.29). Prepare the solutions Heavy metals (2.4.8)
immediately bejore use. Keep the te111perature oj the autosampler M:lXirrium 20 ppm.
at10°C. 1.0 g complies with test C. Prepare the reference solution
Test solution Dissolve 50.0 mg of the substance to be using 2 mL of lead standard solution (lO ppm Pb) R.
examined in 10. O mL of hydroehlorie 111ethanol R. Loss on drying (2.2.32)
Rejerenee solution (a) Dissolve 50.0 mg ofjenbendazole CRS in Maximum 1.0 per cent, determined on 1.000 g by drying in
10.0 mL of hydroehlorie 111ethanol R. Dilute 1.0 mL of the an oven at 105 oC for 3 h.
Vet-74 Fluanisone 2016

Sulfated ash (2.4.14) B. The light absOlption, Appendix II B, in the range 230 to
Maximum 0.3 per cent, determined on 1.0 g. 350 nm of a 0.002% w/v solution in a mixture of 9 volumes
of propan-2-ol and 1 volume of O.lM hydrochloric acid exhibits
ASSAY
a well-defined maximum only at 243 nm. The absorbance at
Dissolve 0.200 g in 30 mL of anhydrous acetic acid R,
243 nm is about 1.1.
warming gently if necessary. Cool and titrate with 0.1 M
perchloric acid, detennining the end-point potentiometrically C. Heat 0.5 mI of chromic-sulphul"l'c acid mixture in a small
(2.2.20). test tube in a water bath for 5 minutes; the solution wets the
side of the tube readily and there is no greasiness. Add 2 to
1 mL of 0.1 M perchloric acid is equivalent to 29.94 mg
3 mg of the substance being examined and again heat in a
of ClsH13N302S.
water bath for 5 minutes; the solution does not wet the side
STORAGE of the tube and does not pour easily from the tube.
Protected from light.
TESTS
IMPURITIES Melting point
Specijied impurities A, B 72° to 76°, Appendix V A.
Related substances
o Carry out the method for thin-layer chromatography,

CX
~ }-OCH3
Appendix III A, using the following solutions.
I ,f-NH
~ N (1) 2.0% w/v of the substance being examined.
(2) 0.010% w/v ofthe substance being examined.
A. methyl (lH-benzimidazol-2-yl)carbamate, (3) 0.020% w/v of 4'-fluoro-4-chlorobutyrophenone BPCRS.
(4) 0.010% w/v of 1-(2-methoxyphenyl)piperazine BPCRS.
CHROMATOGRAPHIC CONDITIONS
(a) Use as the coating silica gel GF2S4 precoated plate
(Merck silica gel 60 plates are suitable).
(b) Use the mobile phase as described below.
B. methyl (5-chloro-1H-benzimidazol-2-yl)carbamate. (c) Apply 1O ~ll of each solution.
___________________________________________ PhE~

(d) Develop the plate to 15 cm.


(e) After removal of the plate, dry in air and expose to iodine
vapour for 15 minutes.
MOBILE PHASE
Fluanisone 10 volumes of ethanol (96%) and 90 volumes of chlorofo1'111.
LIMITS
In the chromatogram obtained with solution (1):
any spots corresponding to 4'-fiuoro-4-chlorobutyrophenone
and 1-(2-methoxyphenyl)piperazine are not more intense
than the spots in the chromatograms obtained with solutions
(3) and (4) respectively (1 % and 0.5%, respectively);
any other secondmy spot in the chromatogram obtained with
356.4 1480-19-9
solution (1) is not more intense than the spot in the
Action and use chromatogram obtained with solution (2) (0.5%).
Dopamine receptor antagonist; neuroleptic. Loss on drying
When dried to constant weight at 40° at a pressure not
DEFINITION exceeding 0.7 kPa, loses not more than 0.5 % of its weight.
Fluanisone is 4'-fiuoro-4-[4-(2-methoxyphenyl)piperazin-1- Use 1 g.
yl]-butyrophenone. It contains not les s than 98.0% and not
Sulphated ash
more than 101.0% of C21H2SFN202, calculated with Not more than 0.1 %, Appendix IX A.
reference to the dried substance.
ASSAY
CHARACTERISTICS
Carry out Method I for non-aqueous titration)
White 01' almost white to buff-coloured crystals 01' powder;
Appendix VIII A, using 0.15 g and clystal violet solution as
odourless 01' almost odourless. It exhibits polymorphism.
indicator. Each mI of O.lM perchloric acid VS is equivalent to
Practically insoluble in water; freely soluble in chlorofonn, in 17.82 mg of C21H2SFN202'
ethanol (96%), in ether and in dilute solutions of organic
acids. STORAGE
Fluanisone should be protected from light.
IDENTIFICATION
A. The infrared absorption spectrum, Appendix II A, is
concordant with the reference spectrum of fiuanisone
(RSV 22). If the spectra are not concordant, dissolve 0.1 g of
the substance being examined in 3 mI of dichloromethane and
evaporate the solvent at room temperature, scratching the
side of the container occasionally with a glass rod and
prepare a new spectrum of the residue.
2016 Flunixin Meglumine Vet-75

Flunixin Meglumine *** - statz"onaJY phase: octadeeylsiljil siliea gel for ehromatography R
*** *** (5 ~lm).
(Flunixin Meglumine for VeterinaJY Use, *** Mobile phase Mix 300 volumes of water R and 700 volumes
Ph Bur 11l0nograph 1696) of aeetonitrile R, and add 0.25 volumes of phosph011e aeid R.
Flow rate 1.0 mUmin.
Deteetion Spectrophotometer at 254 nm.
~' CH 3

t:?!
HO H Jnjection 1O ~lL.
H " Run time 5 times the retention time of fiunixin.
" --OH
HO '. H
HO H Relative retentz"on With reference to fiunixin (retention
HO time = about 3.1 min): impurity A = about 0.4;
impurity C = about 0.6; impurity B = about 0.7;
impurity D = about 4.2.
491.5 42461-84-7 System suitability: reference solution (a):
- resolutz"on: minimum 3.5 between the peaks due to
Action and use
impurity B and fiunixin.
Cyclo-oxygenase inhibitor; analgesic; anti-infiammatory.
Limits:
~E~ ___________________________________________ - eorrection factor: for the calculation of content, multiply the
DEFINITION peak area of impurity C by 1.9,
- impurity A: not more than the area of the corresponding
2- [[2-Methyl-3-(trifiuoromethyl)phenyl] amino ]pyridine-3-
peak in the chromatogram obtained with reference
carboxylic acid, 1-deoxy-1-(methylamino)-D-glucitol.
solution (b) (0.2 per cent),
Content - impurity B: not more than the area of the corresponding
99.0 per cent to 101.0 per cent (dried substance). peak in the chromatogram obtained with reference
CHARACTERS solution (b) (0.2 per cent),
Appearance - i111pUl1ties C, D: for each impurity, not more than the area
White or almost white, crystalline powder. of the peak due to fiunixin in the chromatogram obtained
with reference solution (b) (0.2 per cent),
Solubility
- any other impurity: for each impurity, not more than the
Freely soluble in water and in methanol, practically insoluble
area of the peak due to fiunixin in the chromatogram
in acetone.
obtained with reference solution (b) (0.2 per cent),
IDENTIFICATION - total: not more than 2.5 times the area of the peak due to
A. Specific optical rotation (2.2.7): -12.0 to -9.0 (dried fiunixin in the chromatogram obtained with reference
substance), determined on solution S (se e Tests). solution (b) (0.5 per cent),
B. Infrared absorption spectrophotometry (2.2.24). - disregard limit: 0.25 times the area of the peak due to
C011lparison fiunixin meglumine CRS. fiunixin in the chromatogram obtained with reference
solution (b) (0.05 per cent).
TESTS
Loss on drying (2.2.32)
Solution S Maximum 0.5 per cent, determined on 1.000 g by drying in
Dissolve 2.50 g in earbon dioxide-free water R and dilute to an oven at 105 oC for 4 h.
50.0 mL with the same solvento
Sulfated ash (2.4.14)
Appearance of solution Maximum 0.1 per cent, determined on 1.0 g.
Solution S is clear (2.2.1) and not more intensely coloured
than reference solution Y7 (2.2.2, Method JI). ASSAY
pH (2.2.3) Dissolve 0.175 g in 50 mL of anhydrous aeetie acid R. Titrate
with 0.1 M perehlorie acid, determining the end-point
7.0 to 9.0 for solution S.
potentiometrically (2.2.20).
Related substances
1 mL of 0.1 M perehlorie acid is equivalent to 24.57 mg
Liquid chromatography (2.2.29).
of C21H2SF3N3Ü7'
Test solution Dissolve 50.0 mg of the substance to be
examined in the mobile phase and dilute to 10.0 mL with IMPURITIES
the mobile phase. Speeijied impw1ties: A, B, C, D.
Referenee solution (a) Dissolve 5. O mg of fiunixin
impUl1ty B CRS in 1.0 mL of the test solution and dilute to
50.0 mL with the mobile phase.
Referenee solution (b) Dissolve 5.0 mg of 2-ehloronicotinie
acid R (impurity A) in the mobi1e phase and dilute to
50.0 mL with the mobile phase. To 2.0 mL of this solution
add 2.0 mL of reference solution (a) and dilute to 20.0 mL A. R = H: 2-chloropyridine-3-carboxylic acid,
with the mobile phase. C. R::::;: C 2H s : ethy12-chloropyridine-3-carboxylate,
Referenee solution (e) Dissolve 50 mg of fiunixin
impurity C CRS in the mobile phase and dilute to 100 mL
with the mobile phase.
Column:
size: 1 = 0.125 m, 0 = 4.0 mm,
Vet-76 Gonadotrophin 2016

ASSAY
The potency of equine serum gonadot:rophin is estimated by
comparing under given conditions its effect of increasing the
mass of the ovaries of irnmature female rats with the same
effect of the International Standard of equine serum
B. 2-methyl-3-(trifluoromethyI) aniline, gonadotrophin or of a reference preparation calibrated in
International Units.
The International Unit is the activity contained in a stated
amount of the International Standard, which consists of a
mixture of a freeze-dried extract of equine serum
gonadotrophin from the serum of pregnant mares with
lactose. The equivalence in International Units of the
International Standard is stated by the World Health
Organization.
Use irnmature female rats of the same strain, 21 to 28 days
D. ethyI2-[[2-methyl-3- old, differing in age by not more than 3 days and having
(trifluoromethyl)phenyl] amino] pyridine-3-carboxylate. mas ses such that the difference between the heaviest and the
_____________________________________________ ~Elf
lightest rat is not more than 10 g. Assign the rats at random
to 6 equal groups of not fewer than 5 animals. If sets of
6 litter mates are available, assign one litter mate from each
set to each group and mark according to litter.
Choose 3 doses of the reference preparation and 3 doses of
Serum Gonadotrophin ***
*** ***
the preparation to be examined such that the smallest dose is
sufficient to produce a positive response in sorne of the rats
(Equine Serum Gonadotrophin for VeterinalJi Use, *** and the largest dose does not produce a maximal response in
Ph Eu/" monograph 0719) all the rats. U se doses in geometric progression: as an initial
approximation total doses of 8 IU, 12 IU and 18 IU may be
Action and use
tried, although the dose will depend on the sensitivity of the
Equine serum gonadotrophin.
animals used and may vary widely.
Preparation Dissolve separately the total quantities of the preparation to
Serum Gonadotrophin Injection be examined and of the reference preparation corresponding
PhElf _____________________________________________ to the doses to be used in sufficient of a sterile 9 gIL solution
of sodiui1l chloride R containing 1 mg/mL of bovine albumúz R
DEFINITION such that each single dose is administered in a volume of
Equine serum gonadotrophin for veterinary use is a dry about 0.2 mL. Store the solutions at 5 ± 3 cc.
preparation of a glycoprotein fraction obtained from the
Inject subcutaneously into each rat the dose allocated to its
serum or plasma of pregnant mares. It has follicle-stimulating
group. Repeat the injections 18 h, 21 h, 24 h, 42 h and 48 h
and luteinising activities. The potency is not less than
after the first injection. Not less than 40 h and not more than
1000 IU of gonadotrophin activity per milligram, calculated
72 h after the last injection, euthanise the rats and remove
with reference to the anhydrous substance.
the ovaries. Remove any extraneous fluid and tissue and
PRODUCTION weigh the 2 ovaries irnmediately. Calculate the results by d1e
Equine serum gonadotrophin may be prepared by usual statistical methods, using the combined mass of the
precipitation with alcohol (70 per cent V/V) and further 2 ovaries of each animal as the response.
purification by a suitable form of chromatography. It is The estimated potency is not les s than 80 per cent and not
prepared in conditions designed to minimise microbial more d1an 125 per cent of the stated potency.
contamination. The confidence limits (P = 0.95) of the estimated potency
CHARACTERS are not les s than 64 per cent and not more than 156 per cent
Appearance of the stated potency.
White or pale grey, amorphous powder. STORAGE
Solubility In an airtight container, protected from light, at a
Soluble in water. temperature not exceeding 8 oc. If the substance is sterile,
store in a sterile, airtight, tamper-proof container.
IDENTIFICATION
When administered as prescribed in the assay it causes an LABELLING
increase in the mas s of the ovaries of irnmature female rats. The label states the potency in International Units per
milligram.
TESTS
_____________________________________________ PhElf
Water (2.5.12)
Maximum 10.0 per cent, determined on 80 mg.
Bacterial endotoxins (2.6.14, method C)
Less than 0.035 IU per IU of equine serum gonadotrophin,
if intended for use in the manufacture of parenteral
preparations without a further appropriate procedure for the
removal of bacterial endotoxins.
2016 Levamisole Vet-77

*****
Mobile phase:
Levamisole
** ** -- mobile phase A: dissolve 0.5 g of ammonium dihydrogen
(Levamisole for Vetaina¡y Use, Ph. Eur. monograph *** phosphate R in 90 mL of 'Water R; adjust to pH 6.5 with a
1728) 40 gIL solution of sodium hydroxide R and dilute to
100 mL with 'Water R,
-- mobile phase B: acetonitrile R.

Time Mobile phase A Mobile phase B


(min) (per cent V/V) (per cent V/V)
0-8 90 ~ 30 10 ~ 70
204.3 14769-73-4
8 - 10 30 70

Action and use


Immunostimulant; antihelminthic.
Flo'W mte 1.5 mUmin.
PhEw _____________________________________________ Detection Spectrophotometer at 215 lllTI.

DEFINITION Jnjection 1O ~lL.


(6S)-6-Phenyl-2,3,5,6-tetrahydroimidazo [2, 1-b]thiazole. Jdentijication of impurities Use the chromatogram supplied
Content with levamisole hydrochloride for system suitability CRS and the
98.5 per cent to 101.5 per cent (anhydrous substance). chromatogram obtained with reference solution (a) to
identify the peaks due to impurities A, B, C, D and E.
CHARACTERS
Relative retention With reference to levamisole (retention
Appearance time = about 3 min): impurity A = about 0.9;
White or almost white powder. impurity B = about 1.4; impurity C = about 1.5;
Solubility impurity D = about 1.6; impurity E = about 2.0.
Slight1y soluble in water, freely soluble in ethanol System suitability: reference solution (a):
(96 per cent) and in methanol. -- the chromatogram obtained is similar to the
It shows polymorphism (5.9). chromatogram supplied with levamisole hydrochloride for
syste111 suitability CRS.
IDENTIFICATION
A. Specific optical rotation (see Tests). Limits:
-- correction factors: for the calculation of content, multiply
B. Infrared absorption spectrophotometry (2.2.24).
t11e peak areas of the following impurities by the
Comparison Ph. Eur. refaence spectrum of levamisole. corresponding correction factor: impurity A = 2.0;
If t11e spectra show differences, dissolve the substance to be impurity B = 1.7; impurity C = 2.9; impurity D = 1.3;
examined in l7leth:ylene chloride R, evaporate to dryness and impurity E = 2.7;
record a new spectrum using the residue. -- impurities A, B, C, D, E: for each impurity, not more than
TESTS the area of the principal peak in the chromatogram
obtained with reference solution (b) (0.2 per cent);
Solution S
-- any other impurity: not more than half the area of the
Dissolve 2.50 g in ethanol R and dilute to 50.0 mL with t11e
principal peak in t11e chromatogram obtained with
same solvento
reference solution (b) (0.1 per cent);
Appearance of solution -- total: not more than 1.5 times the area of the principal
Solution S is clear (2.2.1) and not more intensely coloured peak in the chromatogram obtained with reference
than reference solution BY 6 (2.2.2, lVlethod JI). solution (b) (0.3 per cent);
Specific optical rotation (2.2.7) -- disregard limit: 0.25 times the area of the principal peak in
-89 to -85 (anhydrous substance), determined on the chromatogram obtained with reference solution (b)
solution S. (0.05 per cent).
Related substances Water (2.5.12)
Liquid chromatography (2.2.29). Prepare the solutions Maximum 0.5 per cent, determined on 1.00 g.
immediately before use, protect from light and keep belo'W 25 oc. Sulfated ash (2.4.14)
Test solution Dissolve 0.100 g of the substance to be Maximum 0.1 per cent, determined on 1.0 g.
examined in 71lethanol R and dilute to 10.0 mL with the same
ASSAY
solvento
Dissolve 0.150 g in 50 mL of a mixture of 1 volume of
Reference solution (a) Dissolve 10 mg of levamisole anhydrous acetic acid R and 7 volumes of methyl ethyl ketone R.
hydrochloride for system suitability CRS (containing Titrate with 0.1 M perchloric acid, using 0.2 mL of
impurities A, B, C, D and E) in methanol R, add 0.1 mL of naphtholbenzein solution R as indicator.
concentrated ammonia R and dilute to 1.0 mL with
1 mL of 0.1 M perchloric acid is equivalent to 20.43 mg of
methanol R.
CllHlZNzS.
Refaence solution (b) Dilute 1.0 mL of the test solution to
100.0 mL with methanol R. Dilute 5.0 mL ofthe solution to STORAGE
25.0 mL with methanol R. In an airtight container, protected from light.
Column: IMPURITIES
-- size: 1 = 0.10 m, 0 = 4.6 mm, Specijied impurities A, B, C, D, E
-- stationary phase: base-deactivated octadecylsilyl silica gel for
chromatography R (3 ~lm).
Vet-78 Lufenuron 2016

o
H, NH2r\ Solubility
Praetieally insoluble in water, freely soluble in aeetonitrile,
~ N ----/ and enantiomer
soluble in anhydrous ethanol.
U It shows polymorphism (5.9).
mp: about 172 De.
A. 3- [(2RS) - 2-amino-2-phenylethyl] thiazolidin-2-one,
IDENTIFICATION
Infrared absorption speetrophotometry (2.2.24).
Comparison lufenuron CRS.
If the speetra obtained in the solid state show differenees,
dissolve the substanee to be examined and the referenee
substanee separately in 2-propanol R, evaporate to dlyness
and record new speetra using the residues.
B. 3-[ (E)-2-phenylethenyl] thiazolidin-2-imine,
TESTS
Related substances
~~o
H.. I SH Liquid ehromatography (2.2.29).
~ __ N~
u- andenantiomer
Solvent mixture 'water R, aeetonitrile R (30:70 V/V).
Test solution (a) Dissolve 40.0 mg of the substanee to be
examined in the solvent mixture by sonieating for about
C. (4RS)-4-phenyl-1-(2-sulfanylethyl)imidazolidin-2-one, 10 min and dilute to 100.0 mL with the solvent mixture.
Test solution (b) Dilute 1.0 mL of test solution (a) to
10.0 mL with the solvent mixture.
Referenee solution (a) Dilute 1.0 mL of test solution Cb) to
100.0 mL with the solvent mixture.
D. 6-phenyl-2,3-dihydroimidazo [2, 1-b] thiazole, Referenee solution (b) Dissolve 7 mg of lufenuron
impurity G CRS in test solution Ca) and dilute to 50.0 mL
with test solution Ca).
Referenee solution (e) Dissolve the eontents of a vial of
lufenuron for peak identijieation CRS (eontaining impurities B
and C) in 1.0 mL of the solvent mixture.
Referenee solution (d) Dissolve 40.0 mg of lufenuron CRS in
the solvent mixture by sonieating for about 10 min and dilute
E. 1,l'-[(disulfane-1,2-diyl)bis(ethylene)]bis[(4RS)-4-
to 100. O mL with the solvent mixture. Dilute 1. O mL of the
phenylimidazolidin-2-one] .
solution to 10.0 mL with the solvent mixture.
___________________________________________ PhEw
Column:
-- size: l = 0.25 m, 0 = 4.0 mm;
-- stationa1J1 phase: end-eapped oetadeeylsilyl siliea gel for
ehromatography R C5 ~lm).
Anhydrous Lufenuron *** Mobile phase:
*** *** -- mobile phase A: 0.01 per eent V/V of phosphon'e acid R;
(Lufenuron (Anhydrous) for Vetel'7na1Y Use, Ph. Eur. *** -- 1110bile phase B: aeetonitrile R;
monograph 2177)
Time Mobile phase A Mobile phase B
H F el
(min) (per cent V/V) (per cent V/V)

F3e~0~~ Jl)60
FF
F .&"N N ~
and enantiomer
0-5

5 - 15
30

30 -7 10
70

70 -7 90
H H I 15 - 17 10 90
el F ~

511.2 103055-07-8 Flow rate 1.0 mUmin.


Deteetion Speetrophotometer at 255 nm.
Action and use Injection 20 ~lL of test solution Ca) and referenee solutions Ca),
Eetoparasiticide. (b) and Ce).
PhEw ___________________________________________ Identijieation of impurities U se the ehromatogram supplied
with lufenuron for peak identijiéation CRS and the
DEFINITION
ehromatogram obtained with referenee solution Ce) to identify
1-[2,5-Diehloro-4-[ (2RS)-1, 1,2,3,3,3-
the peaks due to impurities B and e.
hexafiuoropropoxy]phenyl]-3-(2,6-difiuorobenzoyl)urea.
Relative retenti07i With referenee to lufenuron Cretention
Content time = about 9 min): impurity B = about 0.3;
98.0 per eent to 102.0 per eent (dried substanee). impurity C = about 0.7; impurity G = about 0.9.
CHARACTERS System suitability: referenee solution Cb):
Appearance -- resolution: minimum 3.0 between the peaks due to
White or pale yellow powder. impurity G and lufenuron.
2016 Lufenuron Vet-79

Limits:
-- correction factors: for the calculation of content, multiply
the peak areas of the following impurities by the
corresponding correction factor: impurity B = 1.3;
impurity C = 1.3;
-- impurity C: not more than 4 times the area of the
principal peak in the chromatogram obtained with B. 1-(2,5-dichloro-4-hydroxyphenyl)-3-(2,6-
reference solution (a) (0.4 per cent); difluorobenzoyl) urea,
-- impUlity B: not more than 3 times the area of the
principal peak in the chromatogram obtained with
reference solution (a) (0.3 per cent);
-- unspecijied impurities: for each impurity, not more than and enantiomer
twice the area of the principal peak in the chromatogram
obtained with reference solution (a) (0.20 per cent);
-- total: not more than 10 times the area of the principal
peak in the chromatogram obtained with reference
solution (a) (1.0 per cent); C. 1-[3-chloro-4- [(2RS)-1, 1,2,3,3,3-
-- disregard limit: the area of the principal peak in the hexafluoropropoxy]phenyl] -3-(2, 6-difluorobenzoyl)urea,
chromatogram obtained with reference solution (a)
H F
(0.1 per cent).
Heavy metals (2.4.8)
Maximum 20 ppm.
F3e~O~~
FF #"N
H
O
)l
N
H
):J
O
~
F

I
and enantiomer

Dissolve 1.0 g in 20 mL of a mixture of 15 volumes of el F ~


'Water R and 85 volumes of dioxan R. 12 mL of the solution
complies with test B. Prepare the reference solution using
lead standard solution (1 ppm Pb) obtained by diluting lead D. 1-[2-chloro-4-[(2RS)-1,1,2,3,3,3-
standard solution (100 ppm Pb) R with a mixture of hexafluoropropoxy]phenyl] -3-(2, 6-difluorobenzoyl)urea,
15 volumes of 'Water R and 85 volumes of dioxan R.
Loss on drying (2.2.32) H)[X ,o~el
F3 e' ~ o o el
Maximum 0.5 per cent, determined on 1.000 g by drying in
an oven at 105 oC.
F F I )l ) : : ) and enantiomer
#" N N ~
H H I
Sulfated ash (2.4.14) el ~
F
Maximum 0.1 per cent, determined on 1.0 g in a platinum
crucible.
E. 1-(2-chloro-6-fluorobenzoyl)-3-[2,5-dichloro-4- [(2RS)-
ASSAY 1,1,2,3,3,3hexafluoropropoxy] phenyl] urea,
Liquid chromatography (2.2.29) as described in the test for

F'CyXO~: 1
related substances with the following modification.
Injection Test solution (b) and reference solution (d).
Calculate the percentage content of C17HsCI2FsN203 from XJO and enantiomer
N N ~
the declared content of hifenuron CRS. H H I
el F ~
IMPURITIES
Specijied impwities B, C
F. 1-[2,5-dichloro-4-[(2RS)-1,1,2,3,3,3-
Other detectable impurities (the following substances would, if
hexafluoropropoxy]phenyl] -3-(2-fluorobenzoyl) urea,
present at a sufficient level, be detected by one or other of
the tests in the monograph. They are limited by the general
acceptance criterion for other/unspecified impurities and/or
by the general monograph Substances for phannaceutical use
(2034). It is therefore not necessary to identify these
impurities for demonstration of compliance. See also 5.10.
Control of impUlities in substances for phannaceutical use): A DJ
J

EJ FJ G, H.
G. 2,5-dichloro-4-[[[(2,6-
o F difluorophenyl)carbonyl] carbamoyl] amino ]phenyl phenyl

H,N~
carbonate,

FJJ
A. 2,6-difluorobenzamide,

H. 1,3-bis [2,5-dichloro-4-( 1,1,2,3,3,3-


hexafluoropropoxy)phenyl] urea.
____________~------~--------------------_PhE~
Vet-80 Marbofloxacin 2016

of sodium octanesulfonate R and previously adjusted to pH 2.5


Marbofloxacin with phosphonc acid R.
(Marbofioxacin for VetelinaJY UseJ Ph. Eur. Flow rate 1.2 mUmin.
monograph 2233) Detection Spectrophotometer at 315 nm.
Injection 10 1lL.
Run time 2.5 times the retention time of marbofloxacin.
Identification of impunties Use the chromatogram supplied
with marbofioxacin for peak identification CRS and the
chromatogram obtained with reference solution (b) to
identify the peaks due to impurities A, B, C, D and E.
Relative retention With reference to marbofloxacin (retention
362.4 115550-35-1 time = about 33 min): impurity B = about 0.5;
impurity A = about 0.7; impurity C = about 0.9;
Action and use impurity D = about 1.3; impurity E = about 1.5.
Fluoroquinolone antibacterial.
System suitability: reference solution (b):
PhE~ ___________________________________________ - resolution: minimum 1.5 between the peaks due to
impurity C and marbofloxacin, and minimum 4.0
DEFINITION between the peaks due to marbofloxacin and impurity D.
9-Fluoro-3-methyl-1 0-(4-methylpiperazin-1-yl)-7 -oxo-2,3-
Limits:
dihydro-7 H-pyrido [3,2,1-ijJ [4,1,2]benzoxadiazine-6-
- correction factor: for the calculation of content, multiply the
carboxylic acid.
peak area of impurity E by 1.5;
Content - impUlities CJ DJ E: for each impurity, not more than twice
99.0 per cent to 101.0 per cent (dried substance). the area of the principal peak in the chromatogram
CHARACTERS obtained with reference solution (a) (0.2 per cent);
Appearance - il11punties AJ B: for each impurity, not more than the area
Light yellow, crystalline powder. of the principal peak in the chromatogram obtained with
reference solution (a) (0.1 per cent);
Solubility - unspecified impunties: for each impurity, not more than
Slightly soluble in water, sparingly soluble or slightly soluble twice the area of the principal peak in the chromatogram
in methylene chloride, very slightly soluble in ethanol obtained with reference solution (a) (0.2 per cent);
(96 per cent). - total: not more than 5 times the area of the principal peak
IDENTIFICATION in the chromatogram obtained with reference solution (a)
Infrared absorption spectrophotometry (2.2.24). (0.5 per cent);
COl11parison marbofioxacin CRS. - disregani limit: the area of the principal peak in the
chromatogram obtained with reference solution (a)
TESTS (0.1 per cent).
Absorbance (2.2.25)
Heavy metals (2.4.8)
Maximum 0.20, determined at 450 nm. Prepare the solution
Maximum 20 ppm.
immediately before use.
Dissolve 0.5 g in dilute acetic acid R and dilute to 30 mL with
Dissolve 0.400 g in 0.1 M ammonium carbonate buffer solution
the same solvento Adding 2 mL of water R instead of 2 mL
pH 10.3 R using sonication and dilute to 10.0 mL with the
of buffer solution pH 3.5 R, the filtrate complies with test E.
same buffer solution.
Prepare the reference solution using 5 mL of lead standard
Related substances solution (2 ppm Pb) R.
Liquid chromatography (2.2.29). Cany out the test protected
Loss on drying (2.2.32)
from light.
Maximum 0.5 per cent, determined on 1.000 g by drying at
Solvent mixture methanol R, water R (23:77 V/V). 105 oC for 4 h.
Test solution To 0.100 g of the substance to be examined add Sulfated ash (2.4.14)
80 mL of the solvent mixture, sonicate until dissolution and Maximum 0.1 per cent, determined on 1.0 g in a platinum
dilute to 100.0 mL with the solvent mixture. crucible.
Reference solution (a) Dilute 5.0 mL of the test solution to
100.0 mL with the solvent mixture. Dilute 1.0 mL ofthis ASSAY
solution to 50.0 mL with the solvent mixture. Dissolve 0.300 g in 80 mL of glacial acetic acid R. Titrate
with 0.1 M perchlonc acid, determining the end-point
Reference solution (b) Dissolve 10 mg of marbofioxacin for peak
potentiometrically (2.2.20).
identification CRS (containing impurities A, B, C, D and E)
in the solvent mixture and dilute to 10 mL with the solvent 1 mL of 0.1 M perchlonc acid is equivalent to 36.24 mg of
mixture. C17H19FN404'
Column: STORAGE
- size: l = 0.15 m, 0 = 4.6 mm; Protected from light.
- stationary phase: end-capped polar-embedded octadecylsilyl
IMPURITIES
amol'phous organosilica polymer R (3.5 /lm);
Specified impunties A, B, C, D, E
- temperature: 40 oC.
Other detectable impunties (the following substances would, if
Mobile phase Mix 230 volumes of methanol R and 5 volumes
present at a sufficient level, be detected by one or other of
of glacial acetic acid R with 770 volumes of a 2.70 gIL
the tests in the monograph. They are limited by the general
solution of sodium dihydrogen phosphate R containing 3.50 giL
2016 Morantel Tartrate Vet-81

acceptance criterion for other/unspecified impurities and/or


by the general monograph Substanees for pharmaeeutieal use
Morantel Tartrate
(2034). It is therefore not necessary to identify these (Morantel Hydrogen Tartate for Veterinary Use)
impurities for demonstration of compliance. See also 5.10. Ph Bur monograph 1546)
Control of impurities in substanees for pharmaeeutieal use): F.
CH3
OH HN/

FyY~'n
F~CO,H
O
370.4 26155-31-7
A. 6,7 -difluoro-8-hydroxy-1-(methylamino )-4-oxo-1,4-
dihydroquinoline-3-carboxylic acid, Action and use
Antihelminthic.
PhEw ___________________________________________

DEFINITION
1-Methyl-2-[(E)-2-(3-methylthiophen-2-yl)ethenyl]-1,4,5,6-
tetrahydropyrimidine hydrogen tartrate.
Content
B. 9,1 O-difluoro-3-methyl-7 -oxo-2,3-dihydro-7H- 98.5 per cent to 101.5 per cent (dried substance).
pyrido [3,2, 1-&] [4, 1,2]benzoxadiazine-6-carboxylic acid, CHARACTERS
Appearance
White or pale yellow, crystalline powder.
Solubility
Very soluble in water and in ethanol (96 per cent), practically
insoluble in ethyl acetate.
IDENTIFICATION
First identifieation B
C. 6,8-difluoro-1-(methylamino )-7 -(4-methylpiperazin-1-yl)-
4-oxo-1 ,4-dihydroquinoline-3-carboxylic acid,
Seeond identifieation A) eD
A. Melting point (2.2.14): 167 oc to 172 oc.
B. Infrared absorption spectrophotometry (2.2.24).
Comparison morantel hydrogen tam'ate CRS.
C. Dissolve about 10 mg in 1 mL of a 5 gIL solution of
ammonium vanadate R. Evaporate to dryness. Add 0.1 mL of
sulfu1'7;e acid R. A purple colour is produced.
D. Dissolve about 10 mg in 1 mL of 0.1 M sodium hydroxide.
D. 6-fluoro-8-hydroxy-1-(methylamino )-7- Transfer to a separating funnel and shake with 5 roL of
(4-methylpiperazin-1-yl)-4-oxo-1 ,4-dihydroquinoline-3- methylene ehloride R. Discard the organic layer. N eutralise the
carboxylic acid, aqueous layer with a few drops of dilute hydroehlorie acid R.
The solution gives reaction (b) of tartrates (2.3.1).
TESTS
Solution S
Dissolve 0.25 g in earbon dioxide-free water R and dilute to
25.0 mL with the same solvento
Appearance of solution
Solution S is clear (2.2.1) and not more intensely coloured
than reference solution GY6 or y 6 (2.2.2) Method JI).
E. 8-ethoxy-6-fluoro-1-(methylamino )-7 -(4-methylpiperazin- pH (2.2.3)
1-yl)-4-oxo-1 ,4-dihydroquinoline-3-carboxylic acid, 3.3 to 3.9 for solution S.
Related substances
Liquid chromatography (2.2.29). Carry out the test protected
from light,
Test solution Dissolve 50.0 mg of the substance to be
examined in the mobile phase and dilute to 100.0 mL with
the mobile phase.
Referenee solution (a) Dilute 1.0mL of the test solution to
F. 4-[6-carboxy-9-fluoro-3-methyl-7 -oxo-2,3-dihydro-7 H- 100.0 mL with the mobile phase.
pyrido [3,2,1-&] [4, 1,2]benzoxadiazin-1 O-yl]-l- Referenee solution (b) Dilute 2.0 mL of reference solution Ca)
methylpiperazine l-oxide. to 100.0 mL with the mobile phase.
___________________________________________ PhEw
Vet-82 Moxidectin 2016

Referenee solution (e) Expose 10 mL of reference solution (a)


to daylight for 15 min before injection.
Referenee solution (d) Dissolve 15.0 mg of tartarie acid R in
the mobile phase and dilute to 100.0 mL with the mobile
phase.
Colu11Zn: B. 1-methyl-2-[ (Z)-2-(3-methylthiophen-2-yl)ethenyl]-
-- size: l = 0.25 m, 0 = 4.6 mm; 1,4,5, 6-tetrahydropyrimidine,
-- stationary phase: base-deaetivated end-eapped oetadeeylsi1yl CH 3
siliea gel for ehromatography R (5 ~lm). I

Mobile phase To a mixture of 0.35 volumes of triethylamine R


and 85 volumes of 'Water R adjusted to pH 2.5 with
(1("'
phospho17:e acid R, add 5 volumes of tetmhydrofuran R and
C. 1,2-dimethyl-1,4,5,6-tetrahydropyrimidine,
10 volumes of methanol R.
H3 C
Flo'W rate 0.75 mUmin.
Deteetion Spectrophotometer at 226 nm.
CN1H3
~S/
- h and enantiomer
Injection 20 ~lL.
Run time Twice the retention time of morantel.
C~ H '-OH S

Systenz suitability: reference solution (c): D. (lRS)-2-(1-methyl-1,4,5,6-tetrahydropyrimidin-2-yl)-1-


-- resolution: minimum of 2 between the principal peak and (3-methylthiophen-2-yl) ethanol,
the preceding peak ((Z)-isomer).
H3 C
Li111its:
-- any inzpurity apart fr0111 the peak due to tartarie acid: not
OHC
)O S
more than 0.5 times the are a of the principal peak in the
chromatogram obtained with reference solution (a) E. 3-methylthiophene-2-carbaldehyde.
(0.5 per cent); ___________________________________________ ~E~

-- total: not more than the area of the principal peak in the
chromatogram obtained with reference solution (a)
(1 per cent);
-- disregard limit: the area of the principal peak in the
Moxidectin ***
*** ***
chromatogram obtained with reference solution (b)
(0.02 per cent).
Heavy metals (2.4.8)
(Moxideetin for Veterina1Y Use) ***
Ph Eur monograph 1656)
Maximum 20 ppm.
1.0 g complies with test C. Prepare the reference solution
using 2 mL of lead standard solution (10 ppm Pb) R.
Loss on drying (2.2.32)
Maximum 1.5 per cent, determined on 1.000 g by drying in
an oven at 105 oc.
Sulfated ash (2.4.14)
Maximum 0.1 per cent, determined on 1.0 g.
ASSAY
Dissolve 0.280 g in 40 mL of anhydrous aeetie acid R. Titrate
with 0.1 M perehlorie acid, determining the end-point
potentiometrically (2.2.20).
1 mL of 0.1 M perehlorie acid is equivalent to 37.04 mg of
C16H22N206S, 640 113507-06-5

STORAGE Action and use


Protected from light. Antihelminthic; ectoparasiticide.
IMPURITIES Preparation
Moxidectin Injection
Moxidectin Oral Solution
Moxidectin Oromucosal Gel
Moxidectin Pour-on
~E~ ___________________________________________

DEFINITION
A. 1-methyl-2-[(E)-2-(4-methylthiophen-2-yl)ethenyl]-
(2aE,2' R,4E,4' E,5' S,6R,6' S,8E,11R,15S,17aR,20R,20aR,
1,4,5,6-tetrahydropyrimidine,
20bS)-6'- [(lE)-1,3-Dimethylbut-l-enyl]-20,20b-dihydroxy-
4'-(methoxyimino)-5' ,6,8, 19-tetramethyl-
3' ,4',5',6,6', 7, 10,11,14,15,17a,20,20a,20b-
tetradecahydrospiro [2H, 17H-11, 15-methanofuro [4,3,2-
pq] [2,6]benzodioxacyclooctadecine-13,2'-pyran] -17 -one
2016 Moxidectin Vet-83

((6R,23E,25S)-5 O-demethyl-28-deoxy-25- [(1E)-1,3- the curve separating this peak from the peak due to
dimethylbut-l-enyl] -6,28-epoxy-23- moxidectin.
(methoxyimino )milbemycin B). Limits:
Semi-synthetic product derived from a fermentation product. - imputity D: not more than 2.5 times the area of the
It may contain suitable stabilisers such as antioxidants. principal peak in the chromatogram obtained with
reference solution (a) (2.5 per cent);
Content
- sum of i711punties E and F: not more than 1.7 times the
92.0 per cent to 102.0 per cent (anhydrous substance).
area of the principal peak in the chromatogram
CHARACTERS obtained with reference solution (a) (1.7 per cent);
Appearance - imputities A, C, G: for each impurity, not more than
White 01' pale yellow, amorphous powder. 1.5 times the area of the principal peak in the
Solubility chromatogram obtained with reference solution (a)
Practically insoluble in water, very soluble in ethanol (1.5 per cent);
(96 per cent), slightly soluble in hexane. - impurity B: not more than 0.5 times the area of the
principal peak in the chromatogram obtained with
IDENTIFICATION reference solution (a) (0.5 per cent);
Infrared absorption spectrophotometry (2.2.24). - any other impurity eluting before impunty G: for each
Comparison moxidectin CRS. impurity, not more than 0.5 times the area of the
principal peak in the chromatogram obtained with
TESTS
reference solution (a) (0.5 per cent);
Appearance of solution
- disregard li711it: 0.1 times the area of the principal peak
The solution is c1ear (2.2.1) and not more intensely coloured
in the chromatogram obtained with reference
than reference solution GYs (2.2.2, Method JI).
solution (a) (0.1 per cent); disregard the peak due to
Dissolve 0.40 g in benzyl alcohol R and dilute to 20 mL with the stabiliser (identify this peak, where applicable, by
the same solvento injecting a suitable reference solution).
Related substances B. Test solution. Dissolve 75.0 mg of the substance to be
Liquid chromatography (2.2.29). examined in acetonitrile R and dilute to 25.0 mL with the
A. Test solution. Dissolve 25.0 mg of the substance to be same solvento
examined in acetonitrile R and dilute to 25.0 mL with the Reference solution (a) Dilute 1.0 mL of the test solution to
same solvento 100. O mL with acetonitrile R.
Reference solution (a) Dilute 1.0 mL of the test solution to Reference solution (b) Dissolve 5 mg of moxidectin for system
100. O mL acetonitrile R. suitability CRS (containing impurities A, B, C, D, E, F, G,
Reference solution (b) Dissolve 5 mg of moxidectin for system H, 1, J and K) in 5 mL of acetonitrz'le R.
suitability CRS (containing impurities A, B, C, D, E, F, G, Column:
H, 1, J and K) in 5 mL of acetonitrile R. - size: 1 = 0.15 m, 0 = 3.9 mm;
ReferelZce SOlUtlOlZ (c) Dissolve 25.0 mg of moxidectin CRS in - stationary phase: end-capped octadecylsilyl silica gel for
acetonitrile R and dilute to 25.0 mL with the same solvento chromatography R (4 ~lm);
Column: - temperature: 35 oC.
- size: 1 = 0.15 m, 0 = 3.9 mm; Mobile phase Dissolve 3.8 g of ammOniUlll acetate R in
statlonaJY phase: end-capped octadecylsilyl silica gel for 250 mL of 'Water R, adjust to pH 4.2 with acetic acid R and
chromatography R (4 ~lm); add 750 mL of acetonim'le R.
- temperature: 50 oC. Flo'W rate 2.0 mUmin.
Mobile phase Dissolve 7.7 g of ammonium acetate R in Detection Spectrophotometer at 242 nm.
400 mL of 'Water R, adjust to pH 4.8 with glacial acetic acid R
Jnjection 1O ~lL.
and add 600 mL of acetonitrile R.
Run time 10 times tlle retention time of moxidectin.
Flo'W rate 2.5 mUmin.
Jdentification of impurities Use the chromatogram supplied
Detection Spectrophotometer at 242 nm.
with Inoxidectin for syste111 suitability CRS and the
Jnjection 1O ~lL of the test solution and reference solutions (a) chromatogram obtained with reference solution (b) to
and (b). identify the peaks due to impurities H + 1, J and K.
Run time 2 times the retention time of moxidectin. Relative retention With reference to moxidectin (retentión
Identification of impurities U se the chromatogram supplied time = about 4 min): impurity G = about 1.4;
with moxidectin for system suitability CRS and the impurities H and 1 = about 2.0; impurity J = about 2.2;
chromatogram obtained with reference solution (b) to impurity K = about 3.4.
identify the peaks due to impurities A, B, C, D, E + F System suitability: reference solution (b):
and G. - resolution: baseline separation between the peaks due
Relative retention With reference to moxidectin (retention to impurities H + 1 and J.
time = about 12 min): impurity A = about 0.5; Limits:
impurity B = about 0.7; impurity C = about 0.75; - SU111 of impurities H and 1: not more than the area of
impurity D = about 0.94; impurities E and , the principal peak in the chromatogram obtained with
F = about 1.3-1.5; impurity G = about 1.6. reference solution (a) (1.0 per cent);
System suitability: reference solution (b): - impw'ities J, K: for eách impurity, not more than
- peak-to-valleJ! ratio: minimum 3.0, where H p = height 0.5 times the area of the principal peak in the
above the baseline of the peak due to impurity D and chromatogram obtained with reference solution (a)
H v = height above the baseline of the lowest point of (0.5 per cent);
Vet-84 Moxidectin 2016

- any other impurity eluting afier impurity G: for each


impurity, not more than 0.5 times the area of the
principal peak in the chromatogram obtained with
reference solution (a) (0.5 per cent);
- disregard limit: 0.1 times the area of the principal peak
in the chromatogram obtained with reference
solution (a) (0.1 per cent); disregard the peak due to
the stabiliser (identify this peak, where applicable, by
injecting a suitable reference solution).
Total of all impurities Calculate the sum of the impurities
eluting from the start of the run to impurity G in test A, and
from impurities H + 1 to the end of the run in test B.
The total of all impurities is not more than 7.0 per cent. D. 2-epi-moxidectin,
Heavy metaIs (2.4.8)
Maximum 20 ppm.
It complies with test A with the following modifications.
Prescribed solution Dissolve 0.50 g in 20 mL of ethanol
(96 per cent) R.
Test solution 12 mL of the prescribed solution.
Reference solution A mixture of 2 mI of the prescribed
solution, 4 mI of water R and 6 mI of lead standard solution
(1 ppm Pb) R.
Blank solution A mixture of 2 mL of the prescribed solution
and 10 mL of ethanol (96 per cent) R.
Use a membrane filter (nominal pore size 0.45 ~lm).
Water (2.5.12)
E. (4S)-2-dehydro-4-hydromoxidectin,
Maximum 1.3 per cent, determined on 0.50 g.
Sulfated ash (2.4.14)
Maximum 0.2 per cent, determined on 1.0 g.
ASSAY
Liquid chromatography (2.2.29) as described in test A for
related substances with the following modification.
InJection Test solution and reference solution (c).
Calculate the percentage content of C37H53NOs using the
declared content of moxidectin CRS.
IMPURITIES
Specified impurities A, B, C, D, E, F, G, H, 1, ], K

G. (23E,25S)-5 O-desmethyl-28-deoxy-25- [(lE)-1 ,3-


dimethylbut-l-enyl] -23-(methoxyimino )milbemycin B,

H
R5

A. R1 = R2 = R3 = R4 = CH3, R5 = R6 = H:
25-des[ (1E)-1 ,3-dimethylbut-1-enyl] -25- [(lE)-l-methylprop-
1-enyl] moxidectin,
B. R1 = R2 = R3 = R5 = R6 = CH3, R4 = H: H. 2,5-didehydro-5-deoxymoxidectin,
24-desmethylmoxidectin,
C. R1 = R2 = R3 = R4 = R5 = CH3, R6 = H:
2 5-des [( 1E) -1 ,3-dimethylbut-l-enyl] -25- [( 1E) -1-methylbut-1-
enyl] moxidectin,
F. one of groups R1 to R6 is C 2 H 5, the others are CH3:
x-desmethyl-x-ethylmoxidectin,
2016 Nandrolone Laurate Vet-85

H e-s
3 "- DEFINITION
0\ H
Nandrolone Laurate is 3-oxo-estr-4-en-17~-yllaurate.
It contains not less than 97.0% and not more than 103.0%
of C30H4S03, calculated with reference to the dried
substance.
CHARACTERISTICS
A white to creamy white, crystalline powder.
Practically insoluble in 'Water; freely soluble in chloroform, in
ethanol (96%), in ether, in fixed oils and in esters of fatty
acids.
IDENTIFICATION
I. (23S)-23-des(methoxyimino )-23- A. The infrared abs01ption spectnl1n, Appendix n A, is
[(methylsulfanyl)methoxy] moxidectin, concordant with the reference spectrum of nandrolone laurate
(RSV 30).
B. Carry out the method for thin-layer chromatography,
Appendix In A, using the following solutions in chloroform.
(1) 0.5% w/v of the substance being examined.
(2) 0.5% w/v of nandrolone laurate BPCRS.
(3) Mix equal volumes of solutions (1) and (2).
CHROMATOGRAPHIC CONDITIONS
(a) Use as the coating silica gel F 254 precoated plate the
surface of which has been modified by chemically-bonded
octadecylsilyl groups (Whatman KC 18F plates are suitable).
(b) U se the mobile phase as described below.
(c) Apply 5 ~lL of each solution.
J. R = CHT S-CH3 , R' = H: 7-0- (d) Develop the plate to 15 cm.
[(methylsulfanyl) methyl] moxidectin, (e) After removal of the plate, dry in air until the odour of
K. R = H, R' = CO-C 6 H r pN0 2 : 5-0- solvent is no longer detectable and heat at 100 0 for
(4-nitrobenzoyl)moxidectin, 10 minutes. Allow to cool and examine under ultraviolet light
(254 nm).
MOBILE PHASE
20 volumes of 'Water, 40 volumes of acetonitrile and
60 volumes of propan-2-01.
CONFIRMA TION
The principal spot in the chromatogram obtained with
solution (1) corresponds in position and colour to that in the
chromatogram obtained with solution (2).
The principal spot in the chromatogram obtained with
solution (3) appears as a single, compact spot.
C. Melting poim, about 47°, Appendix V A.

L. (23Z)-moxidectin. TESTS
___________________________________________ ~E~
Specific optical rotation
In a 2% w/v solution in 1)4-dioxan, +31 to +35, calculated
with reference to the dried substance, Appendix V F.
Nandrolone
Can)' out the method for thin-layer chromatography,
Nandrolone laurate Appendix nI A, using the following solutions in chlorofonn.
(1) 1.50% w/v of the substance being examined.
Me~ (2) 0.030% w/v of nandrolone BPCRS.
CHROMATOGRAPHIC CONDITIONS
(a) Use as the coating silica gel G.
(b) Use the mobile phase as described below.
o (c) Apply 1 ~lL of each solution.
(d) Develop the plate to 15 cm.
456.7 26490-31-3
(e) After removal of the pla~e, dry in air until the solvent has
Action and use evaporated, spray with a 10% v/v solution of sulfuric acid in
Anabolic steroid; androgen. ethanol (96%), heat at 105° for 30 minutes and examine
under ultraviolet light (365 11111).
Preparation
N androlone Laurate Injection
Vet-86 Nitroxinil 2016

lvIOBILE PHASE Inorganic iodide


3 volumes of acetone and 7 volumes of heptane. To 0.40 g add 0.3 5 g of N -methylglucamine and 10 mL of
water, shake to dissolve and add sufficient water to produce
LIMITS
50 mL. To 10 mL of the resulting solution add 4 mL of
In the chromatogram obtained with solution (1): 1M sulfuric acid and extract with three 10 mL quantities of
Any spot cOlTesponding to nandrolone is not more intense chloroform. Add to the aqueous extract 1 mL of hydrogen
than the spot in the chromatogram obtained with solution (2) peroxide solution (100 vol) and 1 mL of chloroform, shake for
(2%). 2 minutes and allow to separate. Any purple colour produced
Loss on drying in the chloroform layer is not more intense than that
When dried over phosphorus pentoxide at a pressure not obtained in a solution prepared in the following manner.
exceeding 0.7 kPa for 24 hours, loses not more than 0.5% of Add 2 mL of a 0.0026% w/v solution of potassium iodide to a
its weight. U se 1 g. mixture of 4 mL of 1M sulfuric acid and 8 mL of water, add
10 mL of chlorofolln, shake for 2 minutes, add to the aqueous
Sulfated ash
layer 1 mL of hydrogen peroxide solution (100 vol) and 1 mL
Not more than 0.1 %, Appendix IX A.
of chloroform, shake for 2 minutes and allow to separate
ASSAY (500 ppm of iodide).
Dissolve 10 mg in sufficient absolute ethanol to produce Loss on drying
100 mL, dilute 5 mL to 50 mL with absolute ethanol and When dried to constant weight at 105°, loses not more than
measure the absorbance of the resulting solution at the 1.0% of its weight. Use 1 g.
maximum at 240 nm, Appendix II B. Calculate the content
of C 30 H 4S 03, taking 380 as the value of A(l %, 1 cm) at the Sulfated ash
maximum at 240 nm. Not more than 0.1 %, Appendix IX A.

STORAGE ASSAY
N androlone Laurate should be protected from light and Carry out the method for oxygen-flask combustion for iodine,
stored at a temperature 2° and 8°. Appendix VIII C, using 25 mg. Each mL of 0.02M sodium
thiosulfate VS is equivalent to 0.9667 mg of C7H3IN203.
STORAGE
Nitroxinil should be protected from light.
Nitroxinil

Orbifloxacin
(Orbifloxacin for Veterinmy Use) Ph Bur nzonograph 2259)

290.0 1689-89-0

Action and use


Antihelminthic.
Preparation
Nitroxinil Injection 395.4 113617-63-3
~E~ ___________________________________________
DEFINITION
Nitroxinil is 4-hydroxy-3-iodo-5-nitrobenzonitrile. It contains DEFINITION
not les s than 98.0% and not more than 101.0% of 1-Cyclopropyl-7 -[ (3R,5S)- 3,5-dimethylpiperazin-1-yl] -5,6,8-
C7H3IN203, calculated with reference to the dried substance. trifiuoro-4-oxo-1 ,4-dihydroquinoline-3-carboxylic acid.
CHARACTERISTICS Content
A yellow to yellowish brown powder. 99.0 per cent to 101.0 per cent (anhydrous substance).
Practically insoluble in 'water; sparingly soluble in ether; CHARACTERS
slightly soluble in ethanol (96%). It dissolves in solutions of Appearance
alkali hydroxides. White or pale yellow, crystals or crystalline powder.
IDENTIFICATION Solubility
A. The infrared absorption spectrum, Appendix II A, is Very slightly soluble in water, soluble in glacial acetic acid,
concordant with the reference specttum of nitroxinil (RSV 31). practically insoluble in anhydrous ethanol.
B. The light absorption, Appendix II B, in the range 220 to It shows polymorphism (5.9).
350 nm of a 0.002% w/v solution in O.OlM sodium hydroxide IDENTIFICATION
exhibits maxima at 225 nm and at 271 nm. The absorbance at Infrared absorption spectrophotometry (2.2.24).
the maximum at 271 nm is about 1.3.
Comparison orbifioxacin CRS.
C. Heat with sulfuric acid; iodine vapour is evolved.
If the spectra obtained in the solid state show differences,
TESTS dissolve 0.1 g of the substance to be examined and 0.1 g of
Melting point the reference substance separately in 12 mL of methanol R.
136° to 139°, Appendix V A. Heat to boiling while shaking. Filter the solutions and let
2016 Orbifloxacin Vet-87

them cool slowly to room temperature. Filter under vacuum corresponding correction factor: impurity A = 2.8;
and wash the residues with cooled methanol R. Dry the impurity D = 1.4;
residues under vacuum and record new spectra using the - impurities A) D: for each impurity, not more than the are a
residues. of the principal peak in the chromatogram obtained with
reference solution (a) (0.2 per cent);
TESTS
- unspeeified impurities: for each impurity, not more than the
Appearance of solution
area of the principal peak in the chromatogram obtained
The solution is clear (2.2.1) and not more intensely coloured
with reference solution (a) (0.20 per cent);
than reference solution GY4 (2.2.2) Method JI).
- total: not more than twice the area of the principal peak in
Dissolve 0.4 g in a 4 gIL solution of sodium hjidroxide R and the chromatogram obtained with reference solution (a)
dilute to 20 mL with the same solution. (0.4 per cent);
Related substances - disregard limit: 0.5 times the area of the principal peak in
Liquid chromatography (2.2.29). the chromatogram obtained with reference solution (a)
Buffer solution Dissolve 5.9 g of sodium eitrate R in 800 mL (0.10 per cent).
of 'Water R, add 90 mL of glacial aeetie acid R and mix. Adjust Water (2.5.12)
to pH 3.5 with a 240 giL solution of sodium hjidroxide R in 1.5 per cent to 2.9 per cent, determined on 0.250 g.
'Water R and dilute to 1000 mL with 'Water R. Sulfated ash (2.4.14)
Test solution Dissolve 10 mg of the substance to be Maximum 0.1 per cent, determined on 1.0 g.
examined in the buffer solution and dilute to 50.0 mL with
the buffer solution. ASSAY
Dissolve 0.300 g in 100 mL of anhjidrous aeetie acid R.
Referenee solution (a) Dilute 1.0 mL of the test solution to
Titrate with 0.1 M perehlorie acid determining the end-point
50.0 mL with the buffer solution. Dilute 1.0 mL of this
potentiometrically (2.2.20).
solution to 10.0 mL with the buffer solution.
1 mL of 0.1 M perehlorie acid is equivalent to 39.54 rng of
Referenee solution (b) Dissolve 10.0 mg of methjil
C19HzoF3N303'
4-aminobenzoate R in the buffer solution and dilute to
100.0 mL with the buffer solution. Mix 10.0 mL ofthe IMPURITIES
solution with 5.0 mL of the test solution and dilute to Speeified impurities A, D.
50.0 mL with the buffer solution. Dilute 1.0 mL of this Other detectable impurities (the following substances would, if
solution to 50.0 mL with the buffer solution. present at a sufficient level, be detected by one or other of
Referenee solution (e) Dissolve the contents of a vial of the tests in the monograph. They are limited by the general
orbijloxacin impuritji mixture CRS (impurities A and D) in acceptance criterion for other/unspecified impurities and/or
1.0 mL of the buffer solution. by the general monograph Substanees for pharmaeeutieal use
Referenee solution (d) Dilute 0.25 mL of reference solution (2034). It is therefore not necessary to identify these
(c) to 1.0 mL of the buffer solution. impurities for demonstration of compliance. See also 5.10.
Colu11Zn: Control of impUlities in substanees for pharmaeeutieal use): B) C)
E) F) G.
- size: 1 = 33 mm, 0 = 4.6 mm;
- statiol1aJY phase: base-deaetivated oetadeejilsil:yl siliea gel for
ehromatographji R (3 !lm).
Mobile phase dioxane R) methanol R) buffer solution
(4:11:86 V/V/V).
Plozu rate 1 mUmin.
Detection Spectrophotometer at 290 nm.
Jl1Jeetion 1O ~lL.
Run time 9 times the retention time of orbifioxacin.
Jdentifieation of the impunties Use the chromatogram
supplied with orbijloxacin impuritji mixture CRS and the
chromatogram obtained with reference solution (c) to identify A. 1-cyclopropyl-5, 7-bis [(3R,5S)- 3,5-dimethylpiperazin-1-
the peaks due to impurities A and D. yl] 6,8-difiuoro-4-oxo-1,4-dihydroquinoline-3-carboxylic acid,
Relative retention With reference to orbifioxacin
(retention time = about 2 min): impurity A = about 0.5;
methyl 4-aminobenzoate = about 1.2;
impurity D = about 2.5.
Sjistem suitabilitji:
resolution: minimum 2.0 between the peaks due to
orbifioxacin and methyl 4-aminobenzoate in the
chromatogram obtained with reference solution (b);
- signal-to-noise ratio: minimum 10 for the peak due to B. 7-[[(2R)-2-aminopropyl]amino]-1-cyclopropyl-5,6-
impurity A in the chromatogram obtained with reference difiuoro-4-oxo-1 ,4-dihydroquinoline-3-carboxylic acid,
solution (d).
Limits:
- eorrection faetors: for the calculation of contents, multiply
the peak areas of the following impurities by the
Vet-88 Oxfendazole 2016

Oxfendazole ***
*** ***
(Oxjendazole jor Veterinary Use) ***
Ph Eur 1110nograph 1458)

QD
~ }-OCH3

c. Rl = C0 2 H, R2 = H : 1-cyclopropyl-7-[(3R,5S)3,5- I I }-NH
.ó'" S ~ N
dimethylpiperazin-l-yl] -6,8-difluoro-4-oxo-l,4- 11

dihydroquinoline-3-carboxylic acid, o
G. Rl = H, R2 = F: 1-cyclopropyl-7-[(3R,5S)-3,5-
dimethylpiperazin-l-yl]-5,6,8-trifluoroquinolin-4(1H)-one, 315.4 53716-50-0

Action and use


Antihelminthic.
Preparation
Oxfendazole Oral Suspension
~E~ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ ___

DEFINITION
Methyl [5-(phenylsulfinyl)-lH-benzimidazol-2-yl] carbamate.
D. 1-cyclopropyl-7 -[ (3R,5S)- 3,5-dimethylpiperazin-l-yl]-6,8- Content
difluoro-5-hydroxy-4-oxo-l ,4-dihydroquinoline-3-carboxylic 97.5 per cent to 100.5 per cent (dried substance).
acid,
CHARACTERS
Appearance
White or almost white powder.
Solubility
Practically insoluble in water, slightly soluble in ethanol
(96 per cent) and in methylene chloride.
It shows polymorphism (5.9).
IDENTIFICATION
Infrared absorption spectrophotometry (2.2.24).
Comparison oxjendazole CRS.
E. 1-cyclopropyl-5-[(3R,5S)-3,5-dimethylpiperazin-l-yl]- If the spectra obtained in the solid state show differences,
6,7 ,8-trifluoro-4-oxo-l ,4-dihydroquinoline-3-carboxylic acid, dissolve tlle substance to be examined and the reference
substance separately in ethanol (96 per cent) R, evaporate to
dryness and record new spectra using the residues.
TESTS
Related substances
Liquid chromatography (2.2.29).
Test solution Dissolve 25.0 mg of the substance to be
examined in the mobile phase and dilute to 100.0 mL with
the mobile phase.
F. 1-cyclopropyl-5 ,6, 7,8-tetrafluoro-4-oxo-l,4-
Rejerenee solution (a) Dilute 1.0 mL of the test solution to
dihydroquinoline-3-carboxylic acid.
100.0 mL with the mobile phase.
_ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ PhEur
Rejerenee solution (b) To 10 mL ofthe test solution add
0.25 mL of strong hydrogen peroxide solution R and dilute to
25 mL with the mobile phase.
Rejerenee solution (e) Dissolve 5.0 mg ofjenbendazole CRS
(impurity A) and 10.0 mg of oxjendazole impurity B CRS Ín
the mobile phase and dilute to 100.0 mL with the mobile
phase. Dilute 1.0 mL of the solution to 20.0 mL with the
mobile phase.
Rejerenee solution (d) Dissolve 5 mg of oxjendazole with
impurity D CRS in the mobile phase and dilute to 20 mL
with the mobile phase (for identification of impurity D).
Column:
- size: 1 = 0.25 m, 0 = 4.6 mm;
- stationary phase: spherical end-eapped octadeeylsilyl siliea gel
jor ehr0111atography R (5 !lm) with a specific surface area of
350 m 2 /g, a pore sÍZe of 10 nm and a carbon loading of
14 per cent.
2016 Oxyclozanide Vet-89

o
Mobile phase Mix 36 volumes of acetonitrile R and 64 volumes

QD
~ }-OCH3
of a 2 giL solution of sodiu11l pentanesulfonate R previously
adjusted to pH 2.7 with a 2.8 per cent V/V solution of
I I }-NH
# S ~ N
SUlfUl7C acid R. // ,\
O O
Flow rate 1 mUmin.
Detection Spectrophotometer at 254 nm. B. methyl [5-(phenylsulfonyl)-lH-benzimidazol-2-
Injection 20 flL. yl] carbamate,
Run time 4 times the retention time of oxfendazole.
Relative retention With reference to oxfendazole (retention
time = about 6.5 min): impurity C = about 0.7;
impurity B = about 1.5; impurity D = about 1.9;
impurity A = about 3.4.
Syste11l suitability: reference solution (b):
- resolution: minimum 4.0 between the peaks due to C. 5-(phenylsulfinyl)-lH-benzimidazol-2-amine,
impurity C and oxfendazole.
Li111its:
- i11lpurity B: not more than the area of the corresponding
peak in the chromatogram obtained with reference
solution (c) (2.0 per cent);
- únpurity A: not more than the area of the corresponding
peak in the chromatogram obtained with reference
solution (c) (1.0 per cent); D. N,N'-bis [5-(phenylsulfinyl)-lH-benzimidazol-2-yl]urea.
- impurities C) D: for each impurity, not more than the area ___________________________________________ PhE~

of the principal peak in the chromatogram obtained with


reference solution (a) (1.0 per cent);
- unspecified i11lpurities: not more than 0.2 times the area of
the principal peak in the chromatogram obtained with
reference solution (a) (0.20 per cent); Oxyclozan ¡de
- total: maximum 3.0 per cent;
- disregard li11lit: 0.1 times the are a of the principal peak in el

0
«1
the chromatogram obtained with reference solution (a)
(0.10 per cent). OH O
Loss on drying (2.2.32)
Maximum 0.5 per cent, determined on 1.000 g by drying in el
~
~~CIOH
an oven at 105 oC at a pressure not exceeding 0.7 kPa for el
2 h.
el
Sulfated ash (2.4.14)
Maximum 0.2 per cent, determined on 1.0 g.
401.5 2277-92-1
ASSAY
Dissolve 0.250 g in 3 mL of anhydrous formic acid R. Action and use
Add 40 mL of anhydrous acetic acid R. Titrate with 0.1 M Antihelminthic.
perchloric acid, determining the end-point potentiometricalIy Preparation
(2.2.20).
Oxyclozanide Oral Suspension
1 mL of 0.1 M perchloric acid is equivalent to 31.54 mg of
ClsH13N303S. DEFINITION
STORAGE Oxyclozanide is 3,3',5,5',6-pentachloro-2'-
hydroxysalicylanilide. It contains not less than 98.0% and not
Protected from light.
more than 101.0% of C 13H 6 ClsN0 3 , ca1culated with
IMPURITIES reference to the dried substance.
Specified i11lpurities A, B, C, D
CHARACTERISTICS
A pale cream or cream coloured powder.
Very slight1y soluble in water; freely soluble in acetone; soluble
in ethanol (96%); slight1y soluble in chloroform.
IDENTIFICATION
A. The infrared absorption spectrum, Appendix II A, is
A. methyl [5-(phenylsulfanyl)-lH-benzimidazol-2- concordant with the reference spectrum of oxyclozanide
yl] carbamate (fenbendazole), (RSV $3).
B. The light absorption, Appendix TI B, in the range 250 to
350 nm of a 0.003% w/v solution in acidified methanol
exhibits a maximum only at 300 nm. The absorbance at the
maximum is about 0.76, Appendix II B.
C. Melting point, about 208°, Appendix V A.
Vet-90 PiperonyI Butoxide 2016

TESTS
Ionisable chIorine
Piperonyl Butoxide
Dissolve 2 g in 100 mL of methanol, add 10 mL of
l.5M nitric acid and titrate with 0.1 M silver nitrate VS
determining the end point potentiometrically. Not more than
1.4 mL is required (0.25%).
Related substances
Carry out the method for liquid chromatography,
Appendix lIT D, using the following solutions.
(1) 0.1 % w/v of the substance being examined prepared by 338.4 51-03-6
dissolving it in a suitable volume of methanol and slowly Action and use
diluting with water containing 0.1 % v/v olthophosphoric acid to Insecticide.
give a solution containing about the same ratio of methanol
to water as the mobile phase. DEFINITION
(2) Dilute 1 volume of solution (1) to 100 volumes with the Piperonyl Butoxide is 5-[2-(2-butoxyethoxy)ethoxyrnethyl]-6-
mobile phase. propyl-1,3-benzodioxole. It contains not les s than 94.0% and
not more than 102.0% of C19H300S'
CHROMATOGRAPHIC CONDITIONS
CHARACTERISTICS
(a) Use a stainless steel column (20 cm x 4.6 mm) packed
with octadecylsilyl silica gel for chro111atography (5 ~lm) (Hypersil A colourless to light yellow, oily liquido
ODS is suitable). Very slightly soluble in water; miscible with ethanol (96%)
with ether and with petroleum oils.
(b) U se isocratic elution and the mobile phase described
below. IDENTIFICATION
(c) Use a fiow rate of 2 mL per minute. The infrared absorption spectrum, Appendix n A, is concordant
with the reference spectrwl1 of Piperonyl Butoxide (RS 478).
(d) Use an ambient column temperature.
(e) Use a detection wavelength of 300 nm. TESTS
Refractive index
(f) Inject 20 ~lL of each solution.
1.494 to 1.504, Appendix V E.
MOBILE PRASE Weight per rnL
A mixture of methanol and water containing 0.1 % v/v of 1.050 to 1.065 g, Appendix V G.
olthophosphoric acid (a mixture of 38 volumes of water and Sulphated ash
62 volumes of methanol is usually suitable). Not more than 0.2%, Appendix IX A, Method n.
LIMITS Related substances
In the chromatogram obtained with solution (1): Carry out the method for gas chromatography,
the area of any secondaJY peak with a retention time less than Appendix In B, using the following solutions in n-hexane.
that of the principal peak is not greater than one third of the (1) 1.2% w/v of the substance being examined.
area of the principal peak in the chromatogram obtained with (2) Dilute 1 volume of solution (1) to 100 volumes and
solution (2) (0.3%); dilute 1 volume of the resulting solution to 10 volumes.
the area of any secondary peak with a retention time greater (3) Dilute 1 volume of solution (2) to 20 volumes.
than that of the principal peak is not greater than the area of (4) 0.16% w/v of piperonyl butoxide impurity standard BPCRS.
the principal peak in the chromatogram obtained with CHROMATOGRAPHIC CONDITIONS
solution (2) (1 %).
(a) Use a fused silica capillary column (25 m x 0.32 mm)
Loss on drying bonded with a 0.52-~lm layer of 100% dimethylpolysiloxane
When dried to constant weight at 60° at a pressure not (DB1 is suitable).
exceeding 0.7 kPa, loses not more than 1.0% ofits weight. (b) Use helium as the carrier gas at 1 mL per minute.
Use 1 g. (c) Use the temperature gradient described below.
Sulfated ash (d) Use an inlet temperature of 250°.
Not more than 0.2%, Appendix IX A. (e) Use a fiame ionisation detector at a temperature of 300°.
ASSAY (f) Use a split ratio of 1:10.
Dissolve 0.25 g in 75 mL of anhydrous pyridine and pass a (g) Inject 1 /lL of each solution.
current of nitrogen through the solution for 5 minutes. Carry
out Method n for non-aqueous titration, Appendix VIn A, Time Temperature oC Comment
maintaining a current of nitrogen through the solution
(Minutes)
throughout the titration, using 0.1 M tetrabutyla11111l0niu11l
hydroxide VS as titrant and determining the end point 0-2 70. isocratic
potentiometrically. Each mL of O.lM tetrabutylam1110niu111
hydroxide VS is equivalent to 20.07 mg of C 13H 6 ClsN0 3. 2-45 70~285 linear gradient

45-60 285 isocratic

SYSTEM SUITABILITY
The test is not valid unless:
in the chromatogram obtained with solution (3), the signal-to-
noise ratio of the principal peak is at least 10;
2016 Potassium Selenate Vet-91

the chromatogram obtained with solution (4) closely


resembles the reference chromatogram supplied with piperonyl
butoxide i11lpurity standard BPCRS;
the resolution between piperonyl butoxide and dipiperonyl
methane is at least 2.
LIMITS
In the chromatogram obtained with solution (1):
determine the amount of each seconda¡y peak by [di-((6-propyl-benzo-[1,3]dioxol-5-yl)methyl)ether]
110 rmalis ation. (dipiperonyl ether).
dipiperonyl methane is not greater than 2.0%;
diethylene glycol butyl ether is not greater than 2.0%;
dipiperonyl ether is not greater than 1.5%;
dihydrosafrole is not greater than 50 ppm; Potassium Selenate
any other seconda¡y peak is not greater than 0.5%; 221.2 7790-59-2
the sum all the seconda¡y peaks is not greater than 2.0%.
Action and use
Disregard any peak with an area les s than principal peak in
Used, with Alpha Tocopheryl Acetate, in the treatment of
solution (2) (0.1 %) except for dihydrosafrole.
nutritional muscular dystrophy.
ASSAY
Carry out the method for gas chromatography, DEFINITION
Appendix III B, using the following solutions in hexane. Potassium Selenate contains not les s than 97.0% and not
(1) 0.16% w/v ofthe substance being examined. more than 100.5 % of K ZSe04, calculated with reference to
the dried substance.
(2) 0.16% w/v ofpiperonyl butoxide BPCRS.
(3) 0.16% w/v ofpiperonyl butoxide impurity standard BPCRS. CHARACTERISTICS
Colourless crystalsor a white, crystalline powder.
CHROMATOGRAPHIC CONDITIONS
Freely soluble in water.
The chromatographic conditions described under Related
substances may be used. IDENTIFICATION
A. Yields the reactions characteristic of potassiu111 salts,
SYSTEM SUIT ABILITY
Appendix VI.
The test is not valid unless the chromatogram obtained with
B. To a solution of 0.1 g in 3 mL of 'lvata add 1 mL of
solution (3) closely resembles the reference chromatogram
hydrochloric acid and 0.5 mL of hydrazine hydrate and boil.
supplied with piperonyl butoxide impurity standard BPCRS and
A red precipitate is produced.
the resolutio11 between piperonyl butoxide and dipiperonyl
methane is at least 2. C. Acidify 1 mL of a 1% w/v solution with 2M hydrochloric
acid and add 0.15 mL of barium chlorz'de solution. Wash the
Calculate the content of C19H300S using the declared
precipitate with water and boil with hydrochloric acid. Chlorine
content of C19H300S in piperonyl butoxide BPCRS.
is evolved.
IMPURITIES
TESTS
Chloride
HO~O~O~ 0.3 g complies with the li111it test for chlorides, Appendix VII
(170 ppm).
2-(2-Butoxyethoxy)ethanol, (diethylene glycol butyl ether); Selenite
Not more than 0.1 %, calculated as Se03' when determined
by the following method. Dissolve 2.0 g in 50 mL of water,
add 50 mL of 9M sulfunc acid, 12 g of disodiu111 hydrogen
orthophosphate and 10 mL of 0.02M potassiu11l permanganate
VS and allow to stand for 20 minutes with occasional
agitation. Titrate the excess of potassium permanganate with
0.1 M a1111110niu111 iron (JI) sulfate VS. Each mL of
5-propyl-1,3-benzodioxole, 1,2-methylendioxy-4- 0.02M potassiu11l per111anganate VS is equivalent to 6.348 mg
propylbenzene, (dihydrosafrole); of Se03'
Loss on drying
When dried to constant weight at 105°, loses not more than
0.1 % of its weight. Use 1 g.
ASSAY
Dissolve 1 g in 60 mL of water, add 15 mL of hydrochlonc
acid anq 5 mL of a 50% w/v solution of hydrazine hydrate,
o
boil, heat on a water bath for 3 hours, and allow to stand
<O ovemight. Transfer the precipitated selenium to a weighed,
sintered-glass crucible, wash with hot water until the washings
are free from chloride ions, rinse with absolute ethanol and dry
[1,1' - di-(6-propyl-benzo-[1,3]-dioxol-5-yl)methane], at 105° to constant weight. Correct for the amount of
(dipiperonyl methane);
Vet-92 Pyrethrum Flower 2016

selenium present as se1enite found in the test for se1enite. potassiul1l hJ!droxide and boi1 under a reflux condenser for
Each mg of se1enium is equiva1ent to 2.801 mg of K2Se04' 45 minutes. Transfer the solution to a beaker and wash the
flask with sufficient hot water, adding the washings to the
beaker, to produce a total vo1ume of 200 mL. Boi1 unti1 the
vo1ume is reduced to 150 mL, coo1 rapid1y and transfer the
solution to a stoppered flask, washing dle beaker with three
Pyrethrum Flower 20 mL quantities of water and transfening any gummy
Da1matian Insect F10wers residue to the flask. Add 1 g of diatomaceous eardl (Filterce1
Preparation is suitab1e) and 10 mL of bariulll chloride solution, swirl gent1y
Pyrethrum Extract and add sufficient water to produce 250 mL. Stopper the
flask, shake vigorous1y until dle separating 1iquid is clear and
DEFINITION filter the suspension through a filter paper (Whatman No.
Pyredlrum F10wer is the dried flower heads of 1 is suitab1e).
Chlysanthemum cineral'iaefolizmz Vis. It contains not 1ess than
F or pyrethrin 1
1.0% of pyrethrins of which not les s dlan ha1f consists of
Transfer 200 mL of the filtrate to a separating funne1, rinsing
pyredrrin I.
the measuring vesse1 with two 5 mL quantities of water, and
CHARACTERISTICS add 0.05 mL of phenolphthalein solutio71 Rl. Neutra1ise dle
Odour, faint but characteristic. solution by the drop wise addition of hydrochloric acid and
Maeroseopieal add 1 mL of hydrochloric acid in excess. Add 5 mL of a
Capitula occuning 100se 01' compressed into mas ses; saturated solution of sodúmz chloride and 50 mL of aromatic-
individual capitula more 01' les s flattened, about 6 to 12 mm free petrolewll spirit (boziing range) 40° to 60°), shake vigorous1y
in diameter, and common1y with a short pie ce of sta1k for 1 minute, allow to separate, remove and retain dle 10wer
attached; receptacle a1most flat, about 5 to 10 mm in 1ayer. Filter dle petro1eum spirit extract through absorbent
diameter, without pa1eae, surrounded by an involucre of 2 01' cotton into a second separating funne1 containing 10 mL of
3 rows of brownish yellow 1anceo1ate bracts; ray florets water. Return dle aqueous 1ayer to the first separating funne1
number about 15 to 23, disc florets about 200 to 300; and repeat the extraction widl 50 mL and then with 25 mL
corollas of ligu1ate florets pa1e brownish and shrivelled, of aro77latic-free petroleuJ1Z spirit (boiling range) 40° to 60"),
ob10ng, about 16 mm long with drree rounded apica1 teeth, reserving dle aqueous 1ayer for dle assay of pyrethrin II, and
the central tooth frequendy smaller than the two lateral ones; filtering the petro1eum spirit extracts t1lrough the same
in the midd1e region of the corolla about 17 veins present; absorbent cotton into the second separating funnel. Shake
corollas of the disc florets tubular, yellow, widl five short the combined petro1eum spirit extracts and water for about
10bes, and enclosing five epipeta10us, syngenesious stamens; 30 seconds and allow to separate; remove the 10wer 1ayer and
each ray and disc floret has an inferior five-ribbed ob10ng add it to the aqueous 1iquid reserved for dle assay of
ovary about 5 mm long with a filiform stv1e and bifid stürma pyrethrin II. Wash the combined petroleum spirit extracts
and sunnounted by a membranous tubuiar ca1yx about b with a furdler 10 mL of water, adding the washings to the
1 mm long; ovaries and 10wer part of the corollas covered reserved aqueous 1iquid.
widl numerous scattered shining oi1 glands. To t1le petro1eum spirit extracts add 5 mL of O.lM sodiz1771
Micro se opic al hydroxide, shake vigorously for 1 minute, allow to separate
Loose1y ananged 1arge-celled sclereids of the receptacle widl and remove the clear 10wer layer, washing the stem of t1le
moderate1y thickened walls and few pits; fragments of the separating hume1 with 1 mL of water. Repeat the extraction
cOl"ollas of the florets, dlose of the ray florets showing by shaking for about 30 seconds with two quantities of
puckered papillae on the inner epidermis and sinuous-walled 2.5 mL and 1. 5 mL of 0.1 M sodium lzydroxide and add t1le
cells with a striated cuticle on the outer epidemlis, those of extracts to the a1ka1ine extracto Add to t1le flask 10 mL of
the tubular florets composed of cells with slighdy dlickened l7lercU/y (11) sulfate solution, stopper, swirl and allow to stand in
walls with papillae occurring on1y on the 10bes; ovoid to the dark at 25° ± 05: for exact1y 60 minutes after t1le
spherica1 glandular trichomes each composed of a ShOlt, addition of the mercury(n) sulfate solution. Add 20 mL of
biseriate sta1k and a biseriate head with two or four cells; acetone and 3 mL of a saturated solution of sodiu711 chlo}'ide,
covering trichomes twisted, T -shaped widl moderate1y heat to boiling on a water bat1l, allow the precipitate to setde
thickened walls; numerous pollen grains, spherica1, 34 to and decant the supernatant liquid through a filter paper
40 ~lm in diameter with three pores and a Walty and spiny (Whatman No. 1 is suitab1e), retaining most of the
exine; groups of small rectangular sclereids, sorne containing precipitate in the flask. Wash the precipitate with 10 mL of
prisms of calcium oxa1ate, from the invo1ucra1 bracts, ovaries acetone, again boi1, allow to sett1e and decant du"ough dle
and basal regio n of the ca1yx; parenchymatous cells of the same filter paper. Repeat t1le washing and decanting with
ca1yx and ovaries containing tabular or diamond-shaped three 10 mL quantities of hot chlorofornl. Transfer the fi1ter
crysta1s of calcium oxa1ate; occasiona1 cells containing cluster paper to the flask, add 50 mL of a cooled mixture of three
crysta1s from the base of the cOl"olla of the disc florets; vo1umes of hydrochloric acid and two vo1umes of water, 1 mL
portions of stigmas with papillose tips. of strong iodine nzonoclzloride reagent and 6 mL of chloroform.
Titrate with O.OlM potassiU711 ~odate VS, running almost all the
Acid-insoluble ash
required vo1ume of titrant into dle flask in one pOltion.
Not more than 1.0%, Appendix XI K.
Continue the titration, shaking dle flask vigorously for
ASSAY 30 secondsaftei" each addition of the titrant, unti1 dle
Transfer 12.5 g in No. 1000 powder to an apparatus for the ch1orofOlID is co10urless. Repeat the Qperation without the
continuous extraction of drugs, Appendix XI F, and extract with extract; the difference between dle titrations represents the
aromatic-free petroleum spirit (boiling range) 40: to 60) for amount of potassium iodate required. Each mL of
7 hours. Evaporate the extract to about 40 mL and allow to O.OlM POtassiU711 iodate VS is equivalent to 5.7 mg of
stand ovemight at 0° to S". Add 20 mL of 0.5M ethanolic pyrethrin I.
2016 Selamectin Vet-93

F or pyrethrin II TESTS
Transfer the combined aqueous liquids reserved in the Assay Colour of solution
for pyrethrin I to a beaker, cover widl a watch glass and A 0.5% w/v solution in methanol is not more intensely
evaporate to 50 mL within 35 to 45 minutes. Cool, washing coloured than reference solution Y6, Appendix IV B, Method n.
dle underside of dle watch glass with not more than 5 mL of (1-Methyl-5-nitroimidazol-2-yl) methanol
water and adding dle washings to the beaker. Filter through Cany out dle medl0d for thin-layer ch1'Omatography,
absorbent cotton into a separating Mmel, washing with Appendix lIT A, using silica gel GF254 as the coating
successive quantities of 10, 7.5, 7.5, 5 and 5 mL of water. substance and a mLxture of 5 volumes of glacial acetic acid,
Saturate the aqueous liquid with sodium chloride, add 10 mL 5 volumes of methanol and 80 volumes of toluene as the
of hyd1'Ochloric acid and 50 mL of ether, shake for 1 minute, mobile phase. Apply separately to the plate 20 ~lL of each of
allow to separate, and remove the lower layer. Repeat the two solutions in acetone containing (1) 1.0% w/v of the
extraction successively widl 50, 25 and 25 mL of ether. Wash substance being examined and (2) 0.0050% w/v of (l-methyl-
dle combined ether extracts with three 10 mL quantities of a 5-nitroimidazol-2-yl)methanol BPCRS. After removal of the
saturated solution of sodiu771 chloride and transfer dle edler plate, allow it to dry in air and examine under ultraviolet light
layer to a flask with the aid of 10 mL of ether. Remove dle (254 1ll1l). Any spot in the chromatogram obtained widl
bulk of the ether by distillation and remove the remainder solution (1) corresponding to (l-methyl-5-nitroimidazol-2-
with a gentle current of air and dry dle residue at 100° for yl)methanol is not more intense than the spot in the
10 minutes, removing any residual acid frunes with a gentle chromatogram obtained with solution (2) (0.5%).
current of airo Add 2 mL of ethanol (96%) previously
neutralised to phenolphthalein solution R1 and 0.05 mL of Water
phenolphthalein solution R1, swirl to dissolve the residue, add Not more than 0.5% w/w, Appendix IX C. Use 5 g.
20 mL of carbon dioxide-free water and titrate rapidly widl Sulfated ash
0.021\,1 sodium hydroxide VS until the colour changes to Not more than 0.1 %, Appendix IX A.
brownish pink and persists for 30 seconds, keeping the flask ASSAY
stoppered between additions of alkali. Repeat dle operation
Carry out Method I for non-aqueous titration,
using dle aqueous liquid reserved for dle repeat operation in
Appendix VIn A, using 0.3 g and detennining the end point
dle Assay for pyrethrin 1. The difference between the
potentiometrically. Each mL of 0.1 M perchloric acid VS is
titrations represents the volume of 0.02M sodiu17l hyd1'Oxide VS
equivalent to 20.02 mg C 6 H sN 40 4 .
required. Each mL of 0.02M sodium hydroxide VS is
equivalent to 3.74 mg of pyred1rin n. STORAGE
Ronidazole should be protected from light.

*****
** **
Me O (Selamectill for Veterillary Use) Ph. Eur. 771onograph ***
02
N-(r°)lNH 2
2268)

200.2 7681-76-7

Action and use


Antiprotozoal.

DEFINITION
Ronidazole is (1-medwl-5-nitroimidazol-2-yl)medlyl
carbamate. It contains not less than 98.5% and not more
than 10l.0% of C6HgN40Lb calculated with reference to dle
anhydrous substance.
770 165108-07-6
CHARACTERISTICS ~E~ _____________________________________________
A white to yellowish brown powder; odourless or almost
odourless. DEFINITION
Slight1y soluble in water, in chlo1'Oforl71 and in ethanol (96%); (2aE,2' R,4E,5' S,6S,6' S,7S,8E,11R,15S,17aR,20Z,20aR,
very slighdy soluble in edler. 20bS)-6/-cyc1ohexyl-7 - [(2,6-dideoxy-3-0-methyl-O:-L-arabino-
hexopyranosyl) oxy] -20b-hydroxy-20-(hydroxyimino)-
IDENTIFICATION 5/,6,8, 19-tetramethyl-
A. The infrared absorption spectrwn, Appendix n A, is 3/,4/,5/,6,6/,7,10,11,14,15,17a,20,20a,20b-
concordant with the reference spectrul71 of ronidazole tetradecahydrospiro [2H, 17 H-11, 15-medlanofuro [4,3,2-
(RSV 36). pq][2,6]benzodioxacyc100ctadecine-13,2/-pyran]-17-one
B. The light absorption, Appendix n B, in the range 230 to ((5Z,21 R,25S)-25-cyc1ohexyl:-4' -O-de (2,6-dideoxy-3-0-
350 nm of a 0.002% w/v solution in O.lM methanolic methyl-O:-L-arabino-hexopyranosyl)-5-demethoxy-25-
hydrochloric acid exhibits a maximum only at 270 nm. The de( 1-met1lylpropyl)-22,23-dihydro-5-
absorbance at dle maximum is about 0.64. (hydroxyimino)avennectin Ala)'
C. Melti71g poim, about 161', Appendix V A. Semi-synthetic product derived from a fennentation producto
Vet-94 Selamectin 2016

Content impurities C, D: for each impurity, not more than


96.0 per cent to 102.0 per cent (anhydrous substance). 1.5 times the area of the principal peak in the
CHARACTERS chromatogram obtained with reference solution (a)
(1.5 per cent);
Appearance
any other impurz'ty: for each impurity, not more than the
White or almost white, hygroscopic powder.
area of the principal peak in the chromatogram obtained
Solubility with reference solution (a) (1.0 per cent);
PracticalIy insoluble in water, freely soluble in isopropyl - total: not more than 4 times the area of the principal peak
alcohol, soluble in acetone and in methylene chloride, in the chromatogram obtained with reference solution (a)
sparingly soluble in methanol. (4.0 per cent);
IDENfIFICATION - disregard limit: 0.2 times the area of the principal peak in
Infrared absorption spectrophotometry (2.2.24). the chromatogram obtained with reference solution (a)
(0.2 per cent).
Comparison selamectin CRS.
Heavy metals (2.4.8)
TESTS
Maximum 20 ppm.
Related substances
Dissolve 2.0 g in ethanol (96 per cent) R and dilute to
Liquid chromatography (2.2.29).
20.0 mL with the same solvento 12 mL of the solution
Solvent mixture water R, acetonitrile R (40:60 V/l!). complies with test B. Prepare the reference solution using
Test solution Dissolve 25 mg of the substance to be examined lead standard solution (2 ppm Pb) obtained by diluting lead
in the solvent mixture and dilute to 50 mL with the solvent standard solution (lOO ppm Pb) R with ethanol (96 per cent) R.
mixture. Filter the solution through a membrane filter (nominal pore
Reference solution (a) Dilute 1.0 mL of the test solution to size 0.45 ~lm). Compare the spots on the filters obtained with
100.0 mL with the solvent mixture. the different solutions. Any brownish-black'colour in the spot
Reference solution (b) Dissolve 2.5 mg of selamectin for peak from the test solution is not more intense than that in the
identification CRS (containing impurities A, C and D) in the spot from the reference solution.
solvent mixture and dilute to 5 mL with the solvent mixture. Water (2.5.12, Method A)
Reference solution (c) Dissolve the contents of a vial of Maximum 7.0 per cent, determined on 0.20 g.
selamectin impurz'ty B CRS in 1.0 mL of reference Sulfated ash (2.4.14)
solution (b). Maximum 0.1 per cent, determined on 1.0 g.
Column:
ASSAY
- size: 1 = 0.15 m, 0 = 3.9 mm;
Liquid chromatography (2.2.29).
- statiol1a/Y phase: end-capped octadecylsilyl silica gel for
chromatogmphy R (4 ~lm); Test solution Dissolve 50.0 mg of the substance to be
- tempemture: 30 oC. examined in the mobile phase and dilute to 250.0 mL with
the mobile phase.
Mobile phase:
- mobile phase A: water R; Reference solution Dissolve 50.0 mg of selamectin CRS in the
- 1110bile phase B: acetonitrile R; mobile phase and dilute to 250.0 mL with the mobile phase.
Column:
- size: 1 = 0.15 m, 0 = 3.9 mm;
Time Mobile phase A Mobile phase B
- stationa/Y phase: end-capped octadecylsiljJI silica gel for
(min) (per cent V/V) (per cent V/V)
chromatography R (4 ~lm);
0-28 40 60
- tempemture: 30 oC.
28 - 45 40 ~ 20 60 ~ 80
Mobile phase water R, acetonitrile R (20:80 V/V).
Flow rate 1.0 mUmin.
Flow rate 2.0 mUmin. Detection Spectrophotometer at 243 nm.
Detectiol1 Spectrophotometer at 243 nm. Iny'ection 2O ~lL.
Iny'ection 2 O ~lL. Run time Twice the retention time of selamectin.
Identification of impurities Use the chromatogram supplied Retention time Selamectin = about 9 min.
with sela111ectin for peak identification CRS and the Calculate the percentage content uf C43H63NOll from the
chromatogram obtained with reference solution (c) to identify declared content of selamectin CRS.
the peaks due to impurities A, B, C and D.
STORAGE
Relative retention With reference to selamectin (retention In an airtight container.
time = about 22 min): impurity A = about 0.2;
impurity B = about 0.4; impurity C = about 0.5; IMPURlTIES
impurity D = about 1.7. Specified impurities A, B, C, D
System suitability Reference solution (c):
- resolution: minimum 2.5 between the peaks due to
impurities B and C.
Limits:
- correction factor: for the calculation of content, multiply the
peak area of impurity D by 1.5;
- impurities A, B: for each impurity, not more than twice
the area of the principal peak in the chromatogram
obtained with reference solution (a) (2.0 per cent);
2016 Sodium Selenite Vet-95

(2, 6-dideoxy-3-0-methyl-cx-L-ambino-hexopyranosyl) -3-0-


methyl-cx-L-ambino-hexopyranosyl] -S-demethoxy-2S-
de( l-methylpropyl)-22,23-dihydro-S-
(hydroxyimino)avermectin AlJ,

Hrc:30yO~30~~-~,
p P
OCH 3 OCH 3
H3 C

A. (2aE,2' R,4E,4' S,S' S,6S,6' R, 7S,8E,IIR,ISS,17aR,20Z,


20aR,20bS)-6'-cyclohexyl-7 -[ (2,6-dideoxy-3-0-methyl-cx-
L-ambino-hexopyranosyl) oxy]-4 ',20b-dihydroxy-20-
(hydroxyimino)-S ',6,8, 19-tetramethyl-
3',4',S',6,6',7,10,11,14,IS,17a,20,20a,20b-
D. (2aE,2'R,4E,5' S,6S,6' S, 7S,8E,IIR,ISS,17aR,20Z,20aR,
tetradecahydrospiro [2H, 17H-ll,IS-methanofuro [4,3,2-
20bS)-6' -cyc1ohexyl-7 - [(2, 6-dideoxy-3-0-methyl-cx-L-
pq] [2,6]benzodioxacyc1ooctadecine-13,2'-pyran]-17-one
ambino-hexopyranosyl-(1--+4)-2, 6-dideoxy-3-0-methyl-cx-
((SZ,21R,23S,2SR)-2S-cyclohexyl-4'-0-de(2,6-dideoxy-3-
L-arabino-hexopyranosyl) oxy] -20b-hydroxy-20-
0-methyl-cx-L-ambino-hexopyranosyl)-S-demethoxy-2S-
(hydroxyimino )-S' ,6,8, 19-tetramethyl-
de(l-methylpropyl)-22,23-dihydro-23-hydroxy-S-
3',4',S',6,6',7,10,11,14,IS,17a,20,20a,20b-
(hydroxyimino)avermectin AlJ,
tetradecahydrospiro [2H, 17 H-ll, IS-methanofuro[4,3,2-
pq] [2,6]benzodioxacyc1ooctadecine-13,2'-pyran]-17-one
((SZ,21 R,2SS) -2S-cyclohexyl-S-demethoxy-2 S-
de( l-methylpropyl)-22,23-dihydro-S-
(hydroxyimino)avermectin AlJ.
_ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ PhEur

Sodium Selenite
172.9 10102-18-8

Action and use


B. (2aE,2' S,4E,S' S,6S,6'R, 7S,8E,IIR,ISS,17aR,20Z,20aR, Used in treatment of selenium deficiency.
20bS)-6'-cyclohexyl-7 - [(2,6-dideoxy-3-0-methyl-cx-L-
ambino-hexopyranosyl) oxy] -20b-hydroxy-20- DEFINITION
(hydroxyimino )-S ',6,8, 19-tetramethyl- Sodium Selenite contains not les s than 44.0% and not more
S',6,6',7,10,11,14,IS,17a,20,20a,20b- than 46.0% of Se, calculated with reference to the dried
dodecahydrospiro [2H, 17H-ll, IS-methanofuro [4,3,2- substance.
pq] [2,6]benzodioxacyc1ooctadecine-13,2'-pyran] -17 -one
CHARACTERISTICS
((SZ,2SR)-2S-cyclohexyl-4'-O-de(2,6-dideoxy-3-0-methyl-
cx-L-ambino-hexopyranosyl)-S-demethoxy-2S- White to slightly greyish pink, granular powder.
de( l-methylpropyl)-S-(hydroxyimino) avermectin Ala), Freely soluble in water; practically insoluble in ethanol (96%)
and in ether.
Dissolve 2 g in caI"bon dioxide-free water prepared from distilled
water and dilute to 100 mL with the same solvent (solution
S).
IDENTIFICATION
A. Dissolve SO mg in S mL of 7M hydrochloric acid, add 2 mL
of O.IM sodiu111 thiosulfate and heat to boiling. A reddish
orange precipitate is produced.
B. 1 mL of solution S yields the reactions characteristic of
sodiu111 salts, Appendix VI.
TESTS
Clarity of solution
C. (2aE,2'R,4E,S' S,6S,6' S,7S,8E,IIR,ISS,17aR,20Z,20aR, Solution S is not more opalescent than reference suspension JI,
20bS)-6' -cyclohexyl-7 ,20b-dihydroxy-20-(hydroxyimino)- Appendix IV A.
S' ,6,8, 19-tetramethyl- AIkalinity
3',4',S',6,6',7,10,11,14,IS,17a,20,20a,20b- To SO mL of solution S add 0.2 mL of thy11lolphthalein
tetradecahydrospiro [2H, 17 H-ll, IS-methanofuro [4,3,2- solution. Not more than 0.2S mL of 1M hydrochloric acid VS is
pq] [2,6]benzodioxacyc1ooctadecine-13 ,2'-pyran] -1 7-one required to change the colour of the solution from dark blue
((SZ,21R,2SS)-2S-cyclohexyl-13-0-de [2,6-dideoxy-4-0- to very pale blue .
Vet-96 Spectinornycin Sulfate Tetrahydrate 2016

Chloride CHARACTERS
Dilute 2.5 mL of solution S to 15 mL with water. Appearance
The solution complies with the limit test for chlorides, White or almost white powder.
Appendix VII (0.1 %). Solubility
Sulfate Freely soluble in water, insoluble in ethanol (96 per cent).
Dilute 3.75 mL of solution S to 15 mL with distilled water.
IDENTIFICATION
The solution complies with the limit test for sulfates,
A. Infrared absorption spectrophotometry (2.2.24).
Appendix VII (0.2%).
Comparison spectinomycin sulfate tetrahydrate CRS.
Loss on drying
0
When dried to constant weight at 100 to 105 loses not 0
,
B. Dilute 1.0 mL of solution S (see Tests) to 10 mL with
more than 1.0% of its weight. Use 1 g. 'luater R. The solution gives reaction (a) of sulfates (2.3.1).

ASSAY TESTS
Dissolve 1 g in 100 mL of 'luater, add 30 mL of hydrochloric Solution S
acid and heat just to boiling. Remove from the heat, add Dissolve 2.50 g in carbon dioxide-free water R and dilute to
carefully, while shaking, 100 mL of sulfur dioxide solution, heat 25.0 mL with the same solvento
the solution to boiling, cover with a watch glass and heat in a Appearance of solution
water bath for 2 hours. Check that the precipitation of Solution S is clear (2.2.1) and colourless (2.2.2, Method JI).
selenium is complete by adding a further 10 mL of sulfur pH (2.2.3)
dioxide solution and then allow to cool. Filter through a tared, 3.8 to 5.6 for solution S.
sintered-glass filter (16) and wash the precipitate with water
Specific optical rotation (2.2. 7)
until the washings are free from chloride. Dry the filter at
0 0
100 to 105 reweigh and determine the weight of Se
,
+ 10.0 to + 14.0 (anhydrous substance).
obtained. Dissolve 2.50 g in an 8 mUL solution of concentrated
ammonia R1 and dilute to 25.0 mL with the same solvento
Allow the solution to stand at room temperature for not less
than 30 min and not more than 2 h prior to determination.
Related substances
Spectinomycin Sulfate Liquid chromatography (2.2.29). Jn order to avoid the
Tetrahydrate formation of anomers, prepare the solutions immediately before use.
Spectinomycin Sulphate Tetrahydrate Test solution Dissolve 15. O mg of the substance to be
examined in the mobile phase and dilute to 100.0 mL with
(Spectinomycin Sulfate Tetrahydrate for Veterinmy Use,
the mobile phase.
Ph Bur monograph 1658)
Reference solution (a) Dissolve 3 mg of spectin0117ycin for system
OH
suitability CRS in the mobile phase and dilute to 20 mL with

/~~:
7 O~H
the mobile phase.
H3C o . .CH 3
Reference solution (b) Dilute 1.0 mL of the test solution to
HO O.
100.0 mL with the mobile phase.
H OH 'R'
HN, R Reference solution (c) Dilute 3.0 mL of reference solution (b)
CH 3 to 10.0 mL with the mobile phase.
Compound R R' Molee. Formula Column:
- size: 1 = 0.25 m, 0 = 4.6 mm;
- stationary phase: octylsilyl silica gel for chro111atography R
(5 ~lm);
- temperature: ambient and constant.
Mobz7e phase Dissolve 4.2 g of oxalic acid R and 2.0 mL of
~E~ ___________________________________________ heptafiuorobutyric acid R in water R and dilute to 1000 mL
with water R; adjust to pH 3.2 with sodium hydroxide
DEFINITION solution R; add 105 mL of acetonitrile R and mix; filter
Mixture of (2R,4aR,5aR,6S,7S,8R,9S,9aR,10aS)-4a,7,9- through a 0.45 ~lm filter and degas with helium for
trihydroxy-2-methyl-6,8-bis (methylamino) decahydro-4H- chro111atography R for 10 min.
pyrano[2,3-b] [1,4]benzodioxin-4-one sulfate tetrahydrate Plow rate 1.0 mUmin.
(spectinomycin sulfate tetrahydrate) and Post-column solution carbonate-free sodium hydroxide solutiol'l R
(2R,4R,4aS,5aR,6S, 7S,8R, 9S, 9aR, 10aS)-2-methyl-6,8- diluted with carbon dioxide-free water R to obtain a final
bis (methylamino )decahydro-2H-pyrano [2,3- concentration ofNaOH of 21 gIL. Degas the solution with
b] [1,4]benzodioxine-4,4a, 7,9-tetrol sulfate tetrahydrate ((4R)- heliu111 for chromatography R for 10 min before use. Add it
dihydrospectinomycin sulfate tetrahydrate). pulse-less to the column efflüent using a 375 ~L polymeric
It is produced by Streptomyces spectabilis or by any other mixing coil.
means. Post-column fiow rate 0.5 mUmin.
Content Detection Pulsed amperometric detection or equivalent with a
- (4R)-dihydrospectinomycin sulfate: maximum 2.0 per cent gold indicator electrode having preferably a diameter of
(anhydrous substance); 1.4 mm or greater, a suitable reference electrode and a
- sum of the contents of spectinomycin sulfate and stainless steel counter electro de, held at + O.12 V detection,
(4R)-dihydrospectinomycin sulfate: 93.0 per cent to + 0.70 V oxidation and -0.60 V reduction potentials
102.0 per cent (anhydrous substance).
2016 Spectinomycin Sulfate Tetrahydrate Vet-97

respectively, with pulse durations according to the instrument System suitability:


used. Keep the detection cell at ambient and constant - repeatability: maximum relative standard deviation of
temperature. Clean the gold indicator electrode with an 3.0 per cent for the principal peak after 6 injections of the
eraser and damp precision wipe prior to start-up of the reference solution.
system to enhance the detector sensitivity and increase the Calculate the sum of the percentage contents of
signal-to-noise ratio. spectinomycin sulfate and (4R)-dihydrospectinomycin sulfate
Injection 20 ~lL. from the declared contents of C14H26Cl2N207 and
Run time 1.5 times the retention time of spectinomycin. C14H2SCl2N207 in spectinomycin hydrochloride CRS, applying
Identification of impurities U se the chromatogram supplied a correction factor of 1.062.
with spectinomycin for system suitability CRS and the STORAGE
chromatogram obtained with reference solution (a) to In an airtight container. If the substance is sterile, store in a
identify the peaks due to impurities A, D and E. sterile, airtight, tamper-proof container.
Relative retention With reference to spectinomycin (retention IMPURITIES
time = 11 min to 20 min): impurity A = about 0.5; Specified impunties A, D, E
impurity D = about 0.7; impurity E = about 0.9;
Other detectable impurities (the following substances would, if
(4R)-dihydrospectinomycin = about 1.3.
present at a sufficient level, be detected by one or other of
System suitability: reference solution (a): the tests in the monograph. They are limited by the general
- resolution: minimum 1.5 between the peaks due to acceptance criterion for other/unspecified impurities and/or
impurity E and spectinomycin. by the general monograph Substances for pharmaceutical use
Limits: (2034). It is therefore not necessary to identify these
- correction factor: for the calculation of content, multiply the impurities for demonstration of compliance. See also 5.10.
peak area of impurity A by 0.4; Control of impunties in substances for phannaceutical use): B, C,
- impunties A, E: for each impurity, not more than the area F, G.
of the principal peak in the chromatogram obtained with
reference solution (b) (1.0 per cent); OH
- impunty D: not more than 4 times the area of the
principal peak in the chromatogram obtained with H3C
/~q: OH
reference solution (b) (4.0 per cent);
Ha "OH
- any other impurity: for each impurity, not more than the
HN,
area of the principal peak in the chromatogram obtained CH 3
with reference solution (b) (1.0 per cent);
- total: not more than 6 times the area of the principal peak
A. 1,3-dideoxy-1,3-bis(methylamino)-myo-inositol
in the chromatogram obtained with reference solution (b)
(actinamine) ,
(6.0 per cent);
- disregard limit: the area of the principal peak in the
chromatogram obtained with reference solution (c)
(0.3 per cent); disregard the peak due to
(4R)-dihydrospectinomycin.
and epimer at C*
Water (2.5.12)
12.0 per cent to 16.5 per cent, detelmined on 0.100 g.
Sulfated ash (2.4.14)
Maximum 1.0 per cent, detelmined on 1.0 g.
Bacterial endotoxins (2.6.14) B. (2S,3RS,5R)-3-hydroxy-5-methyl-2-[[(lr,2R,3S,4r,5R,6S)-
Less than 0.17 IU/mg, if intended for use in the manufacture 2,4,6-trihydroxy-3,5-
of parenteral preparations without a further appropriate bis (methylamino )cyclohexyl] oxy] tetrahydrofuran-3-carboxylic
procedure for the removal of bacterial endotoxins. Prepare acid (actinospectinoic acid),
the solutions using a 0.42 per cent 111/m solution of sodium
hydrogen carbonate R.
ASSAY
Liquid chromatography (2.2.29) as described in the test for
related substances with the following modifications.
Test solution Dissolve 40.0 mg of the substance to be
examined in water R and dilute to 50.0 mL with the same
solvento Allow to stand for not les s than 15 h and not more
than 72 h (formation of anomers). Dilute 5.0 mL of this C. R1 = CH3 , R2 = R4 = H, R3 = OH:
solution to 50.0 mL with the mobile phase. (2R,4S,4aS,5aR,6S,7S,SR,9S,9aR,10aS)-2-methyl-
6,S-bis (methylamino )decahydro-2H-pyrano [2,3-
Reference solution Dissolve 40.0 mg of spectinomycin
b] [1,4]benzodioxine-4,4a, 7,9-tetrol ((4S)-
hydrochloride CRS (containing (4R)-dihydrospectinomycin) in
dihydrospectinomycin) ,
water R and dilute to 50.0 mL with the same solvento Allow
to stand for the same period of time as the test solution D. R1 = CH3 , R2 = H, R3 = R4 = OH:
(formation of anomers). Dilute 5.0 mL of this solution to (2R,3R,4S,4aS,5aR,6S, 7S, SR, 9S, 9aR, 10aS)-2-methyl-6,S-
50.0 mL with the mobile phase. bis (methylamino )decahydro-2H-pyrano [2,3-
b] [1,4]benzodioxine-3,4,4a, 7,9-pentol
(dihydroxyspectinomycin) ,
Vet-98 Spirmnycin 2016

E. R1 = R4 = H, R2 + R3 = o: (spiramycin I; 843). Spiramycin II (4-0-acetylspiramycin I)


(2R,4aR, 5aR, 6S, 7R, 8R, 9S, 9aR, lOaS) -6-amino-4a, 7,9- and spiramycin III (4-0-propanoylspiramycin I) are also
trihydroxy-2-methyl-8-(methylamino) decahydro-4H- presento
pyrano [2,3-b] [1,4 ]benzodioxin-4-one Potency
(N-desmethylspectinomycin) , Mínimum 4100 IU/mg (dried substance).
G. R1 = CH3 , R2 + R3 = 0, R4 = OH: CHARACTERS
(2R,3S,4aR,5aR,6S, 7S,8R, 9S, 9aR, 1OaS)-3,4a, 7,9-
tetrahydroxy-2-methyl-6,8-bis(methylamino)decahydro-4H-
Appearance
White or slight1y yellowish powder, slight1y hygroscopic.
pyrano[2,3-b] [1,4]benzodioxin-4-one
(tetrahydroxyspectinomycin) , Solubility
Slight1y soluble in water, freely soluble in acetone, in ethanol
(96 per cent) and in methanol.
IDENTIFICATION
A. Ultraviolet and visible absorption spectrophotometry
(2.2.25).
Test solution Dissolve 0.10 g of the substance to be examined
in methanol R and dilute to 100.0 mL with the same solvento
F. (2S,4S,6R)-4-hydroxy-6-methyl-2-[[(lr,2R,3S,4r,5R,6S)- Dilute 1.0 mL ofthis solution to 100.0 mL with methanol R.
2,4,6-trihydroxy-3,5- Spectral range 220-350 nm.
bis (methylamino) cyclohexyl] oxy] dihydro-2H-pyran-3(4H)- Absorption maximum At 232 nm.
one (triol spectinomycin).
Specific absorbance at the absorption maximum About 340.
___________________________________________ ~E~

B. Thin-layer chromatography (2.2.27).


Test solution Dissolve 40 mg of the substance to be examined
in methanol R and dilute to 10 mL with the same solvento
Reference solution (a) Dissolve 40 mg of spiramycin CRS in
Spiramycin methanol R and dilute to 10 mL with the same solvento
(Ph. Eur. monograph 0293) Reference solution (b) Dissolve 40 mg of elJlthromycin A CRS
in methanol R and dilute to 10 mL with the same solvento
Plate TLC silica gel G plate R.
Mobile phase The upper layer of a mixture of 4 volumes of 2-
propanol R, 8 volumes of a 150 gIL solution of ammoniu1lZ
acetate R previously adjusted to pH 9.6 with strong sodium
OHC hydroxide solution R, and 9 volumes of ethyl acetate R.
CH 3 H-- Application 5 ~lL.

H3c~1 0
'N/ CH3 O H"
Development Over 3/4 of the plateo
Dlying In airo
HO
CH V O
3
RP,'H Detection Spray with anisaldehyde solution Rl and heat at
OH OH 110 oC for 5 mino
Results The principal spot in the chromatogram obtained with
CH 3 the test solution is similar in position, colour and size to the
principal spot in the chromatogram obtained with reference
Compound R Molee Formula Mr
solution (a). If in the chromatogram obtained with the test
Spiramycin I H C43H74N2014 843.1 solution 1 or 2 spots occur with Rp values slight1y higher
Spiramyein II CO-CH 3 C45H76N2015 885.1 than that of the principal spot, these spots are similar in
Spiramycin III CO-CH 2-CH 3 C46H7SN2015 899.1 position and colo'ur to the secondary spots in the
chromatogram obtained with reference solution (a) and differ
from the spots in the chromatogram obtained with reference
8025-81-8 solution (b).
C. Dissolve 0.5 g in 10 mL of 0.05 M sulfuric acid and add
Action and use 25 mL of water R. Adjust to about pH 8 with 0.1 M sodium
Antibacterial. hydroxide and dilute to 50 mL with water R. To 5 mL of this
~E~ ___________________________________________ solution add 2 mL of a mixture of 1 volume of 'water R and
2 volumes of sulfuric acid R. A brown colour develops.
DEFINITION TESTS
Macrolide antibiotic produced by the growth of certain pH (2.2.3)
strains of Streptomyces ambofaciens or obtained by any other 8.5 to 10.5.
means. The main component is
Dissolve 0.5 g in.s mL of 111ethanol R and dilute to 100 mL
(4R,5S,6S, 7R,9R,10R,11E,13E,16R)-6-[[3,6-dideoxy-4-0-
with carbon dioxide-free water R.
(2,6-dideoxy-3-C-methyl-ex-L-ribo-hexopyranosyl)-3-
(dimethylamino )-~-D-glucopyranosyl] oxy] -4-hydroxy-5- Specific optical rotation (2.2.7)
methoxy-9, 16-dimethyl-7 -(2-oxoethyl)-1 0-[[2,3,4,6- -80 to -85 (dried substance).
tetradeoxy-4-(dimethylamino )-D-elythro-
hexopyranosyl] oxy] oxacyclohexadeca-11, 13-dien-2-one
2016 Spirarnycin Vet-99

Dissolve 1.00 g in a 10 per cent VIV solution of dilute aeetie Li111itS:


acid R and dilute to 50.0 mL with the same acid solution. - impul'ities AJ BJ DJ EJ F J GJ H: for each impurity, not
Composition more than the are a of the principal peak in dle
Liquid chromatography (2.2.29) as described in the test for chromatogram obtained with reference solution (b)
related substances. (2.0 per cent);
- any other impurity: for each impurity, not more than the
1njection Test solution and reference solution (a).
area of the principal peak in the chromatogram obtained
Ca1culate the percentage content using the declared content with reference solution (b) (2.0 per cent);
of spiramycins I, Il and III in spiramycin CRS. - total: not more than 5 times the area of the principal peak
Conzposition of spiramycins (dried substance): in the chromatogram obtained with reference solution (b)
- spiramycin 1: minimum 80.0 per cent, (10.0 per cent);
- spiramycin 11: maximum 5.0 per cent, - disregard limit: 0.05 times the area of the principal peak in
- spiramycin 111: maximum 10.0 per cent, the chromatogram obtained with reference solution (b)
- sum of spiramycins 1J 11 and 111: minimum 90.0 per cent. (0.1 per cent); disregard any peak due to the blank and
Related substances the peaks due to spiramycins I, Il and IlI.
Liquid chromatography (2.2.29). Prepare the solutions Heavy metals (2.4.8)
i11l71lediately befare use. Maximum 20 ppm.
Solvent mixture 11lethanol R, 'Water R (30:70 VIV). 1.0 g complies with test F. Prepare the reference solution
Test solution Dissolve 25.0 mg of the substance to be using 2 mL of lead standard solution (lO ppm Pb) R.
examined in the solvent mixture and dilute to 25.0 mL with Loss on drying (2.2.32)
the solvent mixture. Maximum 3.5 per cent, determined on 0.500 g by drying at
Referenee solution (a) Dissolve 25.0 mg of spiramycin CRS in 80 oC over diphosphorus pentoxide R at a pressure not
the solvent mixture and dilute to 25.0 mL with the solvent exceeding 0.67 kPa for 6 h.
mixture. Sulfated ash (2.4.14)
Referenee solution (b) Dilute 2.0 mL of reference solution (a) Maximum 0.1 per cent, determined on 1.0 g.
to 100.0 mL with the solvent mixture.
ASSAY
Referenee solution (e) Dissolve 5 mg of spiramycin CRS in Carry out the microbiological assay of antibiotics (2.7.2).
15 mL of buffer solution pH 2.2 R and dilute to 25 mL with·
'Water R, then heat in a water-bath at 60 oC for 5 min and STORAGE
cool under cold water. In an airtight container.
Blank solution The solvent mixture. IMPURlTIES
Colunm: Speeified impurities A, B, D, E, F, G, H
- size: 1 = 0.25 m, 0 = 4.6 mm; Other detectable impurities (the following substances would, if
- statiol1my phase: end-eapped polar-embedded octadeeylsilyl present at a sufficient level, be detected by one or other of
a71l0lphous organosiHca polymer R (5 ~l1n) (polar-embedded the tests in the monograph. They are limited by the general
octadecylsilyl methylsilica gel), with a pore size of acceptance criterion for other/unspecified impurities and/or
12.5 nm and a carbon loading of 15 per cent; by the general monograph Substances for pharmaceutical use
- temperature: 70 oc. (2034). It is therefore not necessary to identify these
Mobile phase Mix 5 volumes of a 34.8 gIL solution of impurities for demonstration of compliance. See also 5.10.
dipotassium hydrogen phosphate R adjusted to pH 6.5 with a Control of impurities in substanees for pharmaeeutieal use): C.
27.2 giL solution of potassium dihydrogen phosphate R,
40 volumes of aeetonitrile R and 55 volumes of 'Water R.
Flo'W rate 1.0 mUmin.
Detection Spectrophotometer at 232 nm.
H3 C
1njeetion 20 ~lL of the blank solution, the test solution and H--
reference solutions (b) and (c). R3
Run time 3 times the retention time of spiramycin 1. CH 3 H--

1dentifieation of spiramycins Use the chromatogram supplied ~O O,


with spiramycin CRS and the chromatogram obtained with H3~~N/CH3 ) ~~ ,

reference solution (a) to identify the peaks due to spiramycins R2~ R1 H'
I, Il and IIl. OH
Relative retention With reference to spiramycin I (retention
time = 20 min to 30 min): impurity F = about 0.41; A. R1 = H, R2 = OH, R3 = CHz-CHO:
impurity A = about 0.45; impurity D = about 0.50; (4R,5S,6S, 7R,9R,10R,11E,13E,16R)-6-[[3,6-dideoxy-3-
impurity G = about 0.66; impurity B = about 0.73; (dimethylamino )-~-D-glucopyranosyl] oxy] -4-hydroxy-5-
impurity H = about 0.87; spiramycin Il = about 1.4; methoxy-9, 16-dimethyl-7 -(2-oxoethyl)-1 0-[[2,3,4,6-
spiramycin III = about 2.0; impurity E = about 2.5. tetradeoxy-4- (dimethylamino)- ~-D-erythro­
hexopyranosyl] oxy] oxacyclohexadeca-11, 13-dien-2-one
If necessary adjust the composition of the mobile phase by
(neospiramycin I),
changing the amount of acetonitrile.
System suitability: reference solution (c): B. R1 = H, R2 = osyl, R3 = CHz-CH20H:
- resolution: minimum 10.0 between the peaks due to (4R,5S,6S, 7R,9R,10R,11E,13E,16R)-6-[[3,6-dideoxy-4-0-
impurity A and spiramycin 1. (2, 6-dideoxy-3-C-methyl-cx-L-ribo-hexopyranosyl)-3-
(dimethylamino )-~-D-glucopyranosyl] oxy] -4-hydroxy-7-
(2-hydroxyethyl)-5-methoxy-9, 16-dimethyl-l 0- [[2,3,4,6-
V et-100 Sulfadimidine 2016

tetradeoxy-4- (dimethylamino)- ~-D-elythro­


hexopyranosyl] oxy] oxacyclohexadeca-11, 13-dien-2-one
(spirarnycin IV),
C. R1 = H, R2 = osyl, R3 = C(=CH2 )-CHO:
(4R,5S,6S, 7S,9R,10R,11E,13E,16R)-6-[[3,6-dideoxy-4-0-
(2, 6-dideoxy-3-C-methyl-a-L-ribo-hexopyranosyl) -3-
(dimethylamino )-~-D-glucopyranosyl] oxy]-7-
(1-forrnylethenyl)-4-hydroxy-5-methoxy-9, 16-dimethyl-1 0-
[[2,3,4,6-tetradeoxy-4-(dimethylamino)-~-D-elythro­
hexopyranosyl] oxy] oxacyclohexadeca -11, 13-dien-2-one
(17-methylenespirarnycin I),
E. R1 = H, R2 = osyl, R3 = CHT CH3 :
(4R,5S,6S, 7S,9R,1 OR,11E,13E,16R)-6-[[3,6-dideoxy-4-0-
(2,6-dideoxy-3-C-methyl-a-L-ribo-hexopyranosyl)-3-
(dimethylamino )-~-D-glucopyranosyl] oxy] -7 -ethyl-4-hydroxy-
5-methoxy-9, 16-dimethyl-1 0- [[2,3,4, 6-tetTadeoxy-4-
(dimethylamino )-~-D-elythro-
hexopyranosyl] oxy] oxacyclohexadeca-11, 13-dien-2-one
(18-deoxy-18-dihydrospiramycin I 01' DSPM),
G. R1 = CO-CH3 , R2 = OH, R3 = CHrCHO:
(4R,5S,6S, 7R,9R, 1OR, 11E, 13E, 16R)-6-[[3,6-dideoxy-3-
(dimethylamino )-~-D-glucopyranosyl] oxy]-5-methoxy-9, 16-
dimethyl-2-oxo-7 -(2-oxoethyl)-1 0- [[2,3,4,6-tetradeoxy-4- F. spiramycin dimer.
(dimethylamino )-~-D-elythro- __________________________________________ ~Ew

hexopyranosyl] oxy] oxacyclohexadeca-11, 13-dien-4-yl acetate


(neospiramycin II),
H. R1 = CO-C 2 H s, R2 = OH, R3 = CHTCHO:
(4R,5S,6S, 7R,9R, 1OR, 11E,13E,16R)-6-[[3,6-dideoxy-3-
(dimethylamino )-~-D-glucopyranosyl] oxy] -5-methoxy-9, 16- Sulfadimidine *****
dimethyl-2-oxo-7 -(2-oxoethyl)-1 0-[[2,3 ,4,6-tetradeoxy-4- ** **
(dimethylamino )-~-D-elythro- (Ph. Eur. l1Zonogmph 0295) ***
hexopyranosyl] oxy] oxacyclohexadeca-11, 13-dien-4-yl
propanoatate (neospiramycin III),

OHC
". oZt~:,
H H
278.3 57-68-1
CH 3 H--
Action and use
Sulfonamide antibacterial.
Preparation
o CH 3
Sulfadimidine Injection
CH 3 OH
OH
~Ew __________________________________________
CH 3 DEFINITION
4-Amino-N- (4, 6-dimethylpyrimidin-2-yl) benzenesulfonamide.
D. (4R,5S, 6S, 7R, 9R, 1OR, 11E, 13E, 16R)-6- [[3 ,6-dideoxy-4- Content
0- (2, 6-dideoxy-3-C-methyl-a-L-ribo-hexopyranosyl) -3- 99.0 per cent to 101.0 per cent (dried substance).
(dimethylamino )-~-D-glucopyranosyl] oxy] -1 0- [(2,6-dideoxy-
3-C-methyl-a-L-ribo-hexopyranosyl) oxy] -4-hydroxy-5- CHARACTERS
methoxy-9, 16-dimethyl-7 -(2-oxoethyl)oxacyclohexadeca- Appearance
11,13-dien-2-one (spiramycin V), White 01' almost white powder 01' crystals.
Solubility
Very slightly soluble in water, soluble in acetone, slightly
soluble in etllanol (96 per cent). It dissolves in solutions of
alkali hydroxides and in dilute mineral acids.
IDENTIFICATION
First identijication A.
Second identijication B, C, D.
A. Infrared absorption spectrophotometry (2.2.24).
Comparison sulfadimidine CRS.
B. Thin-Iayer chromatography (2.2.27).
2016 Sulfadimidine Vet-10 1

Solve/u mixture eoneentrated ammonia R, methanol R - stationalY phase: end-eapped oetylsilyl siliea gel fol'
(4:96 V/V). ehromatography R (5 ~lm);
Test solution Dissolve 20 mg of the substance to be examined - temperature: 35 oc.
in 3 mL of the solvent mixture and dilute to 5.0 mL with the lvlobile phase:
solvent mixture. - mobile phase A: mix 10 volumes of aeetonitrile R and
Referenee solution Dissolve 20 mg of sulfadimidine CRS in 90 volumes of a 0.6 per cent V/V solution of aeetie aeid R
3 mL of the solvent mixture and dilute to 5.0 mL with the previously adjusted to pH 6.5 with a 250 giL solution of
solvent mixture. ammonia R;
- mobile phase B: mix equal volumes of aeetonitrile R and a
Plate TLC siliea gel GF254 plate R.
0.6 per cent V/V solution of aeetie acid R previously
lvlobile phase dilute ammonia Rl, water R, nitromethane R, adjusted to pH 6.5 with a 250 gIL solution of ammonia R;
dioxan R (3:5:40:50 V/V/V/Tl).
Applieation 5 ~lL.
Time Mobile phase A Mobile phase B
Development Over 2/3 of the plateo (mio) (per ceot V/V) (per ceot V/V)
Dlyúzg At 100-105 oC for 30 mino 0-25 100 O
Detection Examine in ultraviolet light at 254 nm. 25 - 35 100.¿ O O.¿ 100
Results The principal spot in the chromatogram obtained with
35 - 45 O 100
the test solution is similar in position and size to the principal
spot in the chromatogram obtained with the reference
solution. Flow rate 1.3 mUmin.
C. Place 3 g in a dry tube. Immerse the lower part of the Detection Spectrophotometer at 241 nm.
tube, inclined at 45°, in a silicone-oil bath and heat to about Jnjeetion 20 ~lL.
270 oc. The substance to be examined decomposes and a
Jdentifieation of impurities U se the chromatogram supplied
white or yellowish-white sublimate is formed which, after
recrystallisation from toluene R and drying at 100 oC, melts with sulfadimidine for peak identification CRS and the
(2.2.14) at 150 oC to 154 oc. chromatogram obtained with reference solution (c) to identify
the peak due to impurity G.
D. Dissolve about 5 mg in 10 mL of a 103 giL solution of
Relative retention Widl reference to sulfadimidine (retention
hydroehlorie acid R. Dilute 1 mL of the solution to 10 mL
time = about 20 min): impurity E = about 0.13;
with water R. The solution, without further acidification,
impurity C = about 0.15; impurity D = about 0.2;
gives the reaction of primary aromatic amines (2.3.1).
impurity G = about 1.7.
TESTS System suitabilit:y: reference solution (a):
Appearance of solution - resolution: minimum 2.0 between the peaks due to
The solution is not more intensely coloured than reference impurities E and C.
solution Y5, BY5 or GY5 (2.2.2) lvlethod JI). Limits:
Dissolve 0.5 g in a mixture of 5 mL of dilute sodium hydroxide - impurities C) D) G: for each impurity, not more than the
solution R and 5 mL of water R. area of the principal peak in the chromatogram obtained
Acidity with reference solution (b) (0.10 per cent);
To 1.25 g of the finely powdered substance to be examined, unspeeified impurities: for each impurity, not more than
add 25 mL of earbon dioxide-free water R. Heat at about 0.5 times the area of the principal peak in the
70 c C for 5 mino Cool in iced water for about 15 min and chromatogram obtained with reference solution (b)
filter. To 20 mL of the filtrate add 0.1 mL of bromothymol (0.05 per cent);
blue solution Rl. Not more than 0.2 mL of 0.1 M sodiu11l total: not more than 5 times dle area of the principal peak
hydroxide is required to change the colour of the indicator. in the chromatogram obtained with reference solution (b)
(0.5 per cent);
Related substances
- disregard limit: 0.3 times rile area of the principal peak in
Liquid chromatography (2.2.29).
the chromatogram obtained with reference solution (b)
Solvent mixture 40 gIL solution of sodium hydroxide R,
(0.03 per cent).
aeetonitrile R, water R (2.5:25:75 V/V/T!).
Heavy metals (2.4.8)
Test solution Dissolve 50.0 mg of dle substance to be
Maximum 20 ppm.
examined in 41 mL of dle solvent mixture and dilute to
50.0 mL with water R. 1.0 g complies with test D. Prepare the reference solution
using 2 mL of lead standard solution (lO ppm Pb) R.
Referenee solution (a) Dissolve 5 mg of sulfaeetamide
sodium CRS (impurity E) and 5 mg of sulfaguanidine CRS Loss on drying (2.2.32)
(impurity C) in 41 mL of the solvent mixture and dilute to Maximum 0.5 per cent, detennined on 1.000 g by drying in
100.0 mL with water R. an oven at 105 oc.
Referenee solution (b) Dilute 1.0 mL of the test solution to Sulfated ash (2.4.14)
100.0 mL with mobile phase B. Dilute 1.0 mL of this Maximum 0.1 per cent, detennined on 1.0 g.
solution to 10.0 mL with mobile phase B. ASSAY
Referenee solution (e) Dissolve 20 mg of sulfadimidine for peak Dissolve 0.250 g in a mixture of 20 mL of dilute hydroehlone
identifieation CRS (containing impurity G) in 16.4 mL ofthe acid R and 50 mL of water R. Cool the solution in iced
solvent mixture and dilute to 20.0 mL with water R. water. Carry out the detennination of primary aromatic
Column: amino- nitro gen (2.5.8), detennining the end-point
- size: 1 = 0.25 m, 0 = 4.6 mm; electrometrically.
Vet-102 Sulfadimethoxine Sodium 2016

1 mL of 0.1 M sodium nitrite is equivalent to 27.83 mg of


C12H14N402S.
STORAGE
Protected from light.
IMPURlTIES
Specified impw'ities C, D, G
Other detectable i11lpurities (the following substances would, if G. 4-amino-2-chloro-N-(4,6-dimethylpyrimidin-2-
present at a sufficient level, be detected by one or other of yl) benzenesulfonamide or 4-amino-3-chloro-N-(4,6-
the tests in the monograph. They are limited by the general dimethylpyrirnidin-2-yl)benzenesulfonamide.
___________________________________________
acceptance criterion for other/unspecified impurities and/or ~E~

by the general monograph Substances for pharmaceutical use


(2034). It is therefore not necessary to identify these
impurities for demonstration of compliance. See also 5.10.
Control of impurities in substances for pha17naceutical use): A, B,
E,F. ***
Sulfadimethoxine Sodium
*** ***
(Sulfadimethoxine Sodium for VeterinalY Use, ***
Ph. Eur. 1110nograph 2745)

A. 4-amino-N-(4-methylpyrimidin-2-yl)benzenesulfonamide
(sulfamerazine),

1037-50-9

Action and use


Sulfonamide antibacterial.
~E~ ___________________________________________
B. 4-amino-N-pyrimidin-2-ylbenzenesulfonamide
(sulfadiazine), DEFINITION
Sodium [(4-arninophenyl) sulfo nyl] (2,6-dimethoxypyrimidin-
4-yl)azanide.
Content
97.5 per cent to 102.0 per cent (anhydrous substance).
CHARACTERS
Appearance
c. (4-aminophenylsulfonyl)guanidine (sulfaguanidine), White or almost white, crystalline powder, hygroscopic.
Solubility
Freely soluble in water, slighdy soluble in ethanol
(96 per cent), practically insoluble in rnethylene chloride.
IDENTIFICATION
A. Infrared absorption spectrophotornetry (2.2.24).
Preparation Dissolve 1 g in 20 mL of water R; add 0.20 rnL
D. 4-aminobenzenesulfonamide (sulfanilamide),
of glacial acetic acid R; filter the precipitate, wash wid1 1 mL
of water R and dry at 105 oC for 2 h.
Comparison sulfadimethoxine CRS.
B. 2 mL of the filtra te obtained in identification test A gíves
reaction (a) of sodiurn (2.3.1).
TESTS
E. N- [( 4-aminophenyl) sulfonyl] acetamide (sulfacetamide), Appearance of solution
The solution is clear (2.2.1) and not more intensely coloured
than reference solution BYs (2.2.2, Method JI).
Dissolve 0.5 g in water R and dilute to 10 rnL with the sarne
solvento
pH (2.2.3)
F. 4-aminobenzenesulfonic acid (sulfanilic acid), 8.5 to 10.0.
Dissolve 0.2 g in carbon dioxide-free water R and dilute to
20 mL with the sarne solvento
2016 Sulfadimethoxine Sodium Vet-103

Re1ated substances Heavy metals (2.4.8)


Liquid chromatography (2.2.29). Maximum 20 ppm.
Solution A Dissolve 6.0 g of sodium dihydrogen phosphate R in Solvent 'Water R.
950 mL of 'Water R, adjust to pH 7.0 with dilute sodium 0.5 g complies with test H. Prepare the reference solution
hydroxide solution R and dilute to 1 L with 'Water R. using 1 mL of lead standard solution (lO ppm Pb) R.
Test solution Dissolve 22.0 mg of the substance to be Water (2.5.12)
examined in 76 mL of solution A and dilute to 100.0 mL : maximum 5.0 per cent, determined on 0.200 g.
with methanol R.
ASSAY
Reference solution (a) Dissolve 20.0 mg of
Liquid chromatography (2.2.29) as described in the test for
sulfadimethoxine CRS in 25 mL of methanol R and dilute to
100.0 mL with solution A. related substances with the following modification.
Injection Test solution and reference solution (a).
Reference solution (b) Dilute 2.0 mL of the test solution to
100.0 mL with mobile phase A. Dilute 1.0 mL of this Calculate the percentage content of C12H13N4Na04S taking
solution to 10.0 mL with mobile phase A. into account the assigned content of sulfadimethoxine CRS
Reference solutiol1 (c) Dissolve 4 mg of sulfadimethoxine for and a conversion factor of 1.071.
peak identification CRS (containing impurities A and F) in STORAGE
5 mL of methanol R and dilute to 20 mL with solution A. In an airtight container, protected from light.
Column: IMPURITIES
- size: 1 = 0.25 m, 0 = 4.6 mm; Specified impurities A) F
- statiOl1a1Y phase: end-capped octadecylsilyl a1110rphous
organosilica polymer R (5 ~lm); Other detectable i111punties (the following substances would, if
- temperature: 25 oC. present at a sufficient level, be detected by one or other of
the tests in the monograph. They are limited by the general
Mobile phase:
acceptance criterion for other/unspecified impurities and/or
- mobile phase A: methal101 R, solution A (25:75 VlV);
by the general monograph Substances for pharmaceutical use
- mobile phase B: methanol R, acetonitrile R, solution A
(2034). It is therefore not necessary to identify these
(25:35:40 V/V/V);
impurities for demonstration of compliance. See also 5.10.
Control of impurities in substances for pharmaceutical use): B) C)
Time Mobile phase A Mobile phase B D) E.
(min) (per cent V/V) (per cent V/V)
0- 10 100 O

10 - 30 100~0 O ~ 100

30 - 35 O 100

Flo'W rate 1.0 mUmin. A. 2,6-dimethoxypyrimidin-4-amine,


Detection Spectrophotometer at 254 nm.
Il1Jection 1O ~lL of the test solution and reference solutions (b)
and (c).
Identification of impunties Use the chromatogram supplied
with sulfadimethoxine for peak identification CRS and the
chromatogram obtained with reference solution (c) to identify
the peaks due to impurities A and F.
Relative retention With reference to sulfadimethoxine B. N-[4-[(2,6-dimethoxypyrimidin-4-
(retention time = about 11 min): impurity F = about 0.4; yl) sulfamoyl] phenyl] acetamide,
impurity A = about 1.2.
System suitability:
- resolution: minimum 2.5 between the peaks due to
sulfadimethoxine and impurity A in the chromatogram
obtained with reference solution (c);
- signal-to-noise ratio: minimum 40 for the principal peak in
the chromatogram obtained with reference solution (b). C. 4-( acetylamino) benzene-1-sulfonic acid,
Calculation of percentage contents:
- CO't7'ection factors: multiply the peak areas of the following
impurities by the corresponding correction factor:
impurity A = 1.4; impurity F = 1.7;
- for each impurity, use the concentration of
sulfadimethoxine in reference solution (b). D. 4-aminobenzene-1-sulfonic acid (sulfanilic acid),
Limits:
- impurities A) F: for each impurity, maximum 0.2 per cent;
- unspecified impurities: for each impurity, maximum
0.20 per cent;
- total: maximum 0.5 per cent;
- reporting threshold: 0.10 per cent.
E. 4-aminobenzene-1-sulfonamide (sulfanilamide),
Vet-104 Sulfamerazine 2016

TESTS
Appearance of solution
Dissolve 0.8 g in a mixture of 5 mL of dilute sodium hydroxide
solution R and 5 mL of water R. The solution is not more
intensely coloured than reference solution Y4, BY4 01' GY4
(2.2.2, Method JI).
F. 4-amino-N-(2-hydroxy-6-methoxypyrimidin-4-yl) benzene- Acidity
l-sulfonamide. To 1.25 g, finely powdered, add 40 mL of cal'bon dioxide-free
___________________________________________ PhE~
water R and heat at about 70 oC for 5 mino Cool for about
15 min in iced water and filter. To 20 mL of the filtrate add
0.1 mL of bromothymol blue solution Rl. Not more than
0.2 mL of 0.1 M sodium hydroxide is required to change the
colour of the indicator.
Sulfamerazine Related substances
Examine by thin-Iayer chromatography (2.2.27) using silica
(Ph. Eur. monograph 0358) gel GF254 R as the coating substance.
Test solution (a) Dissolve 0.10 g of the substance to be
examined in 3 mL of a mixture of 2 volumes of concentrated
ammonia R and 48 volumes of methanol R and dilute to 5 mL
with the same mixture of solvents.
Test solution (b) Dilute 1 mL of test solution (a) to 10 mL
with a mixture of 2 volumes of concentrated al1ul1onia R and
48 volumes of methanol R.
264.3 127-79-7 Reference solution (a) Dissolve 10 mg of sulfamerazine CRS in
3 mL of a mixture of 2 volumes of concentrated ammonia R
Action and use and 48 volumes of methanol R and dilute to 5 mL with the
Sulfonamide antibacterial. same mixture of solvents.
PhE~ ___________________________________________
Reference solution (b) Dilute 2.5 mL of test solution (b) to
50 mL with a mixture of 2 volumes of concentrated
DEFINITION ammonia R and 48 volumes of methanol R.
Sulfamerazine contains not less than 99.0 per cent and not Apply to the plate 5 ~lL of each solution. Develop over a
more than the equivalent of 101.0 per cent of 4-amino-N- path of 15 cm with a mixture of 3 volumes of dilute
(4-methyl-2-pyrimidinyl) benzenesulfonamide, calculated with am1110nia Rl, 5 volumes of water R, 40 volumes of
reference to the dried substance. nitromethane R and 50 volumes of dioxan R. Dry the plate at
CHARACTERS 100 oC to 105 oC and examine in ultraviolet light at 254 nm.
White, yellowish-white or pinkish-white, crystalline powder or Any spot in the chromatogram obtained with test
crystals, very slightly soluble in water, sparingly soluble in solution (a), apart from the principal spot, is not more
acetone, slightly soluble in alcohol, very slightly soluble in intense that the spot in the chromatogram obtained with
methylene chloride. It dissolves in solutions of alkali reference solution (b) (0.5 per cent).
hydroxides and in dilute mineral acids. Heavy metals (2.4.8)
It melts at about 235 oC, with decomposition. 1.0 g complies with test C for heavy metals (20 ppm).
Prepare the reference solution using 2 mL of lead standard
IDENTIFICATION solution (lO ppm Pb) R.
First identification A, B.
Loss on drying (2.2.32)
Second identification B, C, D. Not more than 0.5 per cent, determined on 1.000 g by
A. Examine by infrared absorption spectrophotometry drying in an oven at 105 oC.
(2.2.24), comparing with the spectrum obtained with
Sulfated ash (2.4.14)
sulfamerazine CRS. Examine the substances as discs.
Not more than 0.1 per cent, determined on 1.0 g.
B. Examine the chromatograms obtained in the test for
related substances. The principal spot in the chromatogram ASSAY
obtained with test solution (b) is similar in position, colour Dissolve 0.2500 g in a mixture of 20 mL of dilute hydrochlonc
and size to the principal spot in the chromatogram obtained acid R and 50 mL of water R. Cool the solution in iced
with reference solution (a). water. Carry out the determination of primary aromatic
amino- nitro gen (2.5.8), determining the end-point
C. Place 3 g in a dry tube. Incline the tube by about 45°,
electrometrically.
immerse the bottom of the tube in a silicone-oil bath and
heat to about 270 oc. The substance decomposes, producing 1 mL of 0.1 M sodium nitl1te is equivalent to 26.43 mg of
a white 01' yellowish-white sublimate which, after CllH12N402S,
recrystallisation from toluene R and drying at 100 oC, melts STORAGE
(2.2.14) at 157 oC to 161°C. Store protected from light.
D. Dissolve about 20 mg in 0.5 mL of dilute hydrochloric ___________________________________________ PhE~

acid R and add 1 mL of water R. The solution gives, without


further addition of acid, the identification reaction of primary
aromatic amines (2.3.1).
2016 Sulfanilamide Vet-1 05

*****
Acidity
Su Ifametoxypyridazine
** ** To 1.25 g, finely powdered, add 25 mL of cm'bon dioxide-free
(Sulfamethoxypyn'dazine for Veten'nary Use) *** water R. Heat at 70 oC for 5 min. Cool in iced water for
Ph Bur monograph 0638) about 15 min and filter. To 20 mL of the filtrate add 0.1 rnL
of br01110thY11101 blue solution R1. Not more than 0.5 mL of
0.1 M sodiu111 hydroxide is required to charige the colour of
the indicator.
Related substances
Examine by thin layer chromatography (2.2.27), using
TLC silica gel GF254 plate R.
Test solution (a) Dissolve 0.10 g of the substance to be
280.3 80-35-3 examined in acetone R and dilute to 5 mL with the same
solvento
Action and use
Test solution (b) Dilute 1 mL of test solution (a) to 10 mL
Sulfonamide antibacterial.
with acetone R.
___________________________________________
PhE~
Reference solution (a) Dissolve 20 mg of
DEFINITION sulfa111ethoxypyridazine CRS in acetone R and dilute to 10 rnL
with the same solvento
Sulfamethoxypyridazine for veterinary use contains not les s
than 99.0 per cent and not more than the equivalent of Reference solution (b) Dilute 2.5 mL of test solution (b) to
101.0 per cent of 4-amino-N-(6-methoxypyridazin-3- 5 O rnL with acetone R.
yl)benzenesulfonamide, calculated with reference to the dried Apply separately to the plate 5 ~lL of each solution. Develop
substance. over a path of 15 cm using a mixture of 1 volume of dilute
a1111110nia R1, 9 volumes of water R, 30 volumes of 2-
CHARACTERS
propanol R and 50 volumes of ethyl acetate R. Dry the plate at
A white or slightly yellowish, crystalline powder, colouring
100-105 oC and examine in ultraviolet light at 254 nm.
slowly on exposure to light, practically insoluble in water,
Any spot in the chromatogram obtained with test
sparingly soluble in acetone, slightly soluble in alcohol, very
solution (a), apart from the principal spot, is not more
slightly soluble in methylene chloride. It dissolves in solutions
intense than the spot in the chromatogram obtained with
of alkali hydroxides and in dilute mineral acids.
reference solution (b) (0.5 per cent).
It melts at about 180 oC, with decomposition.
Heavy metals (2.4.8)
IDENTIFICATION 1.0 g complies with test D for heavy metals (20 ppm).
First identification A) B Prepare the reference solution using 2 mL of lead standard
Second identification B) C) D solution (lO ppm Pb) R.
A. Examine by infrared absorption spectrophotometry Loss on drying (2.2.32)
(2.2.24), comparing with the spectrum obtained with Not more than 0.5 per cent, determined on 1.000 g by
sulfamethoxypyridazine CRS. Examine the substances prepared drying in an oven at 105 oC.
as discs. Sulfated ash (2.4.14)
B. Examine the chromatograms obtained in the test for Not more than 0.1 per cent, determined on 1.0 g.
related substances. The principal spot in the chromatogram
ASSAY
obtained with test solution (b) is similar in position and size
Carry out the assay of primary aromatic amino-nitrogen
to the principal spot in the chromatogram obtained with
(2.5.8), using 0.2500 g, determining the end-point
reference solution (a).
electrometrically.
e. Dissolve 0.5 g in 1 mL of a 40 per cent V/V solution of 1 mL of 0.1 M sodiwn nitrite is equivalent to 28.03 mg of
sulfuric acid R, heating gently. Continue heating until a
C ll H 12N 4 0 3 S.
crystalline precipitate appears (about 2 min). Cool and add
10 mL of dilute sodium hydroxide solution R. Cool again, add STORAGE
25 mL of ether R and shake the solution for 5 min. Separate Protected from light.
the ether layer, dry over anhydrous sodium sulfate R and filter. ___________________________________________ PhE~

Evaporate the ether by heating in a water-bath. An oily


residue is obtained which becomes crystalline on cooling;
if necessary, scratch the wall of the container with a glass
rod. The residlle melts (2.2.14) at 102 oC to 106 0e.
Sulfanilamide ***
D. Dissolve about 5 mg in 10 mL of 1 M hydrochloric acid.
Dilute 1 mL of the solution to 10 mL with water R.
*** ***
The solution, without further acidification, gives the reaction
(Ph. Bur. 1110nograph 1571) ***
of primary aromatic amines (2.3.1).
TESTS
Appearance of solution
Dissolve 1.0 g in a mixture of 10 mL of 1 M sodium
hydroxide and 15 mL of water R. The solution is clear (2.2.1)
and not more intensely coloured than reference solution Y 4 172.2 63-74-1
or BY4 (2.2.2) Method JI).
Action and use
Sulfonamide antibacterial.
Vet-106 Sulfaquinoxaline 2016

PhE~ ___________________________________________ mixture of 3 volumes of dilute a1111110nia Rl, 5 volumes of


DEFINITION water R, 40 volumes of nitromethane R and 50 volumes of
dioxan R. Dry the plate at 100 oC to 105 oC and examine in
Sulfanilamide contains not less than 99.0 per cent and not
ultraviolet light at 254 nm. Any spot in the chromatogram
more than the equivalent of 101.0 per cent of
obtained with test solution (b), apart from the principal spot,
4-aminobenzenesulfonamide, calculated with reference to the
is not more intense than the spot in the chromatogram
dried substance.
obtained with reference solution (b) (0.5 per cent). The test
CHARACTERS is not valid unless the chromatogram obtained with reference
White 01' yellowish-white crystals 01' fine powder, slightly solution (c) shows two clearly separated principal spots.
soluble in water, freely soluble in acetone, sparingly soluble Heavy metals (2.4.8)
in alcohol, practically insoluble in methylene chloride. 12 mL of solution S complies with test A for heavy metals
It dissolves in solutions of alkali hydroxides and in dilute (20 ppm). Prepare the reference solution using lead standard
mineral acids. solution (1 ppm Pb) R.
IDENTIFICATION Loss on drying (2.2.32)
First identification B Not more than 0.5 per cent, determined on 1.000 g by
Second identification A, C, D drying in an oven at 105 oc.
A. Melting point (2.2.14): 164.5 oC to 166.0 oc. Sulfated ash (2.4.14)
B. Examine by infrared absorption spectrophotometry Not more than 0.1 per cent, determined on 1.0 g.
(2.2.24), comparing with the spectmm obtained with ASSAY
sulfanilamide CRS. Examine the substances prepared as discs. Carry out the determination of primary aromatic amino-
C. Examine the chromatograms obtained in the test for nitrogen (2.5.8), using 0.140 g and determining the
related substances. The principal spot in the chromatogram end-point electrometrically.
obtained with test solution (a) is similar in position and sÍZe 1 mL of 0.1 M sodium nitrite is equivalent to 17.22 mg of
to the principal spot in the chromatogram obtained with C 6 H sN 2 0 2 S.
reference solution (a).
STORAGE
D. Dissolve about 5 mg in 10 mL of 1 M hydroehlorie acid.
Dilute 1 mL of the solution to 10 mL with water R. Store protected from light.
___________________________________________ ~E~

The solution, without further acidification, gives the reaction


of primary aromatic amines (2.3.1).
TESTS
Solution S
To 2.5 g add 50 mL of eal'bon dioxide-free water R. Heat at Sulfaquinoxaline
about 70 c C for about 5 mino Cool in iced water for about
15 min and filter.
Acidity
To 20 mL of solution S add 0.1 mL of bromothymol blue
solution Rl. Not more than 0.2 mL of 0.1 NI sodium hydroxide
is required to change the colour of the indicator.
Related substances
Examine by thin-layer chromatography (2.2.27), using a
TLC silica gel F 254 plate R. 300.3 59-40-5
Test solution (a) Dissolve 20 mg of the substance to be
Action and use
examined in 3 mL of a mixture of 2 volumes of eoneentrated
Sulfonamide antibacterial.
ammonia R and 48 volumes of methanol R and dilute to 5 mL
with the same mixture of solvents. DEFINITION
Test solution (b) Dissolve 0.10 g of the substance to be Sulfaquinoxaline is N 1-quinoxalin-2-ylsulphanilamide.
examined in 0.5 mL of eoncentrated ammonia R and dilute to It contains not les s than 98.0% and not more than 101.0%
5 mL with methanol R. If the solution is not clear, heat gently of C14H12N402S, calculated with reference to the dried
until dissolution is complete. substance.
Referenee solution (a) Dissolve 20 mg of sulfanilamide CRS in CHARACTERISTICS
3 mL of a mixture of 2 volumes of eoncentrated a1111110nia R
A yellow powder.
and 48 volumes of 111ethanol R and dilute to 5 mL with the
same mixture of solvents. Practically insoluble in water; very slightly soluble in ethanol
(96%); practically insoluble in ether. It dissolves in aqueous
Reference solution (b) Dilute 1.25 mL of test solution (a) to
solutions of alkalis.
50 mL with a mixture of 2 volumes of coneentrated
ammonia R and 48 volumes of methanol R. IDENTIFICATION
Referenee solution (e) Dissolve 20 mg of the substance to be A. The infrared absorption spectnl111, Appendix II A, is
examined and 20 mg of sulfamerazine CRS in 3 mL of a concordant with the referenee speetru11l of sulfaquinoxaline
mixture of 2 volumes of conee1Ztrated a71ZJ11onia R and (RSV 41).
48 volumes of methanol R and dilute to 5 mL with the same B. The light abs01ption, Appendix II B, in the range 230 to
mixture of solvents. 350 nm of a 0.001 % w/v solution in O.OIM sodium hydroxide
Apply to the plate 5 ~lL of each solution. Develop over a exhibits a maximum only at 252 run. The abs01'banee at
path corresponding to two-thirds of the plate height using a 252 nm is about 1.1.
2016 Sulfathiazole Sodium Vet-107

C. Yields the reaction characteristic of primaJy aromatic


amines, Appendix VI, dissolving 4 mg in 2 mI of warm
Sulfathiazole Sodium
2M hydrochlm'ic acid. An orange-red precipitate is produced.
TESTS
Acidity
To 2 g add 100 mI of water, heat at 70° for 5 minutes, cool
to 20° and filter. 50 mI of the filtrate reqUires for titration to
pH 7.0 not more than 0.2 mI of O.lM sodium hydroxide VS,
Appendix V L.
CgHsN3Na02S2,11/zH20 304.3 144-74-1 (anhydrous)
Heavy meta1s
Dissolve the residue obtained in the test for Sulphated ash in CgHsN3Na02S2,5H20 367.4 6791-71-5
1 mI of 2M hydrochloric acid and dilute to 14 mI with water.
Action and use
12 mI of the solution complies with limit test A for heavy
Sulfonarnide antibacterial.
metals, Appendix VII. Use lead standard solution (2 ppm Pb) to
prepare the standard (20 ppm). DEFINITION
Related substances Sulfathiazole Sodium is the hydrated sodium salt of N I _
Can-y out the method for thin-layer chro11latography, thiazol-2-yl sulfanilarnide, either the sesquihydrate or the
Appendix III A, using the following solutions. pentahydrate. It contains not less than 99.0% and not more
(1) Dissolve 0.40 g of the substance being examined in 4 mI than 101.0% of CgHsN3Na02S2' ca1culated with reference to
of 1M sodium hydroxide and add sufficient methanol to produce the dried substance.
100 mI. CHARACTERISTICS
(2) 0.012% w/v ofNI,N2 -diquinoxalin-2- A white or yellowish white, crystalline powder or granules.
ylsulphanilamide BPCRS in methanol. Freely soluble in water; soluble in ethanol (96%).
(3) 0.0040% w/v of sulphanilamide in methanol.
IDENTIFICATION
CHROMATOGRAPHIC CONDITIONS A. The infrared absmption spectnt111 of the dried substance,
(a) Use a silica gel F 254 precoated plate (Mere k silica gel 60 Appendix II A, is concordant with the reference spectrum of
F 254 plates are suitable). sulfathiazole sodium (RSV 42).
(b) Use the mobile phase as described below. B. Dissolve 1 g in 25 mL of water and add 2 mL of 6M acetic
(e) Apply 5 ~Ll of each solution. acid. The melting poim of the precipitate, after washing with
water and drying for 4 hours at 105 0 , is about 201 0,
(d) Develop the plate to 15 cm.
Appendix V A.
(e) After removal of the plate, allow it to dry in air until the
C. The precipitate obtained in test B yields the reaction
solvent has evaporated and examine under ultraviolet Nght
(25417m).
characteristic of primary aromatic amines, Appendix VI,
giving an m"ange-red precipita te.
MOBILE PHASE
TESTS
20 volumes of IBM am71l0nia, 40 volumes of 71lethanol and
All,alinity
60 volumes of chloroform.
pH of a solution containing the equivalent of 1.0% w/v of the
LIMITS anhydrous substance, 9.0 to 10.0, Appendix V L.
In the chromatogram obtained with solution (1): Heavy metals
any spot corresponding to NI ,N2-diquinoxalin-2- Dissolve a quantity containing the equivalent of 2.5 g of the
ylsulphanilamide is not more intense than the spot in the anhydrous substance in 10 mL of water, add 15 mL of
chromatogram obtained with solution (2) (3%); 2M acetic acid, shake for 30 minutes and filter. 12 mL of the
any other secondaJY spot is not more intense than the spot in resulting solution complies with limit test A for heavy metals,
the chromatogram obtained with solution (3) (1 %). Appendix VII. Use lead standard solution (2 ppm Pb) to
Loss on drying prepare the standard (20 ppm).
When dried to constant weight at 105°, loses not more than Related substances
1.0% of its weight. Use 1 g. Carry out the method for thin-layer chromatography,
Sulphated ash Appendix III A, using silica gel H as the coating substance
0 and a mixture of 18 volumes of 10M ammonia and
Not more than 0.1 %, Appendix IX A. Ignite at 600 and use
90 volumes of butan-1-ol as the mobile phase. Apply
1.5 g.
separately to the plate 1O ~lL of each of two solutions in a
ASSAY mixture of 1 volume of 13.5M am11l0nia and 9 volumes of
Dissolve 0.65 g in 10 mI of a mixture of equal volumes of ethanol (96%) containing (1) 1.0% w/v of the substance
1M sodium hydroxide and water. Add 20 mI of glycerol, 20 mI being exarnined and (2) 0.0050% w/v of sulfanilamide. After
of 9M sulphuric acid and 5 g of potassium bromide, cool in ice 0
removal of the plate, heat it at 105 for 10 minutes and spray
and titrate slowly with O.lM sodill11Z nitrite VS, stirring with a 0.1 % w/v solution of 4-dimethylaminobenzaldehyde in
constantly and determining the end point electrometrically. ethanol (96%) containing 1 % v/v of hydrochloric acid. Any
Each mI of O.lM sodiu11l nitrite VS is equivalent to 30.03 mg secondaJY spot in the chromatogram obtained with solution (1)
of C14H12N402S. is not more intense than the spot in the chromatogram
STORAGE obtained with solution (2) (0.-5%).
Sulfaquinoxaline should be protected from light. Loss on drying
0
When dried to constant weight at 105 loses not less than
,

6.0% and not more than 10.0% of its weight (sesquihydrate)


Vet-108 Testosterone Phenylpropionate 2016

or not less than 22.0% and not more than 27.0% of its Loss on drying
weight (pentahydrate). When dried to constant weight at 105°, loses not more than
ASSAY 0.5% of its weight. Use 1 g.
Dissolve 0.5 g in a mixture of 75 mL of water and 10 mL of Sulfated ash
hydrochlO1ic acid, add 3 g of potassium bromide, cool in ice and Not more than 0.1 %, Appendix IX A.
titrate slowly with O.1M sodium nit/ite VS, stining constantly ASSAY
and determining the end point electrometrically. Each mL of Dissolve 10 mg in sufficient absolute ethanol to produce
O.lM sodium nitrite VS is equivalent to 27.73 mg of 100 mL, dilute 5 mL to 50 mL with absolute ethanol and
C9HsN3Na02S2' measure the absorbance of the resulting solution at the
STORAGE maximum at 240 nm, Appendix n B. Calculate the content
Sulfathiazole Sodium should be protected from light. of C2sH3603 taking 395 as the value of A(l %, 1 cm) at the
maximum at 240 nm.
LABELLING
The label sta tes whether the substance is the sesquihydrate or STORAGE
the pentahydrate. Testosterone Phenylpropionate should be protected from
light.

Testosterone Phenylpropionate Tiamulin ***


*** ***
(Tiamulin for Vete1inaly Use) ***
Me~ Ph Eur monograph 1660)
OCOCH 2 CH 2 Ph

(CH 3

H3C'-./N~S~O
o O

420.6 1255-49-8

Action and use


Androgen. 493.8 55297-95-5

DEFINITION Action and use


Testosterone Phenylpropionate is 3-oxo-androst-4-en-17 ~-yl Antibacterial.
3-phenylpropionate. It contains not less than 97.0% and not ~E~ ___________________________________________
more than 103.0% of C2sH3603, calculated with reference to
the dried substance. DEFINITION
(3aS,4R,5S,6S,8R, 9R, 9aR, 10R)-6-Ethenyl-5-hydroxy-
CHARACTERISTICS
4,6,9,1 0-tetramethyl-1-oxodecahydro-3a,9-propano-3 aH-
A white to almost white, crystalline powder. cyclopentacycloocten-8-yl [[2-
Practically insoluble in water; sparingly soluble in ethanol (diethylamino )ethyl] sulfanyl] acetate.
(96%). Semi-synthetic product derived from a fermentation producto
IDENTIFICATION Content
A. The infrared abs01ption spectrum, Appendix n A, is 96.5 per cent to 102.0 per cent (dried substance).
concordant with the reference spectrum of testosterone
phenylpropionate (RSV 43). If the spectra are not CHARACTERS
concordant, dissolve the substances in the minimum volume Appearance
of dichloromethane, evaporate to dryness and prepare a new Sticky, translucent yellowish mass, slightly hygroscopic.
spectrum of the residue. Solubility
B. Complies with the test for identification of steroids, Practically insoluble in water, very soluble in methylene
Appendix In A, using impregnating solvent IJI and mobile chloride, freely soluble in anhydrous ethanol.
phase F. IDENTIFICATION
C. Dissolve 25 mg in 1 mL of methanol, add 2 mL of Infrared absorption spectrophotometry (2.2.24).
senzicarbazide acetate solution, heat under a reflux condenser Comparison Ph. Eur. reference spectrum of tiamulin.
for 30 minutes and cool. The melting point of the resulting
precipitate is about 218°, Appendix V A. TESTS
Appearance of solution
TESTS The solution is clear (2.2.1) and its absorbance (2.2.25) at
Melting point 420 nm is not.greater than 0.050.
0
114 to 117°, Appendix V A.
Dissolve 2.5 g in 50 mL of methanol R.
Specific optical rotation
Related substances
In a 1 % w/v solution in 1)4-dioxan) +86 to +91, calculated
Liquid chromatography (2.2.29).
with reference to the dried substance, Appendix V F.
Ammonium carbonate buffer solution pH 100 Dissolve 10. O g of
ammonium carbonate R in water R, add 22 mL of perchlO1ic
2016 Tiamulin Vet-109

acid solution R and dilute to 1000.0 mL with water R. Adjust STORAGE


to pH 10.0 with eoneentrated ammonia R1. Protected from light.
Solvent mixture aeetonitrile R1, arnmonium carbonate buffer IMPURITIES
solution pH 10.0 (50:50 V/V). Speeijied impurities A, B, C, D, E, F
Test solution Dissolve 0.200 g of the substance to be Other detectable impurities (the following substances would, if
examined in the solvent mixture and dilute to 50.0 mL with present at a sufficient level, be detected by one or other of
the solvent mixture. the tests in the monograph. They are limited by the general
Referenee solution (a) Dissolve 0.250 g of tia11lulin hydrogen acceptance criterion for other/unspecified impurities and/or
fumarate CRS in the solvent mixture and dilute to 50.0 mL by the general monograph Substances for pharmaceutical use
with the solvent mixture. (2034). It is therefore not necessary to identify these
Referenee solution (b) Dilute 1.0 mL of the test solution to impurities for demonstration of compliance. See also 5.10.
100.0 mL with the solvent mixture. Control of impurities in substanees for pharmaeeutieal use): G, H,
Referenee solution (e) Dilute 0.1 mL of toluene R to 100 mL 1, J, K, L, l\II, N, O, P, Q, R.
with aeetonittz'le R. Dilute 0.1 mL of this solution to
100.0 mL with the solvent mixture.
Column:
- size: 1 = 0.15 m, 0 = 4.6 mm;
- statiol1a1Y phase: end-eapped oetadeeylsilyl siliea gel for
ehr0111atography R (5 ~lm);
- temperature: 30 oc.
Mobile phase aeetonitrile R1, arnmonium carbonate buffer R2
solution pH 10.0, 111ethanol R1 (21:30:49 V/V/V).
Flow rate 1.0 mUmin. A. R1 = R2 = H: (3aS,4R,5S,6S,8R,9R,9aR,10R)-6-ethenyl-
5,8-dihydroxy-4,6, 9,1 O-tetramethyloctahydro-3a, 9-propano-
Deteetion Spectrophotometer at 212 nm.
3aH-cyc1opentacyc100cten-1 (4H)-one (mutilin),
Il1Jection 2 O ~lL.
G. R1 = CO-CHzOH, R2 = H:
Run time 3 times the retention time of tiamulin. (3aS,4R,5S, 6S, 8R, 9R, 9aR, 10R)-6-ethenyl-5-hydroxy-
Relative retention With reference to tiamulin (retention 4,6,9,1 0-tetramethyl-1-oxodecahydro-3a, 9-propano-3 aH-
time = about 18 min): impurity A = about 0.22; cyc1opentacyc100cten-8-yl hydroxyacetate (pleuromutilin),
impurity B = about 0.5; impurity C = about 0.66; J. R1 = CO-CH 3 , R2 = H: (3aS,4R,5S,6S,8R,9R,9aR,10R)-
impurity D = about 1.1; impurity F = about 1.6; 6-ethenyl-5-hydroxy-4, 6,9,1 0-tetramethyl-1-oxodecahydro-
impurity E = about 2.4. 3a,9-propano-3aH-cyclopentacycloocten-8-yl acetate (mutilin
System suitability: reference solution (a): I4-acetate) ,
- baseline separation between the peaks due to tiamulin
K. RI = H, R2 = CO-CH 3 :
and impurity D.
(3aS,4R,5S,6S,8R,9R,9aR,IOR)-6-ethenyl-8-hydroxy-
Limits: 4,6,9,1 0-tetramethyl-1-oxodecahydro-3a, 9-propano-3aH-
- il1lpurities A, B, C, D, E, F: for each impurity, not more cyc1opentacycloocten-S-yl acetate (mutilin I1-acetate),
than the area of the principal peak in the chromatogram
L. R1 = CO-CHz-O-SOZ-C6H4-pCH3, R2 = H:
obtained with reference solution (b) (1.0 per cent);
(3aS,4R,5S,6S,8R, 9R, 9aR, 10R)-6-ethenyl-5-hydroxy-
- any other impurity: for each impurity, not more than
4,6,9,1 0-tetramethyl-1-oxodecahydro-3a, 9-propano-3 aH-
0.2 times the area of the principal peak in the
cyc1opentacycloocten-8-yl
chromatogram obtained with reference solution (b)
[[ (4-methylphenyl)sulfonyl] oxy] acetate (pleuromutilin
(0.2 per cent);
22-tosylate),
- total: not more than 3 times the area of the principal peak
in the chromatogram obtained with reference solution (b) M. R1 = R2 = CO-CH3 : (3aS,4R,5S,6S,8R,9R,9aR,10R)-6-
(3.0 per cent); ethenyl-4,6, 9,1 0-tetramethyl-1-oxodecahydro-3a, 9-propano-
- disregard limit: 0.1 times the area of the principal peak in 3aH-cyclopentacycloocten-5,8-diyl diacetate (mutilin 11,14-
the chromatogram obtained with reference solution (b) diacetate),
(0.1 per cent); disregard any peak present in the P. R1 = CO-CHz-O-SOz-C 6 Hs, R2 = H:
chromatogram obtained with reference solution (c). (3 aS,4R,5S, 6S, 8R, 9R, 9aR, 1OR)-6-ethenyl-5-hydroxy-
Loss on drying (2.2.32) 4,6,9,1 0-tetramethyl-1-oxodecahydro-3a,9-propano-3aH-
Maximum 1.0 per cent, determined on 1.000 g by drying in cyc1opentacyc100cten-8-yl [(phenylsulfonyl) oxy] aceta te,
an oven at 80 oc.
Bacteria! endotoxins (2.6.14, Method D)
Less than 0.4 IU/mg, determined in a 1 mg/mL solution in
anhydrous ethanol R (endotoxin free) diluted 1:40 with water
for bacterial endotoxins test.
B. R = CHz-C 6 Hs: 2-(benzylsulfanyl)-N,N-
ASSAY diethylethanamine,
Liquid chromatography (2.2.29) as described in the test for C. R = S-CHz-CHz-N(CzHs)z: 2,2'-(disulfane-1,2-
related substances with the following modification. diyl) bis (N,N-diethylethanamine),
Injection Test solution and reference solution (a).
O. R = H: 2-(diethylamino)ethanethiol,
Calculate the percentage content of CZSH47N04S, from the
dec1ared content of tiamulin hydrogen fumarate CRS.
Vet-110 Tiamulin Hydrogen Fumarate 2016

(CH 3 o
H3C'-./N~S~O H02C~O~O
O
o

D. (3aR,4R,6SJ 8R,9R,9aR,10R)-6-ethenylhyelroxy-4,6,9,10- N. (2E)-4-[2-[[(3aS,4R,5S,6S,8R,9R,9aR,10R)-6-ethenyl-5-


tetramethyl-5-oxodecahyelro-3a, 9-propano-3 aH- hyelroxy-4, 6, 9,1 O-tetramethyl-l-oxodecahydro-3a, 9-propano-
cyc1opentacyc1oocten-8-yl [[2- 3aH-cyclopentacycloocten-8-yl]oxy]-2-oxoethoxy]-4-oxobut-
(diethylamino )ethyl] sulfanyl] acetate, 2-enoic acid (pleuromutilin 22-fumarate),

(CH 3

H3C'-./N~s~O
O OH

E. (3aS,4R,6S,8R,9R,9aR,10R)-6-ethenyl-4,6,9,10- Q. (3aS,4R,5S,6S,8R,9R, 10R)-6-ethenyl-2,5-dihydroxy-


tetramethyl-l ,5-dioxodecahydro-3a, 9·-propano-3 aH- 4,6,9,1 0-tetramethyl-2,3,4,5,6, 7,8, 9-octahyelro-3a,9-propano-
cyc1opentacyc1oocten-8-yl [[2- 3aH-cyclopentacycloocten-8-yl [[2-
(diethylamino )ethyl] sulfanyl] acetate (11-oxotiamulin), (diethylamino )ethyl] sulfanyl] acetate (3,4-didehyelro-2-
hydroxytiamulin) ,
(CH 3

~ ~:rCH'
H3C'-./N~S~O
o

~N~CH3
I

R. N-benzyl-N,N-dibutylbutan-l-aminium.
F. (lRS,3aR,4R,6S,8R,9R,9aR,10R)-6-ethenyl-l-hydroxy-
__________________________________________ ~E~

4,6,9,10-tetramethyl-5-oxodecahydro-3a,9-propano-3aH-
cyc1opentacyc1oocten-8-yl [[2-
(diethylamino )ethyl] sulfanyl] acetate (l-hydroxy-ll-
oxotiamulin) ,
Tiamulin Hydrogen Fumarate
(Tiamulin Hydrogen Fumarate for VeterillaJY use,
Ph Eu/' mOllograph 1659)

H. (2E)-4-[(2RS)-2-[(3aS,4R,5S,6R,8R,9R,9aR,10R)-8-[[[[2-
(diethylamino )ethyl] sulfanyl] acetyl] oxy] -5-hyelroxy-4, 6, 9,10-
tetramethyl-l-oxodecahyelro-3a, 9-propano-3 aH-
cyc1opentacyc1oocten-6-yl] -2-hyelroxyethoxy] -4-oxo but-2-
610 55297-96-6
enoic acid (19,20-dihydroxytiamulin 20-fumarate),
Action and use
Antibacterial.
~E~ __________________________________________

DEFINITION
(3aS,4R,5S,6S,8R,9R,9aR,10R)-6-Ethenyl-5-hyelroxy-
4,6,9,10-tetramethyl-l-oxodecahyelro-3a, 9-propano-3 aH-
cyc1opentacyc1o'octen-8-yl [[2-
(diethylamino )ethyl] sulfanyl] acetate hydrogen
1. (2E)-4-[[(3aS,4R,5S,6S,8R,9R,9aR, 10R)-8-[[[[2-
(E)-butenedioate.
(diethylamino )ethyl] sulfanyl] acetyl] oxy]-6-ethenyl-l,5-
Semi-synthetic product derived from a fermentation product.
dihydroxy-4,6,9,10-tetramethyldecahydro-3a,9-propano-3aH-
cyc1opentacyc1oocten-2-yl] oxy]-4-oxobut-2-enoic acid Content
(2,3-dihyelroxytiamulin 2-fumarate), 96.5 per cent to 102.0 per cent (dried substance).
2016 Tiamulin Hydrogen Fumarate Vet-111

CHARACTERS System suitability: reference solution (a):


Appearance - baseline separation between the peaks due to tiamulin
White or light yellow, clystalline powder. and impurity D.
Solubility Limits:
Soluble in water, freely soluble in anhydrous ethanol and - impurities B, H: for each impurity, notmore than
soluble in methanol. 1.5 times the are a of the principal peak in the
chromatogram obtained with reference solution (b)
IDENTIFICATION (1.5 per cent),
Infrared absorption spectrophotomeuy (2.2.24). - impurities A, C, D, E, F, G, 1, ], K, L, M, S, T: for each
Comparison tiamulin hydrogen fumarate CRS. impurity, not more than the area of the principal peak in
TESTS the chromatogram obtained with reference solution (b)
pH (2.2.3) (1.0 per cent),
- any other impurity: for each impurity, not more than
3.1 to 4.1.
0.2 times the area of the principal peak in the
Dissolve 0.5 g in carbon dioxide-free 'Water R and dilute to
chromatogram obtained with reference solution (b)
50 mL with the same solvento
(0.2 per cent),
Related substances - total: not more than 3 times the area of the principal peak
Liquid chromatography (2.2.29). in the chromatogram obtained with reference solution (b)
Ammoniu111 carbonate buffer solution pH 100 Dissolve 10. O g of (3.0 per cent),
ammonium carbonate R in 'Water R, add 22 mL of perchl017c - disregard limit: 0.1 times the area of the principal peak in
acid solution R and dilute to 1000.0 mL with 'Water R. Adjust the chromatogram obtained with reference solution (b)
to pH 10.0 with concentrated ammonia R1. (0.1 per cent); disregard any peak present in reference
Solvent mixture Ammonium carbonate buffer solution solution (c).
pH 10.0, acetonitl'ile R1 (50:50 V/V). Loss on drying (2.2.32)
Test solution Dissolve 0.200 g of the substance to be Maximum 0.5 per cent, detelmined on 1.000 g by drying in
examined in the solvent mixture and dilute to 50.0 mL with an oven at 105 oC.
the solvent mixture. ASSAY
Reference solution (a) Dissolve 0.200 g of tiamulin hydrogen Liquid chromatography (2.2.29) as described in the test for
fumarate CRS in the solvent mixture and dilute to 50.0 mL related substances with the following modification.
with the solvent mixture. Injection Test solution and reference solution (a).
Reference solution (b) Dilute 1.0 mL of the test solution to Calculate the percentage content of C 32H'lNOsS from the
100.0 mL with the solvent mixture. declared content of tia71lulin hydrogen fumarate CRS.
Reference solution (c) Dissolve 40.0 mg offumaric acid R in
STORAGE
the solvent mixture and dilute to 50.0 mL with the solvent
Protected from light.
mixture.
Reference solution (d) Dissolve 4 mg of tiamulin for peak IMPURITIES
identification CRS (tiamulin hydrogen fumarate containing Specified impurities A, B, C, D, E, F, G, H, I, J, K, L, M, S,
impurities B, C, D, F, H and I) in the solvent mixture and T
dilute to 1 mL with the solvent mixture. Other detectable impurities (the following substances would, if
Column: present at a sufficient level, be detected by one or other of
- size: 1 = 0.15 m, 0 = 4.6 mm, the tests in the monograph. They are limited by the general
- stationaIY phase: end-capped octadecylsilyl silica gel for acceptance criterion for other/unspecified impurities and/or
chromatography R (5 ~lm), by the general monograph Substances for pharmaceutical use
- temperature: 30 oC. (2034). It is therefore not necessary to identify these
Mobile phase acetonitrile R1, ammonium carbonate buffer impurities for demonstration of compliance. See also 5.10.
solution pH 10.0, methanol R1 (21:30:49 V/V/V). Control of impw7ties in substances for pharmaceutical use): N, 0,
P, Q, R.
Flo'W rate 1.0 mLlmin.
Detection Specu'ophotometer at 212 nm.
Injection 2O ~lL.
Run time 3 times the retention time of tiamulin.
Identification of impunties U se the chromatogram supplied
with tiamulin for peak identification CRS and the
chromatogram obtained with reference solution (d) to
identify the peaks due to impurities B and H. R2

Relative retention With reference to tiamulin (retention


A. R1 = R2 = H: (3aS,4R,5S,6S,8R,9R,9aR,lOR)-6-ethenyl-
time = about lS min): impurity G = about 0.2;
5,S-dihydroxy-4,6,9,10-teu'amethyloctahydro-3a,9-propano-
impurity A = about 0.22; impurity H = about 0.23;
3aH-cyclopentacycloocten-1 (4H)-one (mutilin),
impurity I =about 0.3; impurity J = about 0.4;
impurity K = about 0.45; impurity B = about 0.5; G. R1 = CO-CH2 0H, R2 = H:
impurity L = about 0.65; impurity C = about 0.66; (3aS,4R, SS, 6S, SR, 9R, 9aR, 10R)-6-ethenyl-5-hydroxy-
impurity F = about O.S; impurity M = about 0.85; 4,6,9,1 0-tetramethyl-1-oxodecahydro-3a,9-propano-3aH-
impurity D = about 1.1; impurity S = about 1.4; cyclopentacycloocten-S-yl hydroxyacetate (pleuromutilin),
impurity T = about 1.6; impurity E = 2.4. J. R1 = CO-CH3 , R2 = H: (3aS,4R,5S,6S,SR,9R,9aR,10R)-
6-ethenyl-5-hydroxy-4,6, 9,1 0-tetramethyl-1-oxodecahydro-
Vet-112 Tiamulin Hydrogen Fumarate 2016

3a, 9-propano-3aH-cyclopentacycloocten-8-yl acetate (mutilin


14-acetate),
K. R1 = H, R2 = CO-CH 3 :
(3aS,4R,5S,6S,8R,9R,9aR,10R)-6-ethenyl-8-hydroxy-
4,6,9,1 0-tetramethyl-1-oxodecahydro-3a,9-propano-3 aH-
cyclopentacycloocten-5-yl acetate (mutilin 11-acetate),
L. R1 = CO-CHz-O-SOZ-C6H4-pCH3, R2 = H:
(3aS, 4R, 5S, 6S, 8R, 9R, 9aR, 1OR) -6-ethenyl-5-hydroxy-
4,6,9,1 0-tetramethyl-1-oxodecahydro-3a,9-propano-3 aH- H. (2E)-4-[(2RS)-2-[(3aS,4R,5S,6R,8R,9R,9aR,10R)-8-[[[[2-
cyclopentacycloocten-8-yl (diethylamino )ethyl] sulfanyl] acetyl] oxy] -5-hydroxy-4,6, 9,10-
[[ (4-methylphenyl)sulfonyl] oxy] acetate (pleuromutilin tetramethyl-1-oxodecahydro-3a, 9-propano-3 aH-
22-tosylate), cyclopentacycloocten-6-yl] -2-hydroxyethoxy] -4-oxo but-2-
enoic acid (19,20-dihydroxytiamulin 20-fumarate),
M. R1 = R2 = CO-CH3 : (3aS,4R,5S,6S,8R,9R,9aR,10R)-6-
ethenyl-4, 6, 9,1 0-tetramethyl-1-oxodecahydro-3a, 9-propano-
3aH-cyclopentacycloocten-5,8-diyl diacetate (mutilin 11,14- (CH 3
diacetate),
H3C'-../N~SITO
P. R1 = CO-CHz-O-SOz-C6Hs, R2 = H:
(3aS,4R,5S, 6S,8R, 9R, 9aR, 10R)-6-ethenyl-5-hydroxy- O
4,6,9,1 0-tetramethyl-1-oxodecahydro-3a,9-propano-3 aH-
cyclopentacycloocten-8-yl [(phenylsulfonyl) oxy] acetate,
T. R1 = CO-CHz-[S-CHz-CHz-hN(C2Hs)2' R2 = H:
(3aS,4R,5S, 6S,8R, 9R, 9aR, 10R)-6-ethenyl-5-hydroxy-
1. (2E)-4-[[(3aS,4R,5S,6S,8R,9R,9aR,10R)-8-[[[[2-
4,6,9,1 0-tetramethyl-1-oxodecahydro-3a, 9-propano-3 aH-
(diethylamino )ethyl] sulfanyl] acetyl] oxy]-6-ethenyl-1,5-
cyclopentacycloocten-8-yl [[2-[[2-
dihydroxy-4, 6,9,10-tetramethyldecahydro-3a, 9-propano-3 aH-
(diethylamino )ethyl] sulfanyl] ethyl] sulfanyl] acetate,
cyclopentacycloocten-2-yl] oxy]-4-oxobut-2-enoic acid
(2,3-dihydroxytiamulin 2-fumarate),

H02C~OITO
B. R = CHz-C6Hs: 2-(benzylsulfanyl)-N,N- O
diethylethanamine,
C. R = S-CHz-CHz-N(CzHsh 2,2'-(disulfane-1,2-
diyl) bis (N,N-diethylethanamine ),
O. R = H: 2-(diethylamino)ethanethiol,
N. (2E)-4-[2-[[(3aS,4R,5S,6S,8R,9R,9aR,10R)-6-ethenyl-5-
hydroxy-4, 6, 9,1 0-tetramethyl-1-oxodecahydro-3a, 9-propano-
3aH-cyclopentacycloocten-8-yl]oxy]-2-oxoethoxy]-4-oxobut-
2-enoic acid (pleuromutilin 22-fumarate),

OH
D. (3aR,4R,6S,8R,9R,9aR,10R)-6-ethenylhydroxy-4,6,9,10-
tetramethyl-5-oxodecahydro-3a, 9-propano-3aH-
cyclopentacycloocten-8-yl [[2-
(diethylamino )ethyl] sulfanyl] acetate,
Q. (3aS,4R,5S,6S,8R, 9R, 10R)-6-ethenyl-2,5-dihydroxy-
4,6,9,1 0-tetramethyl-2,3,4,5,6, 7,8, 9-octahydro-3a,9-propano-
3aH-cyclopentacycloocten-8-yl [[2-
(diethylamino )ethyl] sulfanyl] acetate (3,4-didehydro-2-
hydroxytiamulin) ,

E. (3aS,4R,6S,8R,9R,9aR,10R)-6-ethenyl-4,6,9,10-
tetramethyl-1,5-dioxodecahydro-3a, 9-propano-3 aH-
cyclopentacycloocten-8-yl [[2-
(diethylamino )ethyl] sulfanyl] acetate (ll-oxotiamulin), R. N-benzyl.,.N,N-dibutylbutan-1-aminium,
F. impurity of unknown structure with a relative retention of
about 0.8,
2016 Triclabendazole Vet-113

(CH 3 -- stationalY phase: base-deactivated end-capped octadecylsilyl


H3C'-../N~s~a
silica gel for chromatography R (5 ~lm).
Mobile phase Dissolve 0.77 g of ammonium acetate R in
a
800 mL of 'Water fol' chromatography R, add 1 mL of
and epimer at C* triethylamine R and mix; adjust to pH 4.5with glacial acetic
acid R and dilute to 1 L with 'Water for chromatography R.
Mix 40 volumes of this solution and 60 volumes of
S. (lRS,3aR,4R,5S,6S,8R,9R,9aR,10R)-6-ethenyl-1-ethyl- acetonitrile R.
1,5-dihydroxy-4,6,9,1 O, 12, 12-hexamethyldecahydro-3a,9- Flo'W rate 1.0 mUmin.
propano-3aH-cyclopentacycloocten-8-yl [[2- Detection Spectrophotometer at 305 nm.
(diethylamino )ethyl] sulfanyl] acetate.
Injection 2O ~L.
___________________________________________ PhEw
Run time 2.5 times the retention time of triclabendazole.
Identification of impurities Use the chromatogram supplied
with triclabendazole for system suitability CRS and the
chromatogram obtained with reference solution (b) to
Triclabendazole **** identify the peaks due to impurities A, B and D.
*** *
****
Relative retention With reference to triclabendazole (retention
Tricl~bendazolefor Veterinary Use time = about 10 min): impurity A = about 0.6;
(Ph Bur monograph 2609) impurity B = about 0.7; impurity D = about 1.9.
System suitability: reference solution (b):
CI~ -- resolution: minimum 2.5 between the peaks due to
impurities A and B.
CI~ Calculation of percentage contents:
H
anN -- con'ection factors: multiply the peak areas of the following
);-s/CH
3
I impurities by the corresponding correction factor:
CI ~ N impurity A = 1.9; impurity D = 2.7;
-- for each impurity, use the concentration of
68786-66-3 triclabendazole in reference solution (a).
Limits:
Action and use -- impurities A, D: for each impurity, maximum 0.3 per cent;
Benzimidazole antihelminthic -- unspecified impurities: for each impurity, maximum
~Ew ___________________________________________
0.20 per cent;
-- total: maximum 1.0 per cent;
DEFINITION -- repol'ting threshold: 0.10 per cent.
5-Chloro-6-: (2,3-dichlorophenoxy)-2-(methylsulfanyl)-l H- Loss on drying (2.2.32)
benzimidazole. Maximum 0.5 per cent, determined on 1.000 g by drying in
Content an oven at 105 oC for 6 h.
99.0 per cent to 101.0 per cent (dried substance). Sulfated ash (2.4.14)
CHARACTERS Maximum 0.1 per cent, determined on 1.0 g.
Appearance ASSAY
White or almost white, crystalline powder. Dissolve 0.280 g in 50 mL of anhydrous acetic acid R. Allow
Solubility to cool and titrate with 0.1 M perchloric acid, determining the
Practically insoluble in water, soluble in acetone, sparingly end-point potentiometrically (2.2.20).
soluble in ethanol (96 per cent). 1 mL of 0.1 M perchloric acid is equivalent to 35.97 mg of
IDENTIFICATION CIJf9Cl3N20S.
Infrared absorption spectrophotometry (2.2.24). STORAGE
Comparison triclabendazole CRS. Protected from light.
TESTS IMPURITIES
Related substances Specified impurities A, D
Liquid chromatography (2.2.29). Prepare the solutions protected Other detectable impurities (the following substances would, if
fr011Z light. present at a sufficient level, be detected by one or other of
Test solution Dissolve 50.0 mg of!he substance to be the tests in the monograph. They are limited by the general
examined in 10 mL of acetonitrile R and dilute to 25.0 mL acceptance criterion for other/unspecified impurities and/or
with the mobile phase. by the general monbgraph Substances for pharmaceutical use
Reference solution (a) Dilute 1.0 mL of the test solution to (2034). It is therefore not necessary to identify these
50.0 mL with the mobile phase. Dilute 1.0 mL of this impurities for demonstration of compliance. See also 5.10.
solution to 10.0 mL with the mobile phase. Control of impurities in substances for pharmaceutical use): B.
Reference solution (b) Dissolve the contents of a vial of
triclabendazole for system suitability CRS ( impurities A, B
and D) in 1.0 mL of the mobile phase.
Colu111n:
-- size: l = 0.25 m, 0 = 4.6 mm;
Vet-114 Tylosin 2016

methyl-~-D-allopyranosyl) oxy] methyl] -6- [[3, 6-dideoxy-4-0-


(2, 6-dideoxy-3-C-methyl-ex-L-ribo-hexopyranosyl) -3-
(dimethylamino )-~-D-glucopyranosyl] oxy] -16-ethyl-4-
hydroxy-5,9,13-trimethyl-7-(2-oxoethyl)oxacyc1ohexadeca-
11,13-diene-2,10-dione (tylosin A, M r 916). Tylosin B
(desmycosin, M r 772), tylosin C (macrocin, M r 902) and
tylosin D (relomycin, NIr 918) may also be presento They
A. 5-chloro-6-(2 ,3-dichlorophenoxy)-2- (methylsulfinyI) -lH- contribute to the potency of the substance to be examined.
benzimidazole, Potency
Minimum 900 IU/mg (dried substance).
CIY)
CHARACTERS
CI~ Appearance
Almost white or slightly yellow powder.
H

°ÚN
CI ~
I ~SH
N
Solubility
Slightly soluble in water, freely soluble in anhydrous ethanol
and in methylene chloride. It dissolves in dilute solutions of
B. 5-chloro-6-(2,3-dichlorophenoxy)-lH-benzimidazole-2- mineral acids.
thiol, IDENTIFICATION
A. Infrared absorption spectrophotometry (2.2.24).
CIY) Comparison tylosin CRS.
B. Examine the chromatograms obtained in the test for
CI~ composition.
O~NH2 Results The principal peak in the chromatogram obtained
CI~N02 with the test solution is similar in retention time and size to
the principal peak in the chromatogram obtained with
reference solution (a).
D.4-chloro-5-(2,3-dichlorophenoxy)-2-nitroaniline.
___________________________________________ ~E~
C. Dissolve about 30 mg in a mixture of 0.15 mL of water R,
2.5 mL of acetic anhydride R and 7.5 mL of pyridine R. Allow
to stand for about 10 mino No green colour develops.
TESTS
Tylosin *** pH (2.2.3)
*** *** 8.5 to 10.5.
(Tylosin for Vete1'1na¡y Use) Ph Eur monogmph 1273) *** Suspend 0.25 g in 10 mL of carbon dioxide-free water R.
Composition
Liquid chromatography (2.2.29): use the normalisation
I procedure. Prepare the solutionsimmediately before use.
o osyl
Solvent mixture acetonitrile R, 'Water R (50:50 V/V).
R3 Test solution Dissolve 20.0 mg of the substance to be
CH 3 H-- examined in the solvent mixture and dilute to 100.0 mL with

H3c~1 0
'N/ CH3 O H" :' HQH30O
the solvent mixture.
Reference solution (a) Dissolve 2 mg of tylosin phosphate for
--H
O HO " peak identification CRS (containing tylosins A, B, C and D) in
I H
R1 OH H C HO the solvent mixture and dilute to 10 mL with the solvent
3 R2 OCH 3 mixture.
Name Mol. Formula R1 R2 R3 Reference solution (b) Dissolve 2 mg of tylosin CRS and 2 mg
tylosin A C46H77N017 osyl OCH 3 CHO of tylosin D CRS in the solvent mixture and dilute to 10 mL
tylosin B C39Hs5N014 H OCH 3 CHO with the solvent mixture.
tylosin C C45H75N017 osyl OH CHO Colu111n:
tylosin D C4sH7gN017 osyl OCH 3 CH 20H - size: 1 = 0.20 m, 0 = 4.6 mm;
- stationa¡y phase: octadecylsilyl silica gel for chromatography R
(5 11m);
Action and use - temperature: 35 oC.
Macrolide antibacterial. Mobile phase Mix 40 volumes of acetonitrile R and 60 volumes
Preparation of a 200 giL solution of sodium perchlorate R previously
Tylosin Injection adjusted to pH 2.5 using 1 M hydrochloric acid.
Flo'W mte 1.0 mU1,TIin.
PhE~ ___________________________________________
Detection Spectrophotometer at 290 nm.
DEFINITION Injection 20 ~lL.
Mixture of macrolide antibiotics produced by a strain of Retention time Tylosin A = about 12 mino
Streptomyces fradiae or by any other means. The main
Identification of peaks U se the chromatogram supplied with
component of the mixture is
tylosin phosphate for peak identification CRS and the
(4R,5S,6S, 7R,9R,11E,13E,15R, 16R)-15-[[(6-deoxy-2,3-di-O-
2016 Tylosin Phosphate Vet-115

chromatogram obtained with reference solution (a) to


identify the peaks due to tylosins A, B, C and D.
System suitability: reference solution (b):
-- resalutian: minimum 2.0 between the peaks due to tylosins
A and D.
Limits:
-- tylasin A: minimum 80.0 per cent;
- - SU11l al tylasins A) B) C and D: minimum 95.0 per cent.

Tyramine
Maximum 0.35 per cent and maximum 0.15 per cent, if
intended for use in the manufacture of parenteral
preparations. B. tylosin A aldol.
In a 25.0 mL volumetric fiask, dissolve 50.0 mg in 5.0 mL of ___________________________________________ ~E~

a 3.4 gIL solution of phaspharic acid R. Add 1.0 mL of


pyridine R and 2.0 mL of a saturated solution of ninhydrin R
(about 40 giL). Close the fiask with a piece of aluminium foil
and heat in a watér-bath at 85 oC for 30 mino Cool the
solution rapidly and dilute to 25.0 mL with water R. Mix and Tylosin Phosphate
measure irnmediately the absorbance (2.2.25) of the solution
at 570 nm using a blank solution as the compensation liquido (Tylasin Phosphate Bulk Solutiol1 101" Veteril1a1Y Use)
The absorbance is not greater than that of a standard Ph Bur 1110nograph 1661)
prepared at the same time and in the same manner using
5.0 mL of a 35 mg/L solution of tyra111ine R in a 3.4 gIL
solution of phaspharic acid R. If intended for use in the
manufacture of parenteral preparations, the absorbance is not
greater than that of a standard prepared at the same time and R3
in the same manner using 5.0 mL of a 15 mg/L solution of

H
CH 3 H--
tyramine R in a 3.4 gIL solution of phaspharic acid R. I O O ,
Loss on drying (2.2.32) H3C~/CH3
'N/ '
O HO :
Maximum 5.0 per cent, determined on 1.000 g by drying in
I H
an oven at 60 oC at a pressure not exceeding 0.7 kPa for 3 h. R1 OH

Sulfated ash (2.4.14)


Maximum 3.0 per cent, determined on 1.0 g. Tylosin R1 R2 R3 Mol. Formula Mr

ASSAY osyl OCH 3 CHO

Carry out the microbiological assay of antibiotics (2.7.2). B H OCH 3 CHO C39H65N014 772
Use tylasin CRS as the chemical reference substance. C osyl OH CHO C45H75N017 902
D osyl OCH 3 CH 2 0H C46H79N017 918
STORAGE
Protected from light.
IMPURITIES Action and use
Macrolide antibacterial.
o
PhE~ ___________________________________________

OHC DEFINITION
CH 3 H-- Solution of the dihydrogen phosphate of a mixture of
h oo , macrolide antibiotics produced by a strain of Streptomyces

Hv:'~q~~H'
fradiae or by any other means.
The main component is the phosphate of
CH 3
OH
OH (4R,5S,6S, 7R,9R, 11E,13B, 15R, 16R)-15-[[(6-deoxy-2,3-di-O-
methyl-~-D-allopyranosyl)oxy]methyl]-6-[[3,6-dideoxy-4-0-
(2, 6-dideoxy-3-C-methyl-cx-L-ribo-hexopyranosyl) -3-
CH 3
(dimethylamino )-~-D-glucopyranosyl] oxy] -16-ethyl-4-
hydroxy-5, 9, 13-trimethyl-7-(2-oxoethyl)oxacyclohexadeca-
A. desmycinosyltylosin, 11,13-diene-2,10-dione (tylosin A phosphate).
The phosphates of tylosin B (desmycosin phosphate), tylosin
C (macrocin phosphate) and tylosin D (relomycin
phosphate) may also be presento The solution also contains
sodium dihydrogen phosphate.
Potency
Minimum 800 IU per milligram of dry residue.
Tylosins A, B, C and D contribute to the potency.
CHARACTERS
Appearance
Yellow or brownish-yellow, viscous liquido
Vet-116 Tylosin Phosphate 2016

Solubility chromatogram obtained with reference solution (a) to


Miscible with water. identify the peaks due to tylosins A, B, C and D.
IDENTIFICATION Relative retention With reference to tylosin A (retention
A. Vltraviolet and visible absorption spectrophotometry time = about 12 min): impurity A = about 0.35;
(2.2.25). tylosin C = about 0.5; tylosin B = about 0.6;
tylosin D = about 0.85; impurity B = about 0.9.
Test solution Dilute an amount of the preparation to be
examined equivalent to 400 000 IV of tylosin phosphate to System suitability: reference solution (b):
100.0 mL with water R. Dilute 1.0 mL ofthis solution to - resolution: minimum 2.0 between the peaks due to
100.0 mL with water R. tylosin D and tylosin A.
Speetral range 230-350 nm. Limits:
- tylosin A: minimum 80.0 per cent;
Abs01ption maximum At 290 nm. - sum of tylosins A) B) C and D: minimum 95.0 per cent;
Absorbanee at the abs01ption 711aximum Minimum 0.70. - disregard limit: area of the principal peak in the
B. Examine the chromatograms obtained in the test for chromatogram obtained with reference solution (c).
composition. Tyramine
Results The principal peak in the chromatogram obtained In a 25.0 mL volumetric flask, dissolve an amount of the
with the test solution is similar in retention time and size to preparation to be examined equivalent to 50 000 IV of
the principal peak in the chromatogram obtained with tylosin phosphate in 5.0 mL of a 3.4 giL solution of
reference solution (a). phosphorie acid R. Add 1.0 mL of pyridine R and 2.0 mL of a
C. Dilute an amount of the preparation to be examined saturated solution of ninhydrin R (about 40 gIL). Close the
equivalent to 400 000 IV of tylosin phosphate in 10 mL of flask with aluminium foil and heat in a water-bath at 85 oC
'water R. The solution gives reaction (a) of phosphates for 20-30 mino Cool the solution rapidly and dilute to
(2.3.1). 25.0 mL with water R. Mix and measure immediately the
absorbance (2.2.25) of the solution at 570 nm using a blank
TESTS
solution as the compensation liquido
pH (2.2.3)
5.5 to 6.5. The absorbance is not greater than that of a standard
prepared at the same time and in the same manner using
Dilute 1.0 g in 10 mL of ea/'bon dioxide-free water R.
5.0 mL of a 35 mg/L solution of tyramine R in a 3.4 giL
Composition solution of phosphorz'e acid R.
Liquid chromatography (2.2.29): use the nonnalisation Phosphate
procedure. Prepare the solutio1lS immediately before use. 8.5 per cent to 10.0 per cent of P0 4 , calculated with
Test solution Dilute an amount of the preparation to be reference to the dry residue (see Assay).
examined equivalent to 50 000 IV of tylosin phosphate to Test solution Dissolve an amount of the preparation to be
200 mL with a mixture of equal volumes of aeetonitrile R and examined equivalent to 200 000 IV of tylosin phosphate in
'Water R.
50 mL of water R. Add 5.0 mL of dilute sulfurie acid R and
Referenee solution (a) Dissolve 2 mg of tylosin phosphate for dilute to 100.0 mL with water R. To 2.0 mL ofthis solution
peak identifieation CRS (containing tylosins A, B, C and D) in add successively, mixing after each addition, 10.0 mL of
a mixture of equal volumes of aeetonitrile R and 'Water R and water R, 5.0 mL of ammonium molybdate reagent R2, 1.0 mL
dilute to 10 mL with the same mixture of solvents. of hydroquinone solution R and 1.0 mL of a 200 gIL solution
Referenee solution (b) Dissolve 2 mg of tylosin CRS and 2 mg of sodium metabisulfite R. Allow to stand for at Ieast 20 min
of tylosin D CRS in a mixture of equal volumes of and dilute to 50.0 mL with water R. Mix thoroughly.
aeetonitrile R and water R and dilute to 10 mL with the same Referenee solutiol1 (a) To 1.0 mL of a standard solution
mixture of solvents. containing O.430 giL of potassium dihydrogen phosphate R
Referenee solution (e) Dilute 1.0 mL of reference solution (a) (con"esponds to 300 ppm of P0 4 ) add successively, mixing
to 100.0 mL with a mixture of equal volumes of aeetonitrile R after each addition, 10.0 mL of water R, 5.0 mL of
and water R. Dilute 1. O mL of this solution to 10. O mL with ammonium molybdate reagent R2, 1.0 mL of hydroquinone
a mixture of equal volumes of aeetonitrile R and water R. solution R and 1.0 mL of a 200 giL solution of sodium
Column: nzetabisulfite R. Allow to stand for at least 20 min and dilute
- size: l = 0.20 m, 0 = 4.6 mm; to 50.0 mL with water R. Mix thoroughly.
- stationary phase: oetadeeylsilyl siliea gel for ehromatography R Referenee solutiol1 (b) Prepare as reference solution (a) but
(5 ~lm); using 2.0 mL of the standard solution.
- tempera tu re: 35 oc. Referenee solution (e) Prepare as reference solution (a) but
Mobile phase Mix 40 volumes of aeetonitrile R and 60 volumes using 5. O mL of the standard solution.
of a 200 giL solution of sodiu111 perehlorate R previously Compensation liquid Prepare as reference solution (a) but
adjusted to pH 2.5 using a 36.5 gIL solution of hydroehlorie omitting the standard solution.
aeid R.
Measure the absorbarice (2.2.25) of the test solution and of
Flow rate 1.0 mIJmin. the reference solutions at 650 nm. Draw a calibration curve
Deteetion Spectrophotometer at 290 nm. with the absorbances of the 3 reference solutions as a
Injection 20 ~lL. function of the quantity of phosphate in the solutions and
Run time 1.8 times the retention time of tylosin A. read from the curve the quantity of pho_sphate in the test
solution. Determine the percentage content of P0 4,
Identifieation of tylosins Vse the chromatogram supplied with
calculated with reference to the dry residue (see Assay).
tylosin phosphate for peak identifieation CRS and the
2016 Tylosin Tartrate Vet-117

ASSAY
Tylosin Tartrate ***
Carry out the microbiological assay of antibiotics (2.7.2). *** ***
U se tylosin CRS as the reference substance. Calculate the (Tylosin T artrate for Veterilla/Y Use) ***
potency from the mass of the dry residue and the activity of Ph Eur monograph 1274)
the solution.
Dr:y residue Dry 3.0 g of the preparation to be examined O
in vacuo at 60 oC for 3 h and weigh.

~
STORAGE H3
Protected from light, at a temperature of 2 oC to 8 oc.
CH 3 /~o
LABELLING --H O H

The label states the concentration of the solution in --H


O
International Units per milligram of preparation.
H3 C
IMPURITIES CH 3
O
o

OHC R2 OCH 3 2
CH 3 H--

h oo ,

H~:'CfL{ ~~ H'
CH 3 OH
OH

CH 3

Action and U se
A. desmycinosyltylosin A, Macrolide antibacterial.
PhEw ___________________________________________

DEFINITION
Taru'ate of a mixture of macrolide antibiotics produced by a
su'ain of Streptomyces fradiae or by any other means.
The main component of the mixture is
(4R,5S,6S, 7R,9R,IIE,13E,15R,16R)-15-[[(6-deoxy-2,3-di-O-
methyl-~-D-allopyranosyl)oxy]methyl]-6-[[3,6-dideoxy-4-0-
(2, 6-dideoxy-3-C-methyl-Ci-L-ribo-hexopyranosyl) -3-
(dimethylamino )-~-D-glucopyranosyl] oxy]-16-ethyl-4-
hydroxy-5, 9, 13-trimethyl-7-(2-oxoethyl)oxacyclohexadeca-
1l,13-diene-2,10-dione (tylosin A, tartrate M r 1982).
Tylosin B (desmycosin, tartrate M r 1694), tylosin e
(macrocin, tartrate M r 1954) and tylosin D (relomycin,
B. (IR,2S,3S,4R,8R,9R,10E,12E,15R,16RS)-9-[[(6-deoxy-
tat'U'ate }\IIr 1986) may also be presento They contribute to
2,3-di-O-methy-~-D-allopyranosyl)oxy]methyl]-2-[[3,6-
the potency of the substance to be examined.
dideoxy-4-0- (2, 6-dideoxy-3-C-methyl-Ci-L-ribo-
hexopyranosyl)-3-(dimethylamino )-~-D-glucopyranosyl] oxy]- Potency
8-ethyl-4, 16-dihydroxy-3,11, 15-u'imethyl-7- Minimum 800 IU/mg (dried substance).
oxabicyc1o [13.2.1] octadeca-l O, 12-diene-6, 14-dione (tylosin A CHARACTERS
aldol). Appearance
___________________________________________ ~Ew
Almost white or slightly yellow, hygroscopic powder.
Solubility
Freely soluble in water and in methylene chloride, slightly
soluble in anhydrous ethanol. It dissolves in dilute solutions
of mineral acids.
IDENTIFICATION
A. Infrared absorption spectrophotometry (2.2.24).
Comparison Ph. Eur. reference specrrum of tylosin tamate.
B. Examine the chromatograms obtained in the test for
composition.
Results The principal peak in the chromatogram obtained
with the test solution is simÍlar in retention time and size to
the principal peak in the chromatogram obtained with
reference solution (a).
Vet-118 Valnemulin Hydrochloride 2016

C. Dissolve about 30 mg in a mixture of 0.15 mL of water R, Loss on drying (2.2.32)


2.5 mL of acetic anhydride R and 7.5 mL of pyridine R. Allow Maximum 4.5 per cent, determined on 1.000 g by drying at
to stand for about 10 mino A green colour is produced. 60 oC at a pressure not exceeding 0.7 kPa for 3 h.
TESTS Sulfated ash (2.4.14)
pH (2.2.3) Maximum 2.5 per cent, determined on 1.0 g.
5.0 to 7.2. ASSAY
Dissolve 0.25 g in 10 mL of cm'bon dioxide-free water R. Canyr out the microbiological assay of antibiotics (2.7.2).
Composition Use tylosin CRS as the chemical reference substance.
Liquid chromatography (2.2.29): use the normalisation STORAGE
procedure. Prepare the solutions immediately before use. In an airtight container, protected from light.
Solvent mixture acetonitrile R, water R (50:50 VIV).
IMPURITIES
Test solution Dissolve 20.0 mg of the substance to be
examined in the solvent mixture and dilute to 100.0 mL with o
the solvent mixture.
Reference solution (a) Dissolve 2 mg of tylosin phosphate for OHC
peak identification CRS (containing tylosins A, B, C and D) in CH 3 H--
the solvent mixture and dilute to 10 mL with the solvent h oo ,

Hv:'~r\L{ ~~ H
mixture.
Reference solution (b) Dissolve 2 mg of tylosin CRS and 2 mg
of tylosin D CRS in the solvent mixture and dilute to 10 mL CH 3
OH
OH
with the solvent mixture.
Colu111n: CH 3
-- size: 1 = 0.20 m, 0 = 4.6 mm;
-- stationmy phase: octadecylsilyl silica gel fol' chromatography R A. desmycinosyltylosin,
(5 ~lm);
-- tempera tu re: 35 oc.
Mobile phase Mix 40 volumes of acetonitrile R and 60 volumes HO

of a 200 giL solution of sodiu111 perchlorate R previously


adjusted to pH 2.5 using 1 M hydrochloric acid.
Flow rate 1.0 mLlmin.
Detection Spectrophotometer at 290 nm.
,: HQH30O
--H
Injectiol1 20 ~lL.
Retention time Tylosin A = about 12 mino H C HO
3
H3 CO OCH 3
Identification of peaks Use the chromatogram supplied with
tylosin phosphate for peak identificatiol1 CRS and the
clu'omatogram obtained with reference solution (a) to
identify the peaks due to tylosins A, B, C and D. B. tylosin A aldol.
System suitability: reference solution (b): ___________________________________________ ~Ew

-- resolution: minimum 2.0 between the peaks due to


tylosins A and D.
Limits:
-- tylosin A: minimum 80.0 per cent;
Valnemulin Hydrochloride ***
-- sum of tylosins A) B) C and D: minimum 95.0 per cent. ** **
Tyramine (Valnemulin Hydrochloride for Veterinmy Use) *****
Maximum 0.35 per cent and maximum 0.15 per cent, if it is Ph Bur monograph 2137)
intended for use in the manufacture of parenteral
preparations.
In a 25.0 mL volumetric fiask, dissolve 50.0 mg in 5.0 mL of
a 3.4 gIL solution of phosph01'ic acid R. Add 1.0 mL of , HCI
pyrz'dine R and 2.0 mL of a saturated solution of ninhydrin R
(about 40 gIL). Close the fiask with a piece of aluminium foil
and heat in a water-bath at 85 oC for 30 mino Cool the
solution rapidly and dilute to 25.0 mL with water R. Mix and
measure irnmediately the absorbance (2.2.25) of the solution 601 133868-46-9
at 570 nm using a blank solution as the compensation liquido
The absorbance is not greater than that of a standard Action and use
prepared at the same time and in the same manner using Antibacterial.
5.0 mL of a 35 mg/L solution of tyramine R in a 3.4 gIL ~Ew ____________________________ ~ _____________
solution of phosphorz'c acid R. If intended for use in the
manufacture of parenteral preparations, the absorbance is not DEFINITION
greater than that of a standard prepared at the same time and (3aS,4R,5S,6S,8R,9R,9aR,10R)-6-Ethenyl-5-hydroxy-
in the same manner using 5.0 mL of a 15 mg/L solution of 4-,6,9,1 0-tetramethyl-1-oxodecahydro-3a,9-propano-3aH-
tyramine R in a 3.4 gIL solution of phosphoric acid R. cyclopenta [8] annulen-8-yl [[2- [[ (2R)- 2-amino-3-
2016 Valnemulin Hydrochloride Vet-119

methylbutanoyl] amino ]-1, 1-dimethylethyl] sulfanyl] acetate Time Mobile phase A Mobile phase B
hydroch1oride. (min) (per cent V/V) (per cent V/V)
0-2 95 .¿ 55 5.¿ 45
Semi-synthetic product derived from a fermentation producto
Content 2 - 4.5 55.¿ 50 45.¿ 50

96.0 per cent to 102.0 per cent (anhydrous substance). 4.5 - 5.5 50.¿ 35 50.¿ 65

CHARACTERS 5.5 - 6.85 35 65


Appearance 6.85 - 10 35 .¿ O 65 .¿ 100
White 01' yellowish, amorphous powder, hygroscopic.
10 - 13 O 100
Solubility
Free1y soluble in water and in anhydrous ethanol, practically 13 - 14 O.¿ 95 100 .¿ 5
insoluble in tert-butyl methyl ether. 14 - 20 95
IDENTIFICATION
A. Infrared absorption spectrophotometry (2.2.24).
Plow rate 1.5 mUmin.
Comparison valnemulin hydrochlo1"l'de CRS.
Detection Spectrophotometer at 200 nm.
B. It gives reaction (a) of chlorides (2.3.1).
Injection 5 ~lL.
TESTS Identification of impurities U se the chromatogram supplied
pH (2.2.3) with valnemulin for peak identification CRS and the
3.0 to 6.0. chromatogram obtained with reference solution (c) to identify
Dissolve 2.0 g in carbon dioxide-free water R and dilute to the peaks due to impurities A, B and C.
20 mL with the same solvento Relative retention With reference to valnemulin (retention
Specific optical rotation (2.2.7) time = about 7 min): impurity D = about 0.2;
+ 15.5 to + 18.0 (anhydrous substance). impurity A = about 0.7; impurity B = about 0.85;
Dissolve 0.250 g in water R and dilute to 25.0 mL with the impurity E = about 0.9; impurity C = about 1.1.
same solvento System suitability: reference solution (b):
Related substances - resolution: minimum 1.5 between the peaks due to
Liquid chromatography (2.2.29). impurity E and valnemulin.
Limits:
Phosphate buffer solution pH 25 Dissolve 8.0 g of disodium
- correction factors: for the calculation of content multiply
hydrogen phosphate R and 3.0 g of potassium dihydrogen
the peak are as of the following impurities by the
phosphate R in water for chromatogmphy R and dilute to
corresponding correction factor: impurity B = 3.2;
1000.0 mL with the same solvento Adjust to pH 2.5 with
impurity E = 4.2;
phosphoric acid R.
- impurity A: not more than 0.5 times the area of the
Solvem mixture Mix equal volumes of acetonitrile Rl and water principal peak in the chromatogram obtained with
for chromatogmphy R. reference solution (a) (0.5 per cent);
Test solution Dissolve 0.100 g of the substance to be - impurity B: not more than twice the area of the principal
examined in the solvent mixture and dilute to 10.0 mL with peak in the chromatogram obtained with reference
the solvent mixture. solution (a) (2.0 per cent);
Reference solution (a) Dilute 1.0 mL of the test solution to - impurity C: not more than the area of the principal peak
100.0 mL with the solvent mixture. in the chromatogram obtained with reference solution (a)
Reference solution (b) Dissolve 5 mg of valne111ulin (1. O per cent);
impurity E CRS and 5 mg of the substance to be examined in - any other impurity: for each impurity, not more than
the solvent mixture and dilute to 25 mL with the solvent 0.2 times the area of the principal peak in the
mixture. chromatogram obtained with reference solution (a)
(0.2 per cent);
Reference solution (c) Dissolve the contents of a vial of
total: not more than 3 times the area of the principal peak
valnemulin for peak identification CRS (containing impurities
in the chromatogram obtained with reference solution (a)
A, B and C) in 1 mL of the solvent mixture.
(3.0 per cent);
Column: disregard limit: 0.1 times the area of the principal peak in
- size: l = 0.15 m, 0 = 4.6 mm; the chromatogram obtained with reference solution (a)
- statiOl1a1Y phase: octadecylsilyl silica gel for chr0111atogmphy R (0.1 per cent); disregard the peak due to the chloride ion.
(3 ~lm);
- tenzpemture: 50 oC. Water (2.5.12)
Maximum 4.0 per cent, determined on 0.500 g.
Mobile phase:
- mobile phase A: phosphate buffer solution pH 2.5, wate¡" R ASSAY
(25:75 V/V); Liquid chromatography (2.2.29).
- mobile phase B: phosphate buffer solution pH 2.5, Test solution Dissolve 40. O mg of the substance to be
acetonitrile Rl (25:75 V/V); examined in a mixture of equal volumes of acetonitrile Rl and
water R and dilute to 50.0 mL with the same mixture of
solvents.
Refei"ence solution Dissolve 50.0 mg of valne111ulin hydrogen
tartrate CRS in a mixture of equal volumes of acetonitrile Rl
and water R and dilute to 50.0 mL with the same mixture of
solvents.
Vet-120 Vedaprofen 2016

Column:
- size: l = 0.125 m, 0 = 4.6 mm;
- stationmy phase: octadecylsi1yl silica gel for chromatography R
(3 ~lm);
- temperature: 45 oC.
Mobile phase Mix 43 volumes of acetonitrile R1 and
57 volumes of a solution containing 0.94 giL of disodium
hydrogen phosphate R and 8.7 giL of potassiu11l dihydrogen E. (3aS,4R,5S,6S,8R, 9R, 9aR, 10R) 6-ethenyl-5-hydroxy-
phosphate R previously adjusted to pH 2.5 with phosphoric 4,6,9,10-tetramethyl-1-oxodecahydro-3a, 9-propano-3 aH-
acid R. cyc10penta [8] annulen-8-yl 2-hydroxyacetate (pleuromulin).
Flow rate 1.2 mUmin. ___________________________________________ ~E~

Detection Spectrophotometer at 210 nm.


Jnjection 5 ~lL.
Run time 3 times the retention time of valnemulin (retention
time = about 2.4 min). Vedaprofen
Calculate the percentage content of C31Hs3ClN20sS, using
the dec1ared content of valnemulin hydrogen tartJ"ate CRS and (Vedaprofen fo1' VeterinaJY Use)
by multiplying by 0.841. Ph Bu1' monograph 2248)
STORAGE
In an airtight container, protected from light.
IMPURITIES
Specijied impu1'ities A, B, C.
Other detectable impurities (the following substances would, if
present at a sufficient level, be detected by one or other of
the tests in the monograph. They are limited by the general
acceptance criterion for otherlunspecified impurities and/or 282.4 71109-09-6
by the general monograph Substances for pharmaceutical use
(2034). It is therefore not necessary to identify these Action and use
impurities for demonstration of compliance. See also 5.10. Cyc1o-oxygenase inhibitor; analgesic; anti-inflarnmatory.
Control of i11lpurities in substances for pharmaceutical use): D) E. ~E~ ___________________________________________

DEFINITION
(2RS)-2-( 4-Cyc1ohexyl-1-naphthyl)propanoic acid.
Content
98.5 per cent to 101.0 per cent (dried substance).
CHARACTERS
Appearance
White or almost white powder.
A. R = D-Val, X = SO: (3aS,4R,5S,6S,8R,9R,9aR,10R)-6- Solubility
ethenyl-5-hydroxy-4,6, 9,1 0-tetramethyl-1-oxodecahydro-3a, 9- Practically insoluble in water, freely soluble in acetone,
propano-3aH-cyc1openta [8] annulen-8-yl [[2- [[ (2R)-2-amino- soluble in methanol. It dissolves in dilute solutions of alkali
3-methylbutanoyl] amino] -1, 1-dimethylethyl] sulfinyl] acetate hydroxides.
(valnemulin sulfoxide),
IDENTIFICATION
B. R = H, X = S: (3aS,4R,5S,6S,8R,9R,9aR,10R)-6-ethenyl-
Infrared absorption spectrophotometry (2.2.24).
5-hydroxy-4,6,9,1 0-tetramethyl-1-oxodecahydro-3a, 9-
propano-3 aH-cyc1openta [8] annulen-8-yl [(2-amino-1, 1- Compan·son vedaprofen CRS.
dimethylethyl] sulfanyl] acetate (dimethyl cysteaminyl TESTS
pleuromulin) , Appearance of solution
C. R = D-Val-D-Val, X = S: The solution is c1ear (2.2.1) and not more intensely coloured
(3aS,4R,5S,6S,8R,9R,9aR, 1OR) 6-ethenyl-5-hydroxy-4,6,9, 10- than reference solution Ys (2.2.2) Method JI).
tetramethyl-1-oxodecahydro-3a,9-propano-3 aH- Dissolve 2.0 g in acetone R and dilute to 20.0 mL with the
cyc1openta[8]annulen-8-yl [[2-[[(2R)-2-[[(2R)-2-amino-3- same solvento
methylbutanoyl] amino] -3-methylbutanoyl] amino] -1,1-
Related substances
dimethylethyl] sulfanyl] acetate (valyl-valneumulin),
Liquid chromatography (2.2.29).
Test solution Dissolve 25 mg of the substance to be examined
in methanol R and dilute to 50.0 mL with the same solvento
Reference solution, (a) Dilute 1.0 mL of the test solution to
50.0 mL with methanol R. Dilute 1.0 mL of this solution to
10. O mL with methanol R.
D. (2R)-2-amino-3-methylbutanoic acid (D-valine), Refe1'ence solution (b) Dissolve the contents of a vial of
vedap1'ofen impurity mixture CRS (impurities A, B and C) in
1.0 mL of reference solution (a).
2016 Xylazine Hydrochloride Vet-121

Column: the tests in the monograph. They are limited by the general
-- size: 1 = 0.10 m, 0 = 3.0 mm; acceptance criterion for other/unspecified impurities and/or
-- stationalJJ phase: octadecylsilyl silica gel for chromatography R by the general monograph Substances for pharmaceutical use
(5 ~lm); (2034). It is therefore not necessary to identi:fy these
-- tempera tu re: 35 0e. impurities for demonstration of compliance. See also 5.10.
Mobile phase Dissolve 1.70 g of tetrabutylam1110nium hydrogen Control of impurities in substances fo1' pharmaceutical use): C) D.
sulfate R in 1000 mL of a mixture of 20 volumes of water R
and 80 volumes of methanol R.
Flow rate 0.4 mIJmin.
Detection Spectrophotometer at 288 nm.
and enantiomer
Injection 1O ~lL.
Run time 5 times the retention time of vedaprofen.
Identification of imp'w1ties Use the chromatogram supplied
with vedaprofen i111pW1ty mixture CRS and the chromatogram A. R = CH3 : methyl (2RS)-2-(4-cyc1ohexyl-1-
obtained with reference solution (b) to identify the peaks due naphthyl)propanoate,
to impurities A, B and C. B. R = C(CH3)3: 1,1-dimethylethyl (2RS)-2-(4-cyc1ohexyl-1-
Relative retention With reference to vedaprofen (retention naphthyl)propanoate,
time = about 6 min): impurity C = about 0.8;
impurity A = about 1.8; impurity B = about 3.7.
Syste111 suitability: reference solution (b):
-- resolution: minimum 2.0 between the peaks due to o
impurity e and vedaprofen.
Limits:
-- correction factor: for the calculation of content, multiply the
peak area of impurity B by 0.7; e. R = H: (4-cyc1ohexyl-1-naphthyl)acetic acid,
-- impurz'ties A) B: for each impurity, not more than twice D. R = CH3 : methyl (2RS)-2-(4-cyc1ohexyl-1-
the area of the principal peak in the chromatogram naphthyl)acetate.
obtained with reference solution (a) (0.4 per cent); ___________________________________________ PhE~

-- unspecified i111purities: for each impurity, not more than the


area of the principal peak in the chromatogram obtained
with reference solution (a) (0.20 per cent);
-- total: not more than 2.5 times the area of the principal
peak in the chromatogram obtained with reference
Xylazine Hydrochloride ***
solution (a) (0.5 per cent); *** ***
- disregard limit: 0.5 times the area of the principal peak in (Xylazine Hydrochl011de for Veterina¡y Use) ***
the chromatogram obtained with reference solution (a) Ph Bur monograph 1481)
(0.1 per cent).
Heavy metals (2.4.8)
Maximum 10 ppm.
Dissolve 1. O g in a mixture of 15 vol umes of water R and
85 volumes of acetone R and dilute to 20 mL with the same
mixture of solvents. 12 mL of the solution complies with
test B. Prepare the reference solution using lead standard 256.8 23076-35-9
solution (0.5 ppm Pb) obtained by diluting lead standard
solution (100 PP111 Pb) R with a mixture of 15 volumes of Action and use
water R and 85 volumes of acetone R. Analgesic.
Loss on drying (2.2.32) PhE~ ___________________________________________
Maximum 0.5 per cent, determined on 1.000 g by drying in
DEFINITION
an oven at 105 De.
N-(2, 6-Dimethylphenyl) -5, 6-dihydro-4H-1 ,3-thiazin-2-amine
Sulfated ash (2.4.14) hydrochIoride.
Maximum 0.3 per cent, determined on 0.500 g.
Content
ASSAY 98.0 per cent to 102.0 per cent (dried substance).
Dissolve 0.200 g in 50 mL of ethanol (96 per cem) R and add
CHARACTERS
1.0 mL of 0.1 M hydrochlonc acid. Carry out a potentiometric
Appearance
titration (2.2.20), using 0.1 M sodium hydroxide. Read the
White or almost white, crystalline powder, hygroscopic.
volume added between the 2 points of infiexion.
1 mL of 0.1 M sodiu711 hydroxide is equivalent to 28.24 mg of Solubility
Freely ~oluble in water, very soluble in methanol, free1y
C19H2202'
soluble in methylene chloride.
IMPURITIES It shows polymorphism (5.9):
Specified i111purities A, B
IDENTIFICATION
Other detectable impurities (the following substances would, if
present at a sufficient level, be detected by one 01' other of A. Infrared absorption spectrophotometry (2.2.24).
COmpa11S0n xylazine hydrochl011de CRS.
Vet-122 Xylazine Hydrochloride 2016

If the spectra obtained show differences, dissolve the phosphate R adjusted to pH 7.2 with dilute sodium
substance to be examined and the reference substance hydroxide solution R;
separately in the minimum volume of water R, evaporate to - mobile phase B: methanol R, acetonitrile R (30:70 V/V);
dryness at 60 oC and at a pressure of 10-20 kPa, and record
new spectra using the residues.
Time Mobile phase A Mobile phase B
B. It gives reaction (b) of chlorides (2.3.1). (min) (per cent V/V) (per cent V/V)
TESTS 0- 15 89 ~ 28 11 ~ 72

Solution S 15 - 21 28 72
Dissolve 5.0 g in carbon dioxide-free water R prepared from
distilled water R, heating at 60 oC if necessary; allow to cool
and dilute to 50.0 mL with the same solvento Plow rate 1.0 mL/min.
Appearance of solution Detection Spectrophotometer at 230 nm.
Solution S is not more opalescent than reference Equilibration With a mixture of 28 volumes of mobile phase A
suspension II (2.2.1) and is colourless (2.2.2, l\IIethod II). and 72 volumes of mobile phase B for at least 30 mino
pH (2.2.3) Injection 20 ~lL.
4.0 to 5.5 for solution S. Identification of impurities U se the cruomatogram supplied
Impurity A with xylazine impurity mixture CRS and the cruomatogram
Maximum 100 ppm. obtained with reference solution (b) to identi:fy the peaks due
Solution A Dissolve 0.25 g of the substance to be examined to impurities B and D; use the cruomatogram obtained with
in methanol R and dilute to 10 mL with the same solvento reference solution (a) to identi:fy the peaks due to
This solution is used to prepare the test solution. impurities A and C.
Solution B Dissolve 50 mg of 2,6-dimethylaniline R Relative retention With reference to xylazine (retention
(impurity A) in methanol R and dilute to 100 mL with the time = about 8 min): impurity D = about 0.5;
same solvento Dilute 1 mL of the solution to 100 mL with impurity A = about 0.8; impurity B = about 1.3;
lnethanol R. This solution is used to prepare dle reference impurity C = about 2.2.
solution. System suitability: reference solution (a):
- resolution: minimum 4.0 between the peaks due to
U sing 2 flat-bottomed tubes with an ümer diameter of about
impurity A and xylazine.
10 mm, place in the 1st tube 2 mL of solution A, and Ül the
nd
2 tube 1 mL of solution B and 1 mL of methanol R. Calculation of percentage contents:
To each tube add 1 mL of a freshly prepared 10 giL solution - for impurity C, use the concentration of impurity C in
of dimethyla11linobenzaldehyde R in methanol R and 2 mL of reference solution (a);
glacial acetic acid R and allow to stand at room temperature - for impurities other than C, use the concent:ration of
for 10 mino Compare the colours in diffused daylight, xylazine in reference solution (a).
viewing vertically against a white background. Any yellow Linzits:
colour in the test solution is not more intense than that in impU1ities B, D: for each impurity, maximum 0.2 per cent;
the reference solution. - i11lpurity C: maximum 0.2 per cent;
- unspecified impU1ities: for each impurity, maximum
Related substances
0.20 per cent;
Liquid chromatography (2.2.29). Prepare the solutions
- sum of impznities other than B, C and D: maximum
i11lmediately befare use.
0.2 per cent;
Solvent mixture Mix 8 volumes of acetonitrile R, 30 volumes of - reporting threshold: 0.10 per cent; disregard any peak due
methanol R and 62 volumes of a 2.72 gIL solution of to the blank.
potassium dihydrogen phosphate R adjusted to pH 7.2 with
dilute sodium hydroxide solution R. Heavy metals (2.4.8)
Maximum 10 ppm.
Test solution Dissolve 0.100 g of the substance to be
examüled in the solvent mixture and dilute to 20.0 mL with 12 mL of solution S complies with test A. Prepare the
the solvent mixture. reference solution using 10 mL of lead standard solution
(1 ppm Pb) R.
Reference solution (a) Dissolve 5.0 mg of the substance to be
examined, 5.0 mg of 2,6-dimeth:ylaniline R (impurity A) and Loss on drying (2.2.32)
5.0 mg of xylazine impurity C CRS in acetonitrz'le R and dilute Maximum 0.5 per cent, detelmined on 1.000 g by drying in
to 100.0 mL with the same solvento Dilute 1.0 mL ofdtis an oven at 105 oC for 2 h.
solution to 10.0 mL with the solvent mixture. Sulfated ash (2.4.14)
Reference solution (b) Widl the aid of ultrasound, dissolve the Maximum 0.1 per cent, determined on 1.0 g.
contents of a vial of xylazine impurz'ty mixture CRS ASSAY
(impurities B and D) in 1.0 mL of the solvent mixture. Dissolve 0.200 g in 25 mL of ethanol (96 per cem) R.
Column: Add 25 mL of water R. Titrate with 0.1 M sodium hydroxide,
- size: l = 0.15 m, 0 = 3.9 mm; determining the end-point potentiometrically (2.2.20).
- stationa7)! phase: end-capped octylsilyl silica gel for 1 mL of 0.1 M sodiu111 hydroxide is equivalent to 25.68 mg
chromatography with polar inc01porated groups R (5 ~lm);
of ClzH17ClNzS.
- tempera tu re: 40 oc.
Mobile phase: STORAGE
1110bile phase A: mix 30 volumes of methanol R and In an airtight container, protected from light.
70 volumes of a 2.72 gIL solution of potassiu11l dihydrogen IMPURITIES
Specified impurz'ties: A, B, C, D.
2016 Xylazine Hydrochloride Vet-123

A. 2,6-dimethylaniline (2,6-xylidine),

B. N,N' -bis (2, 6-dimethylphenyl)thiourea,

C. 2,6-dimethylphenyl isothiocyanate,

D. N-(2,6-dimethylphenyl)-N' -(3-hydroxypropyl)thiourea.
_ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ PhEur
Monographs

F orl11.ulated Preparations

Attention is drawn to the General N otices of the British Pharmacopoeia


0'eterinary) governing this section
2016 General Monographs Vet-127

FORMULATED PREPARATIONS: Pharmaceutical Preparations ***


*** ***
GENERAL MONOGRAPHS (Ph. Eur. monograph 2619) ***
The general provisions of the European Pharmacopoeia PhE~ _____________________________________________
relating to a specific type of dosage form apply to all
veterinary dosage forms of the type defined, whether or not INTRODUCTION
an individual monograph is included in the British This monograph is intended to be a reference source of
Pharmacopoeia (Veterinary). These provisions are standards in the European Pharmacopoeia on active
reproduced in the general monographs listed below: substances, excipients and dosage forms, which are to be
applied in the manufacture/preparation of pharmaceuticals,
Capsules
but not a guide on how to manufacture as there is specific
Liquids for Cutaneous Application t guidance available covering methods of manufacture and
Ear Preparations associated controls.
Extracts It does not cover investigational medicinal products, but
Eye Preparations competent authorities may refer to phannacopoeial standards
Medicated Foams when authorising clinical trials using investigational medicinal
products.
Gramiles*
Preparations for Inhalation DEFINITION
Pharmaceutical preparations are medicinal products gene rally
Preparations for Irrigation
consisting of active substances that may be combined with
Oral Liquids* excipients, formulated into a dosage form suitable for the
Nasal Preparations intended use, where necessary after reconstitution, presented
Parenteral Preparations* in a suitable and appropriately labelled container.
Topical Powders* Pharmaceutical preparations may be licensed by the
Pressurised Pharmaceutical Preparations competent authority, or unlicensed and made to dle specific
needs of patients according to legislation. There are 2
Rectal Preparations
categories of unlicensed pharrhaceutical preparations:
Topical Semi-solid Preparations -- extemporaneous preparations, i.e. phmmaceutical
Sticks preparations individually prepared for a specific patient or
Tablets* patient group, supplied after preparation;
Medicated Tampons -- stock preparations, i.e. phmmaceutical preparations
prepared in advance and stored until a request for a
Tinctures
supply is received.
Transdermal Patches
In addition to this monograph, pharmaceutical preparations
Vaginal Preparations also comply with the General Notices and widl dle relevant
Where a general monograph listed aboye includes additional general chapters of the Pharmacopoeia. General chapters are
statements and requirements applicable to the individual normally given for infOlmation and become mandatory when
monographs of the British Pharmacopoeia, such statements, referred to in a general or specific monograph, unless such
modified where appropriate as described below, apply unless reference is made in a way that indicates dlat it is not the
otherwise indicated to any individual monograph for that intention to make the text referred to mandatory but rather
dosage form in the British Pharmacopoeia (Veterinary). to cite it for information.
Where relevant, pharmaceutical preparations also comply
*Where justified and authorised) the requiremems of the European
with the dosage form monographs (e.g. Capsules (0016),
Pharmacopoeia reproduced in the general monograph do not necessarily apply
to fOl1nulated preparations for veterina¡y use.
Tablets (0478)) and general monographs relating to
t W72ere justified and auth01ised) the requirements of the European pharmaceutical preparations (e.g. Alle1'gen products (1063),
Pha1'l1zacopoeia reproduced in the generalmonograph do not necessarily apply He1'bal teas (1435), Homoeopathic p1'eparations (1038),
to formulated preparations for systemic and vete¡ina¡y use. Immunosera fo1' human useJ animal (0084), Iml11unosera fo1'
veterinaJ)' use (0030), Monoclonal antibodies fOI" human
use (2031), Radiopharmaceutical p1'eparations (0125), Vaccines
fo1' human use (0153), Vaccines for vete¡7na¡)' use (0062)).
Where pharmaceutical preparations are
manufactured/prepared using materials of human or animal
origin, the general requirements of general chapters 5.1. 7.
Viral safety, 5.2.6. Evaluation of safety of veterinary vaccines
and immunosera and 5.2.8. Minimising the ¡7s1~ of transmitting
animal spongiform encephalophathy agents via human and
vetennaJ)' medicinal products apply, where appropriate.
ETHICAL CONSIDERATIONS AND GUIDANCE IN
THE PREPARATION OF UNLICENSED
PHARMACEUTICAL PREPARATIONS
The underlying plinciple of legislation for pharmaceutical
preparations is that, subject to specific exemptions, no
pharmaceutical preparation may be placed on the market
without an appropriate marketing authOlisation.
Vet-128 General Monographs 2016

The exemptions from the formallicensing requirement allow Where no specific monographs exist, the required quality
the supply of unlicensed products to meet the special needs must be defined, taking into account the intended use and
of individual patients. However, when deciding to use an the involved risk.
unlicensed preparation all health professionals involved When physicochemical characteristics of active substances
(e.g. the prescribing practitioners and/or the preparing and functionality-related characteristics (FRCs) of excipients
pharmacists) have, within their area of responsibilities, a duty (e.g. particle-size distribution, viscosity, polymorphism) are
of care to the patient receiving the pharmaceutical critical in relation to their role in the manufacturing process
preparation. and quality attributes of the pharmaceutical preparation, they
In considering the preparation of an unlicensed must be identified and controlled.
pharmaceutical preparation, a suitable level of risk assessment Detailed information on FRCs is given in general chapter
is undertaken. 5.15. Functionality-I"elated characteristics of excipients.
The risk assessment identifies: Microbiological quality
- the criticality of different parameters (e.g. quality of active The formulation of the pharmaceutical preparation and its
substances, excipients and containers; design of the container must ensure that the microbiological quality is
preparation process; extent and significance of testing; suitable for the intended use.
stability of the preparation) to the quality of the
During development, it shall be demonstrated that the
preparation; -and
antimicrobial activity of the preparation as such or, if
- the risk that the preparation may present to a particular
necessary, with the addition of a suitable preservative or
patient group.
preservatives, or by the selection of an appropriate container,
Based on the risk assessment, the person responsible for the provides adequate protection from adverse effects that may
preparation must ensure, with a suitable level of assurance, arise from microbial contamination or proliferation during
that the pharmaceutical preparation is, throughout its shelf- the storage anduse of the preparation. A suitable test
life, of an appropriate quality and suitable and fit for its method together with criteria for evaluating the preservative
purpose. For stock preparations, storage conditions and shelf- properties of the formulation are provided in general chapter
life have to be justified on the basis of, for example, 5.1.3. Efficacy of antimicrobial preservatíon.
analytical data or professional judgement, which may be
Ifpreparations do not have adequate antimicrobial efficacy
based on literature references.
and do not contain antlmicrobial preservatives they are
PRODUCTION supplied in single-dose containers, or in multidose containers
Manufacture/preparation must take place within the that prevent microbial contarnination of the contents after
framework of a suitable quality system and be compliant with opening.
the standards relevant to the type of product being made. In the manufacture/preparation of non-sterile pharmaceutical
Licensed products must comply with the requirements of preparations, suitable measures are taken to ensure their
their licence. For unlicensed products a risk assessment as microbial quality; recommendations on this aspect are
outlined in the section 'Ethical considerations and guidance provided in general chapters 5.1.4. Micmbiological quality of
in the preparation of unlicensed pharmaceutical preparations' non-sterile pharmaceutícal preparations and substances for
is of special importance, as these products are not previously pharmaceutical use and 5.1.8. Microbiological quality of habal
assessed by the competent authority. medicinal products for oral use and extracts used in their
Formulation preparation.
During pharmaceutical development or prior to Sterile preparations are manufactured/prepared using
manufacture/preparation, suitable ingredients, processes, tests materials and methods designed to ensure sterility and to
and specifications are identified and justified in order to avoid the introduction of contarninants and the growth of
ensure the suitability of the product for the intended micro-organisms; recommendations on this aspect are
purpose. This includes consideration of the properties provided in general chapter 5.1.1. Methods of preparatíon of
required in order to identify whether specific ingredient sterile products.
properties or process steps are critical to the required quality
Containers
of the phalmaceutical preparation.
A suitable container is selected. Consideration is given to the
Active substances and excipients intended use of the preparation, the properties of the
Active substances and excipients used in the formulation of container, the required shelf-life, and productlcontainer
pharmaceutical preparations comply with the requirements of incompatibilities. Where applicable, containers for
the relevant general monographs, e.g. Substances for pharmaceutical preparations comply with the requirements
pharmaceutícal use (2034), Essential oiZs (2098), Extracts for containers (3.2 and subsections) and materials used for
(0765), Herbal drugs (1433), Hel"bal drug pl"eparations (1434), the manufacture of containers (3.1 and subsections).
Hel"bal drugs fOI" h01110eopathic pl"eparations (2045), Mothel"
Stability
tinctul"es fOI" h01110eopathic preparations (2029), Methods of
Stability requirements of pharmaceutical preparations are
pl"eparation of homoeopathíc stocks and potentisation (2371),
dependent on their intended use and on the desired storage
Pmducts of fermentation (1468), Pmducts with risk of
time.
transmitting agents of animal spongiform encephalopathies (1483),
Products of recombinant DNA technology (0784), Vegetable fatty Where applicable, the probability and criticality of possible
oils (1579). degradation products of the active substance(s) and/or
reaction products of the active substance(s) with an excipient
In addition, where specific monographs exist, the quality of
and/or the immediate container must be assessed. Depending
the active substances and excipients used complies with the
on the result of this assessment, limits ·of degradation and/or
corresponding monographs.
reaction products are set and monitored in the
pharmaceutical preparation. Licensed products require a
stability exercise.
2016 General Monographs Vet-129

Methods used for the purpose of stability testing for all used, it must be demonstrated that they are not affected by
relevant characteristics of the preparation are validated as the presence of the excipients and/or by the formulation.
stability indicating, i.e. the methods allow the quantification Reference standards
of the relevant degradation products and physical See Tests.
characteristic changes.
LABELLING AND STORAGE
TESTS The relevant labelling requirements given in the general
Relevant tests to apply in order to ensure the appropriate dosage form monographs apply. In addition, relevant
quality of a particular dosage form are described in the European Union or other applicable regulations apply.
specific dosage form monographs.
GLOSSARY
Where it is not practical, for unlicensed pharmaceutical
Formulation
preparations, to carry out the tests (e.g. batch size, time
The designing of an appropriate formula (including materials,
restraints), other suitable methods are implemented to ensure
processes, etc.) that will ensure that the patient receives the
that the appropriate quality is achieved in accordance with
suitable pharmaceutical preparation in an appropriate form
the risk assessment carried out and any local guidance or
that has the required quality and that will be stable and
legal requirements.
effective for the required length of time.
Stock preparations are nOlmally tested to a greater extent
than extemporaneous preparations. Licensed pharmaceutical preparation
A medicinal product that has been granted a marketing
The following tests are applicable to many preparations and
authorisation by a competent authority. Synonym: authorised
are therefore listed here.
pharmaceutical preparation.
Appearance
Manufacture
The appearance (e.g. size, shape and colour) of the
All operations of purchase of material s and products,
pharmaceutical preparation is controlled.
Production, Quality Control, release, storage, distribution of
Identity and purity tests medicinal products and the related controls.
Where applicable, the following tests are carried out on the
Preparation (of an unlicensed pharmaceutical
pharmaceutical preparation:
preparation)
- identification of the active substance(s)j
The 'manufacture' of unlicensed pharmaceutical preparations
- identification of specific excipientes), such as
by or at the request of pharmacies or other healthcare
preservatives j
establishments (the telm 'preparation' is used instead of
- purity tests (e.g. investigation of degradation products,
'manufacture' in order clearly to distinguish it from the
residual solvents (2.4.24) or other related impurities,
industrial manufacture of licensed phalmaceutical
sterility (2.6.1))j
preparations) .
- safety tests (e.g. safety tests for biological products).
Reconstitution
Uniformity (2.9.40 or 2.9.5/2.9.6).
Manipulation to enable the use or application of a medicinal
Pharmaceutical preparations presented in single-dos e units product with a marketing authorisation in accordance with
comply with the testes) as prescribed in the relevant specific the instructions given in the summary of product
dosage form monograph. If justified and authorised, general characteristics or the patient information leaflet.
chapter 2.9.40 can be applicable only at the time of release.
Risk assessment
Special uniformity requirements apply in the following cases:
The identification of hazards and the analysis and evaluation
- for herbal drugs and herbal drug preparations, compliance
of risks associated with exposure to those hazards.
with general chapter 2.9.40 is not requiredj
- for homoeopathic preparations, the provisions of general Unlicensed pharmaceutical preparation
chapters 2.9.6 and 2.9.40 are nOlmally not appropriate, A medicinal product that is exempt from the need of having
however in certain circumstances compliance with these a marketing authorisation issued by a competent authority
chapters may be required by the competent authorityj but is made for specific patients' needs according to
- for single- and multivitamin and trace-element legislation.
preparations, compliance with general chapters 2.9.6 and _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ PhEur
2.9.40 (content uniJormity only) is not required;
- in justified and authorised circumstances, for other
preparations, compliance with general chapters 2.9.6 and
2.9.40 may not be required by the competent authority.
Reference standards
Reference standards may be needed at various stages for
quality control of pharmaceutical preparations. They are
established and monitored taking due account of general
chapter 5.12. Reference standards.
ASSAY
Unless otherwise justified and authorised, contents of active
substances and specific excipients such as preservatives are
detelmined in pharmaceutical preparations. Limits must be
defined and justified.
Suitable and validated methods are used. If assay methods
prescribed in the respective active substance monographs are
Vet-130 General Monographs 2016

Glossary SPOT-ON PREPARATIONS


A glossmy of terms relating to formulated preparations is DEFINITION
included in the British Phannacopoeia. Spot-on preparations contain one 01' more active substances
for the prevention and treatment of ectoparasitic and/or
endoparasitic infestations of animals. They are applied
in volumes which are usually less than 10 mL, to a small area
on the head 01' back, as appropriate, of the animal.
Veterinary Liquid Preparations tor *****
** ** S PRAY S
Cutaneous Application *** DEFINITION
(Ph. Eur. monograph 1808)
Sprays contain one 01' more active substances that are
Vetenna1Y Liquid Preparations for Cutaneous Application comply intended to be applied extemally for therapeutic 01'
zuith the requirements of the European Pharmacopoeia. These prophylactic purposes. They are delivered in the form of an
requirements are reproduced belozv. aerosol by the actuation of an appropriate valve 01' by means
PhE~ _____________________________________________ of a suitable atomising device that is either an integral part of
Unless otherzvise justified and authorised) veterina1Y liquid the container 01' is supplied separately.
preparations for cutaneous application comply zuith the Sprays may be presented in pressurised container s (see
requirements of the monograph on Liquid preparations for Pressul'ised phannaceutical preparations (0523)). When so
cutaneous application (0927). In addition to these requirements) presented, sprays usually consist of one 01' more active
the follozuing statements apply to veterina1Y liquid preparations for substances in a suitable vehicle held under pressure with
cutaneous application. suitable propellants 01' suitable mixtures of propellants. When
otherwise presented, sprays are supplied in well-closed
DEFINITION
containers.
Veterinary liquid preparations for cutaneous application are
liquid preparations intended to be applied to the skin to PRODUCTION
obtain a local and/or systemic effect. They are solutions, During the development and manufacture of a spray,
suspensions 01' emulsions which may contain one 01' more measures are taken to ensure that the assembled product
active substances in a suitable vehicle. They may be confonns to a defined spray rate and spray pattem.
presented as concentrates in the form of wettable powders,
pastes, solutions 01' suspensions, which are used to prepare TEAT DIPS
diluted suspensions 01' emulsions of active substances. They DEFINITION
may contain suitable antimicrobial preservatives, antioxidants Teat dips contain one 01' more disinfectant active substances,
and other excipients such as stabilisers, emulsifiers and usually in the form of solutions into which the teats of an
thickeners. animal are dipped pre- and post-milking, as appropriate, to
Several categories of veterinary liquid preparations for reduce the population of pathogenic micro-organisms on the
cutaneous application may be distinguished: surfaces. Teat dips may be supplied/presented as ready-to-use
-- cutaneous foams (see Liquid preparations for cutaneous preparations 01' they may be prepared by dilution of teat dip
application (0927)); concentrates. Pre- and post-milking teat dips often differ in
-- dip concentrates; formulation. Teat dips usually contain emollients to promote
-- pour-on preparations; skin hydration, to soften the skin and allow healing of lesions
-- shampoos (see Liquid preparations for cutaneous that would otherwise harbour bacteria.
application (0927));
-- spot-on preparations; TEAT SPRAYS
-- sprays; DEFINITION
-- teat dips; Teat sprays contain one 01' more disinfectant active
-- teat sprays; substances, usually in the form of solutions which are sprayed
-- udder-washes. onto the teats of an animal pre- and post-milking, as
appropriate, to reduce the population of pathogenic micro-
DIP CONCENTRATES organisms on the surfaces. Teat sprays may be
supplied/presented as ready-to-use preparations 01' they may
DEFINITION be prepared by dilution of teat spray concentrates. Pre- and
Dip concentrates are preparationscontaining one 01' more post-milking sprays often differ in formulation. Teat sprays
active substances, usually in the form of wettable powders, usually contain emollients to promote skin hydration, to
pastes, solutions 01' suspensions, which are used to prepare soften the skin and allow healing of lesions that would
diluted solutions, suspensions 01' emulsions of active otherwise harbour bacteria.
substances. The diluted preparations are applied by complete
irnmersion of the animal. UDDER-WASHES
DEFINITION
POUR-ON PREPARATIONS U dder-washes contain one 01' more disinfectant active
substances, usually in the form of solutions which are sprayed
DEFINITION onto the udder and teats of an animal to remove mud and
Pour-on preparations contain one 01' more active substances faecal contamination before the application of teat dips or
for the prevention and treatment of ectoparasitic and/or sprays. Udder-washes are usually prepared by the dilution
endoparasitic infestations of animals. They are applied either of concentrated preparations 01' of ready-to-use teat
in volumes which are usually greater than 5 mL by pouring dips 01' teat sprays.
along the animal's dorsal midline. _____________________________________________ PhE~
2016 General Monographs Vet-131

Liquid Preparations tor Cutaneous INTRAMAMMARY INFUSIONS *****


* *
Application ot the British Pharmacopoeia intramammaty injections *****
(Veterinary) (IlltramaI1Ulla1y Preparations for Vetelina1Y Use,
Ph Eur monograph 0945)
In addition to the above requirenzents of the European
PhE~ _____________________________________________
Pharmacopoeia, the follozuing statenzents apply to any dip
concentrate that is the subject of an individualnzonograph in the DEFINITION
British Pharmacopoeia (VeterinaJ'Y). Intramammary preparations for veterinmy use are sterile
preparations intended for introduction into the mammary
DIP CONCENTRATES gland via the teat canal. There are two main categories: those
intended for administration to lactating animals, and those
DEFINITION intended for administration to animals at the end of lactation
When diluted, Dip Concentrates are used for the prevention or to non-Iactating animals for the treatment or prevention of
and treatment of ectoparasitic infestations of animals. infection.
The diluted preparations are applied by complete immersion
Intramammary preparations for veterinary use are solutions,
of the animal or, where appropriate, by spraying.
emulsions 01' suspensions or semi-solid preparations
containing one 01' more active substances in a suitable
vehic1e. They may contain excipients such as stabilising,
emulsifying, suspending and thickening agents. Suspensions
EXTRACTS may show a sediment which is readily dispersed on shaking.
Emulsions may show evidence of phase separation but are
(Ph Eur monograph 0765)
readily redispersed on shaking.
Extracts comply zuith the requirements of the European
Unless otherwise justified and authorised, intramammary
Pha1'lnacopoeia. These requirements are reproduced in the Blitish
preparations for veterinary use are supplied in containers for
Pharmacopoeia.
use on one occasion only for introduction in a single teat
canal of an animal.
If supplied in multidose containers, aqueous preparations
contain a suitable antimicrobial preservative at a suitable
EYE PREPARATIONS concentration, except where dle preparation itself has
(Ph Eur 11Z0nograph 1163) adequate antimicrobial properties. Precautions for
administration and for storage between administrations must
Eye Preparations comply zuith the requirements of the European
be taken.
Phamzacopoeia. These require11lents are reproduced in the British
Pharl1lacopoeia. Where applicable, containers for intramammary preparations
for veterinary use comply widl the requirements of Materials
L/sed for the manufacture of containers (3.1 and subsections) and
Contaillers (3.2 and subsections).
PRODUCTION
Eye Preparations of the British During the development of a intramammary preparation for
Pharmacopoeia (Veterinary) veterinary use, the formulation for which contains an
In addition to the requirements of the European Pharmacopoeia, antimicrobial preservative, the effectiveness of the chosen
the statements applicable to Eye Preparatio71s of the Blitish preservative shall be demonstrated to the satisfaction of the
Pharmacopoeia apply to those eye ointments that are the subject of competent authority. A suitable test method together with
an individualnzonograph in the B17'tish Phqzrmacopoeia criteria for judging the preservative properties of the
(Veterina1J¡) . formulation are provided in the text on Efficacy of
antimicrobial preservation (5. 1.3) .
Intramammary preparations for veterinary use are prepared
using materials and methods designed to ensure sterility and
to avoid the introduction of contaminants and the growth of
GRANULES micro-organisms; recommendations on this aspect are
(Ph Eur monograph 0499) provided in the text on Methods of preparation of ste1ile products
Unless otherzuise justified and auth01ised, Granules comply zuith (5.1.1).
the approp¡iate requirements of the European Pharmacopoeia. In the manufacture of intramammary preparations for
These requirements are reproduced in the British Pharmacopoeia. veterinary use containing dispersed particles, measures are
taken to ensure a suitable and controlled particle size with
regard to the intended use.
TESTS
Deliverable mass or volume
Squeeze out as much as possible of the contents of ten
containers according to the instructions on the label.
The mean mas s or volume does not differ by more than
10 per cent from the nominal mas s 01' volume.
Vet-132 General Monographs 2016

Sterility (2.6.1) batch, with a minimum of three and a maximum of ten is


Intramammary preparations for veterinary use comply with considered a suitable number assuming that the preparation
the test for sterility; use the technique of membrane filtration has been manufactured under appropriately validated
or, in justified cases, direct inoculation of the culture media. conditions designed to exc1ude contamination.
Squeeze out the contents of ten containers and mix
thoroughly. For each medium, use 0.5 g to 1 g (or 0.5 mL to
1 mL as appropriate) taken from the mixed sample.
STORAGE INTRARUMINAl DEVICES ***
Store in a sterile, airtight, tamper-proof container. ** **
(Ph. Eur. monograph 1228) *****
LABELLING
The label sta tes: Intranuminal Devises comply with the requirements of the
-- the name of the active substance(s) and the mas s or European Pharmacopoeia. These requirements are reproduced
number of Intemational Units of the active substance(s) below.
_____________________________________________
that may be delivered from the container using normal ~Ew

technique; The requirements of this monograph do not apply to preparation


-- whether the preparation is intended for use in a lactating (sometimes known as boluses) J such as large conventional tabletsJ
animal or a non-lactating animal; capsules 01' moulded dosage fOl"Jns which give immediate or
-- in the case of multidose containers, the name of any prolonged 1'elease of the active substance(s). Such preparations
added antimicrobial preservative. comply with the relevant parts of the monographs on Capsules
_____________________________________________ PhEw (0016) 01" Tablets (0478)
DEFINITION
Intraruminal devices are solid preparations each containing
one or more active substances. They are intended for oral
Intramammary Infusions of the British administration to ruminant animals and are designed to be
retained in the rumen to deliver the active substance(s) in a
Pharmacopoeia (Veterinary) continuous or pulsatile manner. The period of release of the
In addition to the above requirements of the European active substance(s) may Va1y from days to weeks according to
PharmacopoeiaJ the following statements apply to those the nature of the formulation and/or the delivery device.
intramam111aJY infusions that are the subject of an individual Intraruminal devices may be administered using a balling
1110nograph in the Brúish Pharmacopoeia (VeterinaJY). gun. Sorne intraruminal devices are intended to float on the
DEFINITION surface of the ruminal fluid while others are intended to
IntramammalY Infusions intended for administration to remain on the floor of the rumen or reticulum. Each device
lactating animals are described as Intramamma1Y Infusions has a density appropriate for its intended purpose.
(Lactating Cow) and those intended for administration to PRODUCTION
animal s at the end of lactation or during the non-lactating For continuous release, the intraruminal device is designed to
period for the prevention or treatment of infections during release the active substance(s) at a defined rate over a
the dry period are described as Intramammary Infusions (Dry defined period of time. This may be achieved by erosion,
Cow). corrosion, diffusion, osmotic pressure or any other suitable
PRODUCTION chemical, physical or physico-chemical means.
Intramammary Infusions are prepared by dissolving or For pulsatile-release, the intraruminal device is designed to
suspending the sterile medicaments in the sterilised vehic1e release a specific quantity of active substance(s) at one or
using aseptic technique, unless a process of terminal several defined intelmediate times. This may be achieved by
sterilisation is employed. corrosion by ruminal fluids of the metallic elements of the
When Intramammary Infusions are supplied in single-dos e intraruminal device which leads to sequential releas e of the
containers, these are sealed so as to exc1ude micro-organisms constituent units which are usually in the form of tablets.
and are fitted with a smooth, tapered nozzle to facilitate the In the manufacture of intraruminal devices, measures are
introduction of the infusion into the teat canal. taken to ensure an appropriate release of the active
The containers are sterilised before being filled aseptically substance(s) .
unless the intramarnmary infusion is to be subjected to a In the manufacture, packaging, storage and distribution of
process of terminal sterilisation. intraruminal devices, suitable measures are taken to ensure
Sterility their microbial quality; recommendations on this aspect are
Guidance to manufacturers on the number of containers to provided in the text on 5.1.4. Micl"obiological quality of non-
be tested is provided in the Annex to this monograph. sterile pharmaceutical preparations and substances for
pha17naceutical use.
ANNEX TESTS
Uniformity of dosage units
Guidance to manufacturers in performing the test for
Constituent tablet units of intraruminal devices comply with
sterility
the test for uniformity of dosage units (2.9.40) or, where
In determining the number of containers to be tested, the
justified and áuthorised, with the tests for uniformity of
manufacturer should have regard to the environmental
content and/or unifonnity of mas s shown below. Herbal
conditions of manufacture, the quantity (volume) of
drugs and herbal drug preparations present in the dosage
preparation per container and any other special
form are not subject to the provisions of this paragraph.
considerations applying to the preparation concemed. With
respect to intramarnmary infusions, 1% of the containers in a
2016 General Monographs Vet-133

Uniformity of content (2.9.6) are taken to ensure dleir microbial quality; recommendations
Unless otherwise justified and authorised, constituent on this aspect are provided in the text on 5.1.4.
tablet units of intraruminal devices in which the active Microbiological quality of non-sterile pharmaceutical preparations
substances are present at levels less than 2 mg 01" less than and substances for pharmaceutical use, see Table 5.1.4.-1. -
2 per cent of the total mas s comply with test A for Cutaneous use.
uniformity of content of single-dos e preparations. If the Sterile intrauterine preparations for veterinary use are
preparation contains more than one active substance, the prepared using materials and methods designed to ensure
requirement applies only to those substances which sterility and to avoid the inu'oduction of contaminants and
correspond to the aboye conditions. the growth of microorganisms; recommendations on this
Uniformity ofmass (2.9.5) aspect are provided in the text on Methods of preparation of
Unless otherwise justified and authorised, the constituent stetile products (5.1.1).
tablet units of intraruminal devices comply with the test for During development, it must be demonstrated that the
uniformity of mass. If the test for uniformity of content is nominal content can be withdrawn from the container of
prescribed for all active substances, the test for uniformity of liquid and semi-solid intrauterine preparations for veterinary
mass is not required. use presented in single-dose containers.
LABELLING TESTS
The label states: Uniformity of dosage units
-- for continuous-release devices, the dos e released per unit Single-dose intrauterine preparations for veterinary use
time; comply with the test for uniformity of dosage units (2.9.40)
-- for pulsatile-release devices, the dose released at 01", where justified and authorised, with the tests for
specified times. uniformity of content and/or uniformity of mass shown
_____________________________________________ PhEw below. Herbal drugs and herbal drug preparations present in
the dosage form are not subject to the provisions of this
paragraph.
Uniformity of content (2.9.6)
Unless otherwise prescribed 01" justified and authorised, solid
INTRAUTERINE PREPARATIONS ***
*** *** single-dose preparations with a content of active substance
less than 2 mg 01" less than 2 per cent of the total mass
(Intrauterine Preparations for Veterina/J! Use, *** comply with test A (inu'auterine tablets) 01" test B
Ph Eur 1110nograph 1806)
(intrauterine capsules) for uniformity of content of single-
Intrauterine Preparatio1lS comply zuith the requirements of the dos e preparations. If the preparation has more than 1 active
European Pha171zacopoeia. These requirements are reproduced substance, dle requirement applies only to those substances
belozu. which correspond to the aboye conditions.
~Ew _____________________________________________
Uniformity ofmass (2.9.5)
DEFINITION Solid single-dose inu'auterine preparations for veterinary use
Intrauterine preparations for veterinary use are liquid, semi- comply with the test for uniformity of mas s of single-dose
solid 01" solid preparations intended for the direct preparations. If rile test for unifOlmity of content is
adminisu'ation to the uterus (cervix, cavity or fundus), prescribed 01" justified and authorised for all the active
usually in order to obtain a local effect. They contain 1 or substances, the test for uniformity of mass is not required.
more active substances in a suitable basis. Dissolution
Where appropriate, containers for intrauterine preparations A suitable test may be carried out to demonstrate the
for veterinary use comply with the requirements for Materials appropriate release of the active substance(s) from solid
used fol' the manufacture of comainers (3.1 and subsections) and single-dose inu'auterine preparations for veterinary use, for
Containers (3.2 and subsections). example one of rile tests described in Dissolution test for salid
Several categories of intrauterine preparations for veterinary dosage forms (2.9.3).
use may be distinguished: When a dissolution test is prescribed, a disintegration test
-- intrauterine tablets; may not be required.
-- intrauterine capsules; Sterility (2.6.1)
-- intrauterine solutions, emulsions and suspensions, Sterile inu'auterine preparations for veterinary use comply
concentrates for intrauterine solutions; with the test for sterility. Applicators supplied with the
-- tablets for inu'auterine solutions and suspensions; preparation also comply with the test for sterility. Remove
-- semi-solid intrauterine preparations; the applicator with aseptic precautions from its package and
-- intrauterine foams; transfer it to a tube of culture medium so that it is
-- intrauterine sticks. completely immersed. Incubate and interpret the results as
PRODUCTION described in the test for sterility.
During the development of an intrauterine preparation for LABELLING
veterinary use, the effectiveness of any added antimicrobial The label states:
preservative shall be demonstrated to the satisfaction of the -- the name of any added antimicrobial preservative;
competent authority. A suitable test method together with -- where applicable, that the preparation is sterile.
criteria for judging the preservative properties of the
formulation are provided under Efficacy of antimicrobial
preservation (5.1.3).
In the manufacture, packaging, storage and distribution of
intrauterine preparations for veterinary use, suitable mea sures
Vet-134 General Monographs 2016

INTRAUTERINE TABLETS TABLETS FOR INTRAUTERINE


DEFINITION SOLUTIONS AND SUSPENSIONS
Intrauterine tablets are solid preparations each containing a DEFINITION
single dose of 1 or more active substances. They generally Tablets intended for the preparation of intrauterine solutions
conform to the definition given in the monograph on and suspensions are single-dose preparations which are
Tablets (0478). dissolved or dispersed in water at the time of administration.
A suitable applicator may be used for application into the They may contain excipients to facilitate dissolution or
uterus. dispersion or to prevent caking.
TESTS Tablets for intrauterine solutions or suspensions confonn
Disintegration with the definition given in the monograph on Tablets (0478).
Unless intended for prolonged local action, they comply with After dissolution or dispersion, they comply with the
the test for disintegration of suppositories and pessaties requirements for intrauterine solutions or intrauterine
(2.9.2). Examine the state of the tablets after 30 min, unless suspensions, as appropriate.
otherwise justified and authorised. TESTS
Disintegration (2.9.1)
INTRAUTERINE CAPSULES Tablets for intrauterine solutions or suspensions disintegrate
within 3 min using zuater R at 15-25 oc.
DEFINITION
Intrauterine capsules are solid, single-dos e preparations. They LABELLING
are generally similar to soft capsules, differing only in their The label states:
shape and size. Intrauterine capsules have various shapes, - the method of preparation of the intrauterine solution or
usually ovoid. They are smooth and have a uniform external suspension;
appearance. - the conditions and duration of storage of the solution or
A suitable applicator may be used for application into the suspension after reconstitution.
uterus.
TESTS SEMI-SOLID INTRAUTERINE
Disintegration PREPARATIONS
Unless intended for prolonged local action, they comply with DEFINITION
the test for disintegration of suppositOlies and pessaries Semi-solid preparations for intrauterine use are ointments,
(2.9.2). Examine the state of the capsules after 30 min, creams or gels.
unless otherwise justified and authOlised. Semi-solid preparations for intrauterine use comply with the
requirements of tlle monograph on Semi-solid preparatiolls for
INTRAUTERINE SOLUTIONS, cutaneOllS applicatio71 (0132).
SUSPENSIONS AND EMULSIONS They are often supplied in single-dose containers.
CONCENTRATESFORINTRAUTERUNE The container is adapted to deliver the preparation to the
SOLUTIONS uterus or it may be accompanied by a suitable applicator.

DEFINITION
Intrauterine solutions, suspensions and emulsions are liquid
INTRAUTERINE FOAMS
preparations. Concentrates for intrauterine solutions are DEFINITION
intended for administration after dilution. Int:rauterine foams comply with the requirements of the
They may contain excipients, for example to adjust the monograph on Medicated foams (1105).
viscosity of the preparation, to adjust or stabilise the pH, to They are supplied in multidose containers. The container is
increase the solubility of the active substance(s) or to stabilise adapted to deliver the preparation to the uterus or it may be
the preparation. The excipients do not adversely affect the accompanied by a suitable applicator.
intended medical action, or, at the concentrations used,
cause undue local ilTitation.
INTRAUTERINE STICKS
Intrauterine emulsions may show evidence of phase
separation, but are readily redispersed on shaking. DEFINITION
Intrauterine suspensions may show a sediment that is readily Intrauterine sticks comply with the requirements of the
dispersed on shaking to give a suspension which remains monograph on Sticks (1154). They often produce a foam
sufficiently stable to enable a homogeneous preparation to be when coming into contact with physiological fluids.
delivered. ___________________________________________ PhE~

They may be supplied in single-dose containers.


The container is adapted to deliver the preparation to the
uterus or it may be accompanied by a suitable applicator.
PRODUCTION ORAL LIQUIDS
In the manufacture of intrauterine suspensions, mea sures are LIQUID PREPARATIONS FOR ORAL USE
taken to ensure a suitable and controlled particle size with (Ph Bur monograph 0672)
regard to the intended use.
Unless otherzuise justified and authorised) Oral Liquids comply
zuith the appropriate requirements of the Buropean Pharmacopoeia
m0110graph for Liquid Preparatiolls for Oral Use. These
requirements are reproduced in the British Pharmacopoeia.
2016 General Monographs Vet-135

Several categories of parenteral preparations may be


Oral Liquids of the British distinguished:
Pharmacopoeia (Veterinary) - injections;
In addition to the requirements of the European Pharmacopoeia) - infusions;
the statements applicable to Oral Liquids of the British - concentrates for injections 01' infusions;
Phar'l12acopoeia apply to any oral solution 01' oral suspension that - powders for injections or infusions;
is the subject of an individual monograph in the British - gel s for injections;
Pharmacopoeia (Veterina1Y). - implants.
PRODUCTION
During the development of a parenteral preparation, the
formulation for which contains an antimicrobial preservative,
PARENTERAL PREPARATIONS *** the effectiveness of the chosen preservative shall be
** ** demonstrated to the satisfaction of the competent authority.
(Ph. Eur. monograph 0520) ***** A suitable test method together with criteria for judging the
Unless otherwise justified and authorised) Parenteral Preparations preservative properties of the formulation are provided under
comply with the appropriate requirements of the European Efficacy of antimicrobial preservation (5.1.3).
Pharmacopoeia. These requirements are reproduced in the British Parenteral preparations are prepared using materials and
Pharmacopoeia 01') where they apply to veterinary preparations methods designed to ensure sterility and to avoid the
only) below. introduction of contaminants and the growth of micro-
Ph Eur _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ __ organisms. Recornmendations on this aspect are provided in
the text Methods of preparation of ster'i!e products (5.1.1).
The requirements of this monograph do not necessarily apply to
Water used in the manufacture of parenteral preparations
products derived fro111 human blood) to im11lunological
complies with the requirements of water for injections in bulk
preparations) 01' radiopharmaceutical preparations. Special
stated in the monograph Water for injectio11S (0169).
requirements 111ay apply to preparati011s for veterinary use
depending on the species of animal for which the preparation is TESTS
intended. Particulate contamination: sub-visible partic1es (2.9.19)
DEFINITION For preparations for human use, solutions for infusion 01'
solutions for injection comply with the test.
Parenteral preparations are sterile preparations intended for
administration by injection, infusion 01' implantation into the In the case of preparations for subcutaneous 01' intramuscular
human 01' animal body. injection, higher limits may be appropriate.
Radiopharmaceutical preparations are exempt from these
Parenteral preparations may require the use of excipients, for
requirements. Preparations for which the label states that the
example to make the preparation isotonic with respect to
product is to be used with a final filter are exempt from these
blood, to adjust the pH, to increase solubility, to prevent
requirements, providing it has been demonstrated that the
deterioration of the active substances or to provide adequate
filter delivers a solution that complies with the test.
antimicrobial properties, but not to adversely affect the
intended medicinal action of the preparation 01', at the For preparations for veterinary use, when supplied in
concentrations used, to cause toxicity or undue local containers with a nominal content of more than 100 mL and
initation. when the content is equivalent to adose of more than
1.4 mL per kilogram of body mass, solutions for infusion or
Containers for parenteral preparations are made as far as
solutions for injection comply with the test for particulate
possible from materials that are sufficiently transparent to
contamination: sub-visible partic1es.
permit the visual inspection of the contents, except for
implants and in other justified and authorised cases. Sterility (2.6.1)
Where applicable, the containers for parenteral preparations Parenteral preparations comply with the test for sterility.
comply with the requirements for Materials used for the STORAGE
manufacture of containers (3.1 and subsections) and Containers In a sterile, airtight, tamper-proof container.
(3.2 and subsections).
LABELLING
Parenteral preparations intended for chronic use or total
The label states:
parenteral nutrition should have appropriate limits for
- the name and concentration of any added antimicrobial
specific components or elements, taking long-term toxicity
preservative;
into account.
- where applicable, that the solution is to be used in
Parenteral preparations are supplied in glass containers conjunction with a final filter;
(3.2.1) or in other containers such as plastic containers - where applicable, that the preparation is free from
(3.2.2) 3.2.2.1 and 3.2.9) and prefi1led syringes. The tightness bacterial endotoxins or that it is apyrogenic.
of the container is ensured by suitable means. Closures
ensure a good seal, prevent the access of micro-organisms
INJECTIONS
and other contaminants and usually permit the withdrawal of
a part 01' the whole of the contents without removal of the DEFINITION
closure. The plastic material s or elastomers (3.2.9) used to Injections are sterile solutions, emulsions or suspensions.
manufacture the closures are sufficiently firm and elastic to They are prepared by dissolving, emulsifying or suspending
allow the passage of a needle with the least possible shedding the active substance(s) and a!ly added excipients in water, in
of particles. Closures for multidose containers are sufficiently a suitable non-aqueous liquid, that may be non-sterile where
elastic to ensure that the puncture is resealed when the justified, or in a mixture of these vehicles.
needle is withdrawn. Solutions for injection, examined under suitable conditions of
visibility, are clear and practically free from partic1es.
Vet-136 General Monographs 2016

Emulsions for injection do not show any evidence of phase Any prepararían Where the label states that the preparation is
separation. Suspensions for injection may show a sediment free from bacterial endotoxins or apyrogenic, respectively, the
which is readily dispersed on shaking to give a suspension preparation complies with a test for bacterial endotoxins
which remains sufficiently stable to enable the correct dose to (2.6.14) or with the test for pyrogens (2.6.8), respectively.
be withdrawn.
Multidose preparations Multidose aqueous injections contain a INFUSIONS
suitable antimicrobial preservative at an appropriate
concentration except when the preparation itself has adequate DEFINITION
antimicrobial properties. When a preparation for parenteral Infusions are sterile, aqueous solutions or emulsions with
adminisu"ation is presented in a multidose container, the water as the continuous phase. They are usually made
precautions to be taken for its administration and more isotonic with respect to blood. They are principally intended
particular1y for its storage between successive withdrawals are for administration in large volume. Infusions do not contain
given. any added antimicrobial preservative.
Antimicrobial preservatives Aqueous preparations which are Solutions for infusion, examined under suitable conditions of
prepared using aseptic precautions and which cannot be visibility, are c1ear and practically free fram partic1es.
terminally sterilised may contain a suitable antimicrobial Emulsions for infusion do not show any evidence of phase
preservative in an appropriate concentration. separation.
No antimicrobial preservative is added when: PRODUCTION
- the volume to be injected in a single dose exceeds 15 mL, In the manufacture of infusions containing dispersed
unless otherwise justified; partic1es, measures are taken to ensure a suitable and
- the preparation is intended for administration by routes conu"olled partic1e size with regard to the intended use.
where, for medical reasons, an antimicrabial preservative
The volume of the infusion in the container is sufficient to
is not acceptable, such as intracisternally, epidurally,
permit the withdrawal and administration of the
intrathecalIy or by any route giving access to the
nominal dose using a normal technique (2.9.17).
cerebrospinal fluid, or intra- or retro-ocularly.
Such preparations are presented in single-dose containers. TESTS
Bacteria! endotoxins - pyrogens
PRODUCTION They comply with a test for bacterial endotoxins (2.6.14) or,
In the manufacture of injections containing dispersed where justified and authorised, with the test for pyrogens
partic1es, measures are taken to ensure a suitable and (2.6.8). For the latter test inject 10 mL per kilogram ofbody
conu"olled partic1e size with regard to the intended use. mass into each rabbit, unless otherwise justified and
Single-dose preparations The volume of the injection in a authorised.
single-dose container is sufficient to permit the withdrawal
and administration of the nominal dos e using a nonnal
technique (2.9.17).
CONCENTRATES FOR IN}ECTIONS OR
INFUSIONS
TESTS
Uniformity of dosage units DEFINITION
Single-dose suspensions for injection comply with the test for Concentrates for injections or infusions are sterile solutions
uniformity of dosage units (2.9.40) or, where justified and intended for injection or infusion after dilution. They are
authorised, with the test for uniformity of content shown diluted to a prescribed volume with a prescribed liquid before
below. Herbal drugs and herbal drug preparations present in administration. After dilution, they comply with the
the dosage form are not subject to the provisions of this requirements for injections or for infusions.
paragraph. TESTS
Uniformity of content (2.9.6) Bacteria! endotoxins - pyrogens
Unless otherwise prescribed or justified and authorised, They comply with the requirements prescribed for injections
single-dos e suspensions for injection with a content of active or for infusions, after dilution to a suitable volume.
substance less than 2 mg or less than 2 per cent of the total
mass comply with test A for uniformity of content of single- POWDERS FOR IN]ECTIONS OR
dose preparations. If the preparation contains more than one
active substance, the requirement applies only to those
INFUSIONS
substances that correspond to the above conditions. DEFINITION
Bacteria! endotoxins - pyrogens Powders for injections or infusions are solid, sterile
A test for bacterial endotoxins (2.6.14) is carried out or, substances distributed in their final containers and which,
where justified and authorised, the test for pyrogens (2.6.8). when shaken with the prescribed volume of a prescribed
Recommendations on the limits for bacterial endotoxins are sterile liquid rapidly form either c1ear and practically partic1e-
given in general chapter 5.1.10. free solutions or uniform suspensions. After dissolution or
suspension, they comply withthe requirements for injections
Preparatiol1s for human use The preparation complies with a
or for infusions.
test for bacterial endotoxins (2.6.14) or with the test for
pyrogens (2.6.8). Freeze-dried products for parenteral adminisu"ation are
considered as powders for injections or infusions.
Preparations for veterinalY use When the volume to be injected
in a single dose is 15 mL or more and is equivalent to adose PRODUCTION
of 0.2 mL or more per kilogram of body mass, the The uniformity of content and uniformity of mass of freeze-
preparation complies with a test for bacterial endotoxins dried products for parenteral administration are ensured by
(2.6.14) or with the test for pyrogens (2.6.8). the in-process control of the amount of the solution prior to
freeze-drying.
2016 General Monographs Vet-137

TESTS
Uniformity of dosage units
Veterinary Oral Pastes
Powders for injections or infusions comply with the test for Unless otherwise authorised and justified~ VeterinalY Oml Pastes
uniformity of dosage units (2.9.40) or, where justified and comply 'lvith the requirements of the monograph on Oromucosal
authorised, with the tests for uniformity of content anci/or Preparations. In addition~ the following statements apply to
uniformity of mas s shown below. Herbal drugs and herbal Veterinary Oral Pastes that are the subject of an individual
drug preparations present in the dosage form are not subject monograph in the British Pharmacopoeia (VeterinaJY).
to the provisions of this paragraph. DEFINITION
Uniformity of content (2.9.6) Veterinary Oral Pastes are semi-solid preparations containing
Unless otherwise prescribed or justified and authorised, one or more active substances in a suitable vehic1e. They are
powders for injections or infusions with a content of active administered to the oral cavity and are intended to be
substance less than 2 mg or less than 2 per cent of the total swallowed for delivery of active substances to the
mas s, or with a unit mass equal to or less than 40 mg comply gastrointestinal tracto
with test A for uniformity of content of single-dose Veterinary Oral Pastes are presented in multi-dose containers
preparations. If the preparation contains more than one which are designed to allow the accurate dosing of animals
active substance, the requirement applies only to those according to their bodyweight.
substances that correspond to the aboye conditions.
TESTS
Uniformity of mass (2.9.5) Dissolution
Powders for injections or infusions comply with the test for A suitable test may be carried out to demonstrate the
uniformity of massof single-dos e preparations. If the test for appropriate release of the active substance(s), for example,
uniformity of content is prescribed for all the active the test using Apparatus 2 described under dissolution test for
substances, the test for uniformity of mass is not required. tablets and capsules, Appendix XII B 1.
Bacterial endotoxins - pyrogens Uniformity of mass of delivered dos es from multi-dose
They comply with the requirements prescribed for injections containers
or for infusions, after dissolution or suspension in a suitable Veterinary Oral Pastes supplied in multi-dose containers
volume of liquido comply with Appendix XII C2.
LABELLING STORAGE
The label states the instructions for the preparation of Veterinary Oral Pastes should be stored in airtight containers
injections and infusions. if they contain water or other volatile ingredients.

GELS FOR INJECTIONS


DEFINITION
Gels for injections are sterile gel s with a viscosity suitable to TOPICAl POWDERS
guarantee a modified release of the active substance(s) at the POWDERS FOR CUTANEOUS APPLICATION
site of injection. (Ph Eur monograph 1166)
Unless otherwise justified and auth01'ised~ Topical Powders comply
IMPLANTS with the requirements of the European Pharmacopoeia monograph
DEFINITION for Powders for Cutaneous Application. These requirements are
reproduced in the British Pharmacopoeia.
Implants are sterile, solid preparations of a size and shape
suitable for parenteral implantation and release of the active
substance(s) over an extended period of time. Each dose is
provided in a sterile container.
TESTS Topical Powders of the British
A suitable test is carried out to demonstrate the appropriate Pharmacopoeia (Veterinary)
release of the active substance(s).
___________________________________________ PhE~
In additiol1 to the requirements of the European Pharmacopoeia~
the statements applicable to Topical Powders of the British
Phannacopoeia apply to any dusting powder that is the subject of
an individual monogmph in the British Phamwcopoeia
(VeterinalY) .
Parenteral Preparations of the British
Pharmacopoeia (Veterinary)
In addition to the requirements of the European Phannacopoeia~
the statements applicable to Parenteml Prepamtiol1s of the British
Phannacopoeia~ modified as stated below~ apply to those injections
and .intmvenous il1fusions that are the subject of an individual
monograph in the British Phannacopoeia (VeterinalY).
DEFINITION
Parenteral Preparations prepared with an oily vehic1e are
suitable for intramuscular or subcutaneous administration
only and are not given intravenously.
Vet-138 General Monographs 2016

-- in the case of a fermentation product that is not the


VETERINARV ORAL POWDERS subject of a monograph of the European Phmmacopoeia,
Unless otherzuise justijied and authorised) VeterinaJY Oral Pozuders complies with the monograph Products of fermentation
comply zuith the requirements of the European Pharmacopoeia (1468), notably with the section Down-stream processing.
monograph for Oral Pozuders (1165). In addition to these
requirements) reproduced in the British Pharmacopoeia) the TESTS
follozuing statements apply to those veterinaJY oral pozuders that Loss on drying (2.2.32)
are the subject of an individualmonograph in the British Unless otherwise justified and authorised, for premixes
Pharmacopoeia (VeterinaJY). occurring in granulated or powdered form, maximum
15.0 per cent, determined on 3.000 g by drying in an oven at
DEFINITION 105 oC for 2 h.
Veterinary Oral Powders are finely divided powders that
contain one or more active ingredients with or without LABELLING
excipients such as antimicrobial preservatives, dispersing, The label states the instructions for the preparation of the
suspending or wetting agents and, where necessary, medicated feeding stuffs from the premix and the basic feed,
authorised flavouring agents and colouring matter. They are inc1uding information on whether or not the premix can be
intended for oral administration, usually after dilution in the granulated with the feed and the critical parameters
feed or drinking water. Veterinary Oral Powders may be in (e.g. maximum temperature) that may be applied during the
the form of soluble or wettable powders. process.
_____________________________________________ ~E~

STORAGE
Veterinary Oral Powders should be stored in airtight
containers.

Premixes of the British Pharmacopoeia


*** (Veterinary)
Premixes
*** *** DEFINITION
(Premixes for lVledicated Feeding Stuffs for VeterinaJJ! *** Premixes may occur in pdletted formo
Use) Ph Eur monograph 1037)
Premixes comply zuith the requirements of the European
Pharmacopoeia monograph for Premises for lVledicated Feeding
Stuffs for VeterinalY Use. These requirements are reproduced
Veterinary Semi .. solid Preparations *****
belozu. ** **
PhE~ _____________________________________________ for Oral Use ***
(Ph. Eur. monograph 2638)
DEFINITION
PhE~ _____________________________________________
Mixtures of one or more active substances, usually in a
suitable basis or vehic1e, that are prepared to facilitate feeding DEFINITION
the active substances to animals. They are used exc1usively in Vetelinary semi-solid preparations for oral use (usually pastes
the preparation of medicated feeding stuffs. or gels) contain one or more active substances dissolved or
Premixes occur in granulated, powdered, semi-solid or liquid dispersed in a suitable basis. They are administered to the
formo Used as powders or granules, they are free-flowing and oral cavity and are intended to be swallowed for delivery of
homogeneous; any aggregates break apart during normal active substances to the gastrointestinal tracto
handling. U sed in liquid form, they are homogeneous Veterinary semi-solid preparations for oral use may contain
suspensions or solutions that may be obtained from suitable antimicrobial preservatives and other excipients such
thixotropic gel s or structured liquids. The partic1e size and as dispersing, suspending, thickening, emulsifying, buffering,
other properties are such as to ensure uniform distribution of wetting, solubilising, stabilising, flavouring and sweetening
the active substance(s) in the final feed. Unless otherwise agents.
justified and authorised, the instructions for use state that the
Veterinary semi-solid preparations for oral use are usually
concentration of a premix in granulated or powdered form is
supplied in multidose containers such as oral syringes, which
at least 0.5 per cent in the medicated feeding stuff.
are designed to allow the accurate dosing of animal s
PRODUCTION according to their bodyweight.
In the manufacture, packaging, storage and distribution of Where applicable, containers for veterinary semi-solid
premixes for medicated feeding stuffs for veterinary use, preparations for oral use comply with the requirements
suitable measures are taken to ensure their microbial quality; described in Materials used for the manufacture of containers
recommendations on this aspect are provided in general (3.1 and subsections) and Containers (3.2 and subsections).
chapter 5.1.4. Mzáobiological quality of non-sterile
PRODUCTION
pharmaceutical preparations and substances for pharmaceutical
use. During the development of vetelinary semi-solid preparations
for oral use whose formulation contains an antimicrobial
Active substance preservative, the need for and the efficacy of the chosen
Unless already otherwise justified and authorised for existing preservative shall be demonstrated to the satisfaction of the
premixes, an active substance intended for incorporation into competent authority. A suitable test method together with
a medicated premix: criteria for judging the preservative properties of the
-- complies with the requirements of the relevant formulation are provided in general chapter 5.1.3. Efficacy of
monograph of the European Pharmacopoeia; antimicrobial preservaríon.
2016 General Monographs Vet-139

In the manufacture, packaging, storage and distribution of


veterinaty semi-solid preparations for oral use, suitable
measures are taken to ensure their microbiological quality;
recommendations on this are provided in general chapter
5.1.4 ..Microbiological qualz"ty of non-sterzle pharmaceutical
preparations and substances for pharmaceutical use.
TESTS
Dissolution
A suitable test may be carried out to demonstrate the
appropriate release of the active substance(s).
Uniformity of mass of delivered dos es from multidose
contamers (2.9.27)
Unless otherzuise Justified and authorised, the minimum and the
maxi71lu71l labelled doses are tested, zuhere possible using the same
container and dosing device.
Discharge once to waste in arder to prime the system, if so
stated on the label and depending on the type and shape of
the container.
Unless otherwise justified and authorised, the preparation
complies with the test.
STORAGE
If the preparation contains water 01' other volatile ingredients,
store in an airtight container.
LABELLING
The label states:
-- the name of any added antimicrobial preservative;
-- if it is neceSSa1y to prime the dosing device befare
administration.
_____________________________________________ ~E~

(Ph Eur /J/07Zograph 0478)


UJlless otherzuise Justzjied and authorised, Tablets C071lply zuith the
appropriate reqllire17lents of the European Pharmacopoeia. These
require771e7Zts are reproduced in the British Pharmacopoeia.

Tablets of the British Pharmacopoeia


(Veterinary)
In addition to the require11lents of the European Pharmacopoeia,
the statements applicable to Tablets of the British Pharmacopoeia,
nlOdified as stated belozu, apply to those tablets that are the subJect
of an individual monograph in the British Pharmacopoeia
(VeterillalY) .
DEFINITION
Tablets of the British Pharmacopoeia (Veterinary) are usuaHy
solid, right circular cylinders the end sllifaces of which are
fiat 01' convex and the edges of which may be bevelled except
that those that weigh 5 g 01' more are frequently elongated 01'
biconical.
Dismtegration
Apply test A 01' test B, Appendix XII Al, as appropriate.
When using test B, test 6 tablets either by using two basket-
rack assemblies in parallel 01' by repeating tlle procedure.
To pass the test, aH six of the tablets must have
disintegrated.
Vet-140 Acepromazine Preparations 2016

STORAGE
FORMULATED PREPARATIONS: Acepromazine Injection should be protected from light.
SPECIFIC MONOGRAPHS LABELLING
The su"ength is stated as the equivalent amount of
acepromazine in a suitable dose-volume.

Acepromazine Injection
Action and use
Dopamine receptor antagonist; neuroleptic. Acepromazine Tablets
DEFINITION Action and use
Acepromazine Injection is a sterile solution of Acepromazine Dopamine receptor antagonist; neuroleptic.
Maleate in Water for Injections. The pH of the solution is
DEFINITION
adjusted to about 5 by the addition of Sodium Hydroxide.
Acepromazine Tablets contain Acepromazine Maleate.
The i11¡"ection complies with the requirements stated under
The tablets comply with the requirements stated under Tablets and
Parenteral Preparations and with the follO'luing requirements.
with the follO'luing requirements.
Content of acepromazine, C19H22N20S
Content of acepromazine, C19H22N20S
92.5 to 107.5% ofthe stated amount.
92.5 to 107.5 % of the stated amount.
IDENTIFICATION
IDENTIFICATION
A. To a volume containing the equivalent of 20 mg of
A. To a quantity of the powdered tablets containing the
acepromazine add 2 mL of water and 3 mL of 2M sodium
equivalent of 20 mg of acepromazine add 2 mL of water and
hyd1'Oxide, extract with two 5 mL quantities of cyclohexane
3 mL of 2M sodium hydroxide. Extract with two 5 mL
and evaporate to dryness under reduced pressure. The
quantities of cyclohexane and evaporate to dryness under
infrared abs01ption spectrum of the residue, Appendix II A, is
reduced pressure. The infrared abs01ption spectrwn of the
concordant with the reference spectrwll of acepromazine
residue, Appendix II A, is concordant with the reference
(RSV 01).
spectru11Z of acepromazine (RSV 01).
E. Complies with the test for identification of phenotlúazines,
B. Complies with the test for identification of phenothiazines,
Appendix III A, applying to the plate 1 ~lL of each of the
Appendix III A, applying to the plate 1 ~lL of each of the
following solutions. For solution (1) extract a volume
following solutions. For solution (1) extract a quantity of the
containing the equivalent of 20 mg of acepromazine with two
powdered tablets containing dle equivalent of 20 mg of
5 mL quantities of chlo1'Ofor771 and use the combined extracts.
acepromazine with two 5 mL quantities of chloroforl7l.
Solution (2) is a solution of aceproma:::ine maleate BPCRS in
Solution (2) is a solution of acep1'Onzazine maleate BPCRS in
chloroforJIz containing the equivalent of 0.2% w/v of
chlo1'Oform containing the equivalent of 0.2% w/v of
acepromazine.
acepromazine.
C. To 5 mg of the residue obtained in test A add 2 mL of
C. To a quantity of the powdered tablets containing the
sulfuric acid. A yellow colour is produced which becomes
equivalent of 5 mg of acepromazine add 2 mL of sulfuric acid.
deep orange on warming for 2 minutes.
A yellow colour is produced which becomes deep orange on
D. To a volume of the injection containing the equivalent of warming for 2 minutes.
25 mg of acepromazine add 2 mL of 5M sodium hydroxide
D. Dissolve as completely as possible a quantity of the
and shake with three 3 mL quantities of ether. Add 2 mL of
powdered tablets containing the equivalent of 25 mg of
bromine solution to the aqueous solution, warm in a water
acepromazine in a mixture of 3 mL of water and 2 mL of
bath for 10 minutes, heat to boiling and coo!. Add 0.25 mL
5M sodium hyd1'Oxide and shake with three 3 mL quantities of
to a solution of 10 mg of resorcinol in 3 mL of sulfuric acid
ether. Add 2 mL of bromine solution to the aqueous solution,
and heat for 15 minutes in a water bath. A bluish black
warm in a water bath for 10 minutes, heat to boiling and
colour develops.
coo!. Add 0.25 mL to a solution of 10 mg of resorcinol in
TESTS 3 mL of sulfuric acid and heat for 15 minutes on a water
Acidity badl. A bluish black colour develops.
pH, 4.5 to 5.5, Appendix V L.
TESTS
ASSAY Related substances
To a volume containing the equivalent of 40 mg of Comply with the test for related substa12ces in phenothiazines,
acepromazine add 5 mL of 1M sodium hydroxide and extract Appendix III A, but using a mixture of 8 volumes of
with 50 mL quantities of chlo1'Ofomz until the chloroform diethylamine, 17 volumes of buran-2-one and 75 volumes of
extract is colourless. Wash the extracts with the same 10 mL n-hexa12e as the mobile phase and using the following
of water and filter through a plug of absorbent cotton solutions. For solution (1) shake a quantity of the powdered
previously moistened with chlo1'Oform. Evaporate the tablets containing the equivalent of 50 mg of acepromazine
combined extracts to dryness and dissolve the residue in with 10 mL of chlo1'Oform, filter, evaporate to dryness and
15 mL of acetic anhydride. Carry out Method I for 12012- dissolve the re~idue in 5 mL of methanol containing 0.5 % v/v
aqueous titration, Appendix VIII A, using 0.02lv1 perchloric acid of 13.5M ammonia. For solution (2) dilute 1 volume of
VS as titrant and Clystal violet solutio71 as indicator. Each mL solution (1) to 100 volumes with metha120l containing
of 0.02M perchloric acid VS is equivalent to 6.529 mg of 0.5% v/v of 13.5M am1110nia.
C 19 H22NzOS.
2016 Albendazole Preparations Vet-141

ASSAY containing 1.0% w/v of Albendazole and dilute 1 volume of


Weigh and powder 20 tablets. To a quantity of the powder the resulting solution to 2 volumes with solvent A.
containing the equivalent of 60 mg of acepromazine add (2) Dilute 1 volume of solution (1) to 100 volumes with
5 mL of water and extract with tlnee or more 50 mL solvent A.
quantities of chloroform or until tlle chloroform extract is (3) Dissolve 25.0 mg of albendazole BPCRS and 25.0 mg of
colourless. Evaporate to dryness and dissolve the residue in oxibendazole BPCRS in 5 mL of 1% v/v solution of methanolic
15 mL of acetic a17hydride. Carry out Method I for 17on- sulfuric acid and dilute to 50 mL with solvent A.
aqueous titration, Appendix VIII A, using 0.02M perchloric acid
VS as titrant and Clystal violet solution as indicator. Each mL CHROMATOGRAPHIC CONDITIONS
of 0.02M perchloric acid VS is equivalent to 6.529 mg of (a) Use a stainless steel column (25 cm x 4.6 mm) packed
C19H22N20S. with end-capped octadecylsilyl silica gel for chl'Omatography
(5 ~lm) (Waters Symmeu-y is suitable).
LABELLING
The quantity of the active ingredient is stated in terms of the (b) Use gradient elution and tlle mobile phase described
equivalent amount of acepromazine. below.
(c) Use a flow rate of 0.7 mL per minute.
(d) Use an ambient column temperature.
(e) Use a detection wavelengtll of 292 nm.
Albendazole Oral Suspension (f) Inject 20 ~lL of each solution.
MOBILE PHASE
Action and use Mobile phase A O.015M ammonium dihydmgen 011hophosphate.
Benzimidazole antihelminthic.
Mobile phase B methanol.
DEFINITION
Albendazole Oral Suspension is a suspension of Albendazole Time Mobile phase A% Mobile phase B% Comment
in a suitable vehicle.
(Minutes)
The oral suspension complies 'lvith the requirements stated Lindel'
0-3 100 O isocratic
Oral Liquids a17d with the following requirements.
3-5 100---+30 0---+70 linear gradient
Content of albendazole, C12HlSN302S
5-70 30 70 isocratic
90.0 to 110.0% of the stated amount.
70-72 30---+100 70-+0 linear gradient
IDENTIFICATION
72-80 100 O re-equilibration
A. To a volume of oral suspension containing 25 mg of
Albendazole add 50 mL of O.lM sodium hJldmxide and shake
with the aid of ultrasound for 10 minutes. Dilute to 100 mL
with the same solvent, filter through a 0.45-~lm filter and
SYSTEM SUIT ABILITY
dilute 1 volume of this solution to 10 volumes with tlle same
solvento The light absorption, Appendix n B, in the range The test is not valid unless, in tlle chromatogram obtained
240 to 340 nm of the resulting solution exhibits a maximum with solution (3), the resoluúolZ factor between the two
at 308 nm, a minimum at 281 nm and a shoulder at principal peaks is at least 7.0.
269 nm. LIMITS
B. To a volume of oral suspension containing 25 mg of In tlle chromatogram obtained with solution (1):
Albendazole add 50 mL of O.lM hydrochloric acid and shake tlle area of any secondaly peak is not greater than the area of
with the aid of ultrasound for 10 minutes. Dilute to 100 mL tlle principal peak in the chromatogram obtained with
with the same solvent, filter through a 0.45-~lm filter and solution (2) (1 %);
dilute 1 volume of this solution to 10 volumes with the same
the sum of the areas of any secondaJY peaks is not greater than
solvento The light absorption, Appendix n B, in the range
twice tlle area of the principal peak in tlle chromatogram
240 to 340 nm of the resulting solution exhibits a maximum
obtained with solution (2) (2%).
at 292 nm, a minimum at 273 nm and a shoulder at
261 nm. Disregard any peak with an area less than 0.05 times the area
of the principal peak in the chromatogram obtained Witll
C. In the Assay, the chromatogram obtained with solution
solution (2) (0.05%).
(1) shows a peak with the same retention time as tlle
principal peak in the chromatogram obtained with ASSAY
solution (2). Can-y out the method for liquid chromatography,
Appendix nI D, using the following solutions.
TESTS
Acidity (1) Add 70 mL of 1% v/v solution of methanolic sulfuric acid
pH, 4.5 to 5.5, Appendix V L. to a quantity of the oral suspension containing 0.10 g of
Albendazole, stir for ·15 minutes, mix with the aid of
Related substances ultrasound for 10 minutes and add sufficient 1% v/v solution
Can-y out the method for liquid chromatography, of methanolic sulfuric acid to produce 100 mL. Allow to stand
Appendix III D, using the following solutions. and dilute 5 volumes of the clear supernatant to 25 volumes
Solvent A 30% of mobile phase A and 70% of mobile with 1% v/v solution of methe)17olic sulfuric acid.
phase B. (2) 0.020% w/v of albendazole BPCRS in 1% v/v solution of
(1) Dilute a quantity of the oral suspension with 1% v/v methanolic sulfurz'c acid.
solution of methanolic sulful'ic acid to give a solution
Vet-142 Albendazole Preparations 2016

CHROMATOGRAPHIC CONDITIONS Solvent A 30% of mobile phase A and 70% of mobile


The chromatographic conditions described under Related phase B.
substances may be used. (1) Dilute a quantity of the oral suspension with metlzanolic
DETERMINATION OF CONTENT sulfuric acid (1%) to give a solution containing 1.0% w/v of
Albendazole and dilute 1 volume to 2 volumes with solvent
Determine the weight per mL of the oral suspension,
A.
Appendix V G, and calculate the content of C12HlSN302S,
weight in volume, using the declared content of (2) Dilute 1 volume of solution (1) to 100 volumes with
C12HlSN302S in albendazole BPCRS. solvent A.
(3) Dissolve 25.0 mg of albendazole BPCRS and 25.0 mg of
oxibendazole BPCRS in 5 mL of 1% v/v solution of methanolic
sulfuric acid and dilute to 50 mL with solvent A.
CHROMATOGRAPHIC CONDITIONS
Albendazole Oral Suspension with
(a) Use a stainless steel column (25 cm x 4.6 mm) packed
Minerals with end-capped octadecylsilyl silica gel j01' chromatography
(5 ~lm) (Waters Symmetry is suitable).
Action and use
(b) Use gradient elution and the mobile phase described
Benzimidazole antihelminthic.
below.
DEFINITION (c) Use a flow rate of 0.7 mL per minute.
Albendazole Oral Suspension with Minerals is a suspension (d) Use ambient column temperature.
of Albendazole in a suitable vehicle containing cobalt and (e) Use a detection wavelength of 292 nm.
selenium.
(f) Inject 20 ~lL of each solution.
The oral suspension complies with the requirements stated under
MOBILE PHASE
Oral Liquids and with the jollowing requirements.
Mobile phase A 0.015M ammonium dihydrogen o/1hophosphate.
Content of albendazole, C12H1SN30ZS
95.0 to 105.0% of the stated amount. Mobile phase B methanol.
Content of Co
90.0 to 110.0% of the stated amount. Time Mobile phase A% Mobile phase S% Comment

Content of Se (Minutes)
90.0 to 115.0% of the stated amount. 0-3 100 O isocratic

IDENTIFICATION 3-5 100->30 0-,70 linear gradient

A. To a volume of oral suspension containing of 25 mg of 5-70 30 70 isocratic


Albendazole add 50 mL of O.IM sodium hydroxide and shake 70-72 30->100 70-,0 linear gradient
with the aid ofultrasound for 10 minutes. Dilute to 100 mL
72-80 100 O re-equilibration
with the same solvent, filter through a 0.45-~lm filter and
dilute 1 volume of this solution to 10 volumes with the same
solvento The light absol'ption, Appendix II B, in the range
240 to 340 nm of the resulting solution exhibits a maximum SYSTEM SurTABILITY
at 308 nm, a minimum at 281 nm and a shoulder at
269 nm. The test is not valid unless, in the chromatogram obtained
with solution (3), the resolution jactor between the two
B. To a volume of oral suspension containing of 25 mg of principal peaks is at least 7.0.
Albendazole add 50 mL of O.IM hydrochloric acid and shake
with the aid of ultrasound for 10 minutes. Dilute to 100 mL LIMITS
with the same solvent, filter through a 0.45-~lm filter and In the chromatogram obtained with solution (1):
dilute 1 volume of this solution to 10 volumes with the same the area of any secondaJY peak is not greater than the area of
solvento The light absorption, Appendix II B, in the range the principal peak in the cln'omatogram obtained widl
240 to 340 nm of the resulting solution exhibits a maximum solution (2) (1 %);
at 292 nm, a minimum at 273 nm and a shoulder at the sum of the are as of any secondary peaks is not greater than
261 nm. twice the area of the principal peak in the cln'omatogram
C. In the Assay for albendazole, the chromatogram obtained obtained with solution (2) (2%).
with solution (1) shows a peak with the same retention time Disregard any peak with an area less than 0.05 times the area
as the principal peak in the chromatogram obtained with of the principal peak in the cln'omatogram obtained with
solution (2). solution (2) (0.05%).
D. In the Assay for cobalt, the oral suspension absorbs
ASSAY
radiation at 240.7 nm.
For albendazole
E. In the Assay for selenium, the oral suspension absorbs Carry out the method for liquid chromatograplzy,
radiation at 196.0 nm. Appendix III D, using the following solutions.
TESTS (1) Add 70mL of 1% v/v solution of methanolic sulfuric acid
Acidity to a quantity of the oral suspension containing 0.10 g of
pH, 4.5 to 6.0, Appendix V L. Albendazole, stir for 15 minutes, mix with dle aid of
Related substances ultrasound for 10 minutes and add sufficient 1% v/v solution
Carry out the method for liquid chromatography, of methanolic sulfuric acid to produce 100 rnL. Allow to stand
Appendix III D, using the following solutions.
2016 Amitraz Preparations Vet-143

and dilute 5 volumes of the clear supernatant to 25 volumes (1) Dilute the dip concentrate with toluel1e to produce a
with 1% v/v solution of methanolic sulfuric add. solution containing 5.0% w/v of Amitraz.
(2) 0.020% w/v of albendazole BPCRS in 1% v/v solution of (2) Dilute 1 volume of solution (1) to 10 volumes with
methanolic sulfuric add. toluene.
CHROMATOGRAPHIC CONDITIONS (3) 0.5% w/v of amitraz BPCRS in toluene.
The chromatographic conditions described under Related (4) 0.10% w/v of amitl'az BPCRS in toluene.
substances may be used. (5) 0.005% w/v of 2)4-dimethylaniline in toluene.
DETERMINATION OF CONTENT CHROMATOGRAPHIC CONDITIONS
Determine the weight per mL of the oral suspension, (a) Use as the coating silica gel HF254 .
Appendix V G, and calcula te the content of C12HlSN302S, (b) Use the mobile phase as described below.
weight in volume, using the declared content of
(c) Stand the pI ate to a depth of 3.5 cm in a solution
C12HlSN302S in albendazole BPCRS.
prepared by dissolving 35 g of acetamide in 100 mI of
Por cobalt methanol, adding 100 mI of triethylamine and diluting to
Add 5 mL of hydrochloric add to a quantity of the oral 250 mI with methanol before standing the plate in a stream of
suspension containing 2.5 mg of cobalt, heat in a water bath cold air for about 30 seconds. Irnmediately apply separately
for 10 minutes, cool and add sufficient 'water to produce to the plate, at a level 1 cm below the top of the impregnated
100 mL. Mix thorough1y and filter. Carry out the method for zone, 2 ~ll of each solution.
atomic absorption spectrophotometlY, Appendix n D, measuring (d) Develop the plate to 15 cm.
at 240.7 nm and using cobalt standard solution (100 ppm Ca),
(e) After removal of the plate, dry in air and examine under
diluted if necessary with water, to prepare the standard
ultraviolet light (254 11m) (visualisation 1). Expose the plate to
solution. Determine the 'lveight pa mL of the oral suspension,
the vapour of hydroch1oric acid until the coating is
Appendix V G, and calculate the content of Co, weight in
impregnated. Expose to the vapour of nitro gen dioxide
volume.
(prepared by the action of nitric acid on zinc) for
Por selenium 10 minutes, remove any excess nitrogen dioxide with air and
Add 5 mL of nitl1'c add to a quantity of the oral suspension spray with a 0.5% w/v solution ofN-(1-naphthyl)
containing 2.5 mg of selenium, heat in a water bath for ethylenediamine dihydrochlol1'de in methanol (50%)
15 minutes, cool and add sufficient water to produce 50 mL. (visualisation 2).
Mix thoroughly and filter. Carry out the method for atomic
J\lIOBILE PHASE
absorptiol1 spectrophotometly, Appendix n D, measuring at
196.0 nm and using selenium standard solution (100 ppm Se), 2 volumes of triethylamine, 3 volun1es of ethyl acetate and
diluted if necessary with water, to prepare the standard 5 volumes of cyclohexane.
solution. Determine the weight per mL of the oral LIMITS
suspension, Appendix V G, and calculate the content of Se, Using method of visualisation 1, in the chromatogram
weight in volume. obtained with solution (1):
any secondaly spot is not more intense than the spot in the
chromatogram obtained with solution (4) (2%).
Using method of visualisaion 2, in the chromatogram
Amitraz Dip Concentrate (Liquid) obtained with solution (1):
any spot corresponding to 2,4-dimethylaniline is not more
Action and use intense than the spot in the chromatogram obtained with
Topical parasiticide; acaricide. solution (5) (0.1 %)
DEFINITION Water
Amitraz Dip Concentrate (Liquid) contains Amitraz in a Not more than 0.15% w/v, Appendix IX C, Method I A.
suitable emulsifiable vehicle. It may contain a suitable U se 5 mI of the dip concentrate and a mixture of equal
stabilising agent. volumes of chlo1'oform and 2-chloroethanol in place of
anhydrous methanoI.
The dip concentrate complies with the requirements stated under
Veterinmy Liquid Preparations for Cutaneous Application al1d ASSAY
with the following requirements. Carry out the method for gas chromatography,
Content of amitraz, C19H23N3 Appendix nI B. Prepare a 2% v/v solution of squalane
94.0 to 106.0% of the stated amount. (internal standard) in methyl acetate (solution A).
(1) Dissolve a quantity of the dip concentrate containing
IDENTIFICATION
0.15 g of Amitraz in sufficient methyl acetate to produce
A. In the test for Related substances, the principal spot in the
30 mI.
chromatogram obtained with solution (2) corresponds to that
in the chromatogram obtained with solution (3). (2) Dissolve a quantity of the dip concentrate containing
0.15 g of Amitraz in 10 mI of solution A and add sufficient
B. In the Assay, the chromatogram obtained with solution
methyl acetate to produce 30 mI.
(1) shows a peak with the same retention time as the peak
due to amitraz in the chromatogram obtained with (3) Dissolve 0.15 g of amitraz BPCRS in 10 mI of solution A
solution (3). and add sufficient 11lethyl acetate to produce 30 mI.
Related substances CHROMATOGRAPHIC CONDITIONS
Carry out the method for thin-layer chromatography, (a) Use a fused silica capillary column (15 m x 0.53 mm)
Appendix In A, using the following solutions. coated with a film (1.5 !lm) of methyl silicone gum
(Chrompack CP-Sil 5 CB is suitable).
Vet-144 Amoxicillin Preparations 2016

(b) Use helium as the carrier gas at 12 mI per minute. !vIDE/LE PHASE
(c) Use isothermaI conditions maintained at 220°. 10 voIumes of acetone and 90 voIumes of a 15.4% w/v
(d) Use an inlet temperature of 230°. soIution of a111111oniUll1 acetate adjusted to pH 5.0 with glacial
acetic acid.
(e) Use a flame ionisation detector at a temperature of 300°.
SYSTEM SUIT ABILITY
(±) Inject 1 ~lI of each soIution.
The test is not valid unIess the chromatogram obtained with
SYSTEM SUITABILITY
soIution (3) shows two c1early separated spots.
The assay is not vaIid unIess, in the chromatogram obtained
CONFIRMATION
with soIution (3), the resolution factor between the peaks
corresponding to squaIane and amitraz is at Ieast 3.0. The principal spot in the chromatogram obtained with
soIution (1) is similar in position, coIour and size to that in
DETERMINATION OF CONTENT
the chromatogram obtained with soIution (2).
Calculate the content of C19H23N3 in the dip concentrate
using the decIared content of C19H23N3 in amitraz BPCRS. TESTS
Pyrogens
The requirement for Pyrogens does not appIy to Amoxicillin
Oily Injections.
ASSAY
Amoxicillin OUy Injections Carry out the method for liquid chromatography,
Action and use Appendix In D, using the following soIutions.
PenicilIin antibacterial. (1) Shake a quantity of the injection containing the
equivaIent of 60 mg of amoxicillin with 15 mL of petroleum
DEFINITION spirit (boiling range) 120° to 160°)) centrifuge and discard the
Amoxicillin OiIy Injections are steriIe suspensions of supernatant liquido Repeat the extraction twice using a
Amoxicillin Trihydrate in oiIy vehic1es appropriate to their further 15 mL of petroleu111 spi1it (boiling range) 120° to 160°)
intended use. Injections intended for use as long acting each time. DissoIve the residue in 20 mL of ether, centrifuge,
preparations are described as Amoxicillin OiIy Injection discard the supematant Iiquid and allow the residue
(Long Acting). remaining to dry in air until the soIvents have evaporated.
The injections comply zuith the require111ents stated under DissoIve the dried residue in mobiIe phase A, add sufficient
Parenteral Preparations and zuith the follozuing require111ents. mobiIe phase A to produce 100 mL, mix and filter
(Whatman GF/C filter paper is suitabIe).
Content of amoxicillin, C16H19N30SS
90.0 to 105.0% of the stated amount. (2) 0.070% w/v of amoxicillin tlihydrate BPCRS in mobile
phase A.
CHARACTERISTICS
(3) 0.0004% w/v of cefadroxil BPCRS and 0.003% w/v of
A white or aImost white, oiIy suspension.
amoxicillin nihydrate BPCRS in mobiIe phase A.
IDENTIFICATION CHROMATOGRAPHIC CONDITIONS
Extract a quantity containing the equivaIent of 0.25 g of
(a) Use a stainIess steeI coIumn (25 cm x 4.6 mm) packed
amoxicillin with three 20-mL quantities of petroleu11l spirit
with octadecylsilyl silica gel for chromatography (5 ~lm) (Hypersil
(boiling range) 1200 to 160°) and discard the extracts. Wash
5 ODS is suitabIe).
the residue with ether and dry in a current of airo The residue
complies with the following tests. (b) Use isocratic eIution and the mobile phase described
beIow.
A. The infrared absmption spectru111, Appendix n A, is
concordant with the reference spectrwn of amoxicillin (c) Use a flow rate of 1 mL per minute.
trihydrate (RSV 05). (d) Use an ambient coIumn temperature.
B. Carry out the method for thin-Iayer chromatography, (e) Use a detection waveIength of 254 nm.
Appendix In A, using the following soIutions. (±) Inject 50 ~lL of each soIution.
(1) DissoIve a quantity of the residue in sufficient sodium !vIDE/LE PHASE
hydrogen carbonate solution to produce a soIution containing
8 voIumes of mobile phase B and 92 voIumes of mobile
the equivaIent of 0.25% w/v of amoxicillin.
phase A.
(2) 0.25% w/v of amoxicillin t1"l'hydrate BPCRS in sodiu111
Mobile phase A 1 voIume of acetonitlile and 99 voIumes of a
hydrogen carbonate solution.
25% v/v soIution of 0.2M potassium dihydrogen orthophosphate
(3) 0.25% w/v of each of amoxicillin tlihydrate BPCRS and adjusted to pH 5.0 with 2M sodium hydroxide.
ampicillin nihydrate BPCRS in sodium hydrogen carbonate
Mobile phase B 20 voIumes of acetonitrile and 80 voIumes of
solution.
a 25% v/v soIution of 0.2M potassiu111 dihydrogen orthophosphate
CHROMATOGRAPHIC CONDITIONS adjusted to pH 5.0 with 2M sodiu111 hydroxide.
(a) Use a TLC silica gel silanised plate (Merck silanised silica SYSTEM SUITABILITY
gel 60 F 254s (RP-18) pIates are suitabIe).
The Assay is not val id unIess, in the chromatogram obtained
(b) Use the mobile phase as described beIow. with soIution (3)., the resolution factor between the peaks due
(c) AppIy 1 ~lL of each soIution. to amoxicillin and cefadroxiI is at Ieast 2.0. If necessary,
(d) Develop the pIate to 15 cm. adjust the composition of the mobile phase to achieve the
(e) After removaI of the pIate allow it to dry in air, expose to required resoIution.
iodine vapour untiI spots appear and examine in daylight.
2016 Amoxicillin Preparations Vet-145

DETERMINATION OF CONTENT water and add 2 mL of a mixture of 2 mL of cupri-ta1"ta1ic


Calculate the content of C16H19N30SS in the injection from solution Rl and 6 mL of water. A magenta colour is produced
the chromatograms obtained and from the declared content irnmediately.
of C16H19N30SS in amoxicillin trihydrate BPCRS. C. Dissolve 0.1 mL of aniline in a mixture of 1 mL of
LABELLING hydrochlO1ic acid and 3 mL of water. Coolthe solution in ice
and add 1 mL of a freshly prepared 20% w/v solution of
The labe1 states (1) the quantity of active ingredient in terms
of the equivalent amount of amoxicillin in a suitable dose-
sodium nitrite. Add the resulting mixture drop wise to a cold
solution of 0.1 g of the residue obtained in test B in 2 mL of
volume; (2) where appropriate, that the preparation is
Amoxicillin Oily Injection (Long Acting). 5M sodium hydroxide. The solution becomes deep cherry-red
and a copious dark broW11 precipitate is produced.
ASSAY
Carry out the method for liquid chromatography,
Appendix III D, using the following solutions.
Amoxicillin Veterinary Oral Powder (1) Add 80 mL of mobile phase A to a quantity of the
Action and use veterinary oral powder containing the equivalent of 60 mg of
Penicillin antibacterial. amoxicillin and shake for 15 minutes. Mix with the aid of
ultrasound for 1 minute, add sufficient mobile phase A to
DEl'INITION produce 100 mL, mix and filter (Whatman GF/C filter paper
Amoxicillin Veterinary Oral Powder is a mixture of is suitable).
Amoxicillin Trihydrate, Lactose or other suitable diluent and (2) 0.070% w/v of amoxicillin trihydrate BPCRS in mobile
a stabilising agent. phase A.
The veterinary oral powder complies with the requirements stated (3) 0.0004% w/v of cefadroxil BPCRS and 0.003% w/v of
under Veterinary Oral Powders and with the following amoxicillin tlihydrate BPCRS in mobile phase A.
requirements. CHROMATOGRAPHIC CONDITIONS
Content of amoxicillin, C16H19N30SS (a) Use a stainless steel column (25 cm x 4.6 mm) packed
90.0 to 110.0% of the stated amount. with octadecylsilyl silica gel for chromatography (5 ~lm) (Hypersil
IDENTIFICATION 5 ODS is suitable).
A. Carry out the method for thin-Iayer chromatography, (b) U se isocratic e1ution and the mobile phase described
Appendix III A, using the following solutions. below.
(1) Dissolve a quantity of the veterinary oral powder (c) Use a flow rate of 1 mL per minute.
containing the equivalent of 0.25 g of amoxicillin in sufficient (d) U se an ambient column temperature.
sodium hydrogen carbonate solution to produce 100 mL. (e) Use a detection wavelength of 254 nm.
(2) 0.25% w/v of amoxicillin trihydrate BPCRS in sodiu17l (f) Inject 50 ~lL of each solution.
hydrogen carbonate solution.
JliIOBILE PHASE
(3) 0.25% w/v of each of amoxicillin trihydrate BPCRS and
ampicillin trihydrate BPCRS in sodiuJ1Z hydrogen carbonate 8 volumes of mobile phase B and 92 volumes of mobile
solution. phase A.
Mobile phase A 1 volume of acetonitrile and 99 volumes of a
CHROMATOGRAPHIC CONDITIONS
25% v/v solution of 0.2M potassium dihydrogen orthophosphate
(a) Use a TLC silica gel silanised plate (Merck silanised silica adjusted to pH 5.0 with 2M sodium hydroxide.
gel 60 F ZS4s (RP-18) plates are suitable).
Mobile phase B 20 volumes of acetonitJile and 80 volumes of
(b) Use the mobile phase as described below. a 25% v/v solution of 0.2M potassium dihydrogen orthophosphate
(c) Apply 1 ~lL of each solution. adjusted to pH 5.0 with 2M sodium hydroxide.
(d) Develop the plate to 15 cm. SYSTEM SUIT ABILITY
(e) After removal of the plate allow it to dry in air, expose it The Assay is not valid unless, in the chromatogram obtained
to iodine vapour until spots appear and examine in daylight. with solution (3), the resolution factor between the peaks due
MOBILE PHASE to amoxicillin and cefadroxil is at least 2.0. If necessary,
10 volumes of acetone and 90 volumes of a 15.4% w/v adjust the compositionof the mobile phase to achieve the
solution of a11lmonium acetate adjusted to pH 5.0 with glacial required resolution.
acetic acid. DETERMINATION OF CONTENT
SYSTEM SUIT ABILITY Calculate the content of C16H19N30SS in the veterinary oral
The test is not valid unless the chromatogram obtained with powder from the chromatograms obtained and from the
solution (3) shows two clearIy separated spots. declared content of C16H19N30SS in a7110xicillin
tlihydrate BPCRS.
CONFIRMATION
The principal spot in the chromatogram obtained with LABELLING
solution (1) is similar in position, colour and size to that in The quantity of active ingredient is stated in terms of the
the chromatogram obtained with solution (2). equivalent amount of amoxicillin.
B. Shake a quantity of the veterinary oral powder containing
the equivalent of 0.5 g of amoxicillin with 5 mL of water for
5 minutes, filter, wash the residue first with absolute ethanol
and then with ether and dry at a pressure not exceeding
0.7 kPa for 1 hour. Suspend 10 mg of the residue in 1 mL of
Vet-146 Amoxicillin Preparations 2016

(1) Add 80 mL of mobile phase A to a quantity of the


Amoxicillin Tablets powdered tablets containing the equivalent of 60 mg of
Action and use amoxicillin and shake for 15 minutes. Mix with the aid of
Penicillin antibacterial. ultrasound for 1 minute, add sufficient mobile phase A to
produce 100 mL, mix and filter (Whatman GF/C filter paper
DEFINITION is suitable).
Amoxicillin Tablets contain Amoxicillin Trihydrate. (2) 0.070% w/v of amoxicillin tl1hydrate BPCRS in mobile
The tablets comply 'With the requirements stated under Tablets and phase A.
'With the follO'luing requirements. (3) 0.0004% w/v of cefadroxil BPCRS and 0.003% w/v of
Content of amoxicillin, C16Hl~305S
amoxicillin tlihydrate BPCRS in mobile phase A.
90.0 to 110.0% of the stated amount. CHROMATOGRAPHIC CONDITIONS

IDENTIFICATION (a) Use a stainless steel column (25 cm x 4.6 mm) packed
A. Carry out the method for thin-Iayer chromatography, with octadecylsilyl silica gel for chromatography (5 ~lm) (Hypersil
Appendix III A, using the following solutions. 5 ODS is suitable).
(1) Dissolve a quantity of the powdered tablets containing (b ) Use isocratic elution and the mobile phase described
the equivalent of 0.25 g of amoxicillin in sufficient sodium below.
hydrogen carbonate solution to produce 100 mL. (c) Use a fiow rate of 1 mL per minute.
(2) 0.25% w/v of amoxiczllin trihydrate BPCRS in sodium (d) Use an ambient column temperature.
hydrogen carbonate solution. (e) Use a detection wavelength of 254 nm.
(3) 0.25% w/v of each of amoxicillin trihydrate BPCRS and (f) Inject 50 ~lL of each solution.
ampicillin trihydrate BPCRS in sodium hydrogen carbonate
MOBILE PHASE
solution.
8 volumes of mobile phase B and 92 volumes of mobile
CHROMATOGRAPHIC CONDITIONS
phase A.
(a) Use a TLC silica gel silanised plate (Merck silanised silica Mobile phase A 1 volume of acetonitrile and 99 volumes of a
gel 60 F 2S4s (RP-18) plates are suitable). 25% v/v solution of 0.2M potassium dihydrogen orthophosphate
(b) Use the mobile phase as described below. adjusted to pH 5.0 with 2M sodium hydroxide.
(c) Apply 1 ~lL of each solution. lvlobile phase B 20 volumes of acetonitrile and 80 volumes of
(d) Develop the plate to 15 cm. a 25% v/v solution of 0.2M potassiWl1 dihydrogen orthophosphate
(e) After removal of the plate allow it to dry in air, expose it adjusted to pH 5.0 with 2M sodium hydroxide.
to iodine vapour until spots appear and examine in daylight. SYSTEM SUIT ABILITY
MOBILE PHASE The Assay is not valid unless, in the chromatogram obtained
10 volumes of acetone and 90 volumes of a 15.4% w/v with solution (3), the resolution factor between the peaks due
solution of a71ll1l0nium acetate adjusted to pH 5.0 with glacial to amoxicillin and cefadroxil is at least 2.0. If necessary,
acetic acid. adjust the composition of the mobile phase to achieve the
required resolution.
SYSTEM SUIT ABILITY
DETERMINATION OF CONTENT
The test is not valid unless the chromatogram obtained with
solution (3) shows two clearly separated spots. Calculate the content of C16H19N30SS in the tablets from
the chromatograms obtained and from the declared content
CONFIRMATION
of C16H19N30SS in amoxicillin tlihydrate BPCRS.
The principal spot in the chromatogram obtained with
solution (1) is similar in position, colour and size to that in LABELLING
the chromatogram obtained with solution (2). The quantity of active ingredient is stated in terms of the
equivalent amount of amoxicillin.
B. Shake a quantity of the powdered tablets containing the
equivalent of 0.5 g of amoxicillin with 5 mL of 'Water for
5 minutes, filter, wash the residue first with absolute ethanol
and then with ether and dry at a pressure not exceeding
0.7 kPa for 1 hour. Suspend 10 mg of the residue in 1 mL of Ampicillin Sodium andCloxacillin
water and add 2 mL of a mixture of 2 mL of cupri-tartaric
solution Rl and 6 mL of 'Water. A magenta colour is produced Sodium Intramammary Infusion
immediately. (Lactating COW)
C. Dissolve 0.1 mL of aniline in a mixture of 1 mL of Ampicillin and Cloxacillin Intramammary Infusion (LC)
hydrochloric acid and 3 mL of 'Wate¡". Cool the solution in ice
and add 1 mL of a freshly prepared 20% w/v solution of Action and use
sodium nitrite. Add the resulting mixture drop wise to a cold Penicillin antibacterial.
solution of 0.1 g of the residue obtained in test B in 2 mL of
5M sodium hydroxide. The solution becomes deep cherry-red DEFINITION
and a copious dark brown precipitate is produced. Ampicillin Sodium and Cloxacillin Sodium Intramammary
Infusion (Lactating Cow) is a sterile suspension of Ampicillin
ASSAY
Sodium and Cloxacillin Sodium in a súitable vehicle
Weigh and powder 20 tablets. Carry out the method for
containing suitable suspending agents.
liquid chromatography, Appendix III D, using the following
solutions.
2016 Ampicillin Preparations Vet-147

The imramam771aJy infusion complies 'With the requirements stated of a 1% w/v solution of iodine in a 4% w/v solution of
under Intramanmzal'Y Infusions and 'With the follo'Wing potassium iodide.
require711ents. lvIOBILE PHASE
Content of ampicillin, C16H19N304S 1 volume of formic acid, 30 volumes of acetone and
90.0 to 110.0% of the stated amount. 70 volumes of 0.05M potassium hydrogen phthalate that has
Content of cloxacillin, C19HISCIN30sS been adjusted first to pH 6.0 with 5M sodium hydroxide and
90.0 to 110.0% of the stated amount. then to pH 9.0 with O.lM sodiu11l hydroxide.
IDENTIFICATION CONFIRMATION
A. Can-y out the method for thin-layer chromatography, The principal spot in the chromatogram obtained with
Appendix nI A, using the following solutions. solution (1) corresponds to that in the chromatogram
(1) Extract a quantity of the infusion containing the obtained with solution (2).
equivalent of 50 mg of ampicillin with three 15-mL C. Extract a quantity containing the equivalent of 50 mg of
quantities of petroleu111 spirit (boiling range, 120° to 160°). ampicillin with three 15-mL quantities of petroleu11l spirit
Discard the extracts, wash the residue with 10 mL of ether (boiling range, 1200 to 160°). Discard the extracts, wash the
and dIy in a current of airo Dissolve the residue in 50 mL of residue with 10 mL of ether and dry the residue at 55°.
phosphate buffer pH 7. O, shake well, filter and use the filtrate. The residue produces an intense, persistent yellowish orange
(2) 0.1 % w/v of anhydrous ampicillin BPCRS in phosphate colour when introduced into a non-Iuminous Bunsen burner
bufferpH 7.0. fiame on a platinum wire moistened with hydrochloric acid.
CHROMATOGRAPHIC CONDITIONS TESTS
(a) Use a TLC slOlica gel plate (Merck silica gel 60 plates are Water
suitable). Impregnate the plate by spraying it with a Not more than 1.0% w/w, Appendix IX C. Use 1.5 g and a
0.1 % w/v solution of disodiul11 edetate in a 5 % w/v solution of mixture of 70 volumes of chlorofol"111 and 30 volumes of
sodium dihydrogen 011hophosphate, allow the plate to rny in air anhydrous methanol as the solvento
and heat at 105° for 1 hour. ASSAY
(b) Use the mobile phase as described below. Express, as far as possible, weigh and mix the contents of 10
(c) Apply 1 ~lL of each solution. containers. Extract a quantity of the mixed contents
containing the equivalent of 50 mg of ampicillin with three
(d) Develop the plate to 15 cm. 0
15-mL quantities of petroleum spiJit (boiling range, 120 to
(e) After removal of the plate allow it to dIy, heat at 105° for 160°) previously saturated with ampicillin sodium and
lOto 15 minutes and spray with a mixture of 100 volumes of cloxacillin sodium. Discard the extract, wash the residue with
starch mucilage, 6 volumes of glacial acetic acid and 2 volumes ether previously saturated with ampicillin sodium and
of a 1% w/v solution of iodine in a 4% w/v solution of cloxacillin sodium, dIy in a current of air, dissolve in 'Water
potassiwJ/ iodide. and dilute to 100 mL with 'Water. Centrifuge and use the
kIOBILE PHASE clear supernatant liquid (solution A).
5 volumes of buta71-1-ol, 10 volumes of a 0.1 % w/v solution For ampicillin
of disodiu71l edetate in a 5% w/v solution of sodium dihydrogen Dilute 2 mL of solution A to 50 mL with buffered copper
ol'thophosphate, 30 volumes of glacial acetic acid and sulfate solution pH 5.2, transfer 10 mL to a stoppered test
50 volumes of butyl acetate. tube and heat in a water bath at 75° for 30 minutes. Rapidly
CONFIRMATION cool to room temperature, dilute to 20 mL with buffered
copper sulfate solution pH 5.2 and measure the absorbance of
The principal spot in the chromatogram obtained with
solution (1) corresponds to that in the chromatogram the resulting solution at the maximum at 320 nm,
Appendix n B, using in the reference cell a solution prepared
obtained with solution (2).
by diluting 2 mL of solution A to 100 mL with buffered
B. Carry out the method for thin-layer chromatography,
copper sulfate solution pH 5.2. Calculate the content of
Appendix nI A, using the following solutions. C16H19N304S in a container of average content from the
(1) Extract a quantity of the infusion containing the absorbance obtained by canying out the operation at the same
equivalent of 0.130 g of cloxacillin with three 15-mL time using 2 mL of a solution prepared by dissolving 50 mg
quantities of petroleum spirit (boiling range, 120° to 160°). of anhydrous ampicillin BPCRS in 100 mL of 'Water, diluting
Discard the extracts, wash the residue with 10 mL of ether to 50 mL with buffered copper sulfate solution pH 5.2 and
and dly in a current of airo Dissolve the residue in 50 mL of beginning at the words 'transfer 10 mL ... ' and from the
phosphate buffer pH 7. O, shake well, filter and use the filtrate. declared content of C16H19N304S in anhydrous
(2) 0.28% w/v of cloxacillin sodiwl1 BPCRS in phosphate buffer ampicillin BPCRS.
pH 7.0. For cloxacillin
CHROMATOGRAPHIC CONDITIONS Dilute 2 mL of solution A to 100 mL with 1M hydrochloric
Ca) Use a TLC silica gel F254 silanised plate (Merck plates are acid. Measure the a.bsorbance of the resulting solution at the
suitable). maximum at 350 nm, Appendix n B, at 20° after exactly
12 minutes using 1M hydrochloric acid in the reference cell.
(b) Use the mobile phase as described below.
Calcu,late the content of C19H1SCIN30SS in a container of
(c) Apply 1 pL of each solution. average content from the absorbance obtained by carrying out
(d) Develop the plate to 15 cm. the operation at the same time using 2 mL of a solution
(e) After removal of the plate allow it to dry, heat at 1050 for prepared by dissolving 0.14 g of cloxacillin sodiu11l BPCRS in
lOto 15 minutes and spray with a mixture of 100 volumes of 100 mL of 'Water and from the declared content of
starch JJ1ucilage, 6 volumes of glacial acetic acid and 2 volumes C19H17CIN3NaOsS in cloxacillin sodium BPCRS. Each mg of
Vet-148 Ampicillin Preparations 2016

C 19H 17 CIN3NaOsS is equivalent to 0.9520 mg of Por ampicillin


C19HlSCIN30SS. Dilute 2 mL of solution A to 50 mL with buffered copper
LABELLING sulfate solution pH 5.2, transfer 10 mL to a stoppered test
tube and heat in a water bath at 75° for 30 minutes. Rapidly
The label states the quantity of Ampicillin Sodium in terms
cool to room temperature, dilute to 20 mL with buffered
of the equivalent amount of ampicillin and the quantity of
copper sulfate solution pH 5.2 and measure the absorbance of
Cloxacillin Sodium in terms of the equivalent amount of
the resulting solution at the maximum at 320 nm,
cloxacillin.
AppendLx Ir B, using in the reference cell the unheated
buffered solution of the infusion. Calculate the content of
C16H19N304S in a container of average content from the
absorbance obtained by carrying out the operation at the same
Ampicillin Trihydrate and Cloxacillin time using 2 rnL of a solution prepared by dissolving 60 rng
of anhydrous ampicillin BPCRS in 100 mL of a 50% v/v
Benzathine Intramammary Infusion solution of methanol, diluting to 50 rnL widl buffered copper
(Dry COW) sulfate solution pH 5.2, and beginning at the words 'transfer
10 mL. ..' and from the declared content of C16H19N304S in
Ampicillin and Cloxacillin Intramarnmary Infusion (DC)
anhydrous ampicillin BPCRS.
DEFINITION Por cloxacillin
Ampicillin Trihydrate and Cloxacillin Benzathine Dilute 2 mL of solution A to 100 mL with 1M hydrochloric
Intramammary Infusion (Dry Cow) is a sterile suspension of acid. Measure the absorbance of dle resulting solution at the
Ampicillin Trihydrate and Cloxacillin Benzathine in a maximum at 350 nm, Appendix Ir B, at 20° after exacdy
suitable vehicle, containing suitable suspending agents. 12 minutes, using 1M hydrochloric acid in the reference cell.
The intramammary infusioll complies with th.e requirements stated Calculate the content of C19HlSC1N30SS in a container of
under IntramammaJY Injusions and with the following average content from the absorbance obtained by canying out
requirements. the operation at the same time using 2 mL of a solution
Content of ampicillin, C16H19N304S prepared by dissolving 0.165 g of cloxacillin
90.0 to 110.0% of the stated amount. benzathine BPCRS in 100 mL of a 50% v/v solution of
methanol and from the declared content of C19HlSCIN30SS
Content of cloxacillin, C19HlSCIN30sS
in cloxacillin benzathine BPCRS.
90.0 to 110.0% of the stated amount.
LABELLING
IDENTIFICATION
The label states the quantity of Ampicillin Trihydrate in
A. Extract a quantity containing the equivalent of 0.25 g of
terrns of dle equivalent amount of ampicillin and the quantity
ampicillin with three 15-rnL quantities of petroleu11Z spirit
0 of Cloxacillin Benzadline in terms of the equivalent amount
(boiling range, 120 to 160'). Discard the extracts, wash the
of cloxacillin.
residue with 10 mL of ether and dry in a current of air. Shake
with 10 mL of chlorof01l1z and filter. Retain the filtra te. Wash
the residue with two 5-mL quantities of chloroform and dry at
room temperature at a pressure of 2 kPa. The infrared
abs01ption specmmz of the residue, Appendix II A, is Aprarnycin Veterinary Oral Powder
concordant with the reference spectrum of ampicillin trihydrate
(RSV06). Action and use
B. Wash the filtrate obtained in test A with two 5-mL Aminoglycoside antibacterial.
quantities of water, dry the chloroform layer with anhydrous
sodium sulfate, filter and dilute the filtra te to 20 mL with DEFINITION
chlorofol'111. The infrared abs01ption spectrum of the resulting Apramycin Veterinaty Oral Powder contains Apramycin
solution, Appendix II A, is concordant with the reference Sulfate.
spectrum of cloxacillin benzathine (RSV 12). The veterinaJY oral powder complies with the requirements stated
TESTS under VeterinaJY Oral Powders and 'lvith the follO'lving
requirements.
Water
Not more than 3.0% w/w, Appendix IX C. Use 1.5 g and a CHARACTERISTICS
mixture of 70 volumes of chlorofor111 and 30 volumes of Light brown, granular powder.
anhydrous methanol as the solvento
IDENTIFICATION
ASSAY A. Can)' out the method for thin-Iayer chro11latography,
Express, as far as possible, weigh and mix the contents of 10 Appendix III A, using the following solutions.
containers. Extract a quantity of the mixed contents (1) Dissolve a quantity of the veterinat)' oral powder
containing the equivalent of 60 mg of ampicillin with three containing the equivalent of 60 mg of apramycin in water and
15-rnL quantities of petroleum spirz"t (boiling range) 1200 to dilute to 100 mL.
160°) previously saturated with ampicillin trihydrate and
(2) 0.06% w/v of apramycin BPCRS in water.
cloxacillin benzathine. Discard the extracts, wash the residue
with ether previously saturated with ampicillin trihydrate and (3) 0.06% w/v of each of apramycin BPCRS and
cloxacillin benzathine, dry in a current of air, dissolve in tobramycin BPCRS in water.
50 mL of methanol and dilute to 100 mL with water. CHROMATOGRAPHIC CONDITIONS
Centrifuge and use the clear supernatant liquid (solution A). (a) Use a silica gel F 2S4 precoated plate (Meró: silica gel 60
F 2S4 plates are suitable).
(b) Use the mobile phase as described below.
2016 Apramycin Preparations Vet-149

(c) Apply 5 ~lL of each solution. 100% ofmobile phase B. Elute for a further 21 minutes
(d) Develop the plate to 10 cm. using 100% of mobile phase B, then step-wise re-equilibrate
(e) After removal of the plate, allow it to dry in air for to a mixture of 75% of mobile phase A and 25% of mobile
10 minutes, heat at 100° for 10 minutes, spray with sodium phase B and elute for at least 10 minutes.
hypochlorite solution whilst hot and cool for 5 minutes. Spray SYSTEM SUITABILITY
with absolute etlzanol, heat at 100° for 5 to 10 minutes, or The test is not valid unless, in the chromatogram obtained
until an area of the plate below the line of application gives with solution (2), the resolutionfactor between compound A
at most a faint blue colour with one drop of a 1% w/v and 3-hydroxyapramycin, identified as indicated in the
solution of potassiu71l iodide containing 1% w/v soluble starch, reference chromatogram supplied with apramycin BPCRS, is
and spray with the potassium iodide-starch solution. at least 0.8.
MOBILE PHASE LIMITS
20 volumes of clzloroform, 40 volumes of 13.5M ammonia and Multiply the areas of all the secondmy peaks by 0.5. [NOTE:
60 volumes of methanol, equilibrated for 1 hour before use. This is to ensure that the impurities are calculated relative to
SYSTEM SUIT ABILITY Apramycin Sulfate (which contains about 50% w/w of
apramycin).]
The test is not valid unless the chromatogram obtained with
solution (3) shows two cleady separated principal spots. In the chromatogram obtained with solution (1):
CONFIRMATION
the areas of any peaks corresponding to 3-hydroxyapramycin,
lividaminel2-deoxystreptamine (combined), compound A and
The principal spot in the chromatogram obtained with
compound B (identified as indicated in the reference
solution (1) corresponds to that in the chromatogram
chromatogram supplied with apramycin BPCRS) are not
obtained with solution (2).
greater than 1.4, 1.0, 0.4 and 0.4 times respectively the area
B. In the test for Related substances, the retention time of of the principal peak in the chromatogram obtained with
the principal peak in the chromatogram obtained with solution (3) (7%, 5%, 2% and 2% respectively);
solution (1) corresponds to that of the principal peak in the
the area of any other secondmy peak is not greater than
chromatogram obtained with solution (2).
0.4 times the area of the principal peak in the chromatogram
C. Yields reaction A characteristic of sulfates, Appendix VI. obtained with solution (3) (2%);
TESTS the sum of the areas of all the secondmy peaks is not greater
Related substances than 3 times the area of the principal peak in the
Carry out the method for liquid chromatograplzy, _chromatogram obtained with solution (3) (15%).
Appendix III D, using the following solutions in water. Disregard any peak with an area les s than the area of the
(1) Dissolve a quantity of the oral powder containing the peak in the chromatogram obtained with solution (4) (0.1 %).
equivalent of 0.3 g of apramycin and dilute to 100 mL. ASSAY
(2) 0.30% w/v of apramyán BPCRS. Carry out the microbiological assay of antibiotics,
(3) Dilute 1 volume of solution (1) to 20 volumes. Appendix XIV A, Method B. The precision of the assay is
(4) Dilute 1 volume of solution (3) to 50 volumes. such that the fiduciallimits of elTor are not less than 95%
and not more than 105% of the estimated potency.
CHROMATOGRAPHIC CONDITIONS
Calculate the content of apramycin in the veterinary oral
(a) Use a column (25 cm x 4 mm) packed with fast cation-
powder taking each 1000 Units found to be equivalent to
exchange polymeric beads (13 ~lm) with sulfonic acid
1 mg of apramycin. The upper fiduciallimit of elTor is not
functional groups (Dionex Fast Cation-1 R is suitable) and a
less than 97.0% and the lower fiduciallimit of elTor is not
stainless steel post-column reaction coil (380 cm x 0.4 mm)
more than 110.0% of the stated contento
with internal baffies. Use in the reaction coil ninhydrin reagent
1 at a flow rate approximately the same as that for the mobile LABELLING
phase. The quantity of active ingredient is stated in terms of the
(b) U se gradient elution and the mobile phase described equivalent amount of apramycin.
below. IMPURITIES
(c) Use a flow rate of 0.8 mL per minute. The impurities limited by the requirements of this
(d) Use a column temperature of 130°. Maintain the post- monograph include those listed under Apramycin Sulfate.
column reaction coil at the same temperature.
(e) Use a detection wavelength of 568 nm.
(f) Inject 20 ~lL of each solution.
MOBILE PHASE Apramycin Premix
lvlobile phase A A solution containing 1.961 % w/v of sodium
Action and use
citrate, 0.08% v/v of liquefied phenol and 0.5% v/v of
Aminoglycoside antibacterial.
thiodiglycol, adjusted to pH 4.25 using hydrochloric aád.
Mobile phase B A solution containing 4.09% w/v of sodiwll DEFINITION
chloride and 3.922% w/v of sodium citrate with 0.08% v/v of ApraJJ;lycin Premix contains Apramycin Sulfate.
liquefied phenol, adjusted to pH 7.4 with hydrochloric acid. The premix complies with the requirements stated under Premixes
Equilibrate the column using a mixture containing 75% of and with the follo'luing requirements.
mobile phase A and 25% of mobile phase B. After each
CHARACTERISTICS
injection elute for 3 minutes using the same mixture and
then carry out a linear gradient elution for 6 minutes to Light brown granules.
Vet-150 Apramycin Preparations 2016

IDENTIFICATION (b) Use gradient elution and the mobile phase described
A. Carry out the method for thin-layer chromatography, below.
Appendix III A, using the following solutions. (c) Use a flow rate of 0.8 mL per minute.
(1) Shake a quantity of the premix containing the equivalent 0
(d) Use a column temperature of 130 Maintain dle post-

of 60 mg of apramycin with 60 mL of O.lM sodium hydrogen column reaction coil at the same temperature.
carbonate at 60°, cool, dilute to 100 mL, filter and use the (e) Use a detection wavelength of 568 nm.
filtrate.
(±) Inject 20 ~lL of each solution.
(2) 0.06% w/v of apramycin BPCRS in 'Water.
IvIOBILE PHASE
(3) 0.06% w/v of each of apramycin BPCRS and
tobramycin BPCRS in 'Water. Mobile phase A A solution containing 1.961 % w/v of sodium
citrate, 0.08% v/v of liquefied phenol and 0.5% v/v of
CHROMATOGRAPHIC CONDITIONS
thiodiglycol, adjusted to pH 4.25 using hydrochloric acid.
(a) Use a silica gel F 254 precoated plate (Merck silica gel 60 Mobile phase B A solution containing 4.09% w/v of sodium
F 254 plates are suitable). chloride and 3.922% w/v of sodium czúate with 0.08% v/v of
(b) Use the mobile phase as described below. liquefied phenol, adjusted to pH 7.4 widl hydrochloric acid.
(c) Apply 5 ~lL of each solution. Equilibrate the column using a mixture containing 75% of
(d) Develop the pI ate to 10 cm. mobile phase A and 25% of mobile phase B. After each
(e) After removal of the plate, allow it to dry in air for injection elute for 3 minutes using dle same mixture and
10 minutes, heat at 100° for 10 minutes, spray with sodium then carry out a linear gradient elution for 6 minutes to
hypochlorite solution whilst hot and cool for 5 minutes. Spray 100 % of mobile phase B. Elute for a further 21 minutes
0
with absolute ethanol, heat at 100 for 5 to 10 minutes, 01" using 100% of mobile phase B, then step-wise re-equilibrate
until an area of the plate below the line of application gives to a mixture of 75% of mobile phase A and 25% of mobile
at most a faint blue colour with one drop of a 1% w/v phase B and elute for at least 10 minutes.
solution of potassiu11l iodide containing 1% w/v soluble starch, SYSTEM SUITABILITY
and spray with the potassium iodide-starch solution. The test is not valid unless, in the chromatogram obtained
MOBILE PHASE with solution (2), the resolution factor' between compound A
20 volumes of chloroform, 40 volumes of 13.5M ammonia and and 3-hydroxyapramycin, identified as indicated in dle
60 volumes of methanol, equilibrated for 1 hour before use. reference chromatogram supplied with apra17lycin BPCRS, is
at least 0.8.
SYSTEM SUIT ABILITY
LIMITS
The test is not valid unless the chromatogram obtained with
solution (3) shows two clearly separated principal spots. Multiply the areas of all the seconda/Ji peal~s by 0.5. [NOTE:
This is to ensure that the impurities are calculated relative to
CONFIRMATION
Apramycin Sulfate (which contains about 50% w/w of
The principal spot in the chromatogram obtained with apramycin).]
solution (1) corresponds to dlat in dle chromatogram
In the chromatogram obtained widl solution (1):
obtained with solution (2).
the areas of any peaks corresponding to 3-hydroxyapramycin,
B. In the test for Related substances, dle retention time of lividamine/2-deoxystreptamine (combined), compound A and
the principal peak in dle chromatogram obtained with
compound B (identified as indicated in dle reference
solution (1) corresponds to dlat of dle principal peak in dle
chromatogram supplied widl apramycin BPCRS) are not
chromatogram obtained with solution (2).
greater than 1.4, 1.0, 0.4 and 0.4 times respectively dle area
C. Yields reaction A characteristic of sulfates, Appendix VI. of the principal peak in the chromatogram obtained widl
TESTS solution (3) (7%, 5%, 2% and 2% respectively);
Related substances the area of any other seconda/y peak is not greater than
Carry out the method for liquid chromatography, 0.4 times the area of the principal peak in the chromatogram
Appendix III D, using the following solutions. obtained with solution (3) (2%);
(1) Shake a quantity of the premix containing the equivalent the sum of the are as of all dle seconda/y peaks is not greater
of 3 mg of apramycin per mL in a 5% w/v solution of sodium than 3 times the area of the principal peak in the
chlorz"de and centrifuge for 5 minutes at 4000 revolutions per chromatogram obtained with solution (3) (15%).
minute. Filter the supernatant liquid using filters of nominal Disregard any peak with an area less than dle area of dle
pore size of 5 ~lm and 0.45 ~lm. peak in the chromatogram obtained with solution (4) (0.1 %).
(2) 0.30% w/v of apramycin BPCRS in 'Water. ASSAY
(3) Dilute 1 volume of solution (1) to 20 volumes with 'water. To about 5 g of the premix, accurately weighed, add 250 mL
(4) Dilute 1 volume of solution (3) to 50 volumes with 'Water. of a solution containing 0.6% w/v of sodium hydroxide and
CHROMATOGRAPHIC CONDITIONS 0.745% w/v of ethylenediaminetetra-acetic acid in 'Water. Shake
for 1 hour and allow to stand for 15 minutes. Decant the
(a) Use a column (25 cm x 4 mm) packed with fast cation-
supernatant liquid and dilute to concentrations equivalent to
exchange polymeric beads (13 ~lm) with sulfonic acid
the solutions of the Standard Preparation widl the pH 8.0
functional groups (Dionex Fast Cation-1 R is suitable) and a
buffer and carry,out the microbiological assay of amibiotics,
stainless steel post-column reaction coil (380 cm x 0.4 mm)
Appendix XIV A, Method B. The precision of the assay is
with internal baffies. Use in the reaction coil ninhydr¡"¡z reagent
such that dlefiduciallimits of enor are not les s dlan 95%
1 at a flow rate approximately the same as that for the mobile
and not more than 105 % of the estimated potency.
phase.
Calculate the content of apramycin in the premix taking each
1000 Units found to be equivalent to 1 mg of apramycin.
2016 CalciulTI Preparations Vet-151

The upper fiduciallimit of error is not less than 97.0% and ASSAY
the lower fiduciallimit of enor is not more than 110.0% of To a volume containing 0.4 g of Azaperone add 25 mL of
the stated contento 0.5M sulfuric acid and sufficient water to produce 250 mL.
STORAGE Mix, transfer 10 mL of the solution to a separating funnel
containing 10 rnL of 0.05M sulfuric acid and shake with
Apramycin Premix should be stored in a dry place.
20 mL of ether. Wash the ether layer with two 10 mL
LABELLING quantities of 0.05M sulfuric acid. Make the combined acid
The quantity of active ingredient is stated in terms of the extract and washings alkaline with 5 mL of 1M sodium
equivalent amount of apramycin. hyd1'Oxide, add 50 mL of ether, shake and allow to separate.
IMPURITIES Extract the aqueous layer with 50 mL of ether. Wash the two
The impurities limited by the requirements of this ether solutions, in succession, with a 20 mL quantity of water
monograph include those listed under Apramycin Sulfate. and extract each of the two ether solutions, in succession,
with two 20 mL quantities and one 5 mL quantity of
0.25M SUlfW7C acid. Combine the acid extracts and add
sufficient 0.25M sulfuric acid to produce 100 mL. To 5 mL of
the resulting solution add 5 mL of methanol and sufficient
Azaperone Injection 0.25M sulfuric acid to produce 100 mL. Measure the
absorbal1ce of the resulting solution at the maximum at
Aetion and use 242 nm, Appendix II B. Dissolve 40 mg of azaperone BPCRS
Dopamine receptor antagonist; neuroleptic (veterinary). in sufficient 711ethanol to produce 250 mL, dilute 5 mL of the
resulting solution to 100 mL with 0.25M sulfuric acid and
DEFINITION measure the absorbance at 242 nm. Calculate the content of
Azaperone Injection is a sterile solution of Azaperone in C19HzzFN30 in the injection from the absorbances obtained
Water for Injections. using the declared content of C 19HzzFN30 in
The injection complies with the requirements stated under azaperone BPCRS.
Parenteral Preparations and with the following requirements. STORAGE
Content of azaperone, C 19H 22 FN3 0 Azaperone Injection should be protected from light.
90.0 to 110.0% of the stated amount.
CHARACTERISTICS
A clear, yellow solution.
IDENTIFICATION Calcium Borogluconate Injection
A. To a volume containing 80 mg of Azaperone add 5 mL of DEFINITION
0.5M sulfuric acid and 20 mL of water. Extract the solution
Calcium Borogluconate Injection is a sterile solution of
with 50 mL of ether, make the aqueous phase alkaline with
Calcium Gluconate and Boric Acid in Water for Injections.
1M sodill77l hyd1'Oxide and extract with 50 mL of ether. Wash
the ether extracts with two 10 mL-quantities of water, shake The injection complies with the requirements stated under
with anhyd1'Ous sodiu71l sulfate, filter and evaporate to dryness. Parenteral Preparations and with the following requirements.
The infrared absorption spectru711 of the residue, Appendix II A, Content of ealcium, Ca
is concordant with the reference spectrulll of azaperone 95.0 to 105.0% of the stated amount.
(RSV08). Content of borie aeid, H 3 B0 3
B. The light abs01ption, Appendix II B, in the range 230 to Not more than 2.3 times the stated content of calcium.
350 nm of a 2-cm layer of the solution obtained in the Assay
IDENTIFICATION
exhibits maxima at 242 nm and at 312 nm. The absorbal1ces
A. To 1 mL add sufficient water to produce a solution
at the maxima are about 1.1 and about 0.38, respectively.
containing about 0.75% w/v of calcium and add 0.05 mL of
TESTS iron(JJJ) chloride solution Rl. An intense yellow 01' yellowish
Acidity green colour is produced.
pH, 3.5 to 5.0, Appendix V L. B. Yields the reactions characteristic of calciu71l salts,
Related substanees Appendix VI.
Carry out in subdued light the method for thin-layer C. To 1 mL add 0.15 mL of sulfuric acid and 5 mL of
ch1'Omatography, Appendix III A, using a silica gel F Z54 methanol and ignite. The mixture burns with a flame tinged
precoated plate (Mere k silica gel 60 F Z54 plates are suitable) with green.
and a mixture of 1 volume of ethanol (96%) and 9 volumes
Acidity
of chlorofor711 as the mobile phase. Apply separately to the
pH of the injection, diluted if necessary with ca/"bon dioxide-
plate 1O ~lL of each of the following two solutions.
free water to contain 1.5% w/v of calcium, 3.0 to 4.0,
For solution (1) dissolve the extracted residue obtained in
Appendix V L.
Identification test A in sufficient chlo1'Oform to produce a
solution containing 1% w/v of Azaperone. For solution (2) ASSAY
dilute 1 volume of solution (1) to 100 volumes with For ealcium
chlo1'Oforl11. After removal of the plate allow it to dry in air Dilute ,a quantity containing the equivalent of 45 mg of
and examine under ultraviolet light (254 71111). Any secol1dary calcium to about 50 mL with water. Titrate with
spot in the chromatogram obtained with solution (1) is not 0.05M disodium edetate VS to-within a few mL of the expected
more intense than the spot in the chromatogram obtained end point, add 4 mL of a 40% w/v solution of sodium
with solution (2) (1 %). hyd1'Oxide and 10 mg of soloch1'Ome dark blue mixture and
continue the titration until the colour changes from pink to
Vet-152 Calcium Preparations 2016

blue. Each mL of 0.05M disodiu11l edetate VS is equivalent to as indicator, until only a faint blue colour remains; add 2 g
2.004 mg of Ca. of potassiu111 thiocyanate and continue the titration until the
For borie acid blue colour disappears. Each mL of O.lM sodium thiosulfate
Dilute a quantity containing 0.1 g of Boric Acid to 50 mL VS is equivalent to 6.354 mg of Cu.
with water, add 3 g of D-mannitol and titrate with 0.1 M sodium LABELLING
hydroxide VS using phenolphthalein solution Rl as indicator. The strength is stated as the equivalent amount of copper in
Each mL of 0.1 M sodiu111 hydroxide VS is equivalent to a suitable dose-volume.
6.183 mg of H3B03'
STORAGE
Calcium Borogluconate Injection should be protected from
light. It may be supplied in containers of glass type IJI, Cefalonium Intramammary Infusion (Dry
Appendix XIX B.
COW)
LABELLING
The strength is stated as the amount of calcium in a suitable Aetion and use
dose-volume. The label also states the proportion of boric Cephalosporin antibacterial.
acid presento
DEFINITION
Cefalonium Inu"amammary Infusion (Dry Cow) is a sterile
suspension of Cefalonium in a suitable non-aqueous vehicle,
containing suitable suspending agents.
Calcium Copperedetate Injection The intramam71lalY infusion complies 'luith the requirements stated
Aetion and use under Intrama11Z11Zary Infusions and with the following
Used in the treatment of copper deficiency. require11lents.
Content of anhydrous eefalonium, C2oH1SN40SS2
DEFINITION 90.0 to 112.0% of the stated amount.
Calcium Copperedetate Injection is a sterile suspension of
Calcium Copperedetate, with suitable stabilising and IDENTIFICATION
dispersing agents, in an oil-in-water emulsiono A. In the Assay, the retention time of the principal peak in
the chromatogram obtained with solution (1) corresponds to
The injection c011lplies with the require11lents stated under
that of the principal peak in the chromatogram obtained with
Parenteral Preparations and with the following requirements.
solution (2).
Content of eopper, Cu B. To a quantity of the intramammary infusion containing
92.0 to 108.0% ofthe stated amount. the equivalent of 20 mg of anhydrous cefalonium add a few
CHARACTERISTICS drops of sulfuric acid (80% v/v) containing 1% v/v of nitric
NIacroscopical A blue, opaque, viscous suspension. acid and mix. A pale green colour is produced which
Microscopical When diluted with glycerol and examined immediately changes to dark green.
microscopically, cubic crystals lOto 30 ~lm in diameter are TESTS
visible, but large plate-like crystals are absent. Related substanees
IDENTIFICATION Carry out the method for liquid chromatography,
Ignite 1 g and dissolve the residue by warming in 10 mL of a Appendix III D, using the following solutions prepared
mixture of equal volumes of hydrochloric acid and water; filter irnmediately before use and stored in a refrigerator between
if necessary. The solution complies with the following tests. injections.
A. Neutralise 2 mL of the solution with 5M al1u1Zonia, and (1) Disperse a quantity of the intramammary infusion
add 1 mL of 6M acetic acid and 2 mL of potassium iodide containing the equivalent of 0.10 g of anhydrous cefalonium
solution. A white precipitate is produced and iodine is in 50 mL of petroleum spirit (boiling range) 60° to 80°)) add
liberated, colouring the supernatant liquid brown. 100 mL of O.lM hydrochloric acid and shake vigorously by
hand for 5 minutes and then mechanically for 30 minutes,
B. To 5 mL of the solution add 25 mL of a 10% v/v solution
filter and use the lower layer.
of mercaptoacetic acid and filter. Make the filtrate alkaline with
5M ammonia and add 5 mL of a 2.5% w/v solution of (2) Dilute 2 volumes of solution(1) to 100 volumes with
a1111110nium oxalate. A white precipitate is produced which is 0.1 M hydrochloric acid.
soluble in hydrochloric acid but only sparingly soluble in (3) 0.0020% w/v of isonicotinamide in O.lM hydrochloric acid.
6M acetic acid. (4) 0.0050% w/v of each of cefalonium BPCRS and
ASSAY isonicotinamide in O.lM hydrochloríc acid.
Evaporate a quantity containing the equivalent of 0.13 g of CHROMATOGRAPHIC CONDITIONS
0
copper to dryness, ignite at 600 to 700°, cool and heat the (a) Use a stainless steel column (10 cm x 4.6 mm) packed
residue with 5 mL of a mixture of equal volumes of witl:Í particles of silica the sillface of which has been modified
hydrochloric acid and water on a water bath for 15 minutes. with chemically-bonded hexylsilyl groups (Spherisorb SS C6
Add 5 mL of water, filter and wash the residue with about is suitable).
20 mL of water. Combine the filtrate and the washings, add (b) U se isocratic elution and the mobile phase described
10 mL of bromine water) boil to remove the bromine, cool below.
and add dilute sodium carbonate solution until a faint
permanent precipitate is produced. Add 3 g of potassiU111 (c) Use a flow rate of 2 mL per minute.
iodide and 5 mL of 6M acetic acid and titrate the liberated (d) Use an ambient column temperature.
iodine with O.lM sodium thiosulfate VS, using starch mucilage (e) Use a detection wavelength of 262 nm.
2016 Chlortetracycline Preparations Vet-153

(f) Inject 1O ~lL of each solution. The veterinalY oral pozuder complies zuith the l'equirements stated
(g) For solution (1), allow the chromatography to proceed under VeterinalY Oral Pozuders and zuith the follozuing
for at least 3.5 times the retention time of the principal peak. requirements.
MOBILE PHASE Content of chlortetracycline hydrochloride,
3 volumes of acetonittile and 97 volumes of a pH 3.4 solution
CzzHz3CINzOg,HCI
90.0 to 110.0% of the stated amount.
containing 5 volumes of O.IM sodiu111 acetate and 95 volumes
of O.IM acetic acid. IDENTIFICATION
SYSTEM SUITABILITY A. Carry out the method for thin-layer chromatography,
Appendix ITI A, using the following solutions.
The test is not valid unless, in the chromatogram obtained
with solution (4), the resolution factor between the two (1) Extract a quantity of the oral powder containing 10 mg
principal peaks is at least 10. of Ch1ortetracyc1ine Hydrochloride with 20 mL of nzethanol
and centrifuge.
LIMITS
(2) 0.05% w/v of chlortetracycline hydrochloride BPCRS in
In the chromatogram obtained with solution (1): 111ethanol.
the area of any peak corresponding to isonicotinamide is not (3) 0.05% w/v of each of chlortetracycline
greater than the area of the principal peak in the hydrochloride BPCRS, tetracycline hydrochloride BPCRS and
chromatogram obtained with solution (3) (2%); metacycline hydrochloride EPCRS in 111ethanol.
the area of any other secondary peak is not greater than half
CHROMATOGRAPHIC CONDITIONS
the area of the principal peak in the chromatogram obtained
with solution (2) (1 %). (a) Use silica gel H as the coating. Adjust the pH of a
10% w/v solution of disodiu111 edetate to 8.0 with 10M sodium
ASSAY hydroxide and spray the solution evenIy onto the plate (about
Express, as far as possible, weigh and mix the contents of 10 10 mL for a pIate 100 mm x 200 mm). Allow the plate to
containers. Carry out the method for liquid chromatography, dry in a horizontal position for at Ieast 1 hour. At the time of
Appendix lIT D, using the following solutions prepared use, dry the pIate in an oven at 110° for 1 hour.
immediately before use and stored in a refrigerator between
(b) Use the mobile phase as described below.
injections.
(c) AppIy 1 ~L of each soIution.
(1) Disperse a quantity of the mixed contents of the 10
containers containing the equivalent of 75 mg of anhydrous (d) DeveIop the pIate to 15 cm.
0
cefalonium in 50 mL of petroleum spirit (boiling range, 60 to (e) After removaI of the plate, allow it to dry in a current of
0
80 add 100 mL of O.IM hydrochlO1'ic acid and shake
),
air and examine under ultraviolet light (365 11m).
vigorously by hand for 5 minutes and then mechanically for MOBILE PHASE
30 minutes. Filter the lower layer and dilute 10 mL of the 6 volumes of zuater, 35 volumes of methanol and 59 volumes
filtrate to 100 mL with 0.1 M hydrochloric acid. of dichloromethane.
(2) 0.0075% w/v of cefalonium BPCRS in O.IM hydrochloric
SYSTEM SUIT ABILITY
acid.
The test is not valid unIess the chromatogram obtained with
CHROMATOGRAPHIC CONDITIONS
soIution (3) shows three c1earIy separated spots.
The chromatographic conditions described under Related
CONFIRMATION
substances may be used.
The principal spot in the chromatogram obtained with
DETERMINATION OF CONTENT
soIution (1) corresponds to that in the chromatogram
Calculate the content of C2oHlSN40SS2 in a container of obtained with solution (2).
average content using the dec1ared content of C2oHlSN40SS2 B. To a quantity of the oral powder containing 10 mg of
in cefalonium BPCRS. ChIortetracyc1ine HydrochIoride add 20 mL of warm ethanol
STORAGE (96%), allow to stand for 20 minutes, filter and evaporate to
Cefalonium Intramammary Infusion (Dry Cow) should be dryness on a water bath. A 0.1 % w/v solution of the residue
0
stored at a temperature not exceeding 30 It should not be
• in phosphate buffer pH 7.6, when heated at 100° for 1 minute,
allowed to freeze. exhibits a strong bIue fluorescence in ultraviolet light.
LABELLING T etracycline hydrochloride and 4-epichlortetracycline
The quantity of active ingredient is stated in terms of the hydrochloride
equivalent amount of anhydrous cefalonium. Not more than 8.0% and 6.0% respectiveIy, determined as
described under the Assay. Inject separateIy soIutions (1) and
(4).
ASSAY
Carry out the method for liquid chro111atography,
Chlortetracycline Veterinary Oral Powder Appendix lIT D, using the following soIutions.
Ch1ortetracyc1ine Soluble Powder (1) Mix a quantity of the oral powder containing 25 mg of
Ch1ortetracyc1ine HydrochIoride with 50 mL of
Action and use
O.OIM'hydrochloric acid, shake for 10 minutes, dilute to
Tetracyc1ine antibacterial.
100 mL with O.OIM hydrochloric acid and filter (GF/C paper
DEFINITION is suitabIe). '
Ch1ortetracyc1ine Veterinary Oral Powder is a mixture of (2) 0.025% w/v of chlortetracycline hydrochloride BPCRS in
Ch1ortetracyc1ine Hydrochloride and Lactose 01' other O.OIM hydrochloric acid.
suitable diluent.
Vet-154 Chlortetracycline Preparations 2016

(3) 0.025% w/v of each of chlortetracycline dry in a horizontal position for at least 1 hour. At the time of
0
hydrochloride BPCRS and 4-epichl011etracycline use, dry the plate in an oven at 110 for 1 hour.
hydrochloride EPCRS in O.OIM hydrochloric acid. (b) Use the mobile phase as described below.
(4) 0.002% w/v of tetracycline hydrochloride BPCRS and (c) Apply 1 ~lL of each solution.
0.0015% w/v of 4-epichlortetracycline hydrochloride EPCRS in
(d) Develop the plate to 15 cm.
O.OIM hydrochloric acid.
(e) After removal of the plate, allow it to dry in a current of
CHROMATOGRAPHIC CONDITIONS air and examine under ultraviolet light (365 nm).
(a) Use a stainless steel column (25 cm x 4.6 mm) packed
MOBILE PHASE
with end-capped octadecylsilyl silica gel for chromatography
(10 ~lm) (Nuc1eosil C18 is suitable). 6 volumes of water, 35 volumes of methanol and 59 volumes
of dichloromethane.
(b) Use isocratic elution and the mobile phase described
below. SYSTEM SUIT ABILITY

(c) Use a fiow rate of 2 mL per minute. The test is not valid unless the chromatogram obtained with
solution (3) shows three clearly separated spots.
(d) Use a column temperature of 40 0 •
(e) Use a detection wavelength of 355 nm. CONFIRMATION

(±) Inject 20 ~lL of each solution. The principal spot in the chromatogram obtained with
solution (1) corresponds to that in the chromatogram
MOBILE PHASE obtained with solution (2).
20 volumes of dimethylformamide and 80 volumes of B. To a quantity ofthe powdered tablets containing 10 mg
O.IM oxalic acid the pH of which has been adjusted to 2.2 of Chlortetracycline Hydrochloride add 20 mL of warm
with tliethylamine. ethanol (96%), allow to stand for 20 minutes, filter and
SYSTEM SUITABILITY evaporate to dryness on a water bath. A 0.1 % w/v solution of
The Assay is not valid unless, in the chromatogram obtained the residue in phosphate buffer pH 7.6, when heated at 100 0
with solution (3), the resolution factor between the two for 1 minute, exhibits a strong blue fiuorescence in ultraviolet
principal peaks is at least 1.5. light.
DETERMINATION OF CONTENT TESTS
Calculate the content of C22H23ClN20s,HCI in the oral Tetracycline hydrochloride and 4-epich1ortetracycline
powder using the dec1ared content of C22H23ClN20s,HCI in hydrochloride
chlortetracycline hydrochloride BPCRS. Not more than 8.0% and 6.0% respectively, determined as
described under Assay. Inject separately solutions (1) and
(4).
Dissolution
Comply with the requirements for Monographs of the British
Chlortetracycline Tablets Pharmacopoeia in the dissolution test for tablets and capsules,
Appendix XII Bl.
Action and use
Teu'acyc1ine antibacterial. TEST CONDITIONS
(a) Use Apparatus 2, rotating the paddle at 50 revolutions
DEFINITION per minute.
Ch1ortetracyc1ine Tablets contain Ch10rtetracycline (b) Use 900 mL of O.IM hydrochloric acid, at a temperature of
Hydrochloride. 37 0 , as the medium.
The tablets comply with the requirements stated under T ablets and
PROCEDURE
with the following requirements.
After 45 minutes withdraw a 10 mL sample of the medium
Content of ch10rtetracycline hydroch1oride, and filter. Measure the absorbance of the filtered sample,
C22H23CIN20s,HCI suitably diluted with the dissolution medium if necessary, at
95.0 to 110.0% ofthe stated amount. the maximum at 266 nm, Appendix n B, using O.IM
IDENTIFICATION hydrochloric acid in the reference cell.
A. Carry out the method for thin-layer chromatography, DETERMINATION OF CONTENT
Appendix nI A, using the following solutions.
Calculate the total content of chlortetracyc1ine hydroch1oride,
(1) Extract a quantity of the powdered tablets containing C22H23CIN20s,HCI, in the medium taking 346 as the value
10 mg of Chlortetracycline Hydroch1oride with 20 mL of of A(1 %, 1 cm) at the maximum at 266 nm.
methanol and centrifuge.
ASSAY
(2) 0.05% w/v of chlortetracycline hydrochlO1ide BPCRS in
Weigh and powder 20 tablets. Carry out the method for
methanol.
liquid chromatography, Appendix In D, using the following
(3) 0.05% w/v of each of chlortetracycline solutions.
hydrochloride BPCRS, tetracycline hydrochloride BPCRS and
(1) Mix a quantity of the powdered tablets containing 25 mg
metacycline hydrochlO1ide EPCRS in methanol.
of Chlortetracycline Hydrochloride with 50 mL of
CHROMATOGRAPHIC CONDITIONS O.OIM hydrochl011'C acid, shake for 10 minutes, dilute to
(a) Use silica gel H as the coating. Adjust the pH of a 100 mL with.O.OlM hydrochloric acid and filter (GF/C papel'
10% w/v solution of disodium edetate to 8.0 with 10M sodium is suitable).
hydroxide and spray the solution evenly onto the plate (about (2) 0.025% w/v of chlortetracycline hydrochloride BPCRS in
10 mL for a plate 100 mm x 200 mm). A1low the plate to O.OIM hydrochlO1ic acid.
2016 Cloprostenol Preparations Vet-155

(3) 0.025% w/v of each of chlortetracycline (b) U se isocratic elution and the mobile phase described
hydrochloride BPCRS and 4-epichlortetracycline below.
hydrochloride EPCRS in O.OlM hydrochloric acid. (c) Use a flow rate of 1.8 mL per minute.
(4) 0.002% w/v of tetracycline hydrochloride BPCRS and (d) Use an ambient column temperature.
0.0015% w/v of 4-epichlortetracycline hydrochloride EPCRS in (e) Use a detection wavelength of 220 nm.
O.OlM hydrochloric acid.
(f) Inject 20 ~lL of each solution.
CHROMATOGRAPHIC CONDITIONS
(g) For solutions (1) and (2) allow the chromatography to
(a) Use a stainless steel column (25 cm x 4.6 mm) packed proceed for 1.5 times the retention time of the principal
with end-capped octadecylsilyl silica gel for éhromatography peak.
(10 pm) (Nuc1eosil C18 is suitable).
lvIOBILE PHASE
(b) U se isocratic elution and the mobile phase described
below. 270 volumes of acetonitrz"le and 730 volumes of a solution
containing 0.24% w/v of sodium dihydrogen 011hophosphate the
(c) Use a flow rate of 2 mL per minute. pH of which has been adjusted to 2.5 with 011hophosphonc
(d) Use a column temperature of 40°. acid.
(e) Use a detection wavelength of 355 nm. SYSTEM SUITABILITY
(f) Inject 20 ~lL of each solution. The test is not valid unless~ in the chromatogram obtained
lvIOBILE PHASE with solution (3) ~ the resolution factor between the peak due to
20 volumes of dimethylformamide and 80 volumes of hydrocortisone acetate (retention time abolit 25 minutes) and
O.lM oxalic acid the pH ofwhich has been adjusted to 2.2 that of c1oprostenol (retention time about 35 minutes) is at
with t1'iethylamine. least 6.
SYSTEM SUIT ABILITY LIMITS

The assay is not valid unless~ in the chromatogram obtained In the chromatogram obtained with solution (1) the sum of
with solution (3)~ the 1'esolution factor between the two the areas of any secondaly peaks is not more than 1.25 times
principal peaks is at least 1.5. the area of the principal peak in the chromatogram obtained
with solution (2) (2.5%).
DETERMINATION OF CONTENT
Calculate the content of C22H23ClN208~HCl in the tablets ASSAY
using the dec1ared content of C22H23ClN208~HCl in Carry out the method for liquid chromatography~
chlortetracycline hydrochloride BPCRS. Appendix III D~ liSing the following solutions.
(1) Dilute the injection~ if necessary~ in absolute ethanol to
contain the equivalent of 0.009% w/v of cloprostenol.
(2) 0.009% w/v of cloprostenol sodium BPCRS in absolute
Cloprostenol Injection ethanol.
(3) Dissolve 5 mg of hydrocortisone acetate BPCRS and
Action and use 2.5 mg of cloprostenol sodium BPCRS in absolute ethanol and
Prostaglandin (PGF 2cJ analogue. dilute to 10 mL with the mobile phase.
CHROMATOGRAPHIC CONDITIONS
DEFINITION
Cloprostenol Injection is a sterile solution of Cloprostenol The chromatographic conditions described under Related
Sodium in Water for Injections. substances may be used.
The injection complies with the requiremenrs stated under SYSTEM SUIT ABILITY
Parenteral Preparations and zuith the follozuing requirements. The test is not valid unless~ in the chromatogram obtained
Content of cloprostenol, C22H29CI06 with solution (3) ~ the resolution factor between the peak due to
90.0 to 110.0% of the stated amount. hydrocortisone acetate (retention time abolit 25 minutes) and
that of c1oprostenol (retention time about 35 minutes) is at
IDENTIFICATION least 6.
In the Assay~ the chromatogram obtained with solution (1)
DETERMINATION OF CONTENT
shows a peak with the same retention time as the peak due to
c1oprostenol in the chromatogram obtained with solution (2). Calculate the content of C22H29Cl06 in the injection using
the declared content of C22H29Cl06 in cloprostenol
Related substances
sodium BPCRS.
Carry out the method for liquid chromatography~
Appendix III D~ using the following solutions. STORAGE
(1) Dilute the injection~ if necessary~ in absolute ethanol to Cloprostenol Sodium Injection should be protected from
contain the equivalent of 0.009% w/v of c1oprostenol. light.
(2) 0.00018 % w/v of cloprostenol sodium BPCRS in absolute LABELLING
ethanol. The quantity of active ingredient is stated in terms of the
(3) Dissolve 5 mg of hydrocortisone acetate BPCRS and equivalent amount of c1oprostenol in a suitable dose-volume.
2.5 mg of cloprostenol sodium BPCRS in absolute ethanol and
dilute to 10 mL with the mobile phase.
CHROMATOGRAPHIC CONDITIONS
(a) Use a stainless steel column (25 cm x 5 mm) packed
with base-deactivated octadecylsilyl silica gel for chromatography
(Waters Symmetry ODS is suitable).
Vet-156 Cloxacillin Preparations 2016

Cloxacillin Benzathine Intramammary Cloxacillin Sodium Intramammary


Infusion (Dry Cow) Infusion (Lactating COW)
Cloxacillin Intramamma1Y Infusion (DC) Cloxacillin IntramammalY Infusion (LC)

Action and use Action and use


Penicillin antibacterial. Penicillin antibacterial.

DEFINITION DEFINITION
Cloxacillin Benzathine Intramammary Infusion (Dry Cow) is Cloxacillin Sodium Intramarnmary Infusion (Lactating Cow)
a sterile suspension of Cloxacillin Benzathine in a suitable is a sterile suspension of Cloxacillin Sodium in a suitable
non-aqueous vehicle containing suitable suspending agents. non-aqueous vehicle containing suitable suspending and
The intramamma/Y il1fusion complies 'With the requirements stated dispersing agents.
under Intrama11Zl1Za/Y Infusions and 'With the follo'Wing The intrama711mary infusion complies with the requirements stated
requirements. undel' Intra711am711a/y Infusions and 'Wz"th the follo'Wing
Content of cloxacillin, C19HlSC1N30SS requil'ements.
90.0 to 110.0% ofthe stated amount. Content of cloxaciIlin, C19HlSC1N30SS
IDENTIFICATION 90.0 to 110.0% of the stated amount.
Extract a quantity of the infusion containing the equivalent of IDENTIFICATION
75 mg of cloxacillin with three 15-mL quantities of petroleum Extract a quantity containing the equivalent of 75 mg of
spirit (boiling range 120° to 160°), discard the extracts, wash
J cloxacillin with three 15-rnL quantities of petl'oleu111 spirit
the residue with ether and dly in a current of airo The residue (boiling 1'Gnge 120° to 160°). Discard the extracts, wash the
J

obtained complies with the following tests. residue with ethel' and dry in a current of airo The residue
A. The infrared absorption spectrwn, Appendix n A, is obtained complies with the following tests.
concordant with the reference spectrum of cloxacillin A. The infral'ed abs01ption spectrum, Appendix II A, is
benzathine (RSV 12). concordant with the refel'ence spectru711 of cloxacillin sodium
B. Shake 50 mg with 1 mL of 1M sodiu111 hydroxide for (RSV 13).
2 minutes, add 2 mL of ether, shake for 1 minute and allow B. Yields the reactions characteristic of sodium salts,
to separate. Evaporate 1 mL of the ether layer to dryness, Appendix VI.
dissolve the residue in 2 mL of glacial acetic acid and add Water
1 mL of dilute potassiunz dichro711ate solution. A golden yellow Not more than 1.0% w/w, Appendix IX C. Use 3 g and a
precipitate is produced. mixture of 70 volumes of chloroform and 30 volumes of
Water anhydrous methanol as the solvento
Not more than 2.0% w/w, Appendix IX C. Use 3 g and a
ASSAY
mixture of 70 volumes of chloroform and 30 volumes of
Express, as far as possible, weigh and mix the contents of 10
anhydrous methanol as solvento
containers. Cany out the method for liquid chromatography,
ASSAY Appendix In D, using the following solutions.
Express, as far as possible, weigh and mix the contents of 10 (1) Extract a quantity of the mixed contents of the 10
containers. Extract a quantity of the mixed contents containers containing the equivalent of 50 mg of cloxacillin
containing the equivalent of 80 mg of cloxacillin, with three with 15 mL of petroleum spil'it (boiling rangeJ 120° to 160°),
15 mL quantities of petroleum spirit (boiling rangeJ 120° to centrifuge and discard the supernatant liquido Repeat the
160°) previously saturated with cloxacillin benzathine. Discard extraction with a further two 15-rnL quantities of petroleum
the extract, wash the residue with ether previously saturated spi1it (boiling rangeJ 1200 to 160°). Shake the residue with
with cloxacillin benzathine, dry in a current of air, dissolve in 20 mL of ether, centrifuge and dry in a current of air until
25 mL of 711ethanol and dilute to 50 mL with 'Water. Dilute the solvents have evaporated. Dissolve the final residue in
2 mL to 100 mL with buffered copper sulfate solution pH 2. O, sufficient of the mobile phase to produce 50 rnL and dilute
transfer 10 mL to a stoppered test tube and heat in a water 5 volumes of the resulting solution to 50 volumes with the
bath at 70° for 20 minutes. Rapidly cool to room mobile phase.
temperature, dilute to 20 mL with absolute ethanol and (2) 0.011 % w/v of cloxacillin sodiüm BPCRS in the mobile
measure the absorbance of the resulting solution at the phase.
maximum at 338 nm, Appendix n B, using in the reference
cell 10 mL of the unheated buffered solution of the (3) 0.01 % w/v of each of cloxacillin sodium BPCRS and
substance being exarnined, diluted to 20 mL with absolute flucloxacillin sodiwl1 BPCRS in the mobile phase.
ethanol. CHROMATOGRAPHIC CONDITIONS
Calculate the content of C19HlSClN30SS in a container of (a) Use a stainless steel column (25 cm x 4.6 mm) packed
average content from the absorbance obtained by canying out with octadecylsilyl silica gel fol' chromatography (5 /lm) (Hypersil
the procedure simultaneously using 2 mL of a solution 5 ODS is suitable).
prepared by dissolving 105 mg of cloxacillin (b) U se isocratic elution and the mobile phase described
benzathine BPCRS in 50 mL of a mixture of equal volumes below.
of lnethanol and 'Water and from the declared content of (c) Use a flow rate of 1 mL per minute.
C19HlSClN30sS in cloxacillin benzathine BPCRS.
(d) U se an ambient column temperature.
LABELLING (e) Use a detection wavelength of 225 nm.
The quantity of active ingredient is stated in terms of the
(f) Inject 20 ~lL of each solution.
equivalent amount of cloxacillin.
2016 Co-trimazine Preparations Vet-157

MOBILE PRASE (b) Use the mobile phase as described below.


25 volumes of acetonitrile and 75 volumes of a 0.27% w/v (c) Apply 1 ~lL of each solution.
solution of potassiu11l dihyd1'Ogen orthophosphate adjusted to (d) Develop the plate to 15 cm.
pH 5.0 with 2M sodium hyd1'Oxide.
(e) After removal of the plate, dry in air and examine under
SYSTEM SUIT ABILITY u1traviolet 1ight (254 nm).
The test is not valid unless, in the chromatogram obtained MOBILE PRASE
with solution (3), the resolution factor between the first peak
5 volumes of water, 15 volumes of dimethylformamide and
(c1oxacillin) and the second peak (fluc1oxacillin) is at least
75 volumes of ethy1 acetate.
2.5.
CONFIRMATION
DETERMINATION OF CONTENT
In the chromatogram obtained with solution (1):
Ca1culate the content of C19HlSC1N30SS in a container of
average content weight using the dec1ared content of one of the principal spots in the chromatogram obtained with
C 19 H 17 CIN3NaOsS in cloxacillin sodiu11l BPCRS. Each mg of solution (1) corresponds to t11e principal spot in the
C19H17CIN3NaOsS is equivalent to 0.9520 mg of chromatogram obtained with solution (2);
C19HlSC1N30SS. the other principal spot corresponds to the principal spot in
the chromatogram obtained with solution (3).
LABELLING
C. To 5 mL of the filtra te obtained in the Assay for
The quantity of active ingredient is stated in terms of the
equivalent amount of c1oxacillin. sulfadiazine add 10 mL of water and 5 mL of thiobarbituric
acid-citrate buffer, mix and heat on a water bath for
30 minutes. A pink colour is produced.
A1kalinity
Co..trimazine Injection pH, 10.0 to 10.5, Appendix V L.
Trimethoprim and Sulfadiazine Injection ASSAY
Por trimethoprún
Action and use
Extract the chloroform solution reserved in the Assay for
Dihydrofolate reductase inhibitor + sulfonamide antibacterial.
sulfadiazine with three quantities, of 100, 50 and 50 mL, of
DEFINITION 1M acetic acid and dilute the combined extracts to 500 mL
Co-trimazine Injection is a sterile suspension in Water for with 1M acetic acid. To 5 mL add 35 mL of 1M acetic acid
Injections containing Trimethoprim and Sulfadiazine in the and sufficient water to produce 200 mL and measure the
proportion one part to five parts. absorbance of the resulting solution at the maximum at
271 nm, Appendix II B. Ca1culate the content of
PRODUCTION C 1-lH 1SN.jD 3 taking 204 as t11e value of A(1 %, 1 cm) at the
Co-trimazine Injection is prepared by the addition, with maximum at 271 nm.
aseptic precautions, of sterile Trimethoplim to a solution of
For sulfadiazine
sulfadiazine sodium previously sterilised by filtration.
Disperse the trimet110plim evenly t11roughout the injection by
The solution of sulfadiazine sodium is prepared by the
gently inverting the container several times, avoiding the
interaction of Sulfadiazine and Sodium Hydroxide.
formation of foam. Transfer a quantity of the injection
The injection complies with the requirements stated under containing t11e equivalent of 2 g of Sulfadiazine to a
Parenteral Preparations and with the following requirements. separating funnel containing 50 mL of O.lM sodiunz hydroxide
Content of trimethoprim, C14H18N403 and extract with two quantities, of 100 mL and 50 mL, of
90.0 to 110.0% of the stated amount. ch101'Oforl11, washing the extract with the same 25 mL quantity
Content of sulfadiazine, CloHloN402S of O.lM sodiu711 hydroxide. Reserve the combined chloroform
90.0 to 110.0% of the stated amount. extracts for the Assay for trimethoprim.
Dilute the combined aqueous solutions and washings to
CHARACTERISTICS
250 mL with water, filter and dilute 5 mL of the filtrate to
A suspension of an almost white solid in a pale straw-
200 mL with water. Dilute 10 mL uf this solution to 100 rnL
coloured solution.
with water. To 3 mL of the resulting solution add 1 mL of
IDENTIFICATION 2M hydroch1oric acid and 1 mL of a 0.1 % w/v solution of
A. The light absorption, Appendix II B, in the range 250 to sodium nitrite and allow to stand for 2 minutes. Add 1 mL of
325 nm of the solution obtained in the Assay for a 0.5% w/v solution of ammoniu111 sulfamate and allow to
trimethoprim exhibits a maximum only at 271 nm. stand for 3 minutes. Add 1 mL of a 0.1 % w/v solution of
B. Carry out the method for thin-Iayer ch1'Omatography, N - (l-naphthyl) ethy1enediamine dihyd1'Och1oride, allow to stand
Appendix III A, using the following solutions. for 10 minutes, add sufficient water to produce 25 mL and
measure the absorbance of the resulting solution at 538 nm,
(1) Ensure that the injection is homogeneous by gent1y
Appendix II B. Repeat the operation using 3 mL of a
inverting the container several times. To 2.5 mL of the well
solution prepared by dissolving 0.2 g of sulfadiazine BPCRS
mixed injection add 4 mL of hydrochloric acid and dilute to
in 50 mL of O.lM sodium hyd1'Oxide, adding sufficient water to
50 mL with l.4M methanolic ammonia.
produce 200 mL, diluting 5.0 mL of the resulting solution to
(2) 2.0% w/v of sulfadiazine BPCRS in 1.4M methano1ic 250 mL Wit11 water and beginning at the words 'add 1 mL of
ammonza. 2M hydroch1oric acid ... '. CaLculate the content of
(3) 0.4% w/v of trimethoprim BPCRS in 1.4M methano1ic C lO H lON 4 0 2 S in the injection from the absorbances obtained
ammonza. using the dec1ared content of C lOH lON 4 0 2 S in
CHROMATOGRAPHIC CONDITIONS sulfadiazine BPCRS.
(a) Use as the coating silica gel F 254 .
Vet-158 Co-trimazine Preparations 2016

When trimethoprim and sulfadiazine injection is prescribed ASSAY


or demanded, Co-trimazine Injection shall be dispensed or Por trimethoprim
supplied. Extract the combined chIoroform extracts from the Assay for
sulfadiazine with four 50-mL quantities of a 5% v/v solution
of 6M acetic acid. Wash the combined aqueous extracts with
5 mL of chlorofol"m, discard the chIoroform layer and dilute to
250 mL with a 5% v/v solution of 6M acetic acid. Dilute
Co-trimazine Veterinary Oral Powder 20 mL to 100 mL with water. Detelmine the absorbance of
Trimethoprim and Sulfadiazine Veterinary Oral Powder the resulting solution at the maximum at 271 nm,
Appendix II B, and calculate the content of
Action and use C14HlSN403 taking 204 as the value of A(1 %, 1 cm) at the
Dihydrofolate reductase inhibitor + sulfonamide antibacterial. maximum at 271 nm.
DEFINITION Por sulfadiazine
Co-trimazine Veterinary Oral Powder consists of Transfer a quantity of the powder containing 0.125 g of
Trimethoprim and Sulfadiazine in the proportion of one part Sulfadiazine to a separating funnel containing 20 mL of
to five parts mixed with suitable wetting, dispersing and O.lM sodium hydroxide and extract with four 50-rnL quantities
suspending agents. of chlorofol"m. Wash each chloroform extract with the same
two 10-mL quantities of O.lM sodium hydroxide. Combine the
The veterinalY oral powder complies with the requirements stated
aqueous washings and the aqueous layer from the separating
under VeterinalY Oral Powders and with the follo'lving
funnel and reserve the combined chIoroform extracts for the
requirements.
Assay for trimethoprim. Dilute the combined aqueous
Content of trimethoprim, C14H1SN403 solutions to 250 mL with water, filter and dilute 10 mL of
92.5 to 107.5% of the stated amount. the filtrate to 200 mL with water. To 2 mL of the resulting
Content of sulfadiazine, C lOH lON 40 2 S solution add 0.5 mL of 4M hydrochloric acid and 1 mL of a
92.5 to 107.5% ofthe stated amount. 0.1 % w/v solution of sodiul1l nitrite and allow to stand for
2 minutes. Add 1 mL of a 0.5% w/v solution of ammoniul1l
IDENTIFICATION
sulfamate and allow to stand for 3 minutes. Add 1 mL of a
Carry out the method for thin-Iayer chromatography,
0.1 % w/v solution of N-(1-naphthyl)-ethylenediamine
Appendix III A, using the following solutions.
dihydrochlol"ide, allow to stand for 10 minutes and dilute to
(1) Shake a quantity of the powder containing 0.2 g of 25 mL with water. Measure the absorbance of the resulting
Sulfadiazine with sufficient l.4M 711ethanolic ammonia to solution at the maximum at 538 nm, Appendix II B, using in
produce 100 mL, centrifuge and use the supernatant liquido the reference cell a solution prepared at the same time and in
(2) Shake a quantity of the powder containing 0.2 g of the same manner using 2 mL of water and beginning at the
Trimethoprim with sufficient 1.4M methanolic ammonia to words 'add 0.5 mL of 4M hydrochloric acid... '. Calculate the
produce 100 mL, centrifuge and use the supernatant liquido content of ClQHlQN40 2 S from the absorba71ce obtained by
(3) 0.2% w/v solution of sulfadiazine BPCRS in repeating the operation widl 2 mL of a 0.0025% w/v solution
1.4M methanolic ammonia. of sulfadiazine BPCRS in 0.0005M sodiu1J1 hydroxide and
(4) 0.2% w/v solution of trimethoprim BPCRS in beginning at the words 'add 0.5 mL of 4M hydrochloric acid
l.4M methanolic am711onia.
(5) Mix equal volumes of solutions (3) and (5). When trimethoprim and sulfadiazine veterinary oral powder
is prescribed 01' demanded, Co-trimazine Veterinary Oral
CHROMATOGRAPHIC CONDITIONS Powder shall be dispensed 01' supplied.
(a) Use as the coating silica gel GF254 .
(b) Use the mobile phase as described below.
(c) Apply 5 ¡..tL of each solution.
(d) Develop the plate to 15 cm. Co..trimazine Tablets
(e) After removal of the plate, dry in a current of air, spray Trimethoprim and Sulfadiazine Tablets
with a 0.1 % w/v solution of 4-dil11ethylaminobenzaldehyde in a
mixture of 1 mL of hydrochl011C acid and 100 mL of ethanol Action and use
(96%), allow to dry and spray with dilute potassiu111 Dihydrofolate reductase inhibitor' + sulfonamide antibacterial.
iodobismuthate solution.
DEFINITION
MOBILE PHASE
Co-trimazine Tablets contain Trimethoprim and Sulfadiazine
5 volumes of water, 15 volumes of dimethylfol"J11amide and
in the proportion one part to five parts.
75 volumes of ethyl acetate.
The tablets comply with the requirements stated under Tablets and
SYSTEM SUIT ABILITY with the following requirements.
The test is not valid unless the chromatogram obtained with
Content of trimethoprim, C14H1SN403
solution (5) shows two c1ear1y separated spots. 92.5 to 107.5% ofthe stated amount.
CONFIRMATION
Content of sulfadiazine, C lO H lON 40 2 S
The spot in the chromatogram obtained with solution (1) 92.5 to 107.5% ofthe stated amount.
with an Rf value of about 0.7 corresponds to the principal
IDENTIFICATION
spot in the chromatogram obtained with solution (3) and the
spot in the chromatogram obtained with solution (2) with Comply with the test described under Co-trimazine Oral
an Rf value of about 0.3 corresponds to the principal spot in Suspension using solutions prepared in the following manner
the chromatogram obtained with solution (4). as solutions (1) and (2). For solution (1) shake a quantity of
2016 Deltamethrin Preparations Vet-159

the finely powdered tablets containing 0.2 g of Sulfadiazine dl'ugs, Appendix XI F, for eight reflux cycles. Cool and add
with sufficient l.4M methanolie ammonia to produce 100 rnL, sufficient ehloroform to produce 100 rnL. Dilute 5 mL to
centrifuge and use the supernatant liquido For solution (2) 100 mL with absolute ethanol. To 5 rnL add 10 mL of
shake a quantity of the finely powdered tablets containing O.1M hydroehlorie acid and dilute to 100 mL with absolute
0.2 g of Trirnethoprirn with sufficient 1.4M 111ethanolie ethanol. Measure the absorbanee of the resulting solution at
ammonia to produce 100 rnL, centrifuge and use the the maximum at 265 nrn, Appendix II B. Dissolve 50 rng of
supernatant liquido deeoquinate BPCRS in 10 rnL of hot ehloroform and, keeping
ASSAY the solution warm, add slowly 70 rnL of absolute ethanol.
Cool, dilute to 100 mL with absolute ethanol and irnmediately
Weigh and finely powder 20 tablets.
dilute 10 mL to 100 mL with absolute ethanol. To 10 rnL of
For trimethoprim the resulting solution add 10 mL of 0.1 M hydroehlorie acid
Carry out the Assay for trirnethoprirn described under and dilute to 100 mL with absolute ethanol. Calculate the
Co-trimazine Veterinary Oral Powder. content of C24H3SNOs in the premix from the absorbances
For sulfadiazine obtained using the declared content of C24H3SNOs in
Carry out the Assay for sulfadiazine described under deeoquinate BPCRS.
Co-trimazine Veterinary Oral Powder using a quantity of the
powdered tablets containing 0.125 g of Sulfadiazine. Repeat
the operation using 2 rnL of a 0.0025% w/v solution of
sulfadiazine BPCRS in 0.0005M sodium hydroxide beginning at
the words 'add 0.5 mL of 4M hydroehl011e acid ... '. Calculate
Deltamethrin Pour-on
the content of C lOH lON 40 2S in the oral suspension frorn the Action and use
absorbances obtained using the declared content of Insecticide (veterinary).
C lOH lON 4 0 2 S in sulfadiazine BPCRS.
When trimethoprim and sulfadiazine tablets are prescribed or DEFINITION
demanded, Co-trimazine Tablets shall be dispensed or Deltarnethrin Pour-on is a pour-on solution. It contains
supplied. Deltarnethrin in a suitable, oily vehicle.
The pour-on eomplies zuith the requirements stated ullder
Veterinmy Liquid Preparations for Cutaneous Application and
'luith the follozuing requirements.
Decoquinate Premix
Content of deltamethrin, CZZH19BrzN03
Action and use 90.0 to 110.0% of the stated amount.
Antiprotozoal (veterinary). IDENTIFICATION
DEFINITION In the Assay, the chromatogram obtained with solution (1)
shows a peak with the sarne retention time as the peak in the
Decoquinate Prernix contains Decoquinate.
chromatogram obtained with solution (2).
The premix eomplies zuith the requirements stated under Premixes
and zuith the follozuing requirements. ASSAY
Carry out the rnethod for liquid ehro11latography,
Content of decoquinate, CZ4H3SNOs
Appendix 111 D, using the following solutions.
95.0 to 105.0% of the stated amount.
(1) Mix a weighed quantity of the preparation being
IDENTIFICATION examined containing 30 rng of Deltarnethrin with sufficient
Carry out the method for thin-Iayer ehromatography, hexane to produce 100 rnL. Dilute 1 volume to 4 volumes
Appendix III A, using the foIlowing solutions. with hexane.
(1) Heat a quantity containing 0.1 g of Decoquinate with (2) 0.0075% w/v of deltamethrin BPCRS in hexane.
40 mL of ehloroform for 20 minutes on a water bath under a
(3) 0.0075% w/v of deltametl111n imp'U11ty standard BPCRS in
reflux condenser, cool and filter.
hexane.
(2) 0.25% w/v of deeoquinate BPCRS in ehlorofor111.
CHROMATOGRAPHIC CONDITIONS
CHROMATOGRAPHIC CONDITIONS
(a) Use a stainless steel column (25 cm x 4.6 rnm) packed
(a) Use as the coating siliea gel GF254. with particles of silica the surface of which has been modified
(b) Use the rnobile phase as described below. with chemically-bonded nitro-phenyl groups (5 ~lm)
(c) Apply 10 ~lL of each solution. (Nucleosil-N02 is suitable).
(d) Develop the plate to 15 cm. (b) Use isocratic elution and the mobile phase described
(e) After rernoval of the plate, dry in air and examine under below.
ultraviolet light (254 nm). (c) Use a flow rate of 2 mL per minute.
MOBILE PHASE (d) Use an ambient column temperature.
30 volumes of ethanol (96%) and 70 volumes of ehloroform. (e) Use a detection wavelength of 230 nm.
(f) Inject 20 ~L of each solution.
CONFIRMATION
The principal spot in the chrornatograrn obtained with MOBILE PHASE
solution (1) corresponds in position and colour to that in the hexane containing 0.25% v/v, of pi'opan-2-0l.
chrornatogram obtained with solution (2). SYSTEM SUIT ABILITY
ASSAY The test is not valid unless, in the chromatograrn obtained
Heat a quantity containing 0.2 g of Decoquinate with 50 rnL with solution (3), a peak due to (R)-deltamethrin appears
of ehlorofo17n in an apparatus for the eontinuous extraetion of irnmediately before the principal peak, as indicated in the
Vet-160 Diprenorphine Preparations 2016

reference chromatogram supplied with deltamethrin tl7methylchlorosilane and allow to stand for 30 minutes.
impurity standard BPCRS. Prepare solution (2) in a similar manner to solution (3) but
DETERMINATION OF CONTENT omitting the addition of solution A. For solution (3) add
2 mL of 5M ammonia to a volume containing the equivalent
Determine the weight pel' mL of the preparation,
of 6 mg of diprenorphlne and extract with three 7 mL
Appendix V G, and calculate the content of C22H19Br2N03,
quantities of chloroform. To the combined extracts add 2 mL
weight in volume, using the declared content of
of solution A, shake with 2 g of anhydrous sodium sulfate, filter
C22H19Br2N03 in deltamet1117n BPCRS.
and evaporate the filtrate to dryness using a rotary
evaporator; treat the dried residue as described for
solution (1).
The chromatographic procedure may be carried out using a
Diprenorphine Injection glass column (1.5 m x 4 mm) packed with acid-'lvashed,
silanised, diatomaceous support (80 to 100 mesh) coated with
Action and use 2% w/w of methyl silicone gum (SE 30 is suitable) and
Opioid receptor antagonist. maintained at 245 0 •
Calculate the content of C26H3SN04 using the declared
DEFINITION
content of C26H3SN04 in dipren01phine BPCRS.
Diprenorphine Injection is a sterile solution of Diprenorphine
Hydrochloride in Water for Injections containing For an injection containing the equivalent of 0.1 % w/v 01' less of
Methylthioninium Chloride (methylene blue). The pH is dip1'enorphine Carry out the Assay described above, but
adjusted to about 4. dissolving 50 mg of phenolphthalein (internal standard) in
sufficient methanol to produce 100 mL (solution A) and using
The injection complies with the l'equil'ements stated undel'
the following solutions. For solution (1) add 2 mL of
Parenteral Preparations and with the following requirements.
solution A to 5 mL of a 0.027% w/v solution of
Content of diprenorphine, CZ6H3SN04 dip1'enorphine BPCRS in methanol and evaporate the solvent
90.0 to 110.0% of the stated amount. using a rotary evaporator; to the dried residue add 1 mL of a
IDENTIFICATION mixture of 8 volumes of dimethylformamide, 2 volumes of
Carry out the method for thin-Iayer chromatography, N,O-bis(tn'methylsi1yl)acetamide arid 1 volume of
Appendix III A, in subdued light using silica gel GF254 as the trimethylchlorosilane and allow to stand for 30 minutes.
coating substance and a mixture of 10 volumes of Prepare solution (2) in a similar manner to solution (3) but
diethylamine, 20 volumes of ethyl acetate and 70 volumes of omitting the addition of solution A. For solution (3) add
toluene as the mobile phase. Apply separately to each half of 2 mL of 5M am1110nia to a volume containing the equivalent
the plate 1O ~lL of each of the following solutions. of 1.36 mg of diprenorphine and extract with three 10 mL
For solution (1) add 3 mL of 5M ammonia to a volume quantities of chloroform; to the combined extracts add 2 mL
containing the equivalent of 0.5 mg of diprenorphine, mix of solution A, shake with 3 g of anhydrous sodiu111 sulfate, filter
and extract with two 5 mL quantities of chloroform. Combine and evaporate the filtrate to dryness; treat the dried residue
the chloroform extracts, shake with 1 g of anhydrous sodium as described for solution (1).
sulfate, filter, evaporate to dryness using a rotary evaporator LABELLING
and dissolve the residue in 0.4 mL of chloroform. Solution (2) The quantity of active ingredient is stateel in terms of the
contains 0.115% w/v of dipren01phine BPCRS in chloroform. equivalent amount of diprenorphine in a suitable dose-
Add to each point of application of solution (2) 1O ~lL of a volume.
mixture of 4 volumes of methanol and 1 volume of
STORAGE
13.5M a11lmonia. After removal of the plate, allow it to dry in
air and examine under ultraviolet light (254 111n). Spray one Diprenorphine Injection should be protected from light.
half of the plate with a mixture of 5 volumes of chloroplatinic
acid solution, 35 volumes of dilute potassium iodide solution and
60 volumes of acetone. Spray the other half of the plate with a
mixture of 1 volume of ira n (m) chloride solution Rl and Etamiphylline Injection
1 volume of dilute potassium hexacyanoferrate(m) solution.
The spot in the chromatogram obtained with solution (1) Action and use
corresponds in position, quenching and colour to that in the Non-selective phosphodiesterase inhibitor (xanthine);
chromatogram obtained with solution (2). treatment of reversible airways obstruction.
TESTS
DEFINITION
Acidity
pH, 3.5 to 4.5, Appenrux V L. Etamiphylline Injection is a sterile solution of Etamiphylline
Camsilate in Water for Injections.
ASSAY The injection complies with the requi1'ements stated under
For an injection containing the equivalent of more than 0.1 % w/v Pa1'enteral P1'eparations and with the following 1'equi1'ements.
of dipren01phine Dissolve 50 mg of phenolphthalein (n1ternal
standard) in sufficient methanol to produce 20 mL (solution Content of etamiphyIline camsilate,
A). Carry out the method for gas chromatography, C13Hz1NsOz, ClOH1604 S
Appendix III B, using the following solutions. For solution 95.0 to 105.0% afthe stated amount.
(1) add 2 mL of solution A to 2 mL of a 0.30% w/v solution IDENTIFICATION
of dipren01phine BPCRS in methanol and evaporate the solvent Prepare a quantity of the residue as described in the Assay.
using a rotary evaporator. To the dried residue add 1 mL of The residue complies with the following tests.
a mixture of 8 volumes of dimethylformamide, 2 volumes of
N, O-bis (tl7methylsi1yl) acetamide and 1 vol ume of
2016 Etorphine Preparations Vet-161

A. The infrared abs01ption specmlJn, Appendix II A, is A. The iJ1jrared abs01ption spectrum, Appendix II A, is
concordant with the reference spectrum of etamiphylline concordant with the reference spectrum of etamiphyl1ine
(RSV 19). (RSV 19).
B. Yields the reactions characteristic of xanthines, B. Yields the reactions characteristic of xanthines,
Appendix VI. Appendix VI.
TESTS Related substances
Acidity Carry out the method for thin-layer chromatography,
pH, 3.9 to 5.4, Appendix V L. Appendix III A, using the following solutions.
Related substances (1) Shake a quantity of the powdered tablets containing 0.2 g
Carry out the method for thin-laye/' chromatography, of Etamiphylline Camsilate with 5 mL of methanol and
Appendix lIT A, using the following solutions. centrifuge.
(1) Dilute the injection with sufficient water to produce a (2) Dilute 1 volume of solution (1) to 500 volumes with
solution containing 3.5% w/v of Etamiphylline Camsilate. methanol.
(2) Dilute 1 volume of solution (1) to 500 volumes with CHROMATOGRAPHIC CONDITIONS
water. (a) Use as the coating silica gel HF254 .
CHROMATOGRAPHIC CONDITIONS (b) Use the mobile phase as described below.
(a) Use as the coating silica gel HF254. (c) Apply 10 ~lL of each solution.
(b) Use the mobile phase as described below. (d) Develop the plate to 15 cm.
(c) Apply 10 ~L1 of each solution. (e) After removal of the plate, allow it to dry in air and
(d) Develop the plate to 15 cm. examine under ultraviolet light (254 nm).
(e) After removal of the plate, allow it to dry in air and MOBILE PHASE
examine under ultraviolet light (254 nm). 1 volume of 13.5M ammonia, 20 volumes of ethanol (96%)
MOBILE PHASE and 80 volumes of chloroform.
1 volume of 13.5M ammonia, 20 volumes of ethanol (96%) LIMITS
and 80 volumes of chloroform. Any seconda1JI spot in the chromatogram obtained with
LIMITS solution (1) is not more intense than the spot in the
chromatogram obtained with solution (2) (0.2%).
Any seconda1y spot in the chromatogram obtained with
solution (1) is not more intense than the spot in the ASSAY
chromatogram obtained with solution (2) (0.2%). Weigh and powder 20 tablets. Dissolve a quantity of the
powder containing 0.5 g of Etamiphylline Camsilate in
ASSAY
30 mL of water, make alkaline with 5M ammonia and extract
To a volume containing 0.7 g of Etamiphylline Camsilate
with tln-ee 25-mL quantities of chloroform, washing each
add 15 mL of water, make alkaline with 5M ammonia and
chloroform extract with the same 5-smL quantity of water.
extract with three 25-mL quantities of chlorofo17n, washing
Evaporate the combined chloroform extracts to dlyness,
each extract with the same 5-mL quantity of water. Evaporate
dissolve the residue in 25 mL of water and titrate with
the combined extracts to dryness, dissolve the residue in
0.05M sulfuric acid VS using bromocresol green solution as
25 mL of water and titrate with 0.05M sulfuric acid VS using
indicator. Each mL of 0.05M sulfuric acid VS is equivalent to
bromocresol green solution as indicator. Each mL of
51.16 mg of C13HzINsOz,ClOH1604S,
0.05M sulfuric acid VS is equivalent to 51.16 mg of
C13HzINsOz,CIOH1604S,

Etorphine and Acepromazine Injection


Etamiphylline Tablets Action and use
Opioid receptor agonist; analgesic.
Action and use
Non-selective phosphodiesterase inhibitor (xanthine); DEFINITION
treatment of reversible airways obstruction. Etorphine and Acepromazine Injection is a sterile solution of
Etorphine Hydrochloride and Acepromazine Maleate in
DEFINITION
Water for Injections containing a suitable antimicrobial
Etamiphylline Tablets contain Etamiphylline Camsilate. They
preservative. The pH is adjusted to about 4.
are coated.
The injection complies with the requirements stated under
The tablets comply with the requirements stated Linda T ablets and
Parenteral Preparations and 'luith the follo'luing requirements.
with the follo'luing requirements.
Content of etorphine hydrochloride, C2sH33N04,HCI
Content of etamiphylline camsilate,
0.22 to 0.27% w/v.
C13H21Ns02,ClOH1604S
95.0 to 105.0% of the stated amount. Content of acepromazine ma1eate, C19H22N20S,C4H404
0.90 to 1.10% w/v.
IDENTIFICATION
Prepare a quantity of the residue as described in the Assay. CHARACTERISTICS
The residue complies with the following tests. A c1ear, yellow solution.
Vet-162 Fenbendazole Preparations 2016

IDENTIFICATION For acepromazine maleate


A. The light abs01ption, Appendix II B, in the range 230 to Protect the solutions from light throughout the Assay.
350 run of a solution prepared by diluting a volume of the To 1 mL add sufficient ethanol (96%) to produce 100 mL.
injection containing 10 mg of Acepromazine Maleate to Measure the absorbance, Appendix II B, of the resulting
500 mL with zuater exhibits well-defined maxima at 242 nm solution at the maximum at 370 nm. Calculate the content
and at 280 run. The absorbances at the maxima are about 1.1 of C19H22N20S,C4H404 taking 46.1 as the value of
and about 0.85, respectively. A(l %, 1 cm) at the maximum at 370 nm.
B. Carry out the method for thin-layer chromatography, STORAGE
Appendix III A, in subdued light using silica gel GF254 as the Etorphine and Acepromazine Injection should be protected
coating substance and ethyl acetate as the mobile phase and from light.
applying separately to each half of the plate 1O ~lL of each of
two solutions containing (1) the injection being examined
and (2) 0.09% w/v of dipren01phine BPCRS in chlorojorm.
Add to each point of application 1O ~lL of a mixture of
4 volumes of methanol and 1 volume of 13.5M anz111onia. After Fenbendazole Granules
removal of the plate, allow it to dry in air and examine under
ultraviolet light (254 nm). Spray one half of the plate with a Action and use
mixture of 5 volumes of chloroplatinic acid solution, Antihelminthic.
35 volumes of dilute potassium iodide solution and 60 volumes
of acetone. Spray the other half of the plate with a mixture of DEFINITION
1 volume of iron(m) chlorzde solution Rl and 1 volume of Fenbendazole Granules contain Fenbendazole mixed with
dilute potassium hexacyanojerrate(m) solution. The principal suitable diluents.
spot in the chromatogram obtained with solution (1) has The granules comply zuith the requirements stated u11der Granules
an Rf value of about 1.15 relative to that of the spot in the and zuith the jollo'lui11g requirements.
chromatogram obtained with solution (2), absorbs ultraviolet Content of fenhendazole, ClsH13N302S
light and yields a reddish violet colour with the iodoplatinate 95.0 to 105.0% of the stated amount.
spray reagent and a blue colour with the iron-
hexacyanoferrate spray reagent.
IDENTIFICATION
A. In the Assay, the retention time of the principal peak in
TESTS the chromatogram obtained with solution (1) is the same as
Acidity that of the principal peak in the chromatogram obtained with
pH, 3.5 to 4.5, Appendix V L. solution (2).
ASSAY B. Carry out the method for thin-layer chromatography,
For etorphine hydrochloride Appendix III A, using the following solutions.
Dissolve 46 mg of dipre7101phine BPCRS (internal standard) in (1) Mix with the aid of ultrasound a quantity of the
sufficient 7nethanol to produce 10 mL (solution A). Carry out powdered granules containing 80 mg of Fenbendazole with
the method for gas chromatography, Appendix III B, using the 80 mL of O.lM metha110lic hydrochloric acid for 90 minutes,
following solutions. For solution (1) add 1 mL of solution A cool, dilute to 100 mL with O.lM methanolic hydrochloric acid,
to 2 mL of the injection and 2 mL of 5M am7nonia, extract mix, filter through a 0.4-~m filter (Whatman GF/C is
with three 7-mL quantities of chlorojorm, shake the combined suitable) and use the filtrate.
extracts with 2 g of anhydrous sodium sulfate, filter and (2) 0.08% w/v ofjenbe11dazole BPCRS in O.lM metha110lic
evaporate the filtrate to a small volume using a rotary hydrochloric acid.
evaporator. Transfer the contents to a stoppered test tube
CHROMATOGRAPHIC CONDITIONS
with the aid of 2 mL of chlorojorm and evaporate to dryness.
To the residue add 1 mL of a mixture of 8 volumes of (a) Use a TLC silica gel F 2S4 plate (Merck silica gel F 2S4
dim.ethyiformamide, 2 volumes of N,O-bis(trimethylsily1)- plates are suitable).
acetamide and 1 volume of trimethylchlorosilane and allow to (b) Use the mobile phase as described below.
stand for 15 minutes. For solution (2) treat a volume of the (c) Apply 5 ~lL of each solution.
injection containing 4.9 mg of Etorphine Hydrochloride in a
(d) Develop the plate to 10 cm.
similar manner but omitting the addition of solution A.
Protect the solutions from light during preparation and (e) After removal of the plate, dry in air for 10 minutes, heat
subsequent use. at 100 0 for 5 minutes and examine under ultraviolet light
(254 11m and 365 mn).
The chromatographic procedure may be carried out using a
glass column (1.5 m x 4 mm) packed with acid-zuashed) MOBILE PHASE
silanised diatomaceous support (80 to 100 mesh) coated with 2.5 volumes of water, 6.5 volumes of acetone, 26 volumes of
2% w/w of methyl silicone gum (SE 30 is suitable) and 13.5M ammo11ia and 65 volumes of toluene.
0
maintained at 245 •
CONFIRMATION
Calculate the content of C2sH33N04 using the declared By each method of visualisation the principal spot in the
content of C26H3SN04 in diprenOlphine BPCRS assuming chromatogram obtained with solution (1) corresponds to that
that the trimethylsilyl derivatives of equal weights of in the chromatogram obtained with solution (2).
etorphine and diprenorphine have the same response in the
fiame ionisation detector and hence calculate the content of TESTS
C2sH33N04,HCl taking each mg of C2sH33N04 to be Re1ated impurities A, B and 1
equivalent to 1.089 mg of C2sH33N04,HCI. Carry out the method for liquid chromatography,
Appendix III D, using the following solutions.
2016 Fenbendazole Preparations Vet-163

(1) Mix, with the aid of ultrasound, a quantity of the (2) 0.01 % w/v ofjenbendazole BPCRS in a mixture of
powdered granules containing 0.1 g of Fenbendazole with 1 volume of O.lM hydrochloric acid and 1 volume of methanol
50 mL of O.lM methanolic hydrochloric acid for 30 minutes, (85%).
cool, dilute to 100 mL with methanol (65%), mix and filter CHROMATOGRAPHIC CONDITIONS
through a glass-fibre filter (Whatman GF/C is suitable).
The chromatographic procedure described under Related
(2) Dilute 1 volume of a 0.001 % w/v solution of jenbendazole substances may be used.
impurity A EPCRS (methyl (1H-benzimidazol-2-
yl)carbamate) in O.lM methanolic hydrochloric acid to DETERMINATION OF CONTENT
2 volumes with methanol (65%). Calculate the content of ClsH13N302S in the granules using
(3) Dilute 1 volume of a 0.001 % w/v solution ofjenbendazole the declared content of ClsH13N302S, in
impurity B EPCRS (methyl (5-chloro-1H-benzimidazol-2- jenbendazole BPCRS.
yl)carbamate) in O.lM methanolic hydrochloric acid to IMPURITIES
2 volumes with 111ethanol (65%). The impurities limited by the requirements of this
(4) Dilute 1 volume of a 0.0010% w/v solution of monograph include impurities A and B listed under
jenbendazole impurity 1 BPCRS (5-phenylthio)-2- Fenbendazole and the following:
amino benzimidazole) in 0.1 M methanolic hydrochlonc acid to
2 volumes with methanol (65%).
(5) Dilute 1 volume of a solution containing 0.002% w/v
each of jenbendazole impurity A EPCRS, jenbendazole
i711purity B EPCRS, jenbendazole impurity 1 BPCRS and
0.20% w/v oj jenbendazole BPCRS in O.lM methanolic
hydrochloric acid to 2 volumes with methanol (65%).
1. (5-phenylthio )-2-aminobenzimidazole.
CHROMATOGRAPHIC CONDITIONS
(a) Use a stainless steel column (25 cm x 4.6 mm) packed
with octadecylsilyl silica gel jor chromatography (5 ~lm)
(Nucleosil C18 is suitable).
(b) U se isocratic elution and the mobile phase described
Fenbendazole Oral Suspension
below. Action and use
(c) Use a flow rate of 1 mL per minute. Antihelminthic.
(d) Use an ambient column temperature.
DEFINITION
(e) Use a detection wavelength of 280 nm.
Fenbendazole Oral Suspension is an aqueous suspension of
(f) Inject 20 ~lL of each solution. Fenbendazole.
MOBILE PHASE The oral suspension complies with the require71le7lts stated under
350 volumes of a 0.5% w/v solution of sodium dihydrogen Oral Liquids and with the jollowing requirements.
orthophosphate and 650 volumes of methanol containing 1.88 g Content of fenbendazole, ClsH13N302S
of sodium hexanesulfonate, the pH of which has been adjusted 95.0 to 105.0% of the stated amount.
to 3.5 with orthophosphoric acid.
IDENTIFICATION
SYSTEM SUIT ABILITY
In the Assay, the retention time of the principal peak in the
The test is not valid unless the chromatogram obtained with chromatogram obtained with solution (1) is the same as that
solution (5) closely resembles the reference chromatogram of the principal peak in the chromatogram obtained with
supplied with jenbendazole BPCRS. solution (2).
LIMITS TESTS
In the chromatogram obtained with solution (1): Related impurities A, B and 1
the areas of any peaks corresponding to fenbendazole Carry out the method for liquid chromatography,
impurity A, fenbendazole impurity B and fenbendazole Appendix III D, using the following solutions.
impurity 1 (5-(phenylthio)-2-aminobenzimidazole) are not (1) Mix with the aid of ultrasound, a quantity of the oral
greater than the areas of the corresponding peaks in the suspension containing 0.1 g of Fenbendazole with 50 mL of
chromatograms obtained with solutions (2), (3) and (4) O.lM methanolic hydrochloric acid for 30 minutes, cool, dilute
respectively (0.5% each). to 100 mL with methanol (65%), mix and filter through a
ASSAY glass-fibre filter (Whatman GF/C is suitable).
Carry out the method for liquid chromatography, (2) Dilute 1 volume of a 0.001 % w/v solution ofjenbendazole
Appendix III D, using the following solutions. impunty A EPCRS (methyl (lH-benzimidazol-2-
(1) Mix with the aid of ultrasound a quantity of the yl) carbamate) in 0.1 M methanolic hydrochloric acid to
powdered granules containing 0.1 g of Fenbendazole with 2 volumes with methanol (65%).
50 mL of O.lM methanolic hydrochloric acid for 30 minutes, (3) Dilute 1 volume of a 0.001 % w/v solution ofjenbendazole
cool, dilute to 100 mL with methanol (65%), mix, filter impunty B EPCRS (methyl(5-chloro-1H-benzimidazol-2-
through a glass-fibre filter (Whatman GF/C is suitable). yl) carbamate) in 0.1 M methanolic hydrochloric acid to
Dilute 5 volumes of the resulting solution to 50 volumes with 2 volumes with methanol (65%).
0.1 M hydrochloric acid in methanol (85%). (4) Dilute 1 volume of a 0.001 % w/v solution ofjenbendazole
impurity 1 BPCRS ((5-phenylthio)-2-aminobenzimidazole) in
O.lM methanolic hydrochloric acid to 2 volumes with methanol
(65%).
V et-164 F enbendazole Preparations 2016

(5) Dilute 1 volume of a solution containing 0.002% w/v DETERMINATION OF CONTENT


each of jenbendazole i111p'wity A EPCRS) jenbendazole Calculate the content of ClsH13N302S in the oral
impurity B EPCRS) jenbendazole impwity 1 BPCRS and suspension from the chromatogram obtained and using the
0.20% w/v ofjenbendazole BPCRS in O.lM 11lethanolic dec1ared content of ClsH13N302S injenbendazole BPCRS.
hydrochloric acid to 2 volumes with methanol (65%).
IMPURITIES
CHROMATOGRAPHIC CONDITIONS The impurities limited by the requirements of this
(a) Use a stainless steel column (25 cm x 4.6 mm) packed monograph inc1ude impurities A and B listed under
with octadecylsilyl silica gel jor chromatography (5 ~lm) Fenbendazole and the following:
(Nuc1eosil C 18 is suitable).
(b) U se isocratic elution and the mobile phase described
below.
(c) Use a flow rate of 1 mL per minute.
(d) Use an ambient column temperature.
(e) Use a detection wavelength of 280 nm.
(f) Inject 20 ~lL of each solution. 1. (5-phenylthio)-2-aminobenzimidazole.
MOBILE PRASE
350 volumes of a 0.5% w/v solution of sodium dihydrogen
orthophosphate and 650 volumes of methanol containing 1.88 g
of sodium hexanesulfonate, the pH of which has been adjusted
to 3.5 with orthophosphonc acid.
Fenbendazole Veterinary Oral Paste
Fenbendazole Veterinary Paste
SYSTEM SUITABILITY
The test is not valid unless the chromatogram obtained with Action and use
solution (5) c10sely resembles the reference chromatogram Antihelminthic.
supplied with jenbendazole BPCRS.
DEFINITION
LIMITS
Fenbendazole Veterinary Oral Paste contains Fenbendazole
In the chromatogram obtained with solution (1): finely dispersed in a suitable basis.
the area of any peak corresponding to fenbendazole The vetelinmy oral paste complies with the requirements stated
impurity A (methyl (1H-benzimidazol-2-yl)carbamate) is not under Vetelinary Oral Pastes and with the jollo'luing requirements.
greater than the area of the corresponding peak in the
Content of fenbendazole, ClsH13N302S
chromatogram obtained with solution (2), (0.5%);
95.0 to 105.0% ofthe stated amount.
the area of any peak corresponding to fenbendazole
impurity B (methyl(5-chloro-1H-benzimidazol-2- IDENTIFICATION
yl)carbamate) is not greater than the area of the In the Assay, the retention time of the principal peak in the
corresponding peak in the chromatogram obtained with chromatogram obtained with solution (1) is the same as that
solution (3) (0.5%); of the principal peak in the chromatogram obtained with
solution (2).
the area of any peak corresponding to fenbendazole
impurity 1 ((5-phenylthio)-2-aminobenzimidazole) is not TESTS
greater than the area of the corresponding peak in the Related impurities A, B and 1
chromatogram obtained with solution (4) (0.5%). Carry out the method for liquid chromatography,
ASSAY Appendix ITI D, using the following solutions.
Carry out the method for liquid chromatography, (1) Mix with the aid of ultrasound, a quantity of the oral
Appendix lIT D, using the following solutions. paste containing 0.1 g of Fenbendazole with 50 mL of
O.lM methanolic hydrochloric acid for 30 minutes, cool, dilute
(1) Mix with the aid of ultrasound a quantity of the oral
to 100 mL with methanol (65%), mix, filter through a glass-
suspension containing 0.1 g of Fenbendazole with 50 mL of
fibre filter (Whatman GF/C is suitable).
O.lM methanolic hydrochlonc acid for 30 minutes, cool, dilute
to 100 mL with methanol (65%), mix and filter through a (2) Dilute 1 volume of a 0.001 % w/v solution ofjenbendazole
glass-fibre filter (Whatman GF/C is suitable). Dilute impurity A EPCRS (methyl (1H-benzimidazole-2-
5 volumes of the resulting solution to 50 volumes with yl)carbamate) in O.lM methanolic hydrochlonc acid to
O.lM hydrochloric acid in 111ethanol (85%). 2 volumes with methanol (65%).
(2) 0.01 % w/v ofjenbendazole BPCRS in a mixture of (3) Dilute 1 volume of a 0.001 % w/v solution ofjenbendazole
1 volume of O.lM hydrochlO1ic acid and 1 volume of methanol impurity B EPCRS (methyl (5-chloro-1H-benzimidazole-2-
(85%). yl)carbamate) in O.lM methanolic hydrochlonc acid to
2 volumes with methanol (65%).
CHROMATOGRAPHIC CONDITIONS
(4) Dilute 1 volume ofa 0.001% w/v solution ofjenbendazole
The chromatographic conditions described under Related impunty 1 BPCRS((5-phenylthio)-2-aminobenzimidazole) in
substances may be used. O.lM methanolic hydrochloric acid to 2 volumes with methanol
SYSTEM SUIT ABILITY (65%).
The test is not valid unless the chromatogram obtained with (5) Dilute 1 volume of a solution containing 0.002% w/v
solution (5) c10sely resembles the reference chromatogram each of jenbendazole impunty A EPCRS) jenbendazole
supplied with jenbendazole BPCRS. impwity B EPCRS) jenbendazole impurity 1 BPCRS and
0.20% w/v ofjenbendazole BPCRS in O.lM methanolic
hydrochlonc acid to 2 volumes with methanol (65%).
2016 Gonadotrophin Preparations Vet-165

CHROMATOGRAPHIC CONDITIONS
(a) Use a stainless steel column (25 cm x 4.6 mm) packed
Serum Gonadotrophin Injection
with octadecylsilyl silica gel jor chromatography (5 ~lm) Action and use
(Nucleosil C18 is suitable). Equine serum gonadotrophin.
(b) U se isocratic elution and the mobile phase described
below. DEFINITION
(c) Use a fiow rate of 1 mL per minute. Serum Gonadotrophin Injection is a sterile solution of Serum
Gonadotrophin in Water for Injections. It is prepared by
(d) Use an ambient column temperature.
dissolving Serum Gonadotrophin for Injection in the requisite
(e) Use a detection wavelength of 280 nm. amount of Water for Injections immediately before use.
(f) Inject 20 ~lL of each solution. The injection complies with the require111ents stated under
MOBILE PHASE Parenteral Preparations.
350 volumes of a 0.5% w/v solution of sodium dihydrogen STORAGE
orthophosphate and 650 volumes of methanpl containing 1.88 g Serum Gonadotrophin Injection should be used immediately
of sodium hexanesulfonate, the pH of which has been adjusted after preparation.
to 3.5 with orthophosphoric acid
SYSTEM SUIT ABILITY
SERUM GONADOTROPHIN FOR
The test is not valid unless the chromatogram obtained with INJECTION
solution (5) closely resembles the reference chromatogram
supplied with jenbendazole BPCRS. DEFINITION
Serum Gonadotrophin for Injection is a sterile material
LIMITS
consisting of Serum Gonadotrophin with or without
In the chromatogram obtained with solution (1): excipients. It is supplied in a sealed container.
the area of any peaks corresponding to fenbendazole The contents oj the sealed container comply with the requirements
impurity A (methyl (IH-benzimidazol-2-yl)carbamate), jor Powders jor Injections 01" Injusions stated unda Parenteral
fenbendazole impurity B (methyl (5-chloro-1H-benzimidazol- Preparations and with the jollowing requirements.
2-yl)carbamate) and fenbendazole impurity 1
Potency
((5-phenylthio)-2-aminobenzimidazole) are not greater than
For each container tested, the estimated potency is not less
the areas of the corresponding peaks in the chromatograms
than 80% and not more than 125% of the stated potency
obtained with solutions (2), (3) and (4) respectively (0.5%
each). CHARACTERISTICS
ASSAY A white or pale grey, amorphous powder.
Carry out the method for liquid chromatograph:y, Soluble in water.
Appendix III D, using the following solutions. IDENTIFICATION
(1) Mix with the aid of ultrasound a quantity of the oral Causes enlargement of the ovaries of immature female rats
paste containing 0.1 g ofFenbendazole with 50 mL of when administered as directed in the Assay.
O.lM methanolic hydrochloric acid for 30 minutes, cool, dilute
TESTS
to 100 mL with methanol (65%), mix, filter through a glass-
Clarity, colour and acidity or aIkalinity of solution
fibre filter (Whatman GF/C is suitable). Dilute 5 volumes of
A solution containing 5000 IU per mL is clear,
the resulting solution to 50 volumes with O.lM hydrochloric
Appendix IV A, Method 1, and colourless, Appendix IV B,
acid in methanol (85%).
Method 1, and has a pH of 6.0 to 8.0, Appendix V L.
(2) 0.01 % w/v ofjenbendazole BPCRS in a mixture of
1 volume of O.IM hydrochloric acid and 1 volume of methanol Water
(85%). Not more than 10.0% w/w, Appendix IX C. Use 80 mg.
Bacterial endotoxins
CHROMATOGRAPHIC CONDITIONS
Carry out the test jor bacterial endotoxins, Appendix XIV C,
The chromatographic conditions described under Related using Method C. Dissolve the contents of the sealed
impurities A, B and 1 may be used. container in water BET to give a solution containing 1000 IU
DETERMINATION OF CONTENT of serum gonadotrophin per mL (solution A). The endotoxin
Calculate the content of ClsH13N302S in the veterinary oral limit concentration of solution A is 35 IU of endotoxin per
paste from the chromatograms obtained and using the mL. Carry out the test using a suitable dilution of solution A
declared content of ClsH13N302S in jenbendazole BPCRS. as described under Method C.
IMPURITIES ASSAY
The impurities limited by the requirements of this Carry out the Assay described under Serum Gonadotrophin.
monograph include impurities A and B listed under The fiduciallimits of error are not less than 64% and not
Fenbendazole and the following: more than 156% of the stated potency.
STORAGE
The sealed container should be protected from light and
stored at a temperature not exceeding 8°.

1. (5-phenylthio)-2-aminobenzimidazole.
Vet-166 Griseofulvin Preparations 2016

11 minutes. The retention times relative to griseofulvin are:


Griseofulvin Premix dechlorogriseofulvin, about 0.6; dehydrogriseofulvin, about
Action and use 1.4.
Antifungal. LIMITS
U sing the chromatogram obtained with solution (3), calculate
DEFINITION the ratio (R) of the area of the peak due to griseofulvin to the
Griseofulvin Premix contains Griseofulvin. The partic1es of area of the peak due to the intemal standard.
Griseofulvin are generally up to 5 ~lm in maximum
In the chromatogram obtained with solution (2):
dimension, although larger partic1es, which may occasionally
exceed 30 ~lm, may be presento the ratio of the area of any peak due to dechlorogriseofulvin
to the area of the peak due to the internal standard is not
The premix complies 'lvitll the requirements stated under Premixes
greater than 0.6R (3%);
alld zuith the follozuing requirements.
the ratio of the area of any peak due to dehydrogriseofulvin
Content of griseofulvin, C17H17CI06
to the area of the peak due to the internal standard is not
90.0 to 110.0% ofthe stated amount.
greater than 0.15R (0.75%).
IDENTIFICATION
ASSAY
A. Shake a quantity of the premix containing 0.1 g of
To a quantity containing 35 mg of Griseofulvin add 60 mL
Griseofulvin with 10 mL of chloroform. Centrifuge, decant the
of ethyl aceta te, mix, heat to 60 0 and shake for 15 minutes.
supernatant liquid, dry with anhydrous sodium sulfate and
Allow to cool and dilute to 100 mL with ethyl acetate.
evaporate the chlorofOlID. The infrared abs01ption spectrum of
Centrifuge and transfer two 5 mL aliquots of the clear
the residue, Appendix n A, is concordant with the reference
supernatant liquid into separate 100 mL graduated flasks.
spectrum of griseofulvin (RSV 24).
To the first flask add 5.0 mL of a 13% v/v solution of
B. Shake a quantity of the premix containing 80 mg of 0
711ethanesulfonic acid in methanol, allow to stand at 20 for
Griseofulvin with 150 mL of ethanol (96%). Dilute to 30 minutes and dilute to 100 mL with methanol (solution A).
200 mL with ethanol (96%) and centrifuge. Dilute 2 mL of Dilute the contents of the second flask to 100 mL with
the supernatant liquid to 100 mL with ethanol (96%). The 711ethanol (solution B). To a third flask add 5.0 mL of the
light abs01ption of the resulting solution, Appendix II B, in the methanolic methanesulfonic acid solution and dilute to
range 240 to 350 nm, exhibits two maxima at 291 nm and at 100 mL with methanol (solution C). Measure the absorbance
325 nm and a shoulder at 250 nm. of each solution at 266 nm, Appendix II B. Calculate the
e. Dissolve 5 mg of the residue obtained in test A in 1 mL content of C 17H 17 Cl0 6 from the difference between the
of sulfuric acid and add 5 mg of powdered potassium absorbance obtained with solution A and the sum of the
dichromate. A wine red colour is produced. absorbances obtained with solutions B and C and from the
Related substances difference obtained by repeating the operation using 35 mg
Carry out the method for gas chromatography, of griseofulvin BPCRS in place of the preparation being
Appendix nI B, using the following solutions. Dissolve examined and from the declared content of C 17 H 17 Cl0 6 in
5 O mg of 9) 1O-diphenylanthracene (internal standard) in griseofulvin BPCRS.
sufficient chloroform to produce 50 mL (solution A).
(1) Add 60 mL of chloroform to a quantity of the premix
containing 50 mg of Griseofulvin, heat at 60 0 with shaking
for 20 minutes, cool and dilute to 100 mL with chloroform. Ivermectin Injection
Centrifuge and evaporate 20 mL of the supernatant liquid to
about 1 mL. Action and use
(2) Prepare solution (2) in the same manner as solution (1) Antihelminthic.
but adding 1 mL of solution A before diluting to 100 mL
DEFINITION
with chloroform.
Ivermectin Injection is a sterile solution of Ivermectin in a
(3) Dissolve 5 mg ofgriseofulvin BPCRS in chloroform, add suitable non-aqueous vehicle.
2 mL of solution A and sufficient chloroform to produce
The injection complies zuith the requirements stated under
200 mL. Evaporate 20 mL of the solution to about 1 mL.
Parenteral Preparations and zuith the follozuing requirements.
CHROMATOGRAPHIC CONDITIONS
Content of ivermectin, calculated as the sum of
(a) Use a glass column (1 m x 4 mm) packed with acid- component H 2B 1a (C4sH74014) and component H2Blb
zuashed) silanised diatomaceous support (100 to 120 mesh) (C47Hn014)
coated with 1% w/w of cyanopropylmethyl phenylmethyl 95.0 to 105.0% of the stated amount.
silicone fluid (OV 225 is suitable).
The ratio of the contents H 2 B 1a / (H2 B 1a + H 2 B 1b) is at least
(b) Use nitrogen as the carrier gas at a flow rate of 50 to 90.0%.
60 mL per minute.
IDENTIFICATION
(c) Use isothermal conditions maintained at 250°.
A. Carry out the method for thin-Iayer chromatography,
(d) Use an inlet temperature of 270°. Appendix nI A, using a silica gel 60 F 254 precoated plate
(e) Use a flame ionisation detector at a temperature of 300°. (Merck silica gel 60. F 254 plates are suitable) and a mixture of
(f) Inject a suitable volume of each solution. 1 volume of concentrated a11l11Zonia Rl, 9 volumes of methanol
(g) For solution (1), allow the chromatography to proceed and 90 volumes of dichloromethane as the mobile phase.
for 3 times the retention time of griseofulvin. Apply separately to the plate 2 ~lL of each of the following
solutions. For solution (1) dissolve a volume of the injection
When the chromatograms are recorded under the prescribed
in sufficient methanol to produce a solution containing
conditions the retention time of griseofulvin is about
0.05% w/v of Ivermectin; filter if necessary. Solution (2)
2016 Ivermectin Preparations Vet-167

contains 0.05% w/v of ivermectin BPCRS in methanol. After


removal of the plate, allow it to dry in air and examine under
Ivermectin Oral Solution
ultraviolet Hght (254 12m and 366 nm). The principal spot in Action and use
the chromatogram obtained with solution (1) is similar in Antihelminthic.
position, colour and size to that in the chromatogram
obtained with solution (2). DEFINITION
B. In the Assay, the chromatogram obtained with solution Ivermectin Oral Solution is a solution of Ivermectin in a
(1) shows two principal peaks with retention times similar to suitable vehicle.
those of the two principal peaks in the chromatogram The oral solution complies with the requiremems stated unda Oral
obtained with solution (2). Liquids and with the following requirements.
TESTS Content of ivermectin, calculated as the sum of
Clarity and colour of solution component H 2B 1a (C4sH74014) and component H2Blb
The injection is clear, Appendix IV A, and not more intensely (C47Hn014)
coloured than reference solution Y4' Appendix IV B, 95.0 to 105.0% of the stated amount.
Method n. The ratio of the contents H 2 B 1a / (H 2 B 1a + H 2 B 1b) is at least
Related substances 90.0%.
Carry out the method for liquid chromatography, IDENTIFICATION
Appendix lIT D, using the following solutions. For solution
A. Carry out the method for thin-layer chromatography,
(1) dissolve a volume of the injection in sufficient methanol to
Appendix lIT A, using the following solutions.
produce a solution containing 0.04% w/v of I~ermectin.
Solutions (2), (3) and (4) contain 0.04% w/v, 0.0004% w/v (1) Dilute a quantity of the oral solution with sufficient
methanol to produce a solution containing 0.05% w/v of
and 0.00002% w/v respectively of ivermectin BPCRS in
methanol. Inject 20 ~lL of each solution.
Ivermectin.
The chromatographic procedure may be calTied out using (2) 0.05% w/v of ivermectin BPCRS in methanol.
(a) a stainless steel column (25 cm x 4.6 mm) packed with CHROMATOGRAPHIC CONDITIONS
octadecylsilyl silica gel for chromatography (5 ~lm) (Apex ODS 1 (a) Use as the coating silica gel F 254 (Merck silica gel 60 F 254
is suitable), (b) as the mobile phase at a flow rate of 1.5 mL plates are suitable).
per minute a mixture of 39 volumes of water, 55 volumes of (b) Use the mobile phase as described below.
methanol and 106 volumes of acetonitrile and (c) a detection
(c) Apply 2 ~lL of each solution.
wavelength of 245 nm.
(d) Develop the plate to 15 cm.
The test is not valid unless, in the chromatogram obtained
with solution (2), the resolution factor between the first peak (e) After removal of the plate, dry in air and examine under
(component H 2B 1b) and the second peak (component ultraviolet light (254 n71l).
H 2 B 1a) is at least 3.0. MOBILE PHASE
In the chromatogram obtained with solution (1) the area of 1 volume of concentrated am71l0nia Rl, 9 volumes of methanol
any peak with a retention time of 1.3 to 1.5 relative to that of and 90 volumes of dichloromethane.
the principal peak is not greater than 2.7 times the area of
CONFIRMA TION
the principal peak in the chromatogram obtained with
solution (3) (2.7%), the area of any other secondaJY peak is The principal spot in dle chromatogram obtained with
not greater than the area of the principal peak in the solution (1) corresponds in position, colour and size to that
chromatogram obtained with solution (3) (1 %) and the sum in the chromatogram obtained with solution (2).
of the areas of any such peaks is not greater than 6 times the B. In the Assay, the chromatogram obtained with solution
area of the principal peak in the chromatogram obtained with (1) shows peaks with dle same retention times as the peaks
solution (3) (6%). Disregard any peak with an area les s than due to Ivermectin H 2 B1a and Ivermectin H2Blb in the
the area of the principal peak in the chromatogram obtained chromatogram obtained with solution (2).
with solution (4) (0.05%). TESTS
ASSAY Related substances
Carry out the method for liquid chromatography, Prepare a 0.0002% w/v solution of moxidectin BPCRS
Appendix lIT D, using the following solutions. For solution (internal standard) in acetonitrile (solution A).
(1) dissolve a volume of the injection in sufficient methanol to Carry out the method for liquid chromatography,
produce a solution containing 0.04% w/v of Ivermectin. Appendix lIT D, using the following solutions prepared
Solution (2) contains 0.04% w/v of ivermectin BPCRS in immediately before use.
methanol. Inject 20 ~lL of each solution. (1) Dilute a weighed quantity of the oral solution containing
The chromatographic conditions described under Related 4 mg of Ivermectin with about 190 mL of acetonitrile, mix,
substances may be used. allow to cool to room temperature and add sufficient
Calculate the content of ivermectin (H 2 B 1a + H 2B 1b) in the acetonitrile to produce 200 mL. Dilute 5 volumes of this
injection and the ratio HZBla / (HZBla + HZBlb) using as the solution to 50 volumes with solution A. Dry a portion of the
declared content the contents of C4sH74014 (HZBla) and resulting solution over anhydrous sodium sulfate and filter
C47H72014 (H2B 1b) in ivermectin BPCRS. through a 0.45-~lm filter.
(2) Dilute 2 volumes of a 0.02% w/v solution of
ivermectin BPCRS in acetonitriie to 200 volumes with solution
A.
(3) Dilute 2 volumes of solution (1) to 200 volumes with
acetonitrile.
Vet-168 Ivermectin Preparations 2016

(4) Dilute 5 volurnes of solution (3) to 50 volurnes with SYSTEM SUITABILITY


acetonitrile. The test is not valid unless, in the chromatogram obtained
Derivatise the solutions prior to analysis using the following with solution (2), the resolution factor between the peaks due
method. to ivennectin HzB 1a and ivennectin HZBlb is at least 3.0.
Transfer 300 ~tL of the solution being examined to a 2-mL DETERMINATION OF CONTENT
HPLC vial and add 130 ~tL of l-methylimidazole and mix for Determine the weight per mI of the oral solution,
5 seconds using a vortex mixer, allow to settle and mix again Appendix V G, and calculate the content of ivennectin
for 5 seconds. Add 200 ~tL of a 50% v/v solution of (H 2 B 1a + H 2 B 1b), weight in volurne, and the ratio H 2 B 1a /
tlifiuoroacetic anhydride in acetonitl'ile and mix for 5 seconds (HZBla + H 2 B 1b) using as the declared content the contents
using a vortex mixer, allow to settle and mix again for of C4sH74014 (HzB 1a) and C47H72014 (H2B 1b) in
5 seconds. ivennectin BPCRS.
CHROMATOGRAPHIC CONDITIONS
(a) Use a stainless steel colurnn (10 cm x 3.0 mm) packed
with octadecylsilyl silica gel fOl" chromatogmphy (2.5 ~tm)
(Phenomenex Luna C18 HST is suitable). Ivermectin Veterinary Oral Paste
(b) U se isocratic elution and the mobile phase described Ivennectin Veterinary Paste
below.
(c) Use a fiow rate of 1.2 mL per minute. Action and use
Anthelminthic.
(d) Use a colurnn temperature of 40°.
(e) Use a fiuorescence detector with an excitation wavelength DEFINITION
of 365 nm and an emission wavelength of 470 nm. Ivennectin Veterinary Oral Paste contains Ivennectin in a
(f) Inject 20 ~tL of each solution. suitable basis.
MOBILE PHASE The veterinaJY oral paste complies with the requirements stated
To 120 mL of water add 1.2 mL of ol1hophosph01ic acid and Lindel' Veterina¡y Oml Pastes and with the following requirements.
1.2 mL of tliethylamine. Add 1880 mL of acetoniilile, mix and Content of ivermectin, calculated as the sum of
filter through a 0.45-~m filter. component H 2B 1a (C4sH74014) and component H2Blb
When the chromatograms are recorded under the prescribed (C47Hn014)
conditions the retention times relative to ivermectin H 2 B 1a 95.0 to 110.0% of the stated amount.
(retention time, about 7 minutes) are: avennectin B1, about The ratio of the contents HZBla / (HZBla + HZBlb) is at least
0.6; ivennectin H 2 B 1b, about 0.8; ivennectin H 4B 1m about 90.0%.
1.2). IDENTIFICATION
SYSTEM SUITABILITY A. Carry out the metllod for thin-Iayer chromatogmphy,
The test is not valid unless: Appendix In A, using a silica gel 60 F 254 precoated plate
in the chromatogram obtained with solution (2), the resolution (Merck silica gel 60 F Z54 plates are suitable) and a mixture of
factor between tlle peaks due to ivennectin HZBla and 1 volume of concentmted ammonia Rl, 9 volumes of methanol
ivennectin HZBlb is at least 3.0; and 90 volumes of dichloromethane as the mobile phase.
Apply separately to the plate 2 ~tL of each of tlle following
in the chromatogram obtained with solution (4), the signal-to-
solutions. For solution (1) add 10 mL of methanol to a
noise ratio of the principal peak is at least 10.
quantity of the oral paste containing 5 mg of Ivennectin and
LIMITS mix with the aid of ultrasound until completely dispersed.
In the chromatogram obtained with solution (1): Solution (2) contains 0.05% w/v of ivermectin BPCRS in
the area of any secondaJY peak is not greater than 2.5 times methanol. After removal of the plate, allow it to dry in air and
the area of the principal peak in the chromatogram obtained examine under ultmviolet light (254 mn and 366 11m). The
with solution (3) (2.5%); principal spot in the chromatogram obtained with solution
(1) is similar in position, colour and size to that in the
the area of not more than one secondaJY peak is greater than
chromatogram obtained with solution (2).
the area of the principal peak in the chromatogram obtained
with solution (3) (1 %); B. In the Assay, the chromatogram obtained with solution
(1) shows two principal peaks with retention times similar to
the surn of tlle areas of all the secolldaJy peaks is not greater
those of the two principal peaks in the chromatogram
than 5 times the area of the principal peak in the
obtained with solution (2).
chromatogram obtained with solution (3) (5%).
Disregard any peak with an area less than the area of the TESTS
principal peak in the chromatogram obtained with solution Related substances
(4) (0.1 %). Carry out the method for liquid chro111atogmphy,
Appendix In D, using the following solutions. For solution
ASSAY (1) disperse a quantity of the oral paste in methanol with the
Carry out the method for liquid chromatography, aid of ultrasound and add sufficient methanol to produce a
Appendix In D, using derivatised solutions (1) and (2) solution containing 0.04% w/v ofIvennectin. Solutions (2),
described under Related substances. (3) and (4) cont'ain 0.04% w/v, 0.0004% w/vand
CHROMATOGRAPHIC CONDITIONS 0.00002% w/v respectively of ivermectin BPCRS in methanol.
The chromatographic conditions described under Related Inject 20 ~tL of each solution.
substances may be used. The chromatographic procedure may be carried out using
(a) a stainless steel column (25 cm x 4.6 mm) packed with
octadecylsilyl silica gel fOl" chromatography (5 ~tm) (Apex ODS 1
2016 Ivermectin Preparations Vet-169

is suitable), (b) as the mobile phase at a fiow rate of 1.5 mL contains 0.05% w/v of ivermectin BPCRS in methanol. After
per minute a mixture of 39 volumes of water, 55 volumes of removal of the pI ate, allow it to dry in air and examine under
methanol and 106 volumes of acetonitrzle and (c) a detection ultraviolet light (254 mn and 366 nm). The principal spot in
wavelength of 245 nm. the chromatogram obtained with solution (1) is similar in
The test is not valid unless, in the chromatogram obtained position, colour and size to that in the chromatogram
with solution (2), the resolution factor between the first peak obtained with solution (2).
(component H 2 B 1b) and the second peak (component B. In the Assay, the chromatogram obtained with solution
H 2 B 1a) is at least 3.0. (1) shows two principal peaks with retention times similar to
In the chromatogram obtained with solution (1) the are a of those of the two principal peaks in the chromatogram
any peak with a retention time of 1.3 to 1.5 relative to that of obtained with solution (2).
the principal peak is not greater than 3 times the area of the TESTS
principal peak in the chromatogram obtained with solution Related substances
(3) (3%), the area of any other secondary peak is not greater Carry out the method for liquid chromatography,
than the area of the principal peak in the chromatogram Appendix ITI D, using the following solutions. For solution
obtained ~ith solution (3) (1 %) and the sum of the are as of (1) dissolve a quantity of the preparation being examined in
any such peaks is not greater than 6 times the area of the sufficient methanol to produce a solution containing
principal peak in the chromatogram obtained with solution 0.04% w/v of Ivermectin. Solutions (2), (3) and (4) contain
(3) (6%). Disregard any peak with an area less than the area 0.04% w/v, 0.0004% w/v and 0.00002% w/v respectively of
of the principal peak in the chromatogram obtained with ivermectin BPCRS in methanol. Inject 20 ~lL of each solution.
solution (4) (0.05%).
The chromatographic procedw'e may be canied out using
ASSAY (a) a stainless steel column (25 cm x 4.6 mm) packed with
Carry out the method for liquid chromatography, octadecylsilyl silica gel for chromatography (5 ~lm) (Apex ODS 1
Appendix lIT D, using the following solutions. For solution is suitable), (b) as the mobile phase at a fiow rate of 1.5 mL
(1) disperse a quantity of the oral p'aste in methanol with the per minute a mixture of 39 volumes of water, 55 volumes of
aid of ultrasound and add sufficient methanol to produce a methanol and 106 volumes of acetonitrile and (c) a detection
solution containing 0.04% w/v of Ivermectin. Solution (2) wavelength of 245 nm.
contains 0.04% w/v of ivermectin BPCRS in nzethanol. Inject The test is not valid unless, in the chromatogram obtained
20 ~lL of each solution. with solution (2), the resolution factor between the fu'st peak
The chromatographic conditions described under Related (component HZBlb) and the second peak (component
substances may be used. HZBla) is at least 3.0.
Calculate the content of ivermectin (HZBla + H 2 B 1b) in the In the chromatogram obtained with solution (1) the area of
oral paste and the ratio HZBla / (HZBla + HZBlb) using as any peak with a retention time of 1.3 to 1.5 relative to that of
the declared content the contents of C4sH74014 (H 2 B 1J and the principal peak is not greater than 2.7 times the area of
C47H72014 (HZBlb) in ivemzectin BPCRS. the principal peak in the chromatogram obtained with
solution (3) (2.7%), the area of any other secondaJY peak is
not greater than the area of the principal peak in the
chromatogram obtained with solution (3) (1 %) and the sum
of the areas of any such peaks is not greater dlan 6 times the
Ivermectin Pour..on area of the principal peak in the chromatogram obtained with
solution (3) (6%). Disregard any peak with an area less than
Action and use the area of the principal peak in the chromatogram obtained
Antihelminthic.
with solution (4) (0.05%).
DEFINITION ASSAY
Ivermectin Pour-on is a pour-on solution. It contains Carry out the method for liquid chromatography,
Ivermectin in a suitable non-aqueous vehicle. Appendix lIT D, using dle following solutions. For solution
The pour-on complies with the requirements stated under (1) dissolve a quantity of the preparation being examined in
VetennalY Liquid Preparations for Cutaneous Application and sufficient methanol to produce a solution containing
with the following requirements. 0.04% w/v of Ivermectin. Solution (2) contains 0.04% w/v of
ivermectin BPCRS in 111ethanol. Inject 20 ~lL of each solution.
Content of ivermectin, calculated as the sum of
component H 2B 1a (C48H74014) and component H2Blb The chromatographic conditions described under Related
(C47Hn014) substances may be used.
95.0 to 105.0% ofthe stated amount. Calculate the content of ivermectin (HzB 1a + HZBlb) in the
The ratio of the contents HZBla / (HzB 1a + H 2 B 1b) is at least preparation being examined and the ratio
90.0%. HZBla / (HZBla + HZBlb) using as the declared content the
contents of C4sH74014 (HZBla) and C47HnOl4 (HZBlb) in
IDENTIFICATION ivermectin BPCRS.
A. Carry out the method for thin-Iayer chromatography,
Appendix lIT A, using a silica gel 60 F Z54 precoated plate
(Merck silica gel 60 F Z54 plates are suitable) and a mixture of
1 volume of concentrated ammonia Rl, 9 volumes of methanol
and 90 volumes of dichloromethane as the mobile phase.
Apply separately to the plate 2 ~lL of each of the following
solutions. For solution (1) dissolve a quantity of the
preparation being examined in sufficient methanol to produce
a solution containing 0.05% w/v of Ivermectin. Solution (2)
Vet-170 Levamisole Preparations 2016

of chloroform and wash the combined extracts with two


Levamisole Injection 10 mL quantities of water. To the combined extracts add
Action and use 5 O mL of anhydrous acetic acid and can)' out Method I for
Irnmunostimulant; antihelminthic. non-aqueous titration, Appendix VIII A, using
l-naphtholbenzein solution as indicator. Each mL of
DEFINITION O.lMperchloric acid VS is equivalent to 24.08 mg of
Levamisole Injection is a sterile solution of Levamisole C 11 H 12N 2 S,HCl.
Hydrochloride in Water for Injections. It may contain STORAGE
suitable colouring matter. Levamisole Injection should be protected from light.
The injection complies with the require111ents stated under
Parenteral Preparatio1ZS and with the following require111ents.
Content oflevamisole hydroch1oride, C n H 12N 2 S,HCl
92.5 to 107.5% ofthe stated amount.
Levamisole Oral Solution
IDENTIFICATION
A. Can)' out the method for thin-Iayer chromatography, Action and use
Appendix nI A, using silica gel G as the coating substance Irnmunostimulant; antihelminthic.
and a mixture of 100 volumes of ethyl aceta te, 10 volumes of
l11ethanol and 1 volume of 13.5M al111110nia as the mobile DEFINITION
phase. Apply separately to the plate 1 ~LL of each of the Levamisole Oral Solution is an aqueous solution of
following solutions in l11ethanol. For solution (1) dilute a Levamisole Hydrochloride.
volume of the injection to produce a solution containing The oral solution complies with the requirements stated under Oral
1% w/v of Levamisole Hydrochloride. Solution (2) contains Liquids and with the follO'lving requirements.
1% w/v of levamisole hydrochloride BPCRS. Mer removal of Content of levamisole hydroch1oride, C n H 12N2 S,HCl
the plate, allow it to dl)' in air and spray with potassium 92.5 to 107.5% ofthe stated amount.
iodoplatinate solution. The principal spot in the chromatogram
obtained with solution (1) corresponds to that in the IDENTIFICATION
chromatogram obtained with solution (2). A. Carry out the method for thin-Iayer chromatography,
Appendix nI A, using silica gel G as the coating substance
B. Dilute a volume of the injection containing 0.75 g of
and a mixture of 1 volume of 13.5M anz11Zonia, 10 volumes of
Levamisole Hydrochloride to 20 mL with water and add
methanol and 100 volumes of ethyl acetate as the mobile
6 mL of 1M sodiU711 hydroxide. Extract with 20 mL of
phase. Apply separately to the plate 1 pL of each of the
dichloromethane, discard the aqueous layer and wash the
following solutions. For solution (1) dilute a volume of the
dichloromethane layer with 10 mL of water. Shake with
oral solution with methanol to produce a solution containing
anhydrous sodium sulfate, filter and evaporate the
1% w/v of Levamisole Hydrochloride. Solution (2) contains
dichloromethane at room temperature. The melting point of
1% w/v of levamisole hydrochlorzde BPCRS in methanol. After
the residue, after dl)'ing over phosphonls pentoxide at a
removal of the plate, allow it to dl)' in air and spray with
pressure of 1.5 to 2.5 kPa at a temperature not exceeding
potassium iodoplatinate solution. The principal spot in the
40°, is about 59°, Appendix V A.
chromatogram obtained with solution (1) corresponds to that
C. The injection is laevorotatory. in the chromatogram obtained with solution (2).
D. Yields reaction B characteristic of chlorides, Appendix VI. B. To a quantity of the oral solution containing 0.3 g of
TESTS Levamisole Hydrochloride add 10 mL of water and 6 mL of
Acidity 1M sodium hydroxide. Extract with 20 mL of dichloromethane,
pH, 3.0 to 4.0, Appendix V L. discard the aqueous layer and wash the dichloromethane
layer with 10 mL of water. Shake with anhydrous sodium
2,3-Dihydro-6-phenylimidazo[2, l-b ]thiazole
sulfate, filter and evaporate the dichloromethane at room
hydroch1oride
temperature. The melting point of the residue, after dl)'ing
Carry out the method for thin-Iayer chromatography,
over phosphorus pentoxide at a pressure of 1.5 to 2.5 kPa at a
Appendix In A, using silica gel G as the coating substance
temperature not exceeding 40°, is about 59°, Appendix V A.
and a mixture of 8 volumes of glacial acetic acid, 16 volumes
of methanol and 90 volumes of toluene as the mobile phase. C. The oral solution is laevorotatOl)T.
Apply separately to the plate 1O ~LL of each of the following 2,3-Dihydro-6-phenylimidazo[2, l-b ]thiazole
two solutions. For solution (1) dilute a volume of the hydrochloride
injection with methanol to produce a solution containing Carry out the method for thin-Iayer chromatography,
5.0% w/v of Levamisole Hydrochloride. Solution (2) contains Appendix In A, using silica gel G as the coating substance
0.021 % w/v of 2,3-dihydro-6-phenylimidazo[2,l- and a mixture of 8 volumes of glacial acetic acid, 16 volumes
b}thiazole BPCRS in methanol. After removal of the plate, of methanol and 90 volumes of toluene as the mobile phase.
allow it to dry in air and spray with potassiu111 iodoplatinate Apply separately to the plate 5O ~LL of solution (1) and 1O ~LL
solution. Any spot in the chromatogram obtained with of solution (2). For solution (1) dilute a volume of the oral
solution (1) corresponding to 2,3-dihydro-6- solution to produce a solution containing 1.0% w/v of
phenylimidazo[2,1-b]thiazole is not more intense than the Levamisole Hydrochloride. Solution (2) contains 0.021 % w/v
spot in the chromatogram obtained with solution (2) (0.5%). of 2,3-dihydro-6-phenylimidazo[2,l-b}thiazole BPCRS in
ASSAY methanol. After removal of the plate, allow it to dry in air and
spray with potassium iodoplatinate solution. Any spot in the
To a volume ofthe injection containing 0.75 g ofLevamisole
chromatogram obtained with solution (1) corresponding to
Hydrochloride add 50 mL of water and 15 mL of 2M sodium
2,3-dihydro-6-phenylimidazo [2, 1-b] thiazole is not more
hydroxide, extract with three quantities, of 25, 20 and 15 mL,
2016 Lincomycin Preparations Vet-171

intense than dle spot in the chromatogram obtained with (3) Prepare in the same manner as solution (1) but using a
solution (2) (0.5%). quantity of the premix containing the equivalent of 90 mg of
ASSAY lincomycin in place of the lincomycin hydrochloride BPCRS.
To a volume of the oral solution containing 0.75 g of CHROMATOGRAPHIC CONDITIONS
Levamisole Hydrochloride add 15 mL of 2M sodium (a) Use a glass column (1.5 m x 3 mm) packed with acid-
hydroxide, extract with three quantities, of 25 mL, 20 mL and zuashed silanised diatomaceous support impregnated with 3 %
15 mL, of chloroform, wash the combined extracts with two w/w of phenyl methyl silicone fluid (50% phenyl) (OV-17 is
10 mL quantities of zuater and discard the washings. To the suitable) and maintained at 260°.
clear chloroform solution, after drying with anhydrous sodiu111 (b) Use heliu111 as the carrier gas at a fiow rate of about
sulfate if necessary, add 50 mL of anhydrous acetic acid. Carry 45 mL per minute.
out Method 1 for non-aqueous titration, Appendix VIII A,
(c) Use an inlet temperature of 260° to 290°.
using l-naphtholbenzein solution as indicator. Each mL of
O.lM perchloric acid VS is equivalent to 24.08 mg of (d) Use a flame ionisation detector at a temperature of 260°
CIIHI2N2S,HCl. to 290°.
(e) Inject 1 ~lL of each solution.
DETERMINATION OF CONTENT
Calculate the content of CIsH34N206S in the premix using
Lincomycin Premix dle declared content of CIsH34N206S in lincomycin
hydrochloride BPCRS.
Action and use LABELLING
Lincosamide antibacterial. The quantity of active ingredient is stated in terms of the
equivalent amount of lincomycin.
DEFINITION
Lincomycin Premix contains Lincomycin Hydrochloride.
The premix c0111plies zuith the requirements stated under Premixes
and 'luith the follozuing requirements.
Content of lincomycin, ClsH34Nz06S Lincomycin Tablets
90.0 to 105.0% ofthe stated amount.
Action and use
IDENTIFICATION Lincosamide antibacterial.
In the Assay the chromatogram obtained with solution (2)
shows a peak with the same retention time as the peak due to DEFINITION
the trimethylsilyl derivative of lincomycin in the Lincomycin Tablets contains Lincomycin Hydrochloride.
clu'omatogram obtained with solution (1). The tablets comply zuith the requirements stated under T ablets and
TESTS zuith the follozuing requirements.
Lincomycin B Content of lincomycin, ClsH34Nz06S
Examine solution (3) as described in the Assay but increasing 90.0 to 105.0% of tlle stated amount.
the sensitivity by eight to ten times while recording the peak IDENTIFICATION
due to the trimethylsilyl derivative of lincomycin B, which is
A. Mix a quantity of the powdered tablets containing the
eluted irnmediately before the trimethylsilyl derivative of
equivalent of 0.2 g of lincomycin with 5 mL of a mixture of
lincomycin.
4 volumes of chloroform and 1 volume of methanol, filter and
LIMITS evaporate the filtra te. Dissolve the oily residue in 1 mL of
The area of the peak due to the trimethylsilyl derivative of zuater, add acetone until precipitation begins and add a further
lincomycin B, when corrected for the sensitivity factor, is not 20 mL of acetone. Filter, wash with two 10-rnL quantities of
more than 5% of the are a of the peak due to the acetone, dissolve the residue in the chloroform-methanol
trimethylsilyl derivative of lincomycin. mixture, evaporate to dryness and dry at 60° at a pressure of
2 kPa for 1 hour. The il1jrared absorption spectrw11 of the
ASSAY
residue, Appendix II A, is concordant with the reference
Carry out the method for gas chromatography, spectrU1n of lincomycin hydrochloride (RSV 52).
Appendix III B, using the following solutions.
B. In the Assay, the chromatogram obtained with solution
(1) Add 10 mL of a 0.8% w/w solution of dotriacontane (2) shows a peak with the same retention time as the peak
(internal standard) in chloroform to 0.1 g of lincomycin
due to the trimethylsilyl derivative of lincomycirt in the
hydrochlonde BPCRS, dilute to 100 mL with a 2% w/v chromatogram obtained with solution (1).
solution of imidazole in chloroform, shake to dissolve and filter.
Place 4 mL of the filtrate in a 15 mL ground-glass-stoppered TESTS
centrifuge tube, add 1 mL of a mixture of 99 volumes of Lincomycin B
N,O-bis(trimethylsilyl)acetamide and 1 volume of Examine solution (3) as described under the Assay but
trúnethylchlorosilane and swirl gently. Loosen the glass stopper increasing the sensitivity by eight to ten times while recording
and heat at 65° for 30 minutes. the peak due to the trimethylsilyl derivative of lincomycin B,
(2) Prepare in the same manner as solution (1) but omitting which is eluted irnmediately before the trimethylsilyl
the internal standard and using a quantity of the premix derivative of lincomycin.
containing the equivalent of 90 mg of lincomycin in place of LIMITS
the lincomycin hydrochlonde BPCRS. The area of the peak due to the trimethylsilyl derivative of
lincomycin B, when corrected for the sensitivity factor, is not
Vet-172 Meloxicam Preparations 2016

more than 5% ofthe area of the peak due to the (2) Dissolve 10 mg of meloxicam BPCRS in 10 mL of acetone)
trimethylsilyl derivative of lincomycin. add 2 mL of zuater and dilute to 20 mL with acetone.
ASSAY CHROMATOGRAPHIC CONDITIONS
Carry out the method for gas chromatogmph)!, (a) Use a silica gel F254 pIate (Merck HPTLC plates are
Appendix lIT B, using the following solutions. suitable).
(1) Add 10 mL of a 0.8% w/w solution of dotriacontane (b) Use the mobile phase described below.
(internal standard) in chIoroform to 0.1 g of Iincomycin (c) Apply 20 ~lL of each solution.
hydrochIoride BPCRS, dilute to 100 mL with a 2% w/v
(d) Develop the plate to 8 cm.
solution of imidazole in chloroform, shake to dissolve and filter.
Place 4 mL of the filtrate in a 15 mL ground-glass-stoppered (e) After removal of the plate, dry in air and examine under
cent:rifuge tube, add 1 mL of a mixture of 99 volumes of ultraviolet light (254 nm).
N,O-bis(trimethylsilyl)acetamide and 1 volume of MOEILE PHASE
trimethylchlorosilane and swirl gently. Loosen the glass stopper 1 volume of 13.5M ammonia, 20 volumes of 111ethanoI and
and heat at 65° for 30 minutes. 80 volumes of dichIor0111ethane.
(2) Prepare in the same manner as solution (1) but omitting CONFIRMATION
the internal standard and using a quantity of the powdered
The principal spot in the chromatogram obtained with
tablets containing the equivalent of 90 mg of lincomycin in
solution (1) corresponds to that in the chromatogram
place of the lincomycin hydrochloride BPCRS.
obtained with solution (2).
(3) Prepare in the same manner as solution (1) but using a
B. In the Assay, the retention time of the principal peak in
quantity of the powdered tablets containing the equivalent of
the chromatogram obtained with solution (1) corresponds to
90 mg of lincomycin in place of the lincomycin
that of the principal peak in the chromatogram obtained with
hydrochlO1ide BPCRS.
solution (2).
CHROMATOGRAPHIC CONDITIONS
TESTS
(a) Use a glass column (1.5 m x 3 mm) packed with acid-
AIkalinity
zuashed silanised diatomaceous SUppOl1 impregnated with 3 %
pH, 8.0 to 9.0, Appendix V L.
w/w of phenyl methyl silicone fluid (50% phenyl) (OV-17 is
suitable) and maintained at 260°. Related substances
Carry out the method for liquid chromatography,
(b) Use heliu771 as the carrier gas at a flow rate of about
Appendix lIT D, using the following solutions.
45 mL per minute.
0 (1) Add 0.3 mL of O.4M sodium hydroxide to a volume of the
(c) Use an inlet temperature of 260 to 290°.
injection containing 40 mg of Meloxicam and dilute with
(d) Use a flame ionisation detector at a temperature of 260 0 71lethanol (40%) to produce 10 mL.
to 290.
(2) Dilute 2 mL of solution (1) to 100 mL with methanol
(e) Inject 1 ~lL of each solution. (40%). Dilute 1 mL of the resulting solution to 10 mL with
DETERMINATION OF CONTENT methanol (40%).
Calculate the content of ClsH34N206S in the tablets using (3) Add 0.3 mL of 0.4M sodium hydroxide to 40 mg of
the declared content of ClsH34N206S in linc071lycin meloxicam impUJity standard BPCRS and dilute with methanol
hydrochloride BPCRS. (40%) to produce 10 mL.
LABELLING CHROMATOGRAPHIC CONDITIONS
The quantity of active ingredient is stated in terms of the (a) Use a stainless steel column (10 cm x 4.0 mm) packed
equivalent amount of lincomycin. with octadecylsilyl silica gel for chromatogmphy (10 ~lm)
(Kromasil 100 C18 is suitable).
(b) U se gradient elution and the mobile phase described
below.
Meloxicam Injection (c) Use a flow rate of 1.0 mL per minute.
(d) Use an ambient column temperature.
Action and use (e) Use detection wavelengths of 260 nm and 350 nm.
Cyclo-oxygenase inhibitor; analgesic; anti-inflammatory.
(f) Inject 1O ~lL of each solution.
DEFINITION MOEILE PHASE
Meloxicam Injection is a sterile solution of Meloxicam in Mobile phase A 0.1 % w/v solution of potassiU111 dihydrogen
Water for Injections. ol1hophosphate adjusted to pH 6.0 with 2M sodiwn hydroxide.
The injection complies zuith the requirements stated undel' Mobile phase B methanol.
Parenteml Prepamtions and zuith the follozuing l'equirements.
Content of meloxicam, C14H13N304S2 Time Mobile phase A Mobile phase B Commenf
95.0 to 105.0% of the stated amount. (Minutes) (%v/v) (%v/v)
IDENTIFICATION 0-2.5 60 40 isocratic
A. Carry out the method for thin-layer chromatogmphy, 2.5-12 60--730 40--770 linear gradient
Appendix lIT A, using the following solutions.
12-25 30 70 isocratic
(1) Dilute a volume of the injection containing 10 mg of
25-26 30--760 70--740 linear gradient
Meloxicam to 20 mL with acetone, stir for 15 minutes, filter
and use the filtrate. 26-30 60 40 re-equilibration
2016 Meloxicam Preparations Vet-173

SYSTEM SUIT ABILITY


The test is not valid unless the chromatogram obtained with
Meloxicam Oral Suspension
solution (3): Action and use
closely resembles the chromatogram supplied with nzeloxicam Cyclo-oxygenase inhibitor; analgesic; anti-infiammatOlY.
impurz'ty standard BPCRS at 260 nm and 350 nm;
the resolution factor between the peaks due to meloxicam and DEFINITION
impurity A at 350 nm is at least 3.0; Meloxicam Oral Suspension is a suspension of Me10xicam in
a suitable vehicle.
the resolution factor between the peaks due to impurity B and
meloxicam at 260 nm is at least 3.0. The oral suspension complies 'With the requirements stated unda
Oral Liquids and 'With the following requirements.
LIMITS
Content of meloxicam, C14H13N304S2
In the chromatogram obtained with solution (1):
95.0 to 105.0% ofthe stated amount.
identify any peak cOlTesponding to impurity A at 350 nm and
multiply the area of this peak by a correction factor of 2.0; IDENTIFICATION
A. Call.Y out the method for thin-Iayer chronzatography,
the are a of any peak corresponding to impurity A at 350 nm
Appendix nI A, using the following solutions.
is not greater than the area of the principal peak in the
chromatogram obtained with solution (2) (0.2%); (1) Dilute a quantity of the oral suspension containing 3 mg
of Me10xicam to 10 mL with acetone, stir for 10 minutes,
the area of any peak corresponding to impurity C at 350 nm
filter and use the filtrate.
is not greater than the area of the principal peak in the
chromatogram obtained with solution (2) (0.2%); (2) Dissolve 3 mg of meloxicam BPCRS in about 5 mL of
acetone, add 0.5 mL of 'Water and dilute to 10 mL with
the area of any peak corresponding to impurity B at 260 nm
acetone.
is not greater than 2.5 times the area of the principal peak in
the chromatogram obtained with solution (2) (0.5%). CHROMATOGRAPHIC CONDITIONS

In both the chromatograms obtained with solution (1) at (a) Use as the coating high-pelformance silica gel F254 (Merck
350 nm and at 260 nm: silica gel 60 F 254 HPTLC plates are suitable).
the area of any other secondary peak is not greater than the (b) Use the mobile phase as described below.
area of the peak in the chromatogram obtained with solution (c) Apply 5 ~lL of each solution.
(2) at that wavelength (0.2%). (d) Develop the plate to 8 cm.
The nominal total content of any such impurities is not (e) After removal of the plate, allow it to dry in air and
greater than 2.0%. examine under ultraviolet light (254 and 365 7lJ1l).
Disregard any peak with an area less than 0.25 times the area MOBILE PHASE
of the principal peak in the chromatogram obtained with
1 volume of 13.51v1 a7JlJllonia, 20 volumes of llIethanol and
solution (2) at the same wavelength (0.05%).
80 volumes of dichloromethane.
ASSAY
CONFIRMATION
Carry out the method for liquid chromatography,
Appendix nI D, using the following solutions. By each method of visualisation the principal spot in the
chromatogram obtained with solution (1) is similar in
(1) To a volume of the injection containing 40 mg of
position, colour and size to that in the chromatogram
Meloxicam add 0.3 mL of 0.4M sodiu111 hydroxide and add
obtained with solution (2).
sufficient methanol (40%) to produce 10 mL. Dilute 1 mL of
the resulting solution to 10 mL with methanol (40%). B. Disperse a quantity of the oral suspension containing
1.5 mg of Me10xicam in 5 mL of O.lM sodium hydroxide,
(2) 0.04% w/v of meloxica111 BPCRS in methanol (40%).
dilute to 100 mL with methanol and filter. The light abs01ption
(3) Add 0.3 mL of O.4M sodiwl1 hydroxide to 40 mg of of the filtrate, Appendix II B, in the range 340 to 450 nm
meloxica111 impurity standard BPCRS and dilute with methanol exhibits a maximum at 362 nm.
(40%) to produce 10 mL.
ASSAY
CHROMATOGRAPHIC CONDITIONS
Carry out the method for liquid chromatography,
The chromatographic procedure described under Related Appendix nI D, using the following solutions.
substances may be used with a detection wavelength of (1) Mix a quantity of the oral suspension containing 15 mg
350 nm. of Meloxicam with sufficient of the mobile phase to produce
SYSTEM SUITABILITY 200 mL, mix with the aid of ultrasound for 30 minutes and
The test is not valid unless the chromatogram obtained with filter.
solution (3): (2) 0.0075% w/v of meloxicam BPCRS in the mobile phase.
closely resembles the chromatogram supplied with meloxicam CHROMATOGRAPHIC CONDITIONS
i111purity standard BPCRS at 350 nm; (a) Use a stainless steel column (10 cm x 4 mm) packed
the resolution factor between the peaks due to meloxicam and with octadecylsilyl siliéa gel for ch1'Omatography (1 O ~lm)
impurity A at 350 nm is at least 3.0. (Nucleosil C18 is suitable) and a pre-column 1-cm long
DETERMINATION OF CONTENT packed with the same material.
Calculate the content of C14H13N304S2 using the declared (b) U se isocratic elution and the mobile phase described
content of C14H13N304S2 in meloxicam BPCRS. below.
IMPURITIES (c) Use a fiow rate of 0.8 mL per minute.
The impurities limited by the requirements of this (d) Use a column temperature of 40°.
monograph include those listed under Meloxicam. (e) Use a detection wavelength of 254 nm.
Vet-1 74 Mepivacaine Preparations 2016

(f) Inject 20 ~lL of each solution and continue the phase. Solution (2) contains 0.0002% w/v of
chromatography for twice the retention time of the principal 2,6-dimethylaniline in the mobile phase.
peak. The chromatographic procedure described under Assay may
MOBILE PHASE be used.
35 volumes of a mixture containing 10 parts of propan-2-ol In the chromatogram obtained with solution (1) the area of
and 65 parts of methanol and 65 volumes of a 0.2% w/v any peak corresponding to 2,6-dimethylaniline is not greater
solution of diam11lonium hydrogen orthophosphate previously than the area of the principal peak in the chromatogram
adjusted to pH 7.0 with 011hophosphoric acid. obtained with solution (2) (0.2% of the content of
mepivacaine hydrochloride).
DETERMINATION OF CONTENT
Determine the weight per 111L of the oral suspension, ASSAY
Appendix V G, and calculate the content of C14H13N304Sb Carry out the method for liquid chromatography,
weight in volume, from the declared content of Appendix lIT D, using the following solutions. For solution
C14H13N304S2 in meloxicam BPCRS. (1) dilute a quantity of the injection containing 0.1 g of
Mepivacaine Hydrochloride to 100 mL with the mobile
phase and dilute 1 volume of this solution to 20 volumes
with the mobile phase. Solution (2) contains 0.005% w/v of
mepivacaine hydrochloride BPCRS in the mobile phase.
Mepivacaine Injection The chromatographic procedure may be carried out using
(a) a stainless steel column (25 cm x 3.2 mm) packed with
Action and use
octadecylsilyl silica gel for chromatography (5 ~lm) (Spherisorb
Local anaesthetic.
ODS 1 is suitable), (b) as the mobile phase with a flow rate
DEFINITION of 0.5 mL per minute a mixture of 1 volume of triethylamine,
400 volumes of acetonitrile and 600 volumes of a 0.2% w/v
Mepivacaine Injection is a sterile solution of Mepivacaine
solution of potassium dihydrogen 011hophosphate, the mixture
Hydrochloride in Water for Injections.
adjusted to pH 4.0 with 011hophosphol'ic acid and (c) a
The injection complies with the requirements stated under detection wavelength of 212 nm.
Parenteral Preparations and 'luith the following requirements.
Calculate the content of ClsH22N20bHCI in the injection
Content of mepivacaine hydrochloride, using the declared content of ClSH22N202,HCI in
ClsHzzNzOz,HCl 111epivacaine hydrochloride BPCRS.
95.0 to 105.0% ofthe stated amount.
IDENTIFICATION
A. Carry out the method for thin-layer chromatography,
Appendix nI A, using a TLC silica gel F254 plate and a
mixture of 1 volume of 13.5M a11lJ1zonia, 5 volumes of
Moxidectin Injection
methanol and 100 volumes of ether as the mobile phase, but Action and use
allowing the solvent front to ascend 12 cm above the line of Antihelminthic; ectoparasiticide
application. Apply separately to the plate 1O ~lL of each of
the following solutions. For solution (1) dilute a quantity of DEFINITION
the injection with sufficient ethanol (96%) to produce a Moxidectin Injection is a sterile solution of Moxidectin. It is
solution containing 0.4% w/v of Mepivacaine Hydrochloride. supplied as a ready-to-use solution.
Solution (2) contains 0.4% w/v of mepivacaine The injection complies with the requirements stated under
hydrochloride BPCRS in ethanol (96%). Solution (3) contains Parenteral Preparations and with the following requirements.
0.4% w/v of each of 111epivacaine hydrochloride BPCRS and
lidocaine hydrochloride BPCRS in ethanol (96%). After removal Content of moxidectin, C37Hs3NOg
of the plate, allow it to dry in air and examine under 90.0 to 110.0% of the stated amount.
ultraviolet light (254 nm). The principal spot in the IDENTIFICATION
chromatogram obtained with solution (1) is similar in A. Carry out the method for thin-layer chromatography,
position and size to the principal spot in the chromatogram Appendix nI A, using the following solutions.
obtained with solution (2). The test is not valid unless the (1) Dilute a volume of the injection with sufficient methanol
chromatogram obtained with solution (3) shows two clearly to produce a solution containing 0.04% w/v of Moxidectin.
separated principal spots.
(2) 0.04% w/v of moxidectin BPCRS in methal1ol.
B. In the Assay, the chromatogram obtained with solution
(1) shows a peak with the same retention time as the CHROMATOGRAPHIC CONDITIONS
principal peak in the chromatogram obtained with (a) Use as the coating silica gel.
solution (2). (b) Use the mobile phase as described below.
TESTS (c) Apply 5 ~L of each solution.
Acidity (d) Develop the plate to 15 cm.
pH, 4.5 to 6.0, Appendix V L. (e) After removal of the plate, dry in air, spray with
2,6-Dinaethylaniline anisaldehyde solution Rl, heat at 105° for 5 to 10 minutes and
Carry out the method for liquid chromatography, allow to cool.
Appendix In D, using the following solutions. For solution MOBILE PHASE
(1) dilute a quantity of the injection containing 0.1 g of
8 volumes of a 15% w/v solution of a11l111onium aceta te
Mepivacaine Hydrochloride to 100 mL with the mobile
adjusted to pH 9.6 with ammol1ia, 19 volumes of propan-2-ol
and 43 volumes of ethyl acetate.
2016 Moxidectin Preparations Vet-175

CONFIRMATION IDENTIFICATION
The principal spot in the chromatogram obtained with A. Carry out the method for thin-layer chromatography,
solution (1) corresponds in position, colour and size to that Appendix nI A, using the following solutions.
in the chromatogram obtained with solution (2). (1) Dilute a quantity of the oral solution with sufficient
B. In the Assay, the retention time of the principal peak in methanol to produce a solution containing 0.04% w/v of
the chromatogram obtained with solution (1) is similar to Moxidectin.
that of the principal peak in the chromatogram obtained with (2) 0.04% w/v of moxidectin BPCRS in methanol.
solution (2).
CHROMATOGRAPHIC CONDITIONS
ASSAY (a) Use as the coating silica gel.
Carry out the method for liquid chromatography, (b) Use the mobile phase as described below.
Appendix nI D, using the following solutions in acetonitrile.
(c) Apply 5 ~lL of each solution.
(1) Dilute a volume of the injection to produce a solution
containing 0.1 % w/v of Moxidectin. If the solution is cloudy, (d) Develop the plate to 15 cm.
shake, allow to settle and use the supernatant. (e) After removal ofthe plate, dry in air, spray with
anisaldehyde solution Rl, heat at 105° for 5 to 10 minutes and
(2) 0.1 % w/v of moxidectinBPCRS.
allow to coo1.
(3) 0.1 % w/v of moxidectinfor system suitability EPCRS.
MOBILE PHASE
CHROMATOGRAPHIC CONDITIONS
8 volumes of a 15% w/v solution of a7111170nium acetate
(a) Use a stainless steel column (15 cm x 3.9 mm) packed adjusted to pH 9.6 with al11111onia, 19 volumes of propan-2-o1
with end-capped octadecylsilyl silica gel for chromatography and 43 volumes of ethyl acetate.
(4 ¡lm) (Waters Nova-Pak and Waters Pico-Tag are suitable).
CONFIRMATION
(b) U se isocratic elution and the mobile phase described
below. The principal spot in the chromatogram obtained with
solution (1) corresponds in position, colour and size to that
(c) Use a fiow rate of 2.5 mL per minute.
in the chromatogram obtained with solution (2).
(d) Use a column temperature of 50°.
B. In the Assay, the retention time of the principal peak in
(e) Use a detection wavelength of 242 nm. the chromatogram obtained with solution (1) is similar to
(f) Inject 1O ~lL of each solution. that of the principal peak in the chromatogram obtained with
MOBILE PHASE solution (2).
40 volumes of a 1.925% w/v solution of anz1110niu111 acetate in ASSAY
water, adjusted to pH 4.8 with glacial acetic acid, and Carry out the method for liquid chromatography,
60 volumes of acetonitrile. Appendix nI D, using the following solutions in acetonitrile.
When the chromatograms are recorded under the prescribed (1) Dilute a weighed quantity of the oral solution to produce
conditions the retention time of moxidectin is about a solution containing 0.05% w/v of Moxidectin, shake and
12 minutes and the retention time of impurity D relative to allow to settle.
that of moxidection is about 0.94. (2) 0.05% w/v of moxidectin BPCRS.
SYSTEM SUIT ABILITY (3) 0.1 % w/v of moxidectin for system suitability EPCRS.
The test is not valid unless, in the chromatogram obtained CHROMATOGRAPHIC CONDITIONS
with solution (3), the peak-to-valley ratio is at least 3.0 where
(a) Use a stainless steel column (15 cm x 3.9 mm) packed
H p is the height above the baseline of the peak due to
withend-capped octadecylsilyl silica gel for chro711atography
impurity D and H v is the height above the baseline of the
(4 ~lm) (Waters Nova-Pak and Water s Pico-Tag are suitable).
lowest point of the curve separating this peak from the peak
due to moxidectin. (b) U se isocratic elution and the mobile phase described
below.
DETERMINATION OF CONTENT
(c) Use a fiow rate of 2.5 mL per minute.
Calculate the content of C37H53NOs in the injection using
(d) Use a column temperature of 50°.
the declared content of C37H53NOs in moxidectin BPCRS.
(e) Use a detection wavelength of 242 nm.
(f) Inject 20 ~lL of each solution.
MOBILE PHASE

Moxidectin Oral Solution 40 volumes of a 1.925% w/v solution of ammonium acetate in


water, adjusted to pH 4.8 with glacial acetic acid, and
Moxidectin Oral Drench
60 volumes of acetonitrile.
Action and use When the chromatograms are recorded under the prescribed
Antihelminthic; ectoparasiticide conditions the retention time of moxidectin is about
12 minutes and the retention time of impurity D relative to
DEFINITION that of moxidectin is about 0.94.
Moxidectin Oral Solution is a solution of Moxidectin in a SYSTEM SUITABILITY
suitable vehicle.
The test is not valid unless, in the chromatogram obtained
The oral solution complies with the requirements stated under Oral with solution (3), the peak-to~valley ratio is at least 3.0 where
Liquids and with the following requirements. H p is the height above the baseline of the peak due to
Content of moxidectin, C37H53NOg impurity D and H v is the height above the baseline of the
90.0 to 110.0% of the stated amount.
Vet-176 Moxidectin Preparations 2016

lowest point of the curve separating this peak from the peak CHROMATOGRAPHIC CONDITIONS
due to moxidectin. (a) Use a stainless steel column (15 cm x 3.9 mm) packed
DETERMINATION OF CONTENT with end-capped octadecylsilyl silica gel for chromatography
(4/lm) (Waters Nova-Pak and Waters Pico-Tag are suitable).
Determine the 'lOeight per mI of the oral solution,
Appendix V G, and calculate the content of C37Hs3NOs, (b) Use isocratic elution and the mobile phase described
weight in volume, using the dec1ared content of C37Hs3NOs below.
in moxidectin BPCRS. (c) Use a flow rate of 2.5 mL per minute.
(d) Use a column temperature of 50°.
(e) Use a detection wavelength of 242 nm.
(f) Inject 1O ~lL of each solution.
Moxidectin Oromucosal Gel MOBILE PHASE
Moxidectin Oral Gel 40 volumes of a 1.925% w/v solution of anunonium acetate in
'lOater, adjusted to pH 4.8 with glacial acetic acid, and
Action and use 60 volumes of acetonitnle.
Antihelminthic; ectoparasiticide
When the chromatograms are recorded under the prescribed
DEFINITION conditions the retention time of moxidectin is about
Moxidectin Oromucosal Gel is a solution of Moxidectin in a 12 minutes and the retention time of impurity D relative to
suitable water-miscible basis. that of moxidectin is about 0.94.
The oromucosal gel complies 'lvith the requirements stated under SYSTEM SUITABILITY
Oromucosal Preparations and 'lOith the follo'lOing requirements. The test is not valid unless, in the chromatogram obtained
Content of moxidectin, C37Hs3NOg with solution (3), the peak-to-valley ratio is at least 3.0 where
90.0 to 110.0% of the stated amount. H p is the height above the baseline of the peak due to
impurity D and H v is the height above the baseline of the
IDENTIFICATION lowest point of the curve separating this peak from the peak
A. Carry out the method for thin-Iayer chromatography, due to moxidectin.
Appendix III A, using the following solutions.
DETERMINATION OF CONTENT
(1) Disperse a quantity of the gel containing 10 mg of
Moxidectin with sufficient methanol to produce a solution Calculate the content of C37Hs3NOs in the oromucosal gel
containing 0.04% w/v of Moxidectin. using the dec1ared content of C37Hs3NOs in
moxidectin BPCRS.
(2) 0.04% w/v of 11Zoxidectin BPCRS in methanol.
CHROMATOGRAPHIC CONDITIONS
(a) Use as the coating silica gel.
(b) Use the mobile phase as described below. Moxidectin Pour-on
(c) Apply 5 ~lL of each solution.
(d) Develop the plate to 15 cm. Action and use
Antihelminthic; ectoparasiticide
(e) After remo val of the plate, dry in air, spray with
anisaldehyde solution Rl, heat at 105° for 5 to 10 minutes and DEFINITION
allow to cool. Moxidectin Pour-on is a pour-on solution. It contains
lvIOBILE PHASE Moxidectin in a suitable vehic1e.
8 volumes of a 15% w/v solution of ammonium acetate The pour-on complies 'lvith the requirements stated under
adjusted to pH 9.6 with ammonia, 19 volumes of propan-2-01 Vetennary Liquid Preparations for Cutaneous Application and
and 43 volumes of ethyl acetate. 'lOith the follo'lOing requirements.
CONFIRMATION Content of moxidectin, C37Hs3NOg
The principal spot in the chromatogram obtained with 90.0 to 110.0% of the stated amount.
solution (1) corresponds in position, colour and size to that IDENTIFICATION
in the chromatogram obtained with solution (2). A. Carry out the method for thin-Iayer chromatography,
B. In the Assay, the retention time of the principal peak in Appendix lIT A, using the following solutions.
the chromatogram obtained with solution (1) is similar to (1) Dilute a quantity of the pour-on solution, with shaking,
that of the principal peak in the chromatogram obtained with in sufficient methanol to produce a solution containing
solution (2). 0.04% w/v of Moxidectin; filter a 5-mL portion through a
ASSAY 0.45-/lm PTFE membrane filter and use the filtrate.
Can)' out the method for liquid chromatography, (2) 0.04% w/v of moxidectin BPCRS in methanol.
Appendix III D, using the following solutions in acetonitrile. CHROMATOGRAPHIC CONDITIONS
(1) Disperse a quantity of the gel containing 25 mg of (a) Use as the coating silica gel.
Moxidectin in sufficient acetonitrile to produce a solution
(b) Use the 'mobile phase as described below.
containing 0.1 % w/v of Moxidectin and allow to settle.
(c) Apply 5 ~lLof each solution.
(2) 0.1 % w/v of moxidectin BPCRS.
Cd) Develop the plate to 15 cm.
(3) 0.1 % w/v of moxidectin for system suitability EPCRS.
Ce) After removal of the plate, dry in air, spray with
anisaldehyde solution Rl, heat at 105° for 5 to 10 minutes and
allow to cool.
2016 Nitroxinil Preparations Vet-177

MOBILE PRASE
8 volumes of a 15% w/v solution of am7110nium acetate
Nandrolone Laurate Injection
adjusted to pH 9.6 with ammonia, 19 volumes of propan-2-01 Action and use
and 43 volumes of ethyl acetate. Anabolic steroid; androgen.
CONFIRMATION
DEFINITION
The principal spot in the chromatogram obtained with
Nandrolone Laurate Injection is a sterile solution of
solution (1) corresponds in position, colour and size to that
Nandrolone Laurate in Ethyl Oleate, 01' other suitable ester,
in the chromatogram obtained with solution (2).
in a suitable fixed oil 01' in any mixture of these.
B. In the Assay, the retention time of the principal peak in
The iny'ection complies 'lOith the requírements stated under
the chromatogram obtained with solution (1) is similar to
Parenteral Preparations and 'luith the follo'lOing requírements.
that of the principal peak in the chromatogram obtained with
solution (2). Content of nandrolone laurate, C30H4S03
92.5 to 107.5% ofthe stated amount.
ASSAY
Carry out the method for liquid chromatography, IDENTIFICATION
Appendix lIT D, using the following solutions in acetonim'le. Carry out the method for thin-Iayer chromatography,
(1) Dilute a weighed quantity of the pour-on solution to Appendix lIT A, using a silica gel F 2S4 precoated plate the
produce a solution containing 0.1 % w/v of Moxidectin, shake surface of which has been modified by chemically-bonded
and mix with the aid of ultrasound for 10 minutes. Allow to octadecylsilyl groups (Whatman KC 18F plates are suitable)
cool and filter the resulting solution through a O.45-~lm and a mixture of 20 volumes of 'lOater, 40 volumes of
membrane filter, discarding the first few mL of filtrate. acetonitrz'le and 60 volumes of propan-2-01 as the mobile phase.
Apply separately to the plate 5 ~lL of each of the following
(2) 0.1 % w/v of moxidectin BPCRS.
solutions. For solution (1) dilute the injection with chlorofor111
(3) 0.1 % w/v of moxidectinfor system suitability EPCRS. to produce a solution containing 0.5% w/v ofNandrolone
CHROMATOGRAPHIC CONDITIONS Laurate. Solution (2) contains 0.5% w/v of nandrolone
(a) Use a stainless steel column (15 cm x 3.9 mm) packed laurate BPCRS in chloroforl11. For solution (3) mix equal
with end-capped octadecylsilyl silica gel for chromatography volumes of solutions (1) and (2). After removal of the plate,
(4 ~m) (Waters Nova-Pak and Waters Pico-Tag are suitable). allow it to dry in air until the solvent has evaporated and
heat at 100° for 10 minutes. Allow to cool and examine
(b) Use isocratic elution and the mobile phase described
under ultraviolet light (254 nm). The principal spot in the
below.
chromatogram obtained with solution (1) corresponds to that
(c) Use a fiow rate of 2.5 mL per minute. in the chromatogram obtained with solution (2).
(d) Use a column temperature of 50°. The principal spot in the chromatogram obtained with
(e) Use a detection wavelength of 242 nm. solution (3) appears as a single, compact spot.
(f) Inject 1O ~lL of each solution. ASSAY
MOBILE PRASE To a volume containing 0.1 g of N androlone Laurate add
40 volumes of a 1.925% w/v solution of ammonium acetate in sufficient chloroforl1l to produce 100 mL. Dilute 3 mL to
50 mL with chloroform. To 5 mL add 10 mL of isoniazid
'lOater, adjusted to pH 4.8 with glacial acetic acid, and
60 volumes of acetonitrile. solution and sufficient methanol to produce 20 mL. Allow to
stand for 45 minutes and measure the absorbance of the
When the chromatograms are recorded under the prescribed resulting solution at the maximum at 380 nm,
conditions the retention time of moxidectin is about Appendix IT B, using as the reference solution 5 mL of
12 minutes and the retention time of impurity D relative to chloroform treated in the same manner. Calculate the content
that of moxidectin is about 0.94.
of C30H4S03 from the absorbance obtained by repeating the
SYSTEM SUIT ABILITY operation using a suitable quantity of nandrolone BPCRS and
The test is not valid unless, in the chromatogram obtained from the declared content of ClsH2602 in
with solution (3), the peak-to-valley ratio is at least 3.0 where nandrolone BPCRS. Each mg oÍ ClsH2602 is equivalent to
H p is the height aboye the baseline of the peak due to 1.664 mg of C30H4S03'
impurity D and H v is the height aboye the baseline of the STORAGE
lowest point of the curve separating this peak from the peak Nandrolone Laurate Injection should be protected from light.
due to moxidectin.
DETERMINATION OF CONTENT
Determine the 'lOeight per mI of the pour-on solution,
Appendix V G, and calculate the content of C37Hs3NOs, Nitroxinil Injection
weight in volume, using the declared content of C37Hs3NOs
in moxídectín BPCRS. Action and use
Antihelminthic.

DEFINITION
Nitroxinil Injection is a sterile solution of the
N-ethylglucamine salt of Nitroxinil in Water for Injections.
The injection complíes 'lOith tlíe requírements stated under
Parenteral Preparations and 'lOith the follo'lOing requirements.
Content of nitroxinil, C 7H 3IN2 0 3
95.0 to 105.0% ofthe stated amount.
Vet-178 Oxfendazole Preparations 2016

IDENTIFICATION A. The infrared absOlption spectrum, Appendix n A, is


A. The light absOlption, Appendix n B, in the range 240 to concordant with the reference specttum of oxfendazole
350 nm of the solution obtained in the Assay exhibits a (RSV 32).
maximum only at 271 nm. B. The light absOlption, Appendix n B, in the range 220 to
E. Heat 0.5 mL with 3 mL of sulfuric acid; iodine vapour is 350 nm of a 0.001 % w/v solution in 1M hydrochloric acid
evolved. exhibits three maxima, at 226, 284 and 291 nm.
TESTS TESTS
Acidity or aIkalinity Acidity
pH, 5.0 to 7.0, Appendix V L, determined using a 20% w/v pH, 4.3 to 5.3, Appendix V L.
solution of N-ethylgluca111ine hydrochloride BPCRS in place of Related substances
a saturated solution of potassium chloride as the liquid junction Carry out the method for thin-Iayer chromatography,
solution. Appendix In A, using silica gel G as the coating substance
Inorganic iodide and a mixture of 5 volurnes of glacial acetic acid and
Dilute a volume containing 0.4 g ofNitroxinil to 100 mL 95 volurnes of ethyl acetate as the mobile phase. Apply
with water. To 10 mL add 4 mL of 1M sulfuric acid and separately to the plate 20 ~lL of each of the following
extract with three 10 mL quantities of chloroform. Add to the solutions. For solution (1) shake a quantity of the oral
aqueous extract 1 mL of hydrogen peroxide solution (100 vol) suspension containing 0.1 g of Oxfendazole with 20 mL of a
and 1 mL of chloroform, shake for 2 minutes and allow to mixture of 4 volurnes of ethyl acetate and 1 volurne of glacial
separate. Any purple colour produced in the chloroform layer acetic acid and filter. For solution (2) dilute 1 volume of
is not more intense than that obtained in a solution prepared solution (1) to 50 volumes with the same solvent mixture.
in the following manner. Add 2 mL of a 0.0026% w/v Solution (3) contains 0.005% w/v offenbendazole BPCRS.
solution of potassiu111 iodide to a mixture of 4 mL of After removal of the pI ate, allow it to dry in air and examine
1M sulfuric acid and 8 mL of water, add 10 mL of chloroform, under ultraviolet light (254 11m). Any spot in the
shake for 2 minutes, add to the aqueous layer 1 mL of chromatogram obtained with solution (1) corresponding to
hydrogen peroxide solution (100 vol) and 1 mL of chloroform, methyl 5-phenylthio-1H-benzimidazol-2-ylcarbamate is not
shake for 2 minutes and allow to separate (0.1 % w/v of more intense than the spot in the chromatogram obtained
iodide) . with solution (3) (1 %) and any other secondalJi spot is not
more intense than the spot in the chromatogram obtained
ASSAY
with solution (2) (2%).
To a volume containing 1.7 g of Nitroxinil add sufficient
O.OlM sodium hydroxide to produce 500 mL and dilute 20 mL ASSAY
ofthis solution to 500 mL with O.OlM sodiu111 hydroxide. Disperse a quantity of the well-mixed oral suspension
To 5 mL ofthis solution add sufficient O.OlM sodium containing 0.1 g of Oxfendazole in 15 mL of water.
hydroxide to produce 100 mL and measure the absorbance of Add 200 mL of methanol and mix Wiril rile aid of ultrasound
the resulting solution at the maximum at 271 nm, for 15 minutes, cool, add sufficient methanol to produce
Appendix n E. Calculate the content of C 7 H 3IN2 0 3 taking 500 mL and filter. Dilute 4 mL of the filtrate to 100 mL
660 as the value of A(1 %, 1 cm) at the maximum at Wiril methanol and measure the absorbance of rile resulting
271 nm. solution at the maximum at 296 nm, Appendix n B.
STORAGE Calculate the content of C15H13N303S taking 550 as the
value of A(1 %, 1 cm) at rile maximurn at 296 nm.
Nitroxinil Injection should be protected from light.

Oxyclozanide Oral Suspension


Oxfendazole Oral Suspension
Action and use
Action and use Antihelminthic.
Antihelminthic.
DEFINITION
DEFINITION Oxyclozanide Oral Suspension is an aqueous suspension of
Oxfendazole Oral Suspension is an aqueous suspension of Oxyclozanide containing suitable suspending and dispersing
Oxfendazole. agents.
The oral suspension complies 'luith the requirements stated under The oral suspension complies with the requirements stated under.
Oral Liquids and with the following requirements. Oral Liquids and with the following requirements.
Content of oxfendazole, ClsH13N303S Content of oxyclozanide, C 13H 6 CIsN0 3
90.0 to 110.0% of the stated amount. 95.0 to 105.0% of the stated amount.
IDENTIFICATION IDENTIFICATION
Shake a quantity of the oral suspension containing 0.1 g of In test A for Related substances the principal spot in the
Oxfendazole with 50 mL of methanol for 15 minutes, chromatogram obtained with 1O ~lL of solution (1)
centrifuge, evaporate the supernatant liquid to a volume of corresponds to that in the chromatogram obtained with
about 2 mL, cool and filter. Wash the residue with a little solution (3).
water and dry at 105° at a pressure not exceeding 2.7 kPa for
Related substances
1 hour. The residue complies with the following tests.
A. Carry out the method for thin-Iayer chromatography,
Appendix lIT A, using silica gel G as the coating substance
and a mixture of 5 volumes of glacial acetic acid, 20 volumes
2016 Oxytetracycline Preparations Vet-179

of acetone and 60 volurnes of petroleum spirit (boiling range, 60° (2) 0.05% w/v of oxytetracycline hydrochloride BPCRS in
to 80°) as the mobile phase. Apply separately to the plate methanol.
40 ~lL and 10 ~lL of solution (1), 4 ~lL of solution (2) and (3) 0.05% w/v of each of oxytetracycline hydrochloride BPCRS
10 ~L of solution (3). For solution (1) dilute the oral and de111eclocycline hydrochloride BPCRS in methanol.
suspension with acetone to contain 1.0% w/v of Oxyclozanide,
CHROMATOGRAPHIC CONDITIONS
centrifuge and use the supernatant liquido Solution (2)
contains 0.050% w/v of 3,5,6-trichloro-2-hydroxybenzoic (a) Use a silica gel precoated plate (Merck silica gel 60 plates
acid BPCRS in acetone. Solution (3) contains 1.0% w/v of are suitable). Adjust the pH of a 10% w/v solution of
oxyclozanide BPCRS in acetone. After removal of the plate, disodiu111 edetate to 7.0 with 10M sodiu11l hydroxide and spray
allow it to dry in air and spray with a 3.0% w/v solution of the solution evenly onto the plate (about 10 mL for a plate
iron(IJI) chloride hexahydrate in methanol. In the chromatogram 100 mm x 200 mm). Allow the plate to dry in a horizontal
obtained with 40 ~lL of solution (1) any spot corresponding position for at least 1 hour. Before use, dry the plate at 110 0
to 3,5,6-trichloro-2-hydroxybenzoic acid is not more intense for 1 hour.
than the spot in the chromatogram obtained with solution (2) (b) Use the mobile phase as described below.
(0.5%). (c) Apply 1 ~lL of each solution.
B. Carry out the method for thin-layer chromatography, (d) Develop the plate to 15 cm.
Appendix lIT A, using silica gel G as the coating substance (e) After removal of the plate, dry it in a current of air and
and a mixture of 1 volurne of 13.5M ammonia as the mobile examine under ultraviolet light (365 12112).
phase, 10 volurnes of 111ethanol and 100 volumes of ethyl
acetate. Apply separately to the plate 40 ~lL of solution (1) MOBILE PHASE
and 4 ~lL of solution (2). For solution (1) dilute the oral 6 volurnes of zuater, 35 volumes of methanol and 59 volumes
suspension with acetone to contain 1.0% w/v of Oxyclozanide, of dichloromethane.
centrifuge and use the supernatant liquido Solution (2) SYSTEM SUITABILITY
contains 0.050% w/v of 2-a111ino-4,6-dichlorophenol
The test is not valid unless the chromatogram obtained with
hydrochloride BPCRS in acetone. After removal of the plate,
solution (3) shows two clearly separated spots.
allow it to dry in air and spray with phosphomolybdotungstic
reagent. In the chromatogram obtained with solution (1) any CONFIRMATION
spot corresponding to 2-amino-4,6-dichlorophenol is not The principal spot in the chromatogram obtained with
more intense than the spot in the chromatogram obtained solution (1) is similar in position, colour and size to that in
with solution (2) (0.4%). the chromatogram obtained with solution (2).
ASSAY B. To a quantity of tlle powder containing 0.4 mg of
Protect the solutions from light throughout the Assay. To a Oxytetracycline Hydrochloride add 5 mL of a 1 % w/v
quantity of the oral suspension containing 60 mg of solution of sodiwn carbonate, shake· and add 2 mL of
Oxyclozanide add 60 mL of acidified methanol and boil gently diazobenzenesulfonic acid solution. A light brown colour is
on a water bath. Shake continuously for 20 minutes, cool to produced.
2° and dilute to 100 mL with acidified methanol. Filter, dilute C. Shake a quantity ofthe powder containing 0.1 g of
5 mL of the filtrate to 100 mL with acidified methanol and Oxytetracycline Hydrochloride with 10 mL of 2M nitric acid
measure the absorbance of the resulting solution at the and filter. Decolourise the filtrate with activated charcoal and
maximurn at 300 nm, Appendix IT B. Calculate the content filter again. The filtrate yields the reactions characteristic of
of C 13 H 6 CI sN0 3 taking 254 as the value of A(l %, 1 cm) at chlorides, Appendix VI.
the maximurn at 300 nm. ASSAY
Can-y out the method for liquid chromatography,
Appendix ITI D, using the following solutions.
(1) Dissolve a quantity of the oral powder in sufficient
Oxytetracycline Veterinary Oral Powder O.OlM hydrochlonc acid to produce a solution containing
0.005% w/v of Oxytetracyc1ine Hydrochloride.
Action and use
(2) 0.005% w/v of oxytetracycline BPCRS in
Tetracyc1ine antibacterial.
O.OlM hydrochloric acid.
DEFINITION (3) 0.1 % w/v of 4-epioxytetracycline EPCRS in
Oxytetracycline Veterinary Oral Powder is a mixture of O. O1M hydrochlonc acid.
Oxytetracycline Hydrochloride and Lactose or other suitable (4) 0.1 % w/v of tetracycline hydrochloride BPCRS in
diluent. O.OlM hydrochloric acid.
Content of oxytetracycline hydrochloride, (5) Dilute a mixture containing 1.5 mL of a 0.1 % w/v
C22H24N209,HCI solution of oxytetracycline BPCRS in O.OlM hydrochlO1ic acid,
90.0 to 110.0% of the stated amount. 1 mL of solution (3) and 3 mL of solution (4) to 25 mL
The vetenna1Y oral pozuder c0111plies zuith the require111ents stated with O.OlM hydrochloric acid.
under Vetenna¡y Oral Pozuders and zuith the follo'lving CHROMATOGRAPHIC CONDITIONS
requirements. (a) Use a stainless steel column (25 cm x 4.6 mm) packed
IDENTIFICATION with styrene-divinylbenzene copolymer (8 to 1O ~lm) (Polymer
A. Carry out the method for thin-Iayer chro111atography, Laboratories, PLRP-S 100A, is suitable).
Appendix lIT A, using the following solutions. (b) U se isocratic elution and the mobile phase described
(1) Extract a quantity of the oral powder containing 10 mg below.
of Oxytetracycline Hydrochloride with 20 mL of 111ethanol, (c) Use a flow rate of 1 mL per minute.
centrifuge and use the supernatant liquido
Vet-180 Pentobarbital Preparations 2016

(d) Use a column temperature of 60°. filter, wash the residue with four 5 mL quantities of 'Water
(e) Use a detection wavelength of 254 nm. and transfer as completely as possible to a small flask.
Add 25 mL of ethanol (96%) and heat under a reflux
(±) Inject 20 ~lL of each solution.
condenser for 10 minutes. The residue, after drying at 105°
MOEILE PHASE for 30 minutes, melts completely between 136° and 155°.
To 50.0 g of 2-methylpropan-2-ol add 200 mL of 'Water,
ASSAY
60 mL of O.33J'iI phosphate buffer pH 7.5, 50 mL of a
To a volume containing 0.5 g of Pentobarbital Sodium
1.0% w/v solution of tetrabutylammonium hydrogen sulfate
diluted to 15 mL with 'Water add 5 mL of 2M hydrochloric
previously adjusted to pH 7.5 with 2M sodium hydroxide and
acid, extract with 50 mL of ether and then with successive
10 mL of a 0.04% w/v solution of disodium edetate previously
25 mL quantities of ether until complete extraction is
adjusted to pH 7.5 with 2M sodium hydroxide and dilute to
effected. Wash the combined extracts with two 5 mL
1 litre with 'water.
quantities of 'Water and wash the combined aqueous washings
SYSTEM SUITABILITY with 10 mL of ether. Add the ether to the main ether extract,
The Assay is not valid unless, in the chromatogram obtained evaporate to low volume, add 2 mL of absolute ethanol,
with solution (5): evaporate to dryness and dry the residue to constant weight
the resolution factor between the first peak at 105°. Each g of residue is equivalent to 1.097 g of
(4-epioxytetracyc1ine) and the second peak (oxytetracyc1ine) CllH17N2Na03'
is at least 4.0; When pentobarbitone injection is prescribed or demanded,
the resolution factor between the second peak and the third Pentobarbital Injection shall be dispensed or supplied.
peak (tetracyc1ine) is at least 5.0 (if necessary reduce the
1 Solutions comaining 20% w/v of Pentobarbital Sodium in 100 mL and
content of 2-methylpropan-2-ol in the mobile phase to
500 mL quantities are also available for purposes other than injectionj such
increase the resolution);
solutions are not necessariÓI sterile but comPÓ' with all the other requirements
the symmetly factor of the peak due to oxytetracyc1ine is not of the 11l0nographj they may be coloured.
more than 1.25.
DETERMINATION OF CONTENT
Calculate the content of C22H24N209,HCl in the oral
powder using the dec1ared content of C22H2~209 in Phenylbutazone Tablets
oxytetracycline BPCRS. Each mg of C22H24N209 is equivalent
to 1.079 mg of C22H24N209,HCl. Action and use
Cyc1o-oxygenase inhibitor; pyrazolone analgesic.

DEFINITION
Phenylbutazone Tablets contain Phenylbutazone. They are
Pentobarbital Injection coated.
Action and use The tablets comply 'With the requiremems stated under Tablets and
Barbiturate. 'With the follo'Wing require111ents.
Content of phenylbutazone, C19H2oN202
DEFINITION 95.0 to 105.0% ofthe stated amount.
Pentobarbital Injection is a sterile 1 solution of Pentobarbital
IDENTIFICATION
Sodium in a suitable vehic1e.
Extract a quantity of the powdered tablets containing 0.2 g of
The injection complies 'With the requirements stated under
Phenylbutazone with 40 mL of warm acetone, filter and
Parenteml Prepamtions and 'With the follo'Wing requirements.
evaporate the filtrate to dryness. The residue complies with
Content of pentobarbital sodium, CUH17N2Na03 the following tests.
95.0 to 105.0% of the stated amount. A. The infrared absorption spectrum, Appendix II A, is
CHARACTERISTICS concordant with the reference spectrum of phenylbutazone
A c1ear, colourless or almost colourless solution. (RSV 35).
IDENTIFICATION B. To 0.1 g of the residue add 1 mL of glacial acetic acid and
A. The infrared absorption spectrum of the residue obtained in
2 mL of hydrochlonc acid and heat. on a water bath for
the Assay, Appendix II A, is concordant with the reference 30 minutes. Cool, add 10 mL of 'Water and filter. Add to the
spectrum of pentobarbital (RSV 34).
filtrate 3 mL of O.lM sodium nitrite; a yellow colour is
produced. Add 1 mL of this solution to 5 mL of 2-naphthol
B. Melting point of the residue obtained in the Assay, about solution; a brownish red precipitate is produced which
128°, Appendix V A. dissolves on the addition of ethanol (96%) yielding a red
C. When introduced on a platinum wire into the flame of a solution.
Bunsen bumer, imparts a yellow colour to the flameo
TESTS
TESTS Dissolution
Alkalinity Comply with the requirements for Monographs of the British
pH, 10.0 to 11.5, Appendix VL. Pharmacopoeia in the dissolution test for tablets and capsules,
Isomer Appendix XII B 1.
To a volume containing 0.3 g of Pentobarbital Sodium TEST CONDITIONS
diluted, if necessary, to 5 mL with 'Water, add 0.3 g of
(a) Use Apparatus 1, rotating the basket at 100 revolutions
4-nitrobenzyl bromide dissolved in 10 mL of ethanol (96%) and
per minute.
heat under a reflux condenser for 30 minutes. Cool to 25°,
2016 Procaine Benzylpenicillin Preparations Vet-181

(b) Use 900 mL of a 0.68% w/v solution of potassium


dihydrogen orthophosphate adjusted to pH 7.5 with 1M sodiu111
Piperazine Citrate Tablets
hydroxideJ at a temperature of 37° J as the medium. Action and use
PROCEDURE Antihelminthic.
After 45 minutes withdraw a 10 mL sample of the medium
and measure the absorbance of the filtered sampleJ suitably DEFINITION
diluted with the dissolution medium if necessaryJ at the Piperazine Citrate Tablets contain Piperazine Citrate.
maximum at 264 nmJ Appendix II B using the dissolution The tablets c0111ply 'luith the requirements stated undel' Tablets and
medium in the reference cell. with the following requirements.
DETERMINATION OF CONTENT Content of anhydrous piperazine citrate,
Calculate the total content of phenylbutazone J C19H20N202J (C4HlONz)3,2C6Hg07
in the medium taking 653 as the value of A(l %J 1 cm) at the 81.6 to 96.8% ofthe stated amount ofPiperazine Citrate.
maximum at 264 nm. IDENTIFICATION
Related substances Extract a quantity of the powdered tablets containing 1 g of
Carry out the method for thin-layer chromatographyJ Piperazine Citrate with 20 mL of water and filter. The filtrate
Appendix III AJ using the following solutions. complies with the following tests.
(1) Shake a quantity of the powdered tablets containing 0.1 g A. Dilute 1 mL to 5 mL with waterJ add 0.5 g of sodiUln
of Phenylbutazone with 3 mL of chloroform containing hydrogen carbonateJ 0.5 mL of potassium hexacyanofen'ate(m)
0.02% w/v of butylated hydroxytolueneJ centrifuge and use the solution and 0.1 mL of mercury, shake vigorously for 1 minute
supernatant liquido and allow to stand for 20 minutes; a reddish colour slowly
(2) Dilute 1 volume of solution (1) with sufficient of the develops.
same solvent mixture to produce a solution containing B. Mix 4 mL with 1 mL of hydrochloric acidJ add 0.5 g of
0.5 mg of Phenylbutazone per mL. sodiu111 nitlite J heat to boilingJ cool in ice for 15 minutes,
scratching the side of the container with a glass rod to induce
CHROMATOGRAPHIC CONDITIONS
crystallisation and filter. The melting point of the crystalsJ after
(a) Use as the coating silica gel GF254 (Machery Nagel plates washing with 10 mL of iced water and drying at 100° to
are suitable). Prior to applying solutions (1) and (2)J pre- 105°J is about 159° J Appendix V AJ Method 1.
treat the plate with the mobile phase allowing the solvent
C. Yield the reactions characteristic of citrates J Appendix VI.
front to ascend 4 cmJ remove the plate and dry it in a
current of cold airo ASSAY
(b) Use fresh mobile phase as described below. Weigh and powder 20 tablets. Dissolve as completely as
possible a quantity of the powder containing 0.2 g of
(c) Without delay and in an atmosphere of carbon dioxide
Piperazine Citrate in 10 mL of watel'J filter and wash the
apply 3 ~lL of each solution. Expose the plate to cal"bon
filter with three 5 mL quantities of water. To the combined
di oxide for 2 minutes.
filtrate and washings add 3.5 mL of 0.5M sulfu1"l'c acid and
(d) Develop the plate to 10 cm. 100 mL of picric acid solution R1 J heat on a water bath for
(e) After removal of the plate J allow it to dry in air and 15 minutes J allow to stand for 1 hourJ filter J wash the residue
examine under ultraviolet light (254 nm). with piperazine dipicrate SOlUtiOll until the washings are free
MOBILE PHASE from sulfate and dry the residue to constant weight at 105°.
10 volumes of glacial acetic acidJ 40 volumes of cyclohexane Each g of residue is equivalent to 0.3935 g of
and 50 volumes of chloroform containing 0.02% v/v of (C4HION2)3J2C6HsÜ7'
butylated hydroxytoluene. STORAGE
LIMITS Piperazine Citrate Tablets should be protected from light.
Any secondaly spot in the chromatogram obtained with
solution (1) is not more intense than the spot in the
chromatogram obtained with solution (2) (1.5%).
ASSAY Procaine Benzylpenicillin Injection
Weigh and powder 20 tablets. Extract a quantity of the
powder containing 0.5 g of Phenylbutazone with successive Action and use
30- J 30-J 15- and 15-rnL quantities of warm acetone. Filter Penicillin antibacterial.
the combined extracts J cool and titrate with O.lM sodium
DEFINITION
hydroxide VS using bromothymol blue solution R3 as indicator
Procaine Benzylpenicillin Injection is a sterile suspension of
and continuing the titration until the blue colour persists for
Procaine Benzylpenicillin in Water for Injections.
at least 30 seconds. Repeat the titration without the
powdered tablets; the difference between the titrations The injection complies with the requirements stated under
represents the amount of alkali required by the Parenteral Preparatiáns and with the following requirements.
phenylbutazone. Each mL of O.lM sodium hydroxide VS is Content of total penicillins, calculated as
equivalent to 30.84 mg of C19H20N202' C13H~oNzOz,C16HlgNz04S, HzO
90.0 to 110.0% of the stated amount of Procaine
Benzylpenicillin.
Content of procaine, C13HzoNzOz
36.0 to 44.0% of the stated amount of Procaine
Benzylpenicillin.
Vet-182 Procaine Benzylpenicillin Preparations 2016

CHARACTERISTICS (f) Inject 20 ~lL of each solution.


A white suspension. (g) For solution (1) allow the chromatography to proceed for
IDENTIFICATION 1.5 times the retention time of the peak due to
A. Dilute a volume of the well-shaken suspension containing benzylpenicillin.
10 mg of Procaine Benzylpenicillin to 10 mL with water and MOBILE PHASE
add 0.5 mL of neutral red solution. Add sufficient 250 volumes of acetonitrile, 250 volumes of water and
O.OlM sodium hydroxide to produce a permanent orange 500 volumes of a freshly prepared solution containing
colour and then add 1 mL of penicillinase solution. A red 1.4% w/v of potassium dihydrogen orthophosphate and
colour is produced rapidly. 0.65% w/v of tetrabutylanz11l0niu111 hydroxide, adjusted to
B. Carry out the method for thin-layer chromatography, pH 7.0 with 1M potassium hydroxide. Adjust the pH of the
Appendix lIT A, using the foIlowing solutions. mixture to 7.2 with 2M orthophosphoric acid, if necessary.
(1) Shake a volume of the weIl-shaken suspension containing For solution (3), when the chromatogram is recorded under
50 mg of Procaine Benzylpenicillin with 5 mL of methanol, the prescribed conditions the substances elute in the
add a small quantity of water to dissolve any residue and following order: 4-aminobenzoic acid, procaine,
dilute to 10 mL with water. benzylpenicillin.
(2) 0.5% w/v of procaine benzylpeniczllin BPCRS in acetone. SYSTEM SUIT ABILITY
CHROMATOGRAPHIC CONDITIONS The test is not valid unIess, in the chromatogram obtained
(a) Use a TLC silica gel silanised plate (Merck silanised silica with solution (3), the resolution factor between the peaks due
gel 60 plates are suitable). to 4-aminobenzoic acid and procaine is at least 2.0.
(b) Use the mobile phase as described below. If necessary, adjust the concentration of acetonitrile in the
mobile phase.
(c) Apply 1 ~lL of each solution.
LIMITS
(d) Develop the plate to 15 cm.
In the chromatogram obtained with solution (1):
(e) After removal of the plate, allow it to dry in air, expose to
iodine vapour until spots appear and examine in daylight. the area of any peak due to 4-aminobenzoic acid is not
greater than 10 times the area of the corresponding peak in
MOBILE PHASE
the chromatogram obtained with solution (2) (0.5%);
30 volumes of acetone and 70 volumes of a 15.4% w/v the area of any other seconda1JI peak is not greater than the
solution of a11Znzonium acetate adjusted to pH 7.0 with area of the peak corresponding to benzylpenicillin in the
10M ammonia. chromatogram obtained with solution (2) (1 %).
SYSTEM SUIT ABILITY
Bacterial endotoxins
The test is not valid unIess the chromatogram obtained with Carry out the test for bacterial endotoxins, Appendix XIV C,
solution (2) shows two clearly separated spots. Method C. Dilute a quantity of the well-shaken suspension,
CONFIRMATION if necessary, with water BET to produce a solution containing
The two principal spots in the chromatogram obtained with 3 mg of Procaine Benzylpenicillin per mL (solution A).
solution (1) are similar in position, colour and size to those The endotoxin limit concentration of solution A is 0.3 IV per
in the chromatogram obtained with solution (2). mL. Carry out the test using a suitable dilution of solution A
as described under Method C.
C. Yields the reaction characteristic of prúna1y aromatic
amines, Appendix VI, producing a bright orange-red ASSAY
precipitate. Carry out the method for liquid chromatography,
Appendix ITI D, using the foIlowing solutions.
TESTS
Related substances (1) Add to a quantity of the well-shaken suspension
Carry out the method for liquid chromatography, containing 70 mg of Procaine Benzylpenicillin sufficient
Appendix lIT D, using the foIlowing solutions. mobile phase to produce 100 mL, mix, filter and use the
filtrate.
(1) To a quantity of the weIl-shaken suspension containing
70 mg of Procaine Benzylpenicillin add sufficient mobile (2) 0.07% w/v of procaine benzylpenicillin BPCRS in the
phase to produce 50 mL, mix, filter and use the filtrate. mobile phase.
(2) Mix 1 mL of solution (1) and 1 mL of a 0.007% w/v (3) Dissolve 4 mg of 4-aminobenzoic acid in 25 mL of
solution of 4-aminobenzoic acid and add sufficient mobile solution (2).
phase to produce 100 mL. CHROMATOGRAPHIC CONDITIONS
(3) Dissolve 4 mg of 4-aminobenzoic acid in 25 mL of a The chromatographic conditions described under Related
solution containing 0.070% w/v of procaine substances may be used.
benzylpenicillin BPCRS in the mobile phase. For solution (3), when the chromatogram is recorded under
CHROMATOGRAPHIC CONDITIONS the prescribed conditions the substances elute in the
(a) Use a stainless steel column (25 cm x 4.6 mm) packed following order: 4-aminobenzoic acid, procaine,
with octadecylsilyl silica gel for chromatography (5 ~m) benzylpenicillin.
(Lichrospher ODS is suitable). SYSTEM SUIT AB¡LITY
(b) U se isocratic elution and the mobile phase described The Assay is not valid unless, in the chromatogram obtained
below. with solution (3), the resolution factor between the peaks due
(c) Use a fiow rate of 1.5 mL per minute. to 4-aminobenzoic acid and procaine is at least 2.0.
If necessary, adjust the concentration of acetonitrile in the
(d) Use an ambient column temperature.
mobile phase.
(e) Use a detection wavelength of 225 nm.
2016 Pyrethrum Preparations Vet-183

DETERMINATION OF CONTENT 150 mL, cool rapidly and transfer the solution to a stoppered
Calculate the content of C13HzoNzOz and of flask, washing the beaker wim three 20 mL quantities of
C13HzoNzOz,C16HlSNz04S,HzO in dle injection from the water and transfen-ing any gummy residue to the flask.
chromatograms obtained and using the declared content of Add 1 g of diatomaceous earm (Filtercel is suitable) and
C13HzoNzOz and of C13HzoNzOz,C16HlSNz04S,HzO in 10 mL of barium ch10ride solution, swirl gently and add
procaine benzylpenicillin BPCRS. sufficient water to produce 250 mL. Stopper the flask, shake
3 g of Procaine Benzylpenicillin is approximately equivalent vigorously until the separating liquid is clear and filter the
to 2 g of benzylpenicillin. suspension through a filter paper (Whatman No. 1 is
suitable).
F or pyrethrin 1
Transfer 200 mL of me filtrate to a separating funnel, rinsing
the measuring vessel with two 5 mL quantities of water, and
Pyrethrum Extract add 0.05 mL of pheno1phthalein solution Rl. Neutralise me
DEFINITION solution by the drop wise addition of hydrochloric acid and
Pyremrum Extract is prepared from Pyrethrum Flower. add 1 mL of hydmch10ric acid in excess. Add 5 mL of a
saturated solution of sodiu111 ch101'ide and 50 mL of aromatic-
Extemporaneous preparation
free petroleum spirit (boiling rangeJ 40° to 60°), shake vigorously
Exhaust Pyrethrum Flower, in coarse powder, by percolation
for 1 minute, allow to separate, remove and retain me lower
widl a suitable hydrocarbon solvent; remove the solvent and
layer. Filter the petroleum spirit extract through absorbent
concentrate at a low temperature. The resulting product may
cotton into a second separating funnel containing 10 mL of
be decolourised by a suitable procedure. Detetmine the
water. Retum the aqueous layer to the first separating funnel
proportion of pyremrins in a portion of the extract by the
and repeat the extraction with 50 mL and men wim 25 mL
Assay. To the remainder add, if necessary, sufficient Light
of aromatic-free petro1eum spirit (boi1ing rangeJ 40° to 60°),
Liquid Paraffin or deodol"ised kerosene to produce an extract of
reserving the aqueous layer for me assay of pyredmn II, and
the required strengm.
filtering me petroleum spirit extracts through the same
The extract complies with the requirements for Labelling stated absorbent cotton into the second separating funnel. Shake
under Extracts and with the following requirements. the combined petroleum spirit extracts and water for about ,
Content of pyrethrins 30 seconds and allow to separate; remove me lower layer and
24.5% to 25.5% w/w, of which not les s than half consists of add it to me aqueous liquid reserved for me assay of
pyremrin 1. pyrethrin II. Wash me combined petroleum spirit extracts
with a furmer 10 mL of water, adding me washings to me
CHARACTERISTICS
reserved aqueous liquido
A dark olive green or brown viscous liquid or, if
decolourised, a pale amber liquido To the petroleum spirit extracts add 5 mL of O.lM sodium
hydroxide, shake vigorously for 1 minute, allow to separate
ASSAY and remove the clear lower layer, washing dle stem of the
To 0.5 g of dle well-mixed extract add 20 mL of separating funnel wim 1 mL of water. Repeat the extraction
0.5M ethanolic potassium hydmxide and boil under a reflux by shaking for about 30 seconds with two quantities of
condenser for 45 minutes. Transfer me solution to a beaker 2.5 mL and 1.5 mL of O.lM sodium hydmxide and add me
and wash me flask wim sufficient hot water, addingthe extracts to the alkaline extract. Add to me flask 10 mL of
washings to the beaker, to produce a total volume of merC'Ll1y(JI) sulfate solution, stopper, swirl and allow to stand in
200 mL. Boil until me volume is reduced to 150 mL, cool the dark at 25° ± 0.5° for exactly 60 minutes after me
rapidly and transfer the solution to a stoppered flask, washing addition of the mercury(n) sulfate solution. Add 20 mL of
dle beaker widl mree 20 mL quantities of water and acetone and 3 mL of a saturated solution of sodium ch101'zde,
transfen-ing any gummy residue to me flask. Add 1 g of heat to boiling on a water bath, allow the precipitate to setde
diatomaceous earm (Filtercel is suitable) and 10 mL of and decant the supematant liquid through a filter paper
barium chlol'ide solution, swirl gently and add sufficient water (Whatman No. 1 is suitable), retaining most of the
to produce 250 mL. Stopper me flask, shake vigorously until precipitate in dle flask. Wash the precipitate with 10 mL of
the separating liquid is clear and filter the suspension mrough acetone, again boil, allow to settle and decant through me
a filter paper (Whatman No. 1 is suitable). same filter paper. Repeat ·the washing and decanting with
If the pyremrum extract is coloured, carry out me following three 10 mL quantities of hot ch1omform. Transfer the filter
preliminary treatment. Transfer 0.5 g of me well-mixed paper to the flask, add 50 mL of a cooled mixture of three
extract to a stoppered flask, add 50 mL of aromatic-free volumes of hyd1'ochloric acid and two volumes of water, 1 mL
petro1eum spirit (boiling range 40° to 60°), swirl, add 1 g of
J of strong iodine monoch101'ide reagent and 6 mL of ch1orofo1'111.
diatomaceous earm (Filtercel is suitable), swirl to mix Titrate with O.OlM potassium iodate VS, running almost all dle
completely, stopper me flask and allow to stand at 20° to 22° required volume of titrant into me flask in one portion.
for 16 hours. Mix the contents of me flask moroughly, filter Continue me titration, shaking me flask vigorously for
with gende suction mrough a sintered-glass filter (ISO 4793, 30 seconds after each addition of me titrant, until me
porosity grade 4, is suitable) and wash the residue with five chloroform is colourless. Repeat the operation wimout the
10 mL quantities of aro111atic-free petroleum spirit (boi1ing range
J extract; me difference between dle titrations represents me
40° to. 60°). Remove me solvent from the combined filtrate amount of potassium iodate required. Each mL of
and washings and evaporate to a volume of 1 to 2 mL. O.OlM potassium iodate VS is equivalent to 5.7 mg of
Add 20 mL of O.5M ethano1ic potassium hydroxide and boil pyrethrin 1.
under a reflux condenser for 45 minutes. Transfer the F or pyrethrin n
solution to a beaker and wash me flask wim sufficient hot Transfer the combined aqueous liquids reserved in me Assay
water, adding me washings to me beaker, to produce a total for pyrethrin I to a beaker, cover with a watch glass and
volume of 200 mL. Boil until me volume is reduced to evaporate to 50 mL widlln 35 to 45 minutes. Cool, washing
Vet-184 Sodium Ca1cium Edetate Preparations 2016

the underside of the watch glass with not more than 5 mL of The concentrate complies with the requirements for Concentrates
wate," and adding the washings to the beaker. Filter through for Injections or Infusions stated under Pal'enteral Preparations
absorbent cotton into a separating funnel, washing with and with the following requirements.
successive quantities of 10, 7.5, 7.5, 5 and 5 mL of water. Content of anhydrous sodium calcium edetate,
Saturate the aqueous liquid with sodiul11 chloride, add 10 mL ClOH12CaN2Na20g
of hydrochloric acid and 50 mL of ether, shake for 1 minute, 22.5 to 27.5% w/v.
allow to separate, and remove the lower layer. Repeat the
extraction successively with 50, 25 and 25 mL of ether. Wash CHARACTERISTICS
the combined ether extracts with three 10 mL quantities of a A colourless solution.
saturated solution of sodiul11 chloride and transfer the ether IDENTIFICATION
layer to a fiask with the aid of 10 mL of ether. Remove the A. Dilute 2.5 mL with 7.5 mL of water, make alkaline to
bulk of the ether by distillation and remove the remainder litmus paper with 5M am1110nia and add 5 mL of a 2.5% w/v
with a gentle current of air and dry the residue at 100° for solution of am11l0niul11 oxalate. Not more than a trace of
10 minutes, removing any residual acid fumes with a gentle precipitate is produced.
current of airo Add 2 mL of ethanol (96%) previously
B. To 10 mL add 2 mL of a 10% w/v solution of lead(n)
neutralised to phenolphthalein solution Rl and 0.05 mL of
nitra te, shake and add 5 mL of dilute potassiul11 iodide solution;
phenolphthalein solution Rl, swirl to dissolve the residue, add
no yellow precipitate is produced. Make alkaline to litmus
20 mL of carbon dioxide-free 'water and titrate rapidly with
paper with 5M ammonia and add 5 mL of a 2.5% w/v solution
0.02M sodiu111 hydroxide VS until the colour changes to
of al11monium oxalate; a white precipitate is produced.
brownish pink and persists for 30 seconds, keeping the fiask
stoppered between additions of alkali. Repeat the operation C. Evaporate to dryness and ignite. The residue yields the
using the aqueous liquid reserved for the repeat operation in reactions characteristic of sodiu111 salts and of calcium salts,
the Assay for pyrethrin 1. The difference between the Appendix VI.
titrations represents the volume of 0.02M sodium hydroxide VS TESTS
required. Each mL of 0.02M sodium hydroxide VS is Acidity or aIkalinity
equivalent to 3.74 mg of pyrethrin n. pH, 6.5 to 8.0, Appendix V L.
STORAGE Bacterial endotoxins
Pyrethrum Extract should be kept in a well-filled container, Carry out the test for bacterial endotoxins, Appendix XIV C.
protected from light and should be thoroughly stirred before The endotoxin limit concentration is not more than 0.125 ID
use. per mg of sodium calcium edetate.
ASSAY
To 2.5 mL add 90 mL of water, 7 g of hexamine and 5 mL
of 2M h_vdrochloric acid and titrate with 0.05M lead nitrate VS
Sodium Calcium Edetate Intravenous using xylenol orange sollltion as indicator. Each mL of
0.05M lead nitrate VS is equivalent to 18.71 mg of
Infusion tor Veterinary Use ClOH 12 CaNzNazOs·
Action and use STORAGE
Chelating agent. Sterile Sodium Calcium Edetate Concentrate for Veterinary
Use should be kept in containers made fram lead-free glass.
DEFINITION
Sodium Calcium Edetate Intravenous Infusion for Veterinary
Use is a sterile solution of Sodium Calcium Edetate. It is
prepared irnmediately before use by diluting Sterile Sodium
Calcium Edetate Concentrate for Veterinary Use with a Sulfadimidine Injection
suitable diluent in accordance with the manufacturer's
Action and use
instructions.
Sulfonamide antibacterial.
The intravenous infusion c0111plies with the requirements stated
under Parenteral Preparations and with the follO'lving requirement. DEFINITION
LABELLING Sulfadimidine Injection is a sterile solution of sulfadimidine
The quantity of active ingredient is stated in terms of the sodium in Water for Injections free from dissolved airo It is
equivalent amount of anhydrous sodium calcium edetate in a prepared by the interaction of Sulfadimidine and Sodium
suitable dose-volume. Hydroxide.
The injection complies with the requirements stated lInder
Parenteral Preparations and with the following requirements.
STERILE SODIUM CALCIUM EDETATE
Content of sulfadimidine sodium, C12H13N4Na02S
CONCENTRATE FOR VETERINARY USE
95.0 to 105.0% ofthe stated amount.
DEFINITION
IDENTIFICATION
Sterile Sodium Calcium Edetate Concentrate for Veterinary
A. Acidify a volume containing 0.1 g of sulfadimidine sodium
Use is a sterile solution of Sodium Calcium Edetate in Water
with 6M acetic acid, filter, reserving the filtra te, wash the
for Injections containing the equivalent of 25.0% w/v of
residue with water and dry at 105°. The infrared absorption
anhydrous sodium calcium edetate.
spectnl111 of the residue, Appendix n A, is concordant with
the reference spectrum of sulfadimidine (RSV 50).
2016 Sulfadoxine Preparations Vet-185

B. The residue obtained in test A yields the reaction CHARACTERISTICS


characteristic of primaly aromatic amines, Appendix VI, A c1ear, yellow solution.
producing a bright orange-red precipitate.
IDENTIFICATION
TESTS A. Evaporate 50 mL of solution B obtained in the Assay for
Alkalinity trimethoprim to about 10 mL, neutralise with 5M sodiu111
pH, 10.0 to 11.0, Appendix V L. hydroxide and then acidify with 2M acetic acid. Dissolve the
Colour of solution precipitate by warming and adding a small volume of ethanol
An injection containing 1 g of sulfadimidine sodium in 3 mL (25%). Cool, recrystallise the precipitate from ethanol (25%),
is not more intensely coloured than reference solution Y4' wash with water and dry at 105°. The infrared absol'ption
Appendix IV B, Method 1. spectrwl1 of the residue, Appendix IT A, is concordant with
the reference SpeCt1'U111 of sulfadoxine (RSV 37).
Related substances
Carry out the method for thin-layer chromatography, B. Evaporate 100 mL of solution A obtained in the Assay for
Appendix lIT A, using silica gel H as the coating substance trimethoprim to about 4 mL, transfer to a test tube with the
and a mixture of 18 volumes of 10M am1110nia and aid of about 4 mL of hot water and allow to cool. Wash the
90 volumes of butan-l-ol as the mobile phase. Apply resulting crystals with water and dry at 105°. The infrared
separately to the plate 1O ~lL of each of the following abs01ption spectrum of the residue, Appendix II A, is
solutions. For solution (1) use the injection being examined concordant with the reference spectl'Ll11l of trimethoprim
diluted with water to contain 0.20% w/v of sulfadimidine (RSV 45).
sodium. Solution (2) contains 0.0020% w/v of sulfanilamide C. Carry out the method for thin-layer chmmatography,
in a mixture of 1 volume of 13.5M a1111110nia and 9 volumes Appendix ITI A, using silica gel G as the coating substance
of ethanol (96%). After removal of the plate, heat it at 105° and a mixture of 19 volumes of chloroform and 1 volume of
for 10 minutes and spray with a 0.1 % w/v solution of methanol as the mobile phase. Apply separately to the plate
4-dimethylaminobenzaldehyde in ethanol (96%) containing 5 ~lL of each of the following solutions. For solution (1)
1 % v/v of hydrochlO1ic acid. Any secondaly spot in the dilute the injection with 111ethanol to contain 2.5% w/v of
chromatogram obtained with solution (1) is not more intense Sulfadoxine. Solution (2) is a 0.040% w/v solution of
than the spot in the chromatogram obtained with solution (2) lidocaine hydrochloride BPCRS in 0.1 M sodium hydroxide in
(1 %). methanol. After removal of the plate, allow it to dry in air and
spray with potassiu111 iodobismuthate solution. For an injection
ASSAY
labelled as containing Lidocaine Hydrochloride the
Dilute a volume containing 0.5 g of sulfadimidine sodium to
chromatogram obtained with solution (1) exhibits a spot
75 mL with water, add 10 mL of hydrochloric acid and pass
corresponding to the spot in the chromatogram obtained with
air slowly through the solution until the vapours do not turn
solution (2); an injection not so labelled exhibits no such
moistened starch iodate papel' blue. Add 3 g of potassium
spot.
bromide, cool in ice and titrate slowly with O.lM sodiu111 nitrite
VS, stilTing constant1y and determining the end point All,alinity
electrometrically. Each mL of O.lM sodiu111 nitJite VS is pH, 9.0 to 10.5, when diluted with an equal volume of
equivalent to 30.03 mg of C12H13N4Na02S. cal'bon dioxide-free water, Appendix V L.

STORAGE ASSAY
Sulfadimidine Injection should be protected from light. For trimethoprim
Prepare an anion exchange column (Dowex 1-X1 is suitable)
LABELLING pre-treated in the following manner. Wash with 100 mL of
The strength is stated as t11e amount of sulfadimidine sodium water, rinse with 50 mL of 1M sodiu111 hydmxide, wash with
in a suitable dose-volume. water until the washings are neutral, rinse with 70 mL of
O.lM hydrochlO1ic acid in methanol (70%) and again wash with
water; activate the column with 50 mL of 1M sodium
hydroxide, again wash with water and finally moisten with
111ethanol (70%).
Sulfadoxine and Trimethoprim Injection
Apply to the anion-exchange column 20 mL of a dilution of
Action and use the injection in methanol (70%) containing 50 mg of
Sulfonamide antibacterial. Trimethoprim and elute with methanol (70%) to a final
elution volume of 200 mL (solution A). Elute the material
DEFINITION remaining on the column with 150 mL of O.lM hydrochlO1ic
Sulfadoxine and Trimethoprim Injection is a sterile solution, acid in methanol (70%) and add sufficient O.lMhydmchlO1ic
in a suitable aqueous vehic1e, containing Sulfadoxine and acid in methanol (70%) to produce 200 mL (solution B).
Trimethoprim in the proportion five parts to one parto Reserve solution B for the Assay for sulfadoxine and
The pH is adjusted to about 10 by the addition of Sodium Identification test A. To 10 mL of solution A add 1 mL of
Hydroxide. It may contain 0.1 % w/v of Lidocaine 1M sodiu111 hydroxide and sufficient 111ethanol (70%) to produce
Hydroch1oride. 100 mL. Measure the absorbance of the resulting solution at
The injection complies with the requirements stated under the maximum at 288 nm, Appendix II B. Calculate the
Parenteral Preparations and with the following require111ents. content of C14HlSN403 taking 250 as the value of
A(1 %, 1 cm) at the maximum at288 nm.
Content of sulfadoxine, C12H14N404S
92.5 to 107.5% ofthe stated amount. For sulfadoxine
To 2 mL of solution B obtained in the Assay for
Content of trimethoprim, C14H18N403
Trimethoprim add sufficient O.lM hydrochlOJic acid in
92.5 to 107.5% ofthe stated amount.
methanol (70%) to produce 250 mL. Measure the absorbance
Vet-186 Tylosin Preparations 2016

of the resulting solution at the maximum at 267 nm, (c) Use a fiow rate of 1 mL per minute.
Appendix Ir B. Calculate the content of C12H14N404S from (d) Use an ambient column temperature.
the absorbance obtained by repeating the procedure using
(e) Use a detection wavelength of 290 llffi.
2 mL of a 0.125% w/v solution of sulfadoxine BPCRS in
O.lM hyd1'Ochloric acid in methanol (70%) in place of (f) Inject 20 ~lL of each solution.
solution B and using the declared content of C12H14N404S MOBILE PHASE
in sulfadoxine BPCRS. 0.85M sodiu111 perchlorate in a 40% v/v solution of acetonitrile,
STORAGE the solution being adjusted to pH 2.5 using 1M hyd1'Ochloric
Sulfadoxine and Trimethoprim Injection should be protected acid.
from light. SYSTEM SUIT ABILITY

LABELLING The chromatogram obtained with solution (2) shows similar


The label states (1) the amount of Sulfadoxine and of resolution to the reference chromatogram supplied with
Trimethoprim in a suitable dose-volume; (2) where tylosin BPCRS. If necessary adjust the molarity of the sodium
applicable, that the injection contains Lidocaine perchlorate or raise the temperature of the column to a
H ydrochloride. maximum of 50°. The order of elution of the six major
components of tylosin BPCRS in the chromatogram obtained
with solution (2) is: desmycinosyltylosin, tylosin C, tylosin B,
tylosin D, an aldol impurity and tylosin A.
The colu111n efficiency, determined using the peak due to
Tylosin Injection tylosin A in the chromatogram obtained with solution (2),
should be at least 22,000 theoretical plates per metre.
Action and use
Macrolide antibacterial. LIMITS
Calculate the percentage content of components by
DEFINITION normalisation. In the chromatogram obtained with solution
Tylosin Injection is a sterile solution of Tylosin in a mixture (1) :
of equal parts by volume of Propylene Glycol and Water for the content of tylosin A is not less than 80%;
Injections.
the total content of tylosins A, B, C and D is not les s than
The injection complies 'luith the requirements stated under 90%.
Parenteral Preparations and with the following requirements.
Tyramine
CHARACTERISTICS Dilute a volume containing 0.1 g of Tylosin with 5 mL of
A pale yellow to amber-coloured solution. 0.03M orthophosphoric acid in a 25 mL graduated flask, add
IDENTIFICATION 1 mL of pyridine and 2 mL of a saturated solution of
ninhydrin (approximately 4% w/v). Close the fiask by
A. Dilute a volume containing 0.1 g of Tylosin with water to
give a solution containing 0.02% w/v of Tylosin. To 5 mL of covering with a pie ce of aluminium foil and heat in a water
this solution add 10 mL of O.lM sodium hydroxide and extract bath at 85° for at least 20 minutes. Cool rapidly and add
with 10 mL of chlo1'Oform. Separate the chloroform layer and sufficient water to produce 25 mL. Measure the absorbance of
extract it with 25 mL of 0.1 M hyd1'Ochloric acid. Discard the the resulting solution without delay at 570 nm,
Appendix Ir B, using in the reference cell a solution prepared
chloroform, wash the aqueous layer with 3 mL of chlo1'Oform,
in the same manner but omitting the injection being
discard the washings and filter. The light abs01ption of the
filtrate, Appendix Ir B, in the range 230 to 350 nm, exhibits examined. The absorbance is not greater than that obtained
a maximum only at 290 nm. The absorbance at the maximum by carrying out the procedure at the same time using 5 mL
is about 0.94. of a solution in 0.03M orthophosphonc acid containing 35 ~lg of
tyramine per mL and beginning at the words 'add 1 mL ... '
B. To 10 mL of the filtra te obtained in test A add 1 mL of (0.175%).
2M sodium hyd1'Oxide, heat on a water bath for 20 minutes and
cool. The light abs01ption, Appendix Ir B, in the range 250 to ASSAY
430 llffi, exhibits a maximum at 332 nm. Carry out the mic1'Obiological assay of antibiotics,
Appendix XN A. The precision of the assay is such that the
TESTS
fiduciallimits of error are not less than 95% and not more
Composition than 105% of the estimated potency.
Carry out the method for liquid ch1'Omatography,
Appendix lIT D, using the following freshly prepared Calculate the content of tylosin in the injection taking each
solutions. 1000 IV found to be equivalent to 1 mg of tylosin.
The upper fiduciallimit of error is not less than 97.0% and
(1) Dilute the injection with sufficient of a mixture of equal the lower fiduciallimit of error is not more than 110.0% of
volumes of water and acetonitrile to produce a solution the stated contento
. containing 0.02% w/v of Tylosin.
(2) 0.02% w/v of tylosin BPCRS in a mixture of equal STORAGE
volumes of water and acetonitrile. Tylosin Injection should be kept in a cool place.

CHROMATOGRAPHlC CONDITIONS
(a) Vse a stainless steel column (20 cm x 5 mm) packed
with octadecylsilyl silica gel for ch1'Omatography (5 ~lm)
(Nucleosil C18 is suitable).
(b) V se isocratic elution and the mobile phase described
below.
Monographs

Itntnunological Products
2016 Veterinary ImlTIUnOSera Vet-189

VETERINARV IMMUNOSERA *** relevant for the donor animals. It may be necessary to test
*** *** the animals in quarantine for freedom from additional agents,
(Immunosera for Veterina1Y Use, *** depending on their known breeding and rearing history 01'
Ph Eur monograph 0030) any lack of information on their source.
Veterinary Irnrnunosera comply with the requirements of the Any routine or therapeutic medicinal u'eatment administered
European Pharmacopoeia monograph for Irnrnunosera for to the animals in quarantine 01' thereafter must be recorded.
Veterinary Use. These requirements are reproduced below. IMMUNISING ANTIGEN
The provisions of this monograph apply to the following The principIes described in the Production section of
irnrnunosera. Vaccines for veterina1Y use (0062) are applied to the
Antitoxic sera production of the immunogen. The antigen used is identified
Clostridium Novyi Alpha Antitoxin* and characterised. The starting materials used for antigen
Clostridium Perfringens Antitoxins preparation must be controlled to minimise the risk of
contamination with extraneous agents. The antigen may be
(incorporating Clostridium Perfringens Beta Antitoxin*
blended with a suitable adjuvant. The immunogen is
and Clostridium Perfringens Epsilon Antitoxin*) produced on a batch basis. The batches must be prepared
Clostridium Tetani Antitoxin* and tested in such a manner that assures that each batch will
be equally safe and free from extraneous agents and will
*Monograph of the European Pharmacopoeia produce a satisfactory, consistent irnrnune response.
PhEif _____________________________________________
IMMUNISAnON
DEFINITION The donor animal s are irnrnunised according to a defined
Irnrnunosera for veterinary use are preparations containing schedule. For each animal, the details of the dose of
irnrnunoglobulins, purified irnrnunoglobulins or irnrnunising antigen, route of administration and dates of
irnrnunoglobulin fragments obtained from serum or plasma administration are recorded. Animals are kept under general
of irnrnunised animals. They may be preparations of crude health surveillance and the development of specific antibodies
polyc1onal antisera 01' purified preparations. are monitored at appropriate stages of the irnrnunisation
The immunoglobulins 01' irnrnunoglobulin fragments have process.
the power of specifically neutralising the antigen used for COLLECTION OF BLOOD OR PLASMA
irnrnunisation. The antigens inc1ude microbial 01' other Animals are thoroughly examined before each collection.
toxins, bacterial and viral antigens, venoms of snakes and Qnly healthy animal s may be used as a donor animal.
hormones. The preparation is intended for parenteral Collection of blood is made by venepuncture or
administration to provide passive irnrnunity. plasmapheresis. The puncture area is shaved, c1eaned and
PRODUCTION disinfected. The method of collection and the volume to be
GENERAL PROVISIONS collected on each occasion are specified. The blood 01'
plasma is collected in such a manner as to maintain sterility
Irnrnunosera are obtained from the serum 01' plasma of
of the producto If the serum or plasma is stored before
healthy animals irnrnunised by administration of one 01' more
further processing, precautions are taken to avoid microbial
suitable antigens.
contamination.
The production method shall have been shown to yield
The blood 01' plasma collection is conducted at a site
consistently batches of irnrnunosera of acceptable safety
separate from the area where the animals are kept 01' bred
(5.2.6) and efficacy (5.2.7).
and the area where the immunoserum is further processed.
DONOR ANIMALS Clear criteria are established for determining the time
The animal s used are exc1usively reserved for production of between immunisation and first collection of blood or plasma
irnrnunoserum. They are maintained under conditions as well as the time between subsequent collections and the
protecting them from the introduction of disease, as far as 1ength of time over which collections are made. The criteria
possible. The donor animals, and any animals in contact with applied must take into account the effect of the collections
them, are tested and shown to be free from a defined list of on the health and welfare of the animal as well as the effect
infectious agents and re-tested at suitable intervals. The list on the consistency of production of batches of the finished
of agents for testing inc1udes not only those agents that are product, over time.
relevant to the donor animal, but also those that are relevant
The rate of c1earance of any residues that may arise from the
to the recipient target species for the producto Where the
immunising antigen or medication given needs to be taken
donor animals have not been demonstrated to be free from a
into account. In the case of the risk of residues from
relevant pathogen, a justification must be provided and a
che mi cal substances, consideration could be given to the
validated inactivation or purification procedure must be
inc1usion of a withdrawal period for the finished producto
inc1uded in the manufacturing procedure. The feed originates
If the immunising agent consists of a live organism, the time
from a controlled source. Where the donor animal s are
between immunisation and collection may need to take into
chickens, use chickens from a fiock free from specified
account the time required for the donor to eliminate the
pathogens (5. 2. 2). Where applicable for the species used,
immunogen, particularly if any residual live organisms might
measures are taken to avoid contamination with agents of
be harrnful to the recipient.
transmissible spongiform encephalopathies.
As far as possible, animals being introduced into the herd are PREPARATION OF THE FINISHED PRODUCT
from a known source and have a known breeding and rearing Several single plasma 01' serUlTI collections from one 01' more
history. The introduction of animals into the herd follows animals may be pooled to form a bulk for preparation of a
specified procedures, inc1uding defined quarantine measures. batch. The number of collections that may be used to
During the quarantine period the animals are observed and produce a bulk and the size of the bulk are defined. Where
tested to establish that they are free from the list of agents pooling is not undertaken, the production procedure must be
Vet-190 Veterinary Immunosera 2016

very carefully controlled to ensure that the consistency of the agents can be omitted on that product with the agreement of
product is satisfactory. the competent authority. If a product is treated by a
The active substance is subjected to a purification anci/or validated procedure for inactivation of mycoplasmas, the test
inactivation procedure unless omission of such a step has for mycoplasmas can be omitted on that product with the
been justifed and agreed with the competent authority. agreement of the competent authority.
The procedure applied must have been validated and be Only a batch that complies with each of the relevant
shown not to adversely impair the biological activity of the requirements given below under Identification, Tests and
producto The validation studies must address the ability of Potency and/or in the relevant specific monograph may be
the procedure to inactivate or remove any potential released for use. With the agreement of the competent
contaminants such as pathogens that could be transmitted authority, certain tests may be omitted where in-process tests
from the donor to the recipient target species and infectious give an equal or better guarantee that the batch would
agents such as those that cause ubiquitous infections in the comply or where alternative tests validated with respect to
donor animals and cannot be readily eliminated from these the Pharmacopoeia method have been carried out.
donor animals. Certain tests, e.g. for antimicrobial preservatives, for foreign
For purified irnmunosera, the globulins containing the proteins and for albumin, may be carried out by the
immune substances may be obtained from the crude manufacturer on the final bulk rather than on the batch,
irnmunoserum by enzyme treatment and fractional batches or sub-batches of finished product prepared from it.
precipitation or by other suitable chemical or physical In sorne circumstances, e.g. when collections are made into
methods plasmapheresis bags and each one is, essentially, a batch,
Antimicrobial preservatives pools of samples may be tested, with the agreement of the
Antimicrobial preservatives are used to prevent spoilage or competent authority.
adverse effects caused by microbial contamination occurring It is recognised that, in accordance with General Notices
during use of a producto Antimicrobial preservatives are not (section 1.1. General statements), for an established
included in freeze-dried products but, if justified, taking into antiserum the routine application of the safety test will be
account the maximum recommended period of use after waived by the competent authority in the interests of animal
reconstitution, they may be included in the diluent for welfare when a sufficient number of consecutive batches have
multidose freeze-dried products. For single-dose liquid been produced and found to comply with this test, thus
preparations, inclusion of antimicrobial preservatives is not demonstrating consistency of the manufacturing process.
normally acceptable, but may be acceptable, for example Significant changes to the manufacturing process may require
where the same product is filled in single-dose and multidose resumption of routine testing to re-establish consistency.
containers and is for use in non-food producing species. The number of consecutive batches to be tested depends on
For multidose liquid preparations, the need for effective a number of factors such as the type of antiserum, the
antimicrobial preservation is evaluated taking into account frequency of production of batches, and experience with the
likely contamination during use and the maximum irnmunoserum during developmental safety testing and
recommended period of use after broaching of the container. during application of the batch safety test. Without prejudice
During development studies the effectiveness of the to the decision of the competent authority in the light of
antimicrobial preservative throughout the period of validity information available for a given antiserum, testing of
shall be demonstrated to the satisfaction of the competent 10 consecutive batches is likely to be sufficient for the
authority. majority of products. For products with an inherent safety
risk, it may be necessary to continue to conduct the safety
The efficacy of the antimicrobial preservative is evaluated as
test on each batch.
described in chapter 5.1.3; for a multidose preparation,
additional samples are taken, to monitor the effect of the Animal tests In accordance with the provisions of the
antimicrobial preservative over the proposed in-use shelf-life. European Convention for the Protection of Vertebrate
If neither the A criteria nor the B criteria can be met, then in Animals U sed for Experimental and Other Scientific
justified cases the following criteria are applied to antisera for Purposes, tests must be carried out in such a way as to use
veterinary use: bacteria, no increase at 24 h and 7 days, 3 the minimum number of animals and to cause the least pain,
10gIO reduction at 14 days, no increase at 28 days; fungi, no suffering, distress or lasting harm. The criteria for judging
increase at 14 days and 28 days. tests in monographs must be applied in the light of this.
For example, if it is indicated that an animal is considered to
Addition of antibiotics as antimicrobial preservative is not
show positive, infected etc. when typical clinical signs occur
acceptable.
then as soon as sufficient indication of a positive result is
Unless otherwise prescribed in the monograph, the final bulk obtained the animal in question shall be either euthanised or
is distributed aseptically into sterile, tamper-proof containers given suitable treatment to prevent unnecessary suffering.
which are then closed so as to exclude contamination. In accordance with the General Notices, alternative test
The preparation may be freeze-dried. methods may be used to demonstrate compliance with the
In-process tests Suitable tests are carried out in-process, such monograph and the use of such tests is particularly
as on samples from collections before pooling to form a bulk. encouraged when this leads to replacement or reduction of
animal use or reduction of suffering.
BATCH TESTS
The tests that are necessary to demonstrate the suitability of pH (2.2.3)
a batch of a product will vary and are infiuenced by a The pH of crude and purified irnmunosera is shown to be
number of factors, including the detailed method of within the limits set for the products.
production. The tests to be conducted by the manufacturer F ormaldehyde
on a particular product are agreed with the competent If formaldehyde is used for production of immunoserum, a
authority. If a product is treated by a validated procedure for test for free formaldehyde is carried out as prescribed under
inactivation of extraneous agents, the test for extraneous Tests.
2016 Veterinary Immunosera Vet-191

Other inactivating agents electrophoretically, purified irnmunosera show not more than
When other inactivation methods are used, appropriate tests a trace of albumin, and the content of albumin is in any case
are carried out to demonstrate that the inactivating agent has not greater than 30 giL of the reconstituted preparation,
been removed or reduced to an acceptable residual level. where applicable.
Batch potency test Total protein
If a specific monograph exists for the product, the test Dilute the preparation to be examined with a 9 gIL solution
described under Potency is not necessarily carried out for of sodium chloride R to obtain a solution containing about
routine testing of batches of antiserum. The type of batch 15 mg of protein in 2 mL. To 2 mL of this solution in a
potency test to be canied out will depend on the claims round-bottomed centrifuge tube add 2 mL of a 75 gIL
being made for the producto Wherever possible, in vitl"O tests solution of sodium molybdate R and 2 mL of a mixture of
must be used. The type of test required may include 1 volume of nitrogen-free sulfuric acid R and 30 volumes of
measurement of antibodies against specific infectious 'water R. Shake, centrifuge for 5 min, discard the supernatant
organisms, determination of the type of antibody and allow the inverted tube to drain on filter paper.
(e.g. neutralising or opsonising). All tests must be validated. Determine the nitrogen in the residue by the method of
The criteria for acceptance must be set with reference to a sulfuric acid digestion (2.5.9) and calculate the content of
batch that has been shown to comply with the requirements protein by multiplying by 6.25. The results obtained are not
specified under Potency if a specific monograph exists for the greater than the upper limit stated on the label.
product, and which has been shown to have satisfactory Antitnicrobial preservative
efficacy, in accordance with the claims being made for the Determine the amount of antimicrobial preservative by a
product. suitable physicochemical method. The amount is not less
Total irnmunoglobulins than the minimum amount shown to be effective and is not
A test for the quantities of total irnmunoglobulins and/or greater than 115 per cent of that stated on the label.
total gammaglobulins and/or specific irnmunoglobulin classes Formaldehyde (2.4.18)
is carried out. The results obtained must be within the limits Where formaldehyde has been used in the preparation, the
set for the product and agreed with the competent authority. concentration of free formaldehyde is not greater than
The batch contains not more than the level shown to be safe 0.5 gIL, unless a higher amount has been shown to be safe.
in the safety studies and, unless the batch potency test
Sterility (2.6.1)
specifically covers all appropriate irnmunoglobulins, the level
Irnmunosera for veterinary use comply with the test for
in the batch is not less than that in the batch or batches
sterility. When the volume of liquid in a container is greater
shown to be effective in the efficacy studies.
than 100 mL, the method of membrane filtration is used
Total protein wherever possible. If this method is used, incubate the media
For products where claims are being made which relate to for not less than 14 days. Where the method of membrane
the protein content, as well as demonstrating that the batch filtration cannot be employed, the method of direct
contains not more than the stated upper limit, the batch shall inoculation may be used. Where the volume of liquid in each
be shown to contain not less than that in the batch or container is at least 20 mL, the minimum volume to be used
batches shown to be effective in the efficacy studies. for each culture medium is 10 per cent of the contents of the
Extraneous agents container or 5 mL, whichever is the least. The appropriate
In addition to the test described under Tests, specific tests number of items to be tested (2.6.1) is 1 per cent of the
may be required depending on the nature of the preparation, batch with a minimum of 4 and a maximum of 10.
its risk of contamination and the use of the product. Mycoplasmas (2.6.7)
In particular, specific tests for important potential pathogens Irnmunosera for veterinary use comply with the test for
may be required when the donor and recipient species are mycoplasmas.
the same and when these agents would not be detected
Safety
reliably by the general screening test described under Tests.
A test is conducted in one of the species for which the
Water product is recommended. Unless an overdose is specifically
Where applicable, the freeze-drying process is checked by a . contraindicated on the label, twice the maximum
determination of water and shown to be within the limits set recommended dose for the species used is administered by a
for the producto recommended route. If there is a warning against
IDENTIFICATION administration of an overdose, a single dos e is administered.
The identity of the product is established by immunological For products to be used in mammals, use 2 animals of the
tests and, where necessary, by determination of biological minimum age for which the product is recommended.
activity. The potency test may also serve for identification. F or avian products, use not fewer than 10 birds of the
minimum age recommended. The birds are observed for
TESTS 21 days. The other species are observed for 14 days.
The following requirements refer to liquid immunosera and No abnormallocal or systernic reaction occurS.
reconstituted freeze-dried im111unosera.
Extraneous agents
F oreign proteins A test for extraneous agents is conducted by inoculation of
When examined by precipitation tests with specific antisera cell cultures sensitive to pathogens of the species of the donor
against plasma proteins of a suitable range of species, only animal. and into cells sensitive to pathogens of each of the
protein from the declared animal species is shown to be recipient target species stated on the label (2.6.25). Observe
presento the cells for 14 days. During-this time, carry out at least one
Albumin passage. The cells are checked daily for cytopathic effect and
Purified irnmunosera comply with a test for albumino Unless are checked at the end of 14 days for the presence of a
otherwise prescribed in the monograph, when examined
Vet-192 Veterinary Immunosera 2016

haemadsorbing agent. The batch complies with the test if administered at the minimum recornmended dose and
there is no evidence of the presence of an extraneous agent. according to the recomrnended schedule(s), provides a
For immunosera of avian origin, if a test in cel1 culture is response 01' responses consistent with the claims made for the
insufficient to detect potential extraneous agents, a test is producto
conducted by inoculation of embryonated eggs from fiocks Batch potency test
free from specified pathogens (5.2.2) 01' by sorne other The test described under Potency is not necessarily carried
suitable method (polymerase chain reaction (PCR) for out for routine testing of batches of antitoxin. It is carried
example). out on 1 01' more occasions as decided by 01' with the
POTENCY agreement of the competent authority. Where the test is not
Carry out a suitable test for potency. canied out, a suitable validated alternative test is camed out,
the criteria for acceptance being set with reference to a batch
Where a specific monograph exists, carry out the biological
of antitoxin that has given satisfactory results in the test
assay prescribed in the monograph and express the result in
described under Potency and that has been shown to be
International Units per millilitre when such exist. satisfactory with respect to irnmunogenicity in the target
STORAGE species. The fol1owing test may be used after a satisfactory
Protected from light, at a temperature of 5 ± 3 oC. Liquid conelation with the test described under Potency has been
irnmunosera must not be allowed to freeze. established.
LABELLING Determine the level of antibodies against C. novyi alpha toxin
The label states: in the batch of antitoxin using a suitable method such as an
- that the preparation is for veterinary use; irnmunochernical method (2.7.1) 01' neutralisation in cel1
- whether 01' not the preparation is purified; cultures. Use a homologous reference serum calibrated in
- the minimum number of International Units per mil1ilitre, International Units of Clostridium novyi alpha antitoxin.
where such exist; The International Unit is the specific neutralising activity for
- the volume of the preparation in the container; C. novyi alpha toxin contained in a stated amount of the
- the indications for the product; International Standard, which consists of a quantity of dried
- the insttuctions for use inc1uding the interval between any irnmune horse serum. The equivalence in International Units
repeat administrations and the maximum number of of the International Standard is stated by the World Health
administrations that is recommended; Organization.
- the recipient target species for the irnmunoserum; The potency of the finished product is expressed in
- the dose recommended for different species; International Units per millilitre and is shown to be not less
- the route(s) of administration; than the minirnum number stated on the label.
- the name of the species of the donor animal;
IDENTIFICATION
- the maximum quantity of total protein;
The antitoxin is shown, by a suitable irnmunochemical
- the name and amount of any antimicrobial preservative or
method (2.7.1), to react specifical1y with the alpha toxin
any other excipient;
formed by C. novyi.
- any contra-indications to the use of the product including
any required warning on the dangers of administration of POTENCY
an overdose; The potency of Clostridium novyi alpha antitoxin is
- for freeze-dried irnmunosera: determined by comparing the dos e necessary to protect mice
- the name 01' composition and the volume of the or other suitable animals against the toxic effects of a fixed
reconstituting liquid to be added; dose of C. novyi alpha toxin with the quantity of a reference
- the period within which the immunoserum is to be preparation of Clostridium novyi alpha antitoxin, calibrated
used after reconstitution. in International Units, necessary to give the same protection.
_ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ PhEur For this comparison, a suitable preparation of C. novyi alpha
toxin for use as a test toxin is required. The dose of the test
toxin is determined in relation to the reference preparation;
the potency of the antitoxin to be examined is determined in
relation to the reference preparation using the test toxin.
Clostridium Novyi Alpha Antitoxin ***** Preparation of test toxin
* * Prepare the test toxin from a sterile filtrate of an
(Clostridium Novyi Alpha Amitoxin ¡or Veterina1Y ***** approximately 5-day culture in liquid medium of C. novyi
Use J Ph Eur monograph 0339)
type B and dry by a suitable method. Select the test toxin by
PhE~ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ ____
determining for mice the L+/10 dose and the LD so , the· -
DEFINITION observation period being 72 h. A suitable alpha toxin
Clostridium novyi alpha antitoxin for veterinary use is a contains not less than one L+110 dose in 0.05 mg and not
preparation containing the globulins that have the power of less than 10 LD so in each L+/10 dose.
specifical1y neutralising the alpha toxin formed by Determination of test dose of toxin
Clostridiu111 novyi. It consists of the serum 01' a preparation Prepare a solution of the reference preparation in a suitable
obtained from the serum of animals irnmunised against liquid so that it contains 1 IU/rnL. Prepare a solution of the
C. novyi alpha toxin. test toxin in a suitable liquid so that 1 mL contains a
PRODUCTION precisely known amount such as 1 mg. Prepare mixtures of
CHOICE OF COMPOSITION the solution of the reference preparation and the solution of
the test toxin such that each mixture contains 1.0 mL of the
The antitoxin is shown to be satisfactory with respect to
solution of the reference preparation (1 IU), one of a series
safety (5.2.6) and efficacy (5.2.7). For the latter, it shall be
of graded volumes of the solution of the test toxin and
demonstrated, for each target species, that the product, when
2016 Veterinary Immunosera Vet-193

sufficient of a suitable liquid to bring the total volume to -- 93 per cent and 108 per cent when 6 animals per dose
2.0 mL. Allow the mixtures to stand at room temperature for are used.
60 mino Vsing not fewer than 2 mice, each weighing 17-22 g, The potency of the finished product is expressed in
for each mixture, inject adose of 0.2 mL intramuscularly or Intemational Vnits per millilitre and is shown to be not less
subcutaneously into each mouse. Observe the mice for 72 h. than the minimum number stated on the label.
If all the mice die, the amount of toxin present in 0.2 mL of _____________________________________________ PhEw
the mixture is in excess of the test dose. If none of the mice
die, the amount of toxin present in 0.2 mL of the mixture is
less than the test dose. Prepare similar fresh mixtures such
that 2.0 mL of each mixture contains 1.0 mL of the solution
of the reference preparation (1 IV) and 1 of a series of
graded volumes of the solution of the test toxin separated
Clostridium Perfringens Antitoxins
from each other by steps of not more than 20 per cent and DEFINITION
covering the expected end-point. Allow the mixtures to stand Clostridium Perfringens Antitoxins are preparations
at room temperature for 60 mino V sing not fewer than containing either the individual antitoxic globulins or a
2 mice for each mixture, inject adose of 0.2 mL combination of the antitoxic globulins that have the specific
intramuscularly or subcutaneously into each mouse. Observe power of neutralising either the beta toxin or the beta and
the mice for 72 h. Repeat the determination at least once and epsilon toxins produced by Clostridium perfringens type B, the
combine the results of the separate tests that have been beta tOXÍ11 produced by Cl. pel'fringens type e or the epsilon
carried out with mixtures of the same composition so that a toxin produced by el. perfringens type D.
series of totals is obtained, each total representing the The name Clostridium Perfringens Beta Antitoxin may be
mortality due to a mixture of a given co:¡:nposition. The test used for preparations stated to contain beta antitoxins only.
dos e of toxin is the amount present in 0.2 mL of that The names Clostridium Perfringens Epsilon Antitoxin or
mixture which causes the death of one half of the total Clostridium Perfringens Type D Antitoxin may be used for
number of mice injected with it. preparations stated to contain epsilon antitoxins only.
Determination of the potency of the antitoxin to be The name Clostridium Perfringens Type B Antitoxin
examined (synonym Lamb Dysentery Antiserurn) may be used for
PreliminaJY test Dissolve a quantity of the test toxin in a preparations stated to contain both beta and epsilon
suitable liquid so that 1 mL contains 10 times the test dose antitoxin.
(solution of the test toxil1). Prepare mixtures of the solution
The antitoxins c0111ply zuith the l'equirements stated under
of the test toxin and of the antitoxin to be examined such
VeterinaJY Immunosera 'luith the modifications belozu and zuith the
that each mixture contains 1.0 mL of the solution of the test
requirements of one 01' both of the follozuing tzuo monographs
toxin, one of a series of graded volumes of the antitoxin to be
accol'ding to the composition of the antitoxin as stated 011 the label.
examined and sufficient of a suitable liquid to bring the final
volume to 2.0 mL. Allow the mixtures to stand at room LABELLING
temperature for 60 mino V sing not fewer than 2 mice for The label states (1) whether the preparation contains beta or
each mixture, inject adose of 0.2 mL intramuscularly or epsilon antitoxin or both; (2) the type or types of el.
subcutaneously into each mouse. Observe the mice for 72 h. perfringens against which the antitoxin will provide protection.
If none of the mice die, 0.2 mL of the mixture contains more
than 0.1 IV. If all the mice die, 0.2 mL of the mixture
contains less than 0.1 IV.
Final test Prepare mixtures of the solution of the test toxin
Clostridium Perfringens Beta ***
and of the antitoxin to be examined such that 2.0 mL of *** ***
each mixture contains 1.0 mL of the solution of the test Antitoxin ***
toxin and one of a series of graded volunles of the antitoxin (Clostridium Perfringens Beta Antitoxin for VeterinaJY
to be examined, separated from each other by steps of not Use) Ph Bur 7110nograph 0340)
more than 20 per cent and covering the expected end-point Ph Eur ___________________________________________
as determined by the preliminary test. Prepare further
mixtures such that 2.0 mL of each mixture contains 1.0 mL DEFINITION
of the solution of the test toxin and one of a series of graded Clostridium perfringens beta antitoxin for veterinary use is a
volumes of the solution of the reference preparation, in order preparation containing principally the globulins that have the
to confirm the test dos e of the toxin. Allow the mixtures to power of specifically neutralising the beta toxin formed by
stand at room temperature for 60 mino Vsing not fewer than elostridium perfringens (types B and C). It consists of the
2 mice for each mixture, proceed as described in the serum or a preparation obtained from the serum of animal s
preliminary test. The test mixture which contains 0.1 IV in immunised against C. peifringens beta toxin.
0.2 mL is that mixture which kills the same or almost the PRODUCTION
same number of mice as the reference mixture containing
CHOICE OF COMPOSITION
0.1 IV in 0.2 mL. Repeat the determination at least once
The antitoxin is shown to be satisfactory with respect to
and calculate the average of all valid estimates. Estimates are
safety (5.2.6) and efficacy (5.2.7). For the latter, it shall be
valid only if the reference preparation gives a result within
demonstrated, for each target species, that the product, when
20 per cent of the expected value.
admmistered at the minimum recommended dose and
The confidence limits (P = 0.95) have been estimated to be: according to the recommended schedule(s), provides a
85 per cent and 114 per cent when 2 animals per dose response or responses consistent with the claims made for the
are used; producto
91.5 per cent and 109 per cent when 4 animals per dose
are used;
Vet-194 Veterinary Immunosera 2016

Batch potency test for each mixture, inject adose of 0.5 mL intravenously 01'
The test described under Potency is not necessarily carried intraperitoneally into each mouse. Observe the mice for 72 h.
out for routine testing of batches of antitoxin. It is cmried If all the mice die, the amount of toxin present in 0.5 mL of
out on 1 01' more occasions as decided by 01' with the the mixture is in excess of the test dose. If none of the mice
agreement of the competent authority. Where the test is not die, the amount of toxin present in 0.5 mL of the mixture is
carried out, a suitable validated altemative test is camed out, less than the test dose. Prepare similar fresh mixtures such
the criteria for acceptance being set with reference to a batch that 5.0 mL of each mixture contains 2.0 mL of the solution
of antitoxin that has given satisfactory results in the test of the reference preparation (10 IV) and 1 of a series of
described under Potency and that has been shown to be graded volumes of the solution of the test toxin separated
satisfactory with respect to immunogenicity in the target from each other by steps of not more than 20 per cent and
species. The following test may be used after a satisfactory covering the expected end-point. Allow the mixtures to stand
correlation with the test described under Potency has been at room temperature for 30 mino U sing not fewer than
established. 2 mice for each mixture, inject adose of 0.5 mL
Determine the level of antibodies against C. perfringens beta intravenously 01' intraperitoneally into each mouse. Observe
toxin in the batch of antitoxin using a suitable method such the mice for 72 h. Repeat the detelmination at least once and
as an immunochemical method (2.7.1) or neutralisation in combine the results of the separate tests that have been
cell cultures. Use a homologous reference serum calibrated in carried out with mixtures of the same composition so that a
Intemational Units of Clostridium perfringens beta antitoxin. series of totals is obtained, each total representing the
mortality due to a mixture of a given composition. The test
The Intemational Unit is the specific neutralising activity for
dose of toxin is the amount present in 0.5 mL of that
C. perfrúzgens beta toxin contained in a stated amount of the
mixture wmch causes the death of one half of the total
Intemational Standard, wmch consists of a quantity of dried
number of mice injected with it.
immune horse serum. The equivalence in Intemational Units
of the Intemational Standard is stated by the World Health Determination of the potency of the antitoxin to be
Organization. examined
The potency of the finished product is expressed in Preli11linmy test Dissolve a quantity of the test toxin in a
Intemational Units per millilitre and is shown to be not les s suitable liquid so that 2.0 mL contains 10 times the test dose
than the minimum number stated on the label. (solution of the test toxin). Prepare mixtures of the solution
of the test toxin and of the antitoxin to be examined such
IDENTIFICATION that each mixture contains 2.0 mL of the solution of the test
The antitoxin is shown, by a suitable immunochemical toxin, one of a series of graded volumes of the antitoxin to be
method (2.7.1), to react specifically with the beta toxin examined and sufficent of a suitable liquid to bring the final
formed by C. perfringens. volume to 5.0 mL. Allow the mixtures to stand at room
POTENCY temperature for 30 mino Using not fewer than 2 mice for
The potency of Clostridium perfringens beta antitoxin is each mixture, inject adose of 0.5 mL intravenously 01'
determined by comparing the dose necessmy to protect mice intraperitoneally into each mouse. Observe the mice for 72 h.
or other suitable animals against the toxic effects of a fixed If none of the mice die, 0.5 mL of the mixture contains more
dose of C. perfringens beta toxin with the quantity of a than 1 IV. If aH the mice die, 0.5 mL of the mixture
reference preparation of Clostridium perfringens beta contains les s than 1 IU.
antitoxin, calibrated in Intemational Units, necessary to give Final test Prepare mixtures of the solution of the test toxin
the same protection. For this comparison, a suitable and of the antitoxin to be examined such that 5.0 mL of
preparation of C. perfringells beta toxin for use as a test toxin each mixture contains 2.0 mL of the solution of the test
is required. The dos e of the test toxin is determined in toxin and one of a series of graded volumes of the antitoxin
relation to the reference preparation; the potency of the to be examined, separated from each other by steps of not
antitoxin to be examined is determined in relation to the more than 20 per cent and covering the expected end-point
reference preparation using the test toxin. as determined by the preliminary test. Prepare further
Preparation of test toxin mixtures such that 5.0 mL of each mixture contains 2.0 mL
Prepare the test toxin from a sterile filtra te of an early culture of the solution of the test toxin and one of a series of graded
in liquid medium of C. perfringens type B 01' type C and dry volumes of the solution of the reference preparation, in order
by a suitable method. Select the test toxin by determining for to confirm the test dos e of the toxin. AHow the mixtures to
mice the L+ dose and the LD so , the observation period stand at room temperature for 30 mino Using not fewer than
being 72 h. A suitable beta toxin contains not less than one 2 mice for each mixture, proceedas described in the
L+ dose in 0.2 mg and not less than 25 LD so in each preliminary test. The test mixture wmch contains 1 IU in
L+ dose. 0.5 mL is that mixture which kills the same 01' almost the
same number of mice as the reference mixture containing,
Determination of test dose of toxin 1 IV in 0.,5 mL. Repeat the determination at least once and
Prepare a solution of the reference preparation in a suitable calculate the average of aH valid estimates. Estimates are valid
liquid so that it contains 5 IU/mL. Prepare a solution of the only if the reference preparation gives a result within
test toxin in a suitable liquid so that 1 mL contains a 20 per cent of the expected value.
precisely known amount such as 10 mg. Prepare mixtures of
The confidence limits (P = 0.95) have been estimated to be:
the solution of the reference preparation and the solution of
85 per cent and 114 per cent when 2 animals per dose
the test toxin such that each mixture contains 2.0 mL of the
are used;
solution of the reference preparation (10 IV), one of a series
91. 5 per cent and 109 per cent when 4 animals per dose
of graded volumes of the solution of the test toxin and
are used; -
sufficient of a suitable liquid to bring the total volume to
- 93 per cent and 108 per cent when 6 animals per dose
5.0 mL. Allow the mixtures to stand at room temperature for
are used.
30 mino Using not fewer than 2 mice, each weighing 17-22 g,
2016 Veterinary Irnrnunosera Vet-195

The potency of the finished product is expressed in POTENCY


Intemational Units per millilitre and is shown to be not less The potency of Clostridium perfringens epsilon antitoxin is
than the minimum number stated on the label. determined by comparing the dose necessary to protect mice
_____________________________________________ PhEw 01' other suitable animal s against the toxic effects of a fixed
dose of C. pelfiingens epsilon toxin with the quantity of a
reference preparation of Clostridium perfringens epsilon
antitoxin, calibrated in Intemational Units, necessary to give
the same protection. For this comparison, a suitable
Clostridium Perfringens Epsilon *** preparation of C. perfringens epsilon toxin for use as a test
*** *** toxin is required. The dos e of the test toxin is determined in
Antitoxin *** relation to the reference preparation, the potency of the
(Clostridiu111 Pelfringens Bpsilon Antitoxin for antitoxin to be examined is determined in relation to the
Veterina1Y Use, Ph Bur monograph 0341) reference preparation using the test toxin.
PhEw _____________________________________________
Preparation of test toxin
DEFINITION Prepare the test toxin from a sterile filtrate of an early culture
Clostridium perfringens epsilon antitoxin for veterinary use is in liquid medium of C. pe¡fringens type D and dry by a
a preparation containing the globulins that have the power of suitable method. Select the test toxin by determining for
specifically neutralising the epsilon toxin formed by mice the L+/10 dose and the LD so, the observation period
Clostridiu111 peJfringens type D. It consists of the serum or a being 72 h. A suitable epsilon toxin contains not less than
preparation obtained from the serum of animals immunised one L+/10 dose in 0.005 mg and not less than 20 LD so in
against C. pelfringens epsilon toxin. each L+/10 dose.
Determination of test dose of toxin
PRODUCTION
Prepare a solution of the reference preparation in a suitable
CHOICE OF COMPOSITION
liquid so that it contains 0.5 IU/mL. Prepare a solution of
The antitoxin is shown to be satisfactory with respect to the test toxin in a suitable liquid so that 1 mL contains a
safety (5.2.6) and efficacy (5.2.7). For the latter, it shall be precisely known amount such as 1 mg. Prepare mixtures of
demonstrated, for each target species, that the product, when the solution of the reference preparation and the solution of
administered at the minimum recommended dose and the test toxin such that each mixture contains 2.0 mL of the
according to the recommended schedule(s), provides a solution of the reference preparation (1 IV), one of a series
response or responses consistent with the claims made for the of graded volumes of the solution of the test toxin and
producto sufficient of a suitable liquid to bring the total volume to
Batch potency test 5.0 mL. A1low the mixtures to stand at room temperature for
The test described under Potency is not necessarily carried 30 mino Using not fewer than 2 mice, each weighing 17-22 g,
out for routine testing of batches of antitoxin. It is carried for each mixture, inject adose of 0.5 mL intravenously or
out on one 01' more occasions as decided by or wiL~ the intraperitoneally into each mouse. Observe the mice for 72 h.
agreement of the competent authority. Where the test is not If all the mice die, the amount of toxin present in 0.5 mL of
carried out, a suitable validated altemative test is carried out, the mixture is in excess of the test dose. If none of the mice
the criteria for acceptance being set with reference to a batch die, the amount of toxin present in 0.5 mL of the mixture is
of antitoxin that has given satisfactory results in the test less than the test dose. Prepare similar fresh mixtures such
described under Potency and that has been shown to be dlat 5.0 mL of each mixture contains 2.0 mL of dle solution
satisfactory with respect to immunogenicity in the target of the reference preparation (1 IV) and 1 of a series of
species. The following test may be used after a satisfactory graded volumes of the solution of the test toxin, separated
correlation with the test described under Potency has been from each other by steps of not more than 20 per cent and
established. covering the expected end-point. Allow the mixtures to stand
Determine the level of antibodies against C. perfringens at room temperature for 30 mino Using not fewer than
epsilon toxin in the batch of antitoxin using a suitable 2 mice for each mixture, inject adose of 0.5 mL
method such as an immunochemical method (2.7.1) or intravenously 01' intraperitoneally into each mouse. Observe
neutralisation in cell cultures. Use a homologous reference the mice for 72 h. Repeat the determination at least once and
serum calibrated in Intemational Units of combine the results of the separate tests that have been made
Clostridium perfringens epsilon antitoxin. with mixtures of the same composition so that a series of
The Intemational Unit is the specific neutralising activity for total s is obtained, each total representing the mortality due to
C. pel'fringens epsilon toxin contained in a stated amount of a mixture of a given composition. The test dose of the toxin
the Intemational Standard, which consists of a quantity of is the amount present in 0.5 mL of that mixture which
dried immune horseserum. The equivalence in Intemational causes the death of one half of the total number of mice
Units of the Intemational Standard is stated by the World injected with it.
Health Organization. Determination of the potency of the antitoxin to be
The potency of the finished product is expressed in examined
Intemational Units per millilitre and is shown to be not less Prelimina1y test Dissólve a quantity of the test toxin in a
than the minimum number stated on the label. suitable liquid so that 2.0 mL contains 10 times the test dose
(solution of the test toxin). Prepare mixtures of the solution
IDENTIFICATION ofthe 'test toxin and of the antitoxin to be examined such
The antitoxin is shown, by a suitable immunochemical that each mixture contains 2_.0 mL of the solution of the test
method (2.7.1), to react specifically with the epsilon toxin toxin, one of a series of graded volumes of the antitoxin to be
formed by C. perfringens. examined and sufficient of a suitable liquid to bring the final
volume to 5.0 mL. Allow the mixtures to stand at room
Vet-196 Veterinary Irnrnunosera 2016

temperature for 30 mino Using not fewer than 2 mice for Demonstration of neurotoxin neutralisation
each mixture, inject adose of 0.5 mL intravenously or The ability of tetanus antitoxin to neutralise the neurotoxin
intraperitoneally into each mouse. Observe the mice for 72 h. of C. tetani is determined by establishing the dose necessary
If none of the mice die, 0.5 mL of the mixture contains more to protect mice (01' guinea-pigs) against the toxic effects of a
than 0.1 IU. If aH the mice die, 0.5 mL of the mixture fixed dos e of tetanus toxin. The test must be conducted in
contains less than 0.1 IU. parallel with a test of a reference preparation of tetanus
Final test Prepare mixtures of the solution of the test toxin antitoxin, calibrated in International Units, using a quantity
and of the antitoxin to be examined such that 5.0 mL of expected to give the same protection. The ability of the test
each mixture contains 2.0 mL of the solution of the test antitoxin to neutralise the neurotoxin (potency) can then be
toxin and one of a series of graded volumes of the antitoxin expressed in International Units. For this study, a suitable
to be examined, separated from each other by steps of not preparation of tetanus toxin for use as a test toxin is
more than 20 per cent and covering the expected end-point required. The dose of the test toxin is determined in relation
as determined by the preliminary test. Prepare further to the reference preparation; the potency of the antitoxin to
mixtures such that 5.0 mL of each mixture contains 2.0 mL be examined is determined in relation to the reference
of the solution of the test toxin and one of a series of graded preparation using the test toxin.
volumes of the solution of the reference preparation to Preparation of test toxin
confirm the test dose of the toxin. AHow the mixtures to Prepare the test toxin from a sterile filtrate of an 8-10 day
stand at room temperature for 30 mino Using not fewer than culture in liquid medium of C. tetard. A test toxin may be
2 mice for each mixture proceed as described in the prepared by adding this filtra te to glycerol R in the proportion
preliminary test. The test mixture which contains 0.1 IU in of 1 volume of filtrate to 1 to 2 volumes of glycerol R.
0.5 mL is that mixture which kills the same or almost the The solution of test toxin may be stored at or slightly below
same number of mice as the reference mixture containing O oC. The toxin may also be dried by a suitable method.
0.1 IU in 0.5 mL. Repeat the determination at least once Select the test toxin by determining for mice the Lp/10 dos e
and calculate the average of aH valid estimates. Estimates are and the paralytic dose 50 per cent. A suitable toxin contains
valid only if the reference preparation gives a result within not less than 1000 times the paralytic dose 50 per cent in
20 per cent of the expected value. 1 Lpll O dose.
The confidence limits (P = 0.95) have been estimated to be: Lp/10 dose (Limes paralyticum)
- 85 per cent and 114 per cent when 2 animals per dose This is the smallest quantity of toxin wruch when mixed with
are used; 0.1 IU of antitoxin and injected subcutaneously into mice (or
91.5 per cent and 109 per cent when 4 animals per dose guinea-pigs) causes tetanic paralysis in the animals on or
are used; before the 4th day after injection.
93 per cent and 108 per cent when 6 animals per dose
Paralytic dose 50 per cent
are used.
This is the quantity of toxin which when injected
The potency of the finished product is expressed in subcutaneously into mice (or guinea-pigs) causes tetanic
International Units per millilitre and is shown to be not less paralysis in one half of the animals on or before the 4th day
than the minimum number stated on the label. after injection.
_____________________________________________ ~E~

Determination of test dos e of toxin


Reconstitute 01' dilute the reference preparation with a
suitable liquid so that it contains 0.5 IU/rnL. Measure 01'
weigh a quantity of the test toxin and dilute with or dissolve

Clostridium Tetani Antitoxin *** in a suitable liquido Prepare mixtures of the solution of the
*** *** reference preparation and the solution of the test toxin so
that each mixture will contain 0.1 IU of antitoxin in the
Tetanus Antitoxin (Veterinary) *** volume chosen for injection and one of a series of graded
(Tetanus Antitoxin fOl" VeterinalY Use,Ph Eur monograph 0343)
volumes of the solution of the test toxin, separated from each
~E~ ____________________________________________
other by steps of not more than 20 per cent and covering the
DEFINITION expected end-point. Adjust each mixture with a suitable
Tetanus antitoxin for veterinary use is a preparation liquid to the same final volume (0.4 mL to 0.6 mL if mice
containing principally the globulins that have the power of are used for the test or 4.0 mL if guinea-pigs are used).
specifically neutralising the neurotoxin formed by AIlow the mixtures to stand at room temperature for 60 mm.
ClostridiuJ1Z tetani. It consists of the serum or a preparation Using not fewer than 2 animals for each mixture, inject the
obtained from the serum of animals immunised against chosen volume subcutaneously into each animal. Observe the
tetanus toxin. animals for 96 h and make daily records of the degree of
tetanus developing in each group of animals. Repeat the test
PRODUCTION at least once and calculate the test dose as the mean of the
CHOICE OF COMPOSITION different tests. The test dose of the toxin is the amount
The antitoxin is shown to be satisfactory with respect to present in that mixture which causes tetanic paralysis in one
safety (5.2.6) and efficacy (5.2.7). For the latter, it shall be half of the total number of animal s injected with it.
demonstrated, for each target species, that the product, when
Determination of the neutralising ability of the
administered at the minimum recommended dose and
antitoxin to be examined
according to the recommended schedule(s), provides a
Prelimina1y test Measure or weigh a quantity of the test toxin
response 01' responses consistent with the claims made for the
and dilute with or dissolve in a suitablé liquid so that the
producto The ability of the product to neutralise the
solution contains 5 test doses per millilitre (solution of the
neurotoxin formed by C. tetani must also be demonstrated,
test toxin). Prepare mixtures of the solution of the test toxin
e.g. by conducting the test in mice as described below.
and of the antitoxin to be examined so that for each mixture
2016 Veterinary Vaccines Vet-197

the volume chosen for injection contains the test dose of


toxin and one of a series of graded volumes of the antitoxin
Veterinary Vaccines
to be examined. Adjust each mixture to the same final (Vaccines for VeterinalY Use,
volume with a suitable liquido Allow the mixtures to stand at Ph Bur 1110nogmph 0062)
room temperature for 60 min. Using not fewer than Veterinary Vaccines comply with the requirements of the
2 animals for each mixture, inject the chosen volume European Pharmacopoeia monograph for Vaccines for
subcutaneously into each animal. Observe the animal s for Veterinary Use. These requirements are reproduced below.
96 h and make daily records of the degree of tetanus
The provisions of this monograph apply to the following
developing in each group of animals. Using the results, select
vaccines.
suitable mixtures for the final test.
Inactivated Bacterial Vaccines
Final test Prepare mixtures of the solution of the test toxin
and of the antitoxin to be examined so that for each mixture Bovine Leptospirosis Vaccine (Inactivated)*
the volume chosen for the injection contains the test dos e of Canine Leptospirosis Vaccine (Inactivated)*
toxin and one of a series of graded volumes of the antitoxin Clostridium Botulinum Vaccine*
to be examined, separated from each other by steps of not Clostridium Chauvoei Vaccine*
more than 20 per cent and covering the eXpected end-point
as determined in the preliminary test. Prepare further Clostridium Novyi Type B Vaccine*
mixtures with the same amount of test toxin and graded Clostridium Perfringens Vaccines*
volumes of the reference preparation, centred on 0.1 IU in Clostridium Septicum Vaccine*
the volume chosen for injection, to confirm the test dose of Clostridium Tetani Vaccines*
the toxin. Adjust each mixture to the same final volume with Feline Chlamydiosis Vaccine (Inactivated)*
a suitable liquido Allow the mixtures to stand at room
temperature for 60 min. U sing not fewer than 2 animal s for Fowl Cholera Vaccine (Inactivated)*
each mixture, inject the chosen volume subcutaneously into Furunculosis Vaccine for Salmonids, Inactivated*
each animal. Observe the animals for 96 h and make daily Mannheimia Vaccine (Inactivated) for Cattle*
records of the degree of tetanus developing in each group of Mannheimia Vaccine (Inactivated) for Sheep*
animals. The test mixture which contains 0.1 IU in the Mycoplasma Gallisepticum Vaccine (Inactivated)*
volume injected is that mixture which causes tetanic paralysis
in the same, or almost the same, number of animal s as the Pasteurella Vaccine (Inactivated) for Sheep*
reference mixture containing 0.1 IU in the volume injected. Porcine Actinobacillosis Vaccine, Inactivated*
Repeat the determination at least once and calculate the Porcine Enzootic Pneumonia Vaccine (Inactivated)*
mean of all valid estimates. Estimates are valid only if the Porcine E. Coli Vaccine, Inactivated*
reference preparation gives a result within 20 per cent of the Porcine Progressive Atrophic Rhinitis Vaccine, Inactivated*
expected value.
Ruminant E. Coli Vaccine, Inactivated*
The confidence limits (P = 0.95) have been estimated to be:
-- 85 per cent and 114 per cent when 2 animals per dose Salmonella Enteritidis Vaccine (Inactivated) for Chickens*
are used; Salmonella Typhimurium Vaccine (Inactivated) for
-- 91.5 per cent and 109 per cent when 3 animals per dose Chickens*
are used; Swine Erysipelas Vaccine, Inactivated*
-- 93 per cent and 108 per cent when 6 animals per dose Vibriosis Vaccine for Salmonids, Inactivated, Cold-water*
are used.
Vibriosis Vaccine for Salmonids, Inactivated*
IDENTIFICATION Living Bactelial Vaccines
The antitoxin is shown, by a suitable irnmunochemical Anthrax Vaccine, Living*
method (2. 7.1), to react specifically with the neurotoxin
Brucella Melitensis (Strain Rev. 1) Vaccine, Living*
formed by C. tetani. The potency test may also serve for
identification. Coccidiosis Vaccine (Live) for Chickens*
Salmonella Dublin Vaccine, Living
POTENCY
Determine the titre of antibodies against the neurotoxin Inactivated Viral Vaccines
formed by C. tetani using a suitable irnmunochemical method Aujesky's Disease Vaccine, Inactivated*
(2.7.1) such as a toxin-binding-inhibition test (ToBI test) and Avian Infectious Bronchitis Vaccine, Inactivated*
a homologous reference serum, calibrated in International Avian Paramyxovirus 3 Vaccine, Inactivated*
Units per millilitre.
Bovine Viral Diarrhoea Vaccine (Inactivated)*
The Intemational Unit is the specific neutralising activity for
Calf Coronavirus Diarrhoea Vaccine (Inactivated)*
tetanus toxin contained in a stated amount of the
International Standard which consists of a quantity of dried Calf Rotavirus Diarrhoea Vaccine (Inactivated)*
irnmune horse serum. The equivalence in Intemational Units Canine Adenovirus Vaccine, Inactivated*
of the Intemational Standard is stated by the World Health Canine Parvovirus Vaccine, Inactivated *
Organization. Egg-drop Syndrome 76 (Adenovirus) Vaccine*
The potency of the finished product is expressed in Equine Herpesvirus Vaccine, Inactivated*
Intemational Units per millilitre and is shown to be not less
Equine Influenza Vaccine*
than the minimum number stated on the label.
Feline Calicivirus Vaccine, I~activated*
_____________________________________________ PhE~

Feline Infectious Enteritis Vaccine, Inactivated*


Feline Leukaemia Vaccine, Inactivated*
Feline Viral Rhinotracheitis Vaccine, Inactivated*
Vet-198 Veterinary Vaccines 2016

Foot and Mouth Disease (Ruminants) Vaccine* 1 DEFINITION


Infectious Bursal Disease Vaccine, Inactivated* Vaccines for veterinary use are preparations containing
Louping-ill Vaccine antigenic substances and are administered for the purpose of
inducing a specific and active immunity against disease
Newcastle Disease Vaccine, Inactivated*
provoked by bacteria, toxins, virus es, fungi 01' parasites.
Ovine Enzootic Abortion Vaccine The vaccines, live 01' inactivated, confer active immunity that
Porcine Parvovirus Vaccine, Inactivated* may be transferred passively via maternal antibodies against
Rabbit Haemorrhagic Disease Vaccine (Inactivated)* the immunogens they contain and sometimes also against
Rabies Veterinary Vaccine, Inactivated* antigenically related organisms. Vaccines may contain
bacteria, toxins, viruses or fungi, living or inactivated,
Swine Influenza Vaccine, Inactivated*
parasites, 01' antigenic fractions or substances produced by
Living Viral Vaccines these organisms and rendered harmless whilst retaining all 01'
Aujesky's Disease Vaccine, Living* part of their antigenic properties; vaccines may also contain
Avian Infectious Bronchitis Vaccine, Living* combinations of these constituents. The antigens may be
Avian Viral Tenosynovitis Vaccine (Live)* produced by recombinant DNA technology. Suitable
adjuvants may be included to enhance the immunising
Bovine Parainfluenza Virus Vaccine, Living*
properties of the vaccines.
Bovine Respiratory Syncytial Vuus Vaccine, Living*
Terminology used in monographs on vaccines for veterinary
Canine Adenovirus Vaccine, Living * use is defined in chapter 5.2.1.
Canine Distemper Vaccine, Living*
1-1 BACTERIAL VACCINES AND BACTERIAL
Canine Parainfluenza Virus Vaccine (Live)* TOXOIDS
Canine Parvovirus Vaccine, Living* Bacterial vaccines and bacterial toxoids are prepared from
Contagious Pustular Dermatitis Vaccine, Living cultures grown on suitable solid or liquid media, 01' by other
Duck Plague Vaccine (Live)* suitable means; the requirements of this section do not apply
to bacterial vaccines prepared in cell cultures or in live
Duck Viral Hepatitis Type I Vaccine (Live)*
animals. The strain of bacterium used may have been
Feline Calicivirus Vaccine, Living* modified by genetic engineering. The identity, antigenic
Feline Infectious Enteritis Vaccine, Living* potency and purity of each bacterial culture used is carefully
Feline Viral Rhinotracheitis Vaccine, Living* controlled.
Ferret and Mink Distemper Vaccine, Living* Bacterial vaccines contain inactivated or live bacteria or their
Fowl Pox Vaccine, Living* antigenic components; they are liquid preparations of various
degrees of opacity or they may be freeze-dried.
Infectious Avian Encephalomyelitis Vaccine, Living*
Bacterial toxoids are prepared from toxins by diminishing
Infectious Bovine Rhinotracheitis Vaccine, Living*
theu" toxicity ro a very low level 01' by completely eliminating
Infectious Bursal Disease Vaccine, Living* ir by physical 01' chemical means whilst retaining adequate
Infectious Chicken Anaemia Vaccine (Live) * immunising potency. The toxins are obtained from selected
Laryngotracheitis Vaccine, Living* strains of specified micro-organisms grown in suitable media
Marek's Disease Vaccine, Living* or are obtained by other suitable means, for example,
chemical synthesis.
Myxomatosis Vaccine (Live) for Rabbits*
The toxoids may be:
Newcastle Disease and Avian Infectious Bronchitis Vaccine,
-- liquid;
Living
-- precipitated with alum or another suitable agent;
Newcastle Disease Vaccine, Living* -- purified and/or adsorbed on aluminium phosphate,
Rabies Vaccine for Foxes, Living* aluminium hydroxide, ca1cium phosphate or another
Swine-Fever Vaccine (Live, Prepared in Cell Cultures), adsorbent prescribed in the monograph.
Classical* Bacterial toxoids are clear or slight1y opalescent liquids.
Hel711inth Vaccine Adsorbed toxoids are suspensions or emulsions. Certain
Lungworm (Dictyocaulus Viviparus) Oral Vaccine, Living toxoids may be freeze-dried.
Unless otherwise indicated, statements and requirements
*Monograph of the European Pharmacopoeia given below for bacterial vaccines apply equally to bacterial
PhE~ _____________________________________________ vaccines, bacterial toxoids and products containing a
combination of bacterial cells and toxoid.
In the case of combined vaccines, for each component that is
the subject of a monograph in the Pharmacopoeia, the 1-2 VIRAL VACCINES
provisions of that monograph apply to that component, Viral vaccines are prepared by growth in suitable cell cultures
modified where necessary as indicated (se e chapters 5.2.6. (5.2.4), in tissues, in micro-organisms, in fertilised eggs 01',
Evaluation of safety of veterinary vaccines and immunosera where no other possibility is available, in live animals, 01' by
and 5.2.7. Evaluation of efficacy ofveterinary vaccines and other suitable means. The strain of virus used may have been
immunosera). If an immunological product for veterinary use modified by genetic engineering. They are liquid or freeze-
is intended for minor use, certain tests may be excluded, dried preparations of one or more virus es or viral subunits or
subject to approval by the competent authorityl). peptides.
Live viral vaccines are prepared from virus es of attenuated
1 Note: Guidance on data requirements for immunological veterinary
virulence or of naturallow virulence for the target species.
medicinal products intended for minor use 01' minor species/limited
markets (EMNCVMP!IWP/123243/2006, including any subsequent
revision of this documento
2016 Veterinary Vaccines Vet-199

Inactivated viral vaccines are treated by a validated pro ce dure in a seed-Iot system wherever possible. Each master seed lot
for inactivation of the virus and may be purified and is tested as described below. A record of the origin, date of
concentrated. isolation, passage history (including purification and
1-3 VECTOR VACCINES characterisation procedures) and storage conditions is
maintained for each master seed loto Each master seed lot is
Vector vaccines are liquid or freeze-dried preparations of one
assigned a specific code for identification purposes.
or more types of live micro-organisms (bacteria or virus es)
that are non-pathogenic or have low pathogenicity for the 2-1-3-1-2 Propagation. The minimum and maximum number
target species and in which have been inserted one or more of sub cultures of each master seed lot prior to the production
genes encoding antigens that stimulate an irnmune response stage are specified. The methods used for the preparation of
protective against other micro-organisms. seed cultures, preparation of suspensions for seeding,
techniques for inoculation of seeds, titre and concentration of
2 PRODUCTION inocula and the media used, are documented. It shall be
2-1 PREPARATION OF THE VACCINE demonstrated that the characteristics of the seed material (for
The methods of preparation, which vary according to the example, dissociation or antigenicity) are not changed by
type of vaccine, are such as to maintain the identity and these subcultures. The conditions under which each seed lot
immunogenicity of the antigen and to ensure freedom from is stored are documented.
contamination with extraneous agents. 2-1-3-1-3 Identity and purity. Each master seed lot is shown
Substances of animal origin used in the production of to contain only the species and su"ain of bacterium stated.
vaccines for veterinary use comply with the requirements of A brief description of the method of identifying each strain
chapter 5.2.5. Other substances used in the preparation of by biochemical, serological and morphological characteristics
vaccines for veterinary use comply with requirements of the and distinguishing it as far as possible from related strains is
Pharmacopoeia (where a relevant monograph exists) and are recorded, as is also the method of detelmining the purity of
prepared in a manner that avoids contamination of the the strain. If the master seed lot is shown to contain living
vaccme. organisms of any kind other than the species and su"ain
2-1-1 Substrates for production stated, then it is unsuitable for vaccine production.
Cell cultures used in the production of vaccines for veterinary 2-1-3-2 Virus seed lots
use comply with the requirements of chapter 5.2.4. 2-1-3-2-1 General requirements. Vu"uses used in manufacture
Where a monograph refers to chicken fiocks free from are handled in a seed-Iot system. Each master seed lot is
specified pathogens (SPF), these fiocks comply with the tested as described below. A record of the origin, date of
requirements prescribed in chapter 5.2.2. isolation, passage history (including purification and
For production of inactivated vaccines, where vaccine characterisation procedures) and storage conditions is
organisms are grown in poultry embryos, such embryos are maintained for each seed loto Each master seed lot is assigned
derived either from SPF fiocks (5.2.2) or from healthy non- a specific code for identification purposes. Production of
SPF fiocks free from the presence of certain agents and their vaccine is not nOlmally undertaken using virus more than 5
antibodies, as specified in the monograph. It may be passages from the master seed loto In the tests on the master
necessary to demonstrate that the inactivation process is seed lot described below, the organisms used are not
effective against specified potential contaminants. For the normally more than 5 passages from the master seed lot at
production of a master seed lot and for all passages of a the start of the tests, unless otherwise indicated.
micro-organism up to and including the working seed lot, Where the master seed lot is contained within a permanently
eggs from SPF fiocks (5.2.2) are used. infected master cell seed, the following tests are carried out
Where it is unavoidable to use animals or animal tissues in on an appropriate volume of virus from disrupted master cell
the production of veterinary vaccines, such animals shall be seed. Where relevant tests have been carried out on disrupted
free from specified pathogens, as appropriate to the source cells to validate the suitability of the master cell seed, these
species and the target animal for the vaccine. tests need not be repeated.
2-1-2 Media used for seed culture preparation and for 2-1-3-2-2 Propagation. The master seed lot and all
production subsequent passages are propagated on cells, on embryonated
At least the qualitative composition must be recorded of eggs 01' in animals that have been shown to be suitable for
media used for seed culture preparation and for production. vaccine production (see aboye), and, where applicable, using
The grade of each named ingredient is specified. Where substances of animal origin that meet the requirements
media or ingredients are claimed as proprietary, this is prescribed in chapter 5.2.5.
indicated and an appropriate description recorded. 2-1-3-2-3 Identification. A suitable method to identify the
Ingredients that are derived from animal s are specified as to vaccine strain and to distinguish it as far as possible from
the source species and country of origin, and must comply related strains must be used.
with the criteria described in chapter 5.2.5. Preparation 2-1-3-2-4 Bacteria and fungi. The master seed lot complies
processes for media used, including sterilisation procedures, with the test for sterility (2. 6.1).
are documented.
2-1-3-2-5 Mycoplasmas (2.6.7). The master seed lot complies
The addition of antibiotics during the manufacturing process with the test for mycoplasmas (culture method and indicator
is normally restricted to cell culture fiuids and other media, cell culture method).
egg inocula and material harvested from skin or other tissues. 2-1-3-2.-6 Absence of extraneous vu-uses. Monographs may
2-1-3 Seed 10ts contain requirements for freedom from exU"aneous agents,
2-1-3-1 Bacterial seed lots otherwise the requirements stated below apply.
2-1-3-1-1 General requirements. The genus and species (and Preparations of monoclonal or polyclonal antibodies
varieties where appropriate) of the bacteria used in the containing high levels of neutralising antibody to the virus of
vaccine are stated. Bacteria used in manufacture are handled the seed lot are made on a batch basis, using antigen that is
Vet-200 Veterinary Vaccines 2016

not derived from any passage level of the virus iso late giving inactivated harvest at the completion of the inactivation
rise to the master seed virus. Each batch of serum is procedure.
maintained at 56 oC for 30 min to inactivate complemento 2-1-4-3 Formaldehyde. If formaldehyde is used as the
Each batch is shown to be free of antibodies to potential inactivating agent, then a test for free formaldehyde is carried
contaminants of the seed virus and is shown to be free of any out as prescribed under Tests.
non-specific inhibiting effects on the ability of virus es to
2-1-4-4 Other inactivating agems. When other inactivation
infect and propagate within cells (01' eggs, where applicable).
methods are used, appropriate tests are carried out to
If such a serum cannot be obtained, other methods are used
demonstrate that the inactivating agent has been removed or
to remove 01' neutralise the seed virus specifically.
reduced to an acceptable residual level.
If the seed lot virus would intelfere with the conduct and
2-1-4-5 Residual live vints/bacteria and/or detoxification testing.
sensitivity of a test for extraneous virus es, a sample of the
A test for complete inactivation and/or detoxification is
master seed lot is treated with a minimum amount of the
performed immediately after the inactivation and/or
monoclonal 01' polyclonal antibody so that the vaccine virus
detoxification procedure and, if applicable, the neutralisation
is neutralised as far as possible 01' removed. The final virus-
or removal of the inactivating or detoxifying agent.
serum mixture shall, if possible, contain at least the virus
content of 10 doses of vaccine per 0.1 mL for avian vaccines 2-1-4-5-1 Bacterial vaccines. The test selected shall be
and per millilitre for other vaccines. For avian vaccines, the appropriate to the vaccine bacteria being used and shall
testing to be carried out on seed lots is given in chapter consist of at least 2 passages in production medium or, if
2.6.24. For mammalian vaccines, the seed lot 01' the mixture solid medium has been used for production, in a suitable
of seed lot and antiserum is tested for freedom from liquid medium or in the medium prescribed in the
extraneous agents as follows. monograph. The product complies with the test if no
evidence of any live micro-organism is observed.
The mixture is inoculated onto cultures of at least 70 cm2 of
the required cell types. The cultures may be inoculated at 2-1-4-5-2 Bacterial toxoids. The test selected shall be
any suitable stage of growth up to 70 per cent confiuency. appropriate to the toxin or toxins present and shall be the
At least 1 monolayer of each type must be retained as a most sensitive available.
control. The cultures must be monitored daily for a week. 2-1-4-5-3 Viral vaccines. The test selected shall be
At the end of this period the cultures are freeze thawed appropliate to the vaccine virus being used and must consist
3 times, centrifuged to remove cell debris and re-inoculated of at least 2 passages in cells, embryonated eggs or, where no
onto the same cell type as above. This is repeated twice. other suitably sensitive method is available, in animals.
The final passage must produce sufficient cells in appropriate The quantity of cell samples, eggs 01' animals shall be
vessels to carry out the tests below. sufficient to ensure appropriate sensitivity of the test.
Cytopathic and haemadsorbing agents are tested for using For tests in cell cultures, not less than 150 cm2 of cell
the methods described in the relevant sections on testing cell culture monolayer is inoculated with 1.0 mL of inactivated
cultures (5.2.4) and teclmiques such as immunofiuorescence harvest. The product complies with the test if no evidence of
are used for detection of specific contaminants for the tests in the presence of any live virus 01' other micro-organism is
cell cultures. The master seed lot is inoculated onto: observed.
- primaly cells of the species of origin of the virus; The final bulk vaccine is prepared by combining one or more
- cells sensitive to vil'uses pathogenic for the species for batches of antigen that comply with all the relevant
which the vaccine is intended; requirements with any auxilimy substances, such as
- cells sensitive to pestiviruses. adjuvants, stabilisers, antimicrobial preservatives and diluents.
If the master seed lot is shown to contain living organisms of 2-2 CHOICE OF VACCINE COMPOSITION AND
any kind, other than the virus of the species and strain stated, CHOICE OF VACCINE STRAIN
or foreign viral antigens, then it is unsuitable for vaccine For the choice of vaccine composition and choice of vaccine
production. strain, important aspects to be evaluated include safety,
2-1-4 Inactivation efficacy and stability. General requirements for evaluation of
Inactivated vaccines are subjected to a validated inactivation safety and efficacy are given in chapter 5.2.6 and
procedure. The testing of the inactivation kinetics described chapter 5.2. 7. These requirements may be made more
below is carried out once for a given production process. explicit or supplemented by the requirements of specific
The rest of this section applies to each production runo When monographs.
conducting tests for inactivation, it is essential to take For live vaccines, a maximum virus titre or bacterial count
account of the possibility that under the conditions of acceptable from the point of view of safety is established
manufacture, organisms may be physically protected from during development studies. This is then used as the
inactivant. maximum acceptable titre for each batch of vaccine at
2-1-4-1 Inactivatioll kinetics. The inactivating agent and the release.
inactivation procedure shall be shown, under conditions of 2-2-1 Potency and hnmunogenicity
manufacture, to inactivate the vaccine micro-organismo The tests given under the headings Potency and .
Adequate data on inactivation kinetics shall be obtained. Immunogenicity in monographs serve 2 purposes:
Normally, the time required for inactivation shall be not - the Potency section establishes, by a well-controlled test
more than 67 per cent of the duration of the inactivation in experimental conditions, the minimum acceptable
process. vaccinating capacity for all vaccines within the scope of
2-1-4-2 Azitidine. If an aziridine compound is used as the the definition, which must be guaranteed throughout the
inactivating agent then it shall be shown that no inactivating period of validity; .
agent remains at the end of the inactivation procedure. This - well-controlled experimental studies are normally a part of
may be accomplished by neutralising the inactivating agent the overall demonstration of efficacy of a vaccine (see
with thiosulfate and demonstrating residual thiosulfate in the chapter 5.2.7); the test referred to in the section
2016 Veterinary Vaccines Vet-201

Irnmunogenicity (to which the section Potency usually maximum recommended period of use after broaching of the
cross-refers) is suitable as a part of this testing. container.
2-2-2 Route of administration During development studies the effectiveness of the
During development of a vaccine, safety and irnmunogenicity antimicrobial preservative dU'oughout the period of validity
are demonstrated for each route of administration to be shall be demonstrated to the satisfaction of the competent
recommended. The following is a non-exhaustive list of such authority.
routes of administration: The efficacy of the antimicrobial preservative is evaluated as
- intramuscular; described in chapter 5.1.3 and in addition samples are tested
- subcutaneous; at suitable intervals over the proposed in-use shelf-life.
- intravenous; If neither the A criteria nor the B criteria can be met, then in
- ocular; justified cases the following criteria are applied to vaccines for
- oral; veterinary use: bacteria, no increase from 24 h to 7 days,
- nasal; 3 10gIO reduction at 14 days, no increase at 28 days; fungi,
- foot-stab; no increase at 14 days and 28 days.
- wingweb; Addition of antibiotics as antimicrobial preservative is
- intradermal; gene rally not acceptable.
- intraperitoneal;
- zn ovo. 2-2-6 Stability
Evidence of stability is obtained to justify the proposed
2-2-3 Methods of administration period of validity. This evidence takes the form of the results
During development of a vaccine, safety and irnmunogenicity of virus titrations, bacterial counts or potency tests carried
are demonstrated for each method of administration to be out at regular intervals until 3 months beyond the end of dle
recommended. The following is a non-exhaustive list of such shelf life on not fewer than 3 representative consecutive
methods of administration: batches of vaccine kept under recommended storage
- injection; conditions together with results from studies of moisture
- drinking water; content (for freeze-dried products), physical tests on the
- spray; adjuvant, chemical tests on substances such as the adjuvant
- eye-drop; constituents and preservatives, and pH, as appropriate.
- scarification;
Where applicable, studies on the stability of the reconstituted
- implantation;
vaccine are carried out, using the product reconstituted in
- immersion.
accordance with the proposed recommendations.
2-2-4 Categories of animal
Monographs may indicate that a given test is to be carried 2-3 MANUFACTURER'S TESTS
out for each category of animal of the target species for Certain tests may be carried out on the final bulk vaccine
which the product is recommended or is to be rather than on the batch or batches prepared from it; such
recommended. The following is a non-exhaustive list of tests include dlose for antimicrobial preservatives, free
categories that are to be taken into account. formaldehyde and d1e potency determination for inactivated
- Mammals: vaccines.
- pregnant animals/non-pregnant animals; 2-3-1 Residuallive viruslbacteria and/or detoxification
- animals raised primarily for breeding/animals raised testing
primarily for food production; For inactivated vaccines, where the auxiliary substances
- animals of the minimum age or size recommended for would intelfere with a test for inactivation and/or
vaccination. detoxification, a test for inactivation or detoxification is
- A vian species: carried out during preparation of the final bulk, after d1e
- birds raised primarily for egg production/birds raised different batches of antigen have been combined but before
primarily for production of meat; addition of auxiliary substances; the test for inactivation or
- birds before point of lay/birds after onset of layo detoxification may then be omitted on the final bulk and the
- Fish: batch.
- broodstock fishlfish raised primarily for food Where there is a risk of reversion to toxicity, the test for
production. detoxification performed at the latest stage of the production
2-2-5 Antimicrobial preservatives process at which the sensitivity of the test is not
Antimicrobial preservatives are used to prevent spoilage or compromised (e.g. after the different batches of antigen have
adverse effects caused by microbial contamination occurring been combined but before the addition of auxiliary
during use of a vaccine which is expected to be no longer substances) is important to demonstrate a lackof reversion to
than 10 h after first broaching. Antimicrobial preservatives toxicity.
are not included in freeze-dried products but, if justified, 2-3-2 Batch potency test
taking into account the maximum recommended period of For most vaccines, the tests cited under Potency or
use after reconstitution, they may be included in the diluent Immunogenicity are-not suitable for the routine testing of
for multi-dose freeze-dried products. For single-dose liquid batches.
preparations, inclusion of antimicrobial preservatives is not
For live vaccines, the minimum acceptable virus titre or
acceptable unless justified and authorised, but may be
bacterial count that gives satisfactory results in the potency
acceptable, for example where the same vaccine is filled in
test and other efficacy studies is established during
single-dos e and multidose containers and is used in non-
development. For routine testing it must be demonstrated for
food-producing species. For multidose liquid preparations,
each batch that the titre or count at release is such that at the
the need for effective antimicrobial preservation is evaluated
end of the period of validity, in the light of stability studies,
taking into account likely contamination during use and the
the vaccine, stored in the recommended conditions, will
Vet-202 Veterinary Vaccines 2016

contain not less than the minimum acceptable virus titre or 2-3-3-4 pH. The pH of liquid products and diluents is
bacterial count detelmined during development studies. measured and shown to be within the limits set for the
For inactivated vaccines, if the test described under Potency producto
is not used for routine testing, a batch potency test is 2-3-3-5 Water. Where applicable, the freeze-drying process is
established during development. The aim of the batch checked by a detelmination of water and shown to be within
potency test is to ensure that each batch of vaccine would, if the limits set for the producto
tested, comply with the test described under Potency and 3 BATCH TESTS
Irnmunogenicity. The acceptance critelia for the batch
The monographs also indicate tests to be canied out OlZ each
potency test are therefore established by correlation with the
particular vaccine.
test described under Potency. Where a batch potency test is
described in a monograph, this is given as an example of a All hen eggs, chickens and chicken cell cultures for use in
test that is considered suitable, after establishment of quality control tests shall be derived from an SPF fiock
cOlTelation with the potency test; other test models can also (5.2.2).
be used. 3-1 Identification
2-3-3 Batch For inactivated vaccines, the identification presclibed in
Unless otherwise prescribed in the monograph or othelwise monographs is usually an antibody induction test since this is
justified and authorised, the final bulk vaccine is distributed applicable to all vaccines.
aseptically into sterile, tamper-proof containers, with or 3-2 Formaldehyde (2.4.18); use Method B if sodium
without freeze-drying, which are then c10sed so as to exc1ude metabisulfite has been used to neutralise excess
contamination. formaldehyde)
Only a batch that complies with each of the requirements Where formaldehyde has been used in the preparation, the
given below under 3 Batch tests or in the relevant individual concentration of free formaldehyde is not greater than
monograph may be released for use. With the agreement of 0.5 gIL, unless a higher amount has been shown to be safe.
the competent authority, certain of the batch tests may be 3-3 Phenol (2.5.15)
omitted where in-process tests give an equal or better When the vaccine contains phenol, the concentration is not
guarantee that the batch would comply or where altemative greater than 5 gIL.
tests validated with respect to the Phmmacopoeia method 3-4 Sterility (2.6.1)
have been calTied out. Under particular circumstances Vaccines comply with the test for sterility. Where the volume
(i.e. significant changes to the manufacturing process, as well of liquid in a container is greater than 100 mL, the method
as reports of unexpected adverse reactions observed in the of membrane filtration is used wherever possible. Where the
field or reports that the final batches do not comply with the method of membrane filtration cannot be used, the method
former data provided during licensing), other tests, inc1uding of direct inoculation may be used. Whe1'e the volume of
tests on animals, may be needed on an ad hoc basis; they are liquid in each container is at least 20 mL, the minimum
carried out in agTeement with or at the request of the volume to be used for each culture medium is 10 pe1' cent of
competent authority. For safety testing, one or more of the the contents or 5 mL, whichever is less. The appropriate
tests described in chapter 5.2.6 may be carried out. number of items to be tested (2.6.1) is 1 per cent of the
The identification test can often be conveniently combined batch with a minimum of 4 and a maximum of 10.
with the batch potency test to avoid the unnecessary use of For live bacterial and for live fungal vaccines, the absence of
animals. For a given vaccine, a validated in vitro test can be micro-organisms other than the vaccine strain is
used to avoid the unnecessary use of animals. demonstrated by suitable methods such as microscopic
2-3-3-1 Animal tests. In accordance with the provisions of the examination and inoculation of suitable media.
European Convention for the Protection of Vertebrate For frozen 01' freeze-dried avian live viral vaccines produced
Animals U sed for Experimental and Other Scientific in embryonated eggs, for non-parenteral use only, the
PUTposes, tests must be carried out in such a way as to use requirement for sterility is usually replaced by requirements
the minimum number of animals and to cause the least pain, for absence of pathogenic micro-organisms and fo1' a
suffering, distress or lasting hann. The clitelia for judging maximum of 1 non-pathogenic micro-organism per dose.
tests in monographs must be applied in light of this.
For example, if it is indicated that an animal is considered to 3-5 Extraneous agents
be positive, infected etc. when typical c1inical signs occur Monographs presClibe a set of measures that, taken togethe1',
then as soon as it is c1ear that result will not be affected the give an acceptable degree of assurance that the final product
animal in question shall be either euthanised or given suitable does not contain infectious extraIieous agents. These
treatment to prevent unnecessary suffeling. In accordance measures inc1ude:
with the General Notices, altemative test methods may be 1) production within a seed-lot system and a cell-seed
used to demonstrate compliance with the monograph and the system, wherever possible;
use of such tests is particularly encouraged when this leads to 2) extensive testing of seed lots and cell seed for extraneous
replacement 01' reduction of animal use or reduction of agents;
suffering. 3) requirements forSPF fiocks used for providing substrates
2-3-3-2 Physical tests. A vaccine with an oily adjuvant is tested for vaccine production;
for viscosity by a suitable method and shown to be within the 4) testing of substances of animal origin, which must,
limits set for the product. The stability of the emulsion shall wherever possible, undergo an inactivation procedure;
be demonstrated.
5) for live vaccines, testing of the fina! product for infectious
2-3-3-3 Chemical tests. Tests for the concentrations of extraneous agents; such tests are less extensive than those
appropriate substances such as aluminium and preservatives carried out at earlier stages because of the guarantees given
are carried out to show that these are within the limits set for by in-process testing.
the producto
2016 Veterinary Vaccines Vet-203

In case of doubt, the tests intended for the seed lot of a live
Anthrax Vaccine, living ***
vaccine may also be applied to the final producto If an *** ***
extraneous agent is found in such a test, the vaccine does not (Anthrax Spore Vaccine (Live) for VeterinalY Use) ***
comply with the monograph. Ph Bur monograph 0441)
Avian live viral vaccines comply with the tests for extraneous ~E~ ____________________________________________
agents in batches of finished product (2.6.25).
1 DEFINITION
3-6 Mycoplasmas (2.6.7)
Anthrax spore vaccine (live) for veterinary use is a
Live viral vaccines comply with the test for mycoplasmas
preparation of live spores of a suitable attenuated, non-
(culture method).
capsulated strain of Bacillus anthracis. This monograph
3-7 Potency applies to vaccines intended for the active irnmunisation of
The vaccine complies with the requirements of the test animals against disease caused by B. anthracis.
mentioned under Irnmunogenicity (section 2-2-1) when
2 PRODUCTION
administered by a recommended route and method.
2-1 PREPARATION OF THE VACCINE
Bxpily date Unless otherwise stated, the expiry date is
B anthracis Is grown in an appropriate medium. At the end
calculated from the beginning of the virus titration or
of growth the spores are suspended in a stabilising solution
bacterial count (for live vaccines) or the beginning of the
and counted. The vaccine may be adjuvanted.
potency test (for other vaccines). For combined vaccines, the
expiry date is that of the component which expires first. 2-2 CHOICE OF VACCINE STRAIN
For vaccines stored by the manufacturer at a temperature The strain used is:
lower than that stated on the label, the stability for the entire - not lethal to the guinea-pig or the mouse,
storage period is demonstrated by an appropriate study. - or lethal to the guinea-pig but not to the rabbit,
The expiry date is then calculated from the date that the - or lethal to sorne rabbits.
vaccine is stored in the conditions stated on the label. The vaccine strain is shown to be satisfactory with respect to
The expiry date applies to vaccines stored in the prescribed safety (5.2.6) and efficacy (5.2.7) for the animals for which it
conditions. is intended.
4STORAGE The following test for irnmunogenicity (2-2-1) may be used
Store protected from light at a temperature of 5 ± 3 oC, during the demonstration of efficacy.
unless otherwise indicated. Liquid preparations are not to be 2-2-1 Irnmunogenicity
allowed to freeze, unless otherwise indicated. For a strain of B. anthracis which is not lethal to the guinea-
pig or the mouse, the test may be carried out in guinea-pigs.
5 LABELLING
For a strain which is lethal to the guinea-pig but not to the
The label states:
rabbit, the test may be carried out in rabbits. For a strain
- that the preparation is for veterinary use;
which is lethal to sorne rabbits, carry out the test in sheep.
- the volume of the preparation and the number of doses in
the container; If the test is carried out in guinea-pigs or in rabbits, use not
- the route of administration; fewer than 13 healthy animals (group a). Inject by the
- the type or types of bacteria (and where applicable the subcutaneous or the intradermal route into each of not fewer
antigenic components) or viruses used and for live than 10 animals 1/10th of the smallest dose to be
vaccines the minimum and the maximum number of live recommended for sheep. Maintain not fewer than 3 animals
bacteria or the minimum and the maximum virus titre; of the same species and the same origin as controls. Observe
- where applicable, for inactivated vaccines, the minimum the animals at least daily for 21 days. If more than 2 animal s
potency in International Units; die from non-specific causes, repeat the test.
- where applicable, the name and amount of antimicrobial If the test is carried out in sheep, use not fewer than
preservative or other substance added to the vaccine; 8 healthy sheep (group b). Vaccinate by the subcutaneous or
- the name of any substance that may cause an adverse the intradermal route each of not fewer than 5 sheep 1/10 of
reaction; the smallest dos e of the vaccine stated on the label for sheep.
- for freeze-dried vaccines: Maintain not fewer than 3 sheep of the same origin as
- the name or composition and the volume of the controls. Observe the sheep at least daily for 21 days.
reconstituting liquid to be added; Challenge each vaccinated animal of group (a) or group (b)
- the period within which the vaccine is to be used after by a subcutaneous route with at least 100 MLD, and
reconstitution; challenge each control animal by a subcutaneous route with
- for vaccines with an oily adjuvant, that if the vaccine is at least 10 MLD of a strain of B. anthracis pathogenic for the
accidentally injected into man, urgent medical attention is species of animal used in the test. Observe all the animal s at
necessary; least daily for 10 days after challenge.
- the animal species for which the vaccine is intended; The vaccine complies with the test if during the observation
- the indications for the vaccine; period after challenge, all the vaccinated animals survive and
- the instructions for use; all the controls die from anthrax. If a vaccinated animal dies
any contra-indications to the use of the product including after the challenge, repeat the test. If in the second test a
any required warning on the dangers of administration of vaccinated animal dies, the vaccine fails the test.
an overdose;
- the doses recommended for different species. 3 BATCH TESTS
____________________________________________ PhE~
3-1 Identification
B. anthracis present in the vaccine is identified by means of
morphological and serological tests, culture and biochemical
tests.
Vet-204 Veterinary Vaccines 2016

3-2 Bacteria and fungi schedule to be recommended requires a 2nd dose, administer
Carry out the test by microscopic examination and by another dose after an interval of at least 14 days. Observe the
inoculation of suitable media. The vaccine, inc1uding where pigs at least daily until 14 days after the last administration.
applicable, the diluent supplied for reconstitution, does not If the test is carried out in pregnant sows, observe the sows
contain contaminating bacteria and fungi. until 1 day after farrowing.
3-3 Live spores The vaccine complies with the test if no pig shows abnormal
Make a count of live spores by plate count. The vaccine local or systemic reactions or dies from causes attributable to
complies with the test if the number of live spores is not les s the vaccine during the test. If d1e test is carried out in
than 80 per cent of that stated on the labe!' pregnant sows, no adverse effects on gestation or the
3-4 Potency offspring are noted.
The vaccine complies with the requirements of the test 2-3-1-1-2 Safety in the pigs used in tests 2-3-2 for
prescribed under Immunogenicity (section 2-2-1). It is not immunogenicity. The pigs used in the tests for
necessary to carry out the potency test for each batch of the immunogenicity are also used to evaluate safety. Measure the
vaccine if it has been carried out on a representative batch body temperature of each vaccinated pig at the time of
using a vaccinating dose containing not more than the vaccination and 6 h, 24 h and 48 h later. Examine the
minimum number of live spores stated on the labe!' injection site at slaughter for local reactions.
_ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ PhEur The vaccine complies with the test if no pig shows:
- a temperature rise greater than 1.5 oC and the number of
pigs showing a temperature greater than 41 oC do es not
exceed 10 per cent of the group;
- other systemic reactions (for example, anorexia);
Aujeszky's Disease Vaccine, ***
*** *** - abnormal local reactions attributable to the vaccine.
Inactivated *** 2-3-1-2 Field studies. The pigs used for field trials are also
(Aujeszky's Disease Vaccine (Inactivated) for Pigs, used to evaluate safety. Carry out a test in each category of
pigs for which the vaccine is intended (sows, fattening pigs).
Ph Eur J1lonograph 0744)
Use not fewer than 3 groups each of not fewer than 20 pigs
Ph Eur _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ __
with corresponding groups of not fewer than 10 controls.
1 DEFINITION Measure the body temperature of each vaccinated pig at the
Aujeszky's disease vaccine (inactivated) for pigs is a time of vaccination and 6 h, 24 b and 48 h later. Examine
preparation of a suitable strain of Aujeszky's disease virus, the injection site at slaughter for local reactions.
inactivated while maintaining adequate immunogenic The vaccine complies with the test if no pig shows:
propelties, or a preparation of an inactivated fraction of the - a temperature rise greater than 1.5 oC and the number of
virus having adequate immunogenic propenies. This pigs showing a temperature greater than 41°C does not
monograph applies to vaccines intended for the active exceed 25 per cent of the group;
immunisation of pigs and for passive protection of their - abnonnal local reactions attributable to the vaccine.
progeny against Aujeszky's disease. 2-3-2 hrununogenicity
2 PRODUCTION A test is carried out for each route and method of
2-1 PREPARATION OF THE VACCINE administration to be recommended, using in each case pigs -
The vaccine virus is grown in cell cultures. The viral of the age to be recommended for vaccination. The vaccine
suspension is harvested and inactivated; it may be treated to administered to each pig is of minimum potency.
fragment d1e virus and the viral fragments may be purified 2-3-2-1 Vaccines intended fa/' active i711711unisatioll. U se for the
and concentrated. The vaccine may be adjuvanted. test not fewer than 15 fattening pigs that do not have
2-2 SUBSTRATE FOR VIRUS PROPAGATION antibodies against Aujeszky's disease virus. The body mass of
none of the pigs differs from the average body mass of the
2-2-1 CeIl cultures
group by more than 20 per cent. Vaccinate not fewer than 10
The cell cultures comply with the requirements for cell
pigs, according to the schedule to be recommended.
cultures for production of veterinary vaccines (5.2.4).
Maintain not fewer than 5 pigs as controls. At the end of the
2-3 CHOICE OF VACCINE COMPOSITION fattening period (80-90 kg), weigh and challenge each pig by
The vaccine is shown to be satisfactory wid1 respect to safety the intranasal route with a sufficient quantity of virulent
(5.2.6) and efficacy (5.2.7) for the pigs for which it is Aujeszky's disease virus (challengewith at least 10 6 CCID so
intended. The following tests for safety (section 2-3-1) and of a virulent strain having undergone not more than
immunogenicity (section 2-3-2) may be used during the 3 passages and administered in not les s than 4 mL of diluent
demonstration of safety and efficacy. has been found to be satisfactory). The titre of excreted virus
2-3-1 Safety is determined in swabs taken from the nasal cavity of each
2-3-1-1 Laborato/JI tests. Carry out d1e tests for each route pig daily from the day before challenge until virus is no
and meth.od of administration to be recommended for longer detected. Each pig is weighed 7 days after challenge or
vaccination and where applicable, in pigs of each category for at the time of death if this occúrs earlier and the average
which the vaccine is intended (sows, fattening pigs), using in daily gain is calculated as a percentage. For each group
each case pigs not older than the minimum age to be (vaccinated and controls), the average of the average daily
recommended for vaccination. Use a batch of vaccine gains is calculated.
containing not less than the maximum potency that may be The test is invalid unless all the controt pigs display signs of
expected in a batch of vaccine. Aujeszky's disease and the average of their daily gains is less
2-3-1-1-1 General safety. For each test, use not fewer than than -0.5 kg. The vaccine complies with the test if:
8 pigs d1at do not have antibodies against Aujeszky's disease
virus. Administer to each pig 1 dose of d1e vaccine. If the
2016 Veterinary Vaccines Vet-205

-- all the vaccinated pigs survive and the difference between strain is not pathogenic for the rabbit, carry out the test in 2
the average s of the daily gains for the 2 groups is not less sheep.
than 1.5 kg; 3-4 Specified extraneous agents
-- the geometrical mean titres and the duration of excretion U se not fewer than 2 pigs that do not have antibodies against
of the challenge virus are significantly lower in vaccinates Aujeszky's disease virus and against pestiviruses. Administer
than in controls. to each pig by a recommended route a double dos e of the
2-3-2-2 Vaccines intended for passive immunisation. If the vaccine, then another dose after 14 days. 14 days after the
vaccine is intended for use in sows for the passive protection last administration, cany out tests for antibodies. The vaccine
of piglets, the suitability of the strain for this purpose may be complies with the test if it does not stimulate the formation
demonstrated by the following method. of antibodies against pestiviruses.
U se for the test not fewer than 12 sows that do not have 3-5 Potency
antibodies against Aujeszky's disease virus. Vaccinate not The vaccine complies with the requirements of the test
fewer than 8 sows, according to the schedule to be described below when administered by a recommended route
recommended. Maintain not fewer than 4 sows as controls. and method.
At 6-10 days of age, challenge the piglets from the sows with U se for the test not fewer than 10 pigs weighing 15-35 kg
a sufficient quantity of virulent Aujeszky's disease virus. and that do not have antibodies against Aujeszky's disease
Observe the piglets at least daily for 21· days. virus or against a fraction of the virus. The body mass of
The test is not valid if the average number of piglets per litter none of the pigs differs from the average body mass of the
for each group is less than 6. The vaccine complies with the group by more than 25 per cent. Vaccinate not fewer than 5
test if not les s than 80 per cent protection against mortality is pigs with 1 dose of the vaccine. Maintain not fewer than 5
found in the piglets from the vaccinated sows compared to pigs as controls. After 3 weeks, weigh each pig, then
those from the control sows. challenge them by the intranasal route with a sufficient
2-4 MANUFACTURER'S TESTS quantity ofvirulent Aujeszky's disease virus. Weigh each
2-4-1 Residuallive virus animal 7 days after challenge 01' at the time of death if dlis
The test for residual live virus is carried out using 2 passages occurs earlier and calculate the average daily gain as a
in the same type of cell culture as that used in the percentage. Por each group (vaccinated and controls),
production of the vaccine or cells shown to be at least as calculate dle average of dle average daily gains.
sensitive. The quantity of inactivated virus ha1\Test used in The test is invalid unless all the control pigs display signs of
the test is equivalent to not les s than 25 doses of the vaccine. Aujeszky's disease and dle average of dleir daily gains is less
The inactivated virus ha1\Test complies with the test if no live dlan -0.5 kg. The vaccine complies with the test if the
virus is detected. vaccinated pigs survive and the difference between the
2-4-2 Batch potency test averages of the daily gains for the 2 groups is not less than
It is not necessary to carry out the potency test (section 3-5) 1.1 kg.
for each batch of vaccine if it has been carried out using a 4 LABELLING
batch of vaccine with a minimum potency. The test The label states whedler the vaccine strain is pathogenic for
described under Potency is carried out for a given vaccine, the rabbit.
on one or more occasions, as decided by or with the _____________________________________________ PhE~

agreement of the competent authority. Where this test is not


carried out, an alternative validated method is used, the
criteria for acceptance being set with reference to a batch of
vaccine that has given satisfactory results in the test described
Aujeszky's Disease Vaccine, ***
*** ***
under Potency.
3 BATCH TESTS Living ***
3-1 Identification (AuJeszky's Disease Vaccine (Live) for Pigs for
In animals that do not have antibodies against Aujeszky's Parenteral Administratiol1, Ph Bur mOl1ograph 0745)
disease virus or against a fraction of the virus, the vaccine
~E~ _____________________________________________
stimulates the production of specific antibodies against
Aujeszky's disease virus or the fraction of the virus used in 1 DEFINITION
the production of the vaccine. Aujeszky's disease vaccine (live) for pigs for parenteral
3-2 Bacteria and fungi administration is a preparation of a suitable strain of
The vaccine, inc1uding where applicable the diluent supplied Aujeszky's disease virus. This monograph applies to vaccines
for reconstitution, complies with the test for sterility intended for the active immunisation of pigs and for passive
ptescribed in the monograph Vaccil1es for veterinalJI use protection of dleir progeny against Aujeszky' s disease.
(0062). The vaccine may be administered after mixing with an
adjuvant.
3-3 Residuallive virus
Wherever possible, carry out a suitable test for residual live 2 PRODUCTION
Aujeszky's disease virus using 2 passages in the same type of 2-1 PREPARATION OF THE VACCINE
cell culture as used in the production of the vaccine or cells The vaccine virus is grown in cell cultures.
shown to be at least as sensitive. Otherwise, inject 1 dose of
2-2 SUBSTRATE FOR VIRUS PROPAGATION
the vaccine subcutaneously into each of 5 healthy non-
2-2-1 Cell cultures
immunised rabbits. ObSel\Te the rabbits for 14 days after the
The cell cultures comply with the requirements for cell
injection. The vaccine complies with the test if no abnormal
cultures for the production of veterinaty vaccines (5.2.4).
reaction (in particular a local rash) occurs. If the vaccine
Vet-206 Veterinary Vaccines 2016

2-3 CHOICE OF VACCINE VIRUS 2-3-1-5 Neurological safet:y for strains other than gE-negative
The vaccine virus is shown to be satisfactory with respect to This test is not necessary for gE-negative strains. Administer
safety (5.2.6) and efficacy (5.2.7) for the pigs for which it is to not fewer than 5 piglets, 3-5 days old, by the intracerebral
intended. The virus may have a genetic marker. route, 104 .5 CCID 50 of vaccine virus.
The following tests for safety (section 2-3-1), virus excretion The vaccine virus complies with the test if none of the piglets
(section 2-3-2), non-transmissibility, including transmission dies or shows signs of neurological disorder.
across the placenta and by semen (section 2-3-3), increase in 2-3-1-6 Absence of latent infections Use for the test not fewer
virulence (section 2-3-4) and immunogenicity (section than 10 piglets, 3-4 weeks old and that do not have
2-3-5), may be used during the demonstration of safety and antibodies against Aujeszky' s disease virus. Administer to
efficacy. each piglet a daily injection of 2 mg of prednisolone per
2-3-1 Safety kilogram of body mass for 5 consecutive days. On the 3rd day
2-3-1-1 Safety test in piglets Carry out the test for each route administer to each piglet a quantity of vaccine virus
and method of administration to be recommended for equivalent to not less than the maximum virus titre likely to
vaccination, using in each case piglets 3-4 weeks old. be contained in 1 dose of the vaccine by a route to be
Use vaccine virus at the least attenuated passage level that recommended. Antimicrobial agents may be administered to
will be present between the master seed lot and a batch of prevent aspecific signs. Observe the piglets at least daily for
the vaccine. at least 21 days.
For each test, use not fewer than 20 piglets that do not have The vaccine virus complies with the test if no piglet shows
antibodies against Aujeszky' s disease virus. Administer to not signs of disease or dies from causes attributable to the
fewer than 10 piglets a quantity of the vaccine virus vaccine virus.
equivalent to not less than 10 times the maximum virus titre 2-3-1-7 Safety in pregnant SO'lUS and absence of tmnsmission
likely to be contained in 1 dose of the vaccine. Maintain not across the placenta U se for the test not fewer than 15 pregnant
fewer than 10 piglets ascontrols. Observe the piglets at least sows that do not have antibodies against Aujeszky' s disease
daily for at least 21 days. virus. Administer to not fewer than 5 sows, by a route to be
The vaccine virus complies with the test if the weight curve recommended, a quantity of vaccine virus equivalent to not
of the vaccinated piglets does not differ significant1y from less than 10 times the maximum virus titre likely to be
that of the controls and if no piglet shows signs of disease or contained in 1 dose of the vaccine during the 4 th or 5th week
dies from causes attributable to the vaccine virus. of gestation. Administer to not fewer than 5 other sows the
2-3-1-2 Safety of the pigs used in tests 2-3-5 for i11Z111unogenicity same dose of the virus by the same route during the 10th or
The pigs used in the tests for immunogenicity are also used 11 th weeks of gestation. Maintain not fewer than 5 other
to evaluate safety. Measure the body temperature of each pregnant sows as controls. For the piglets from vaccinated
vaccinated pig at the time of vaccination and 6 h, 24 h and sows: carry out tests for serum antibodies against Aujeszky's
48 h later. Examine the injection site at slaughter for local disease virus; carry out tests for Aujeszky's disease virus
reactions. antigen in the liver and lungs of those piglets showing
abnormalities and in a qualter of the remaining healthy
The vaccine virus complies with the test if no pig shows:
piglets.
- a temperature rise greater than 1.5 oC and the number of
pigs showing a temperature greater than 41 oC does not The vaccine virus complies with the test if:
exceed 10 per cent of the group; - the number of piglets born to the vaccinated sows, any
- other systemic reactions (far example, anorexia); abnormalities in the piglets and the duration of gestation
- abnormallocal reactions attributable to the vaccine virus. do not differ significant1y from those of t11e controls;
- no Aujeszky's disease virus antigen is found in piglets
2-3-1-3 Safety infield studies The pigs used for field trials are
born to the vaccinated sows;
also used to evaluate safety. Carry out a test in each category
- no antibodies against Aujeszky' s disease virus are found
of pigs for which the vaccine is intended (sows, fattening
in the serum taken before ingestion of the colostrum.
pigs). U se not fewer than 3 groups, each of not fewer than
20 pigs, with corresponding groups of not fewer than 2-3-2 Virus excretion
10 controls. Measure the body temperature of each pig at the Use for the test not fewer than 18 pigs, 3-4 weeks old and
time of vaccination and 6 h, 24 h and 48 h later. Examine that do not have antibodies against Aujeszky' s disease virus.
the injection site at slaughter for local reactions. Administer to not fewer than 14 pigs a quantity of the
vaccine virus equivalent to not less than the maximum virus
The vaccine virus complies with the test if no pig shows:
titre likely to be contained in 1 dose of the vaccine by a route
- a temperature rise greater than 1.5 oC and the number of
and a site to be recommended. Maintain not fewer than
pigs showing a temperature greater than 41 oC does not
4 pigs as contact controls. Carry out suitably sensitive tests
exceed 25 per cent of the group;
for the virus individually on the nasal and oral secretions as
- abnormal local reactions attributable to the vaccine virus.
follows: collect nasal and oral swabs daily from the day
2-3-1-4 Neurological safety Use for the test not fewer than before vaccination until 10 days after vaccination.
10 piglets, 3-5 days old and that do not have antibodies
The vaccine complies with the test if the virus is not isolated
against Aujeszky's disease virus. Administer to each piglet by
from the secretions collected ..
the intranasal route a quantity of the vaccine virus equivalent
to not less than 10 times the maximum virus titre likely to be 2-3-3 Non-transmissibility
contained in 1 dose of the vaccine. Observe the piglets at Carry out the te~t on 4 separate occasions. Each time,
least daily for at least 21 days. administer to not fewer than 4 piglets, 3-4 weeks old and
The vaccine virus complies with the test if none of the piglets that do not have antibodies against Aujeszky' s disease virus,
dies or shows signs of neurological disorder attributable to by a route to be recommended, a quantity of the vaccine
the vaccine virus. virus equivalent to not less than the maximum virus titre
lil,ely to be contained in 1 dos e of the vaccine. After 1 day,
keep not fewer than 2 other piglets of the same age and that
2016 Veterinary Vaccines Vet-207

do not have antibodies against Aujeszky' s disease virus close daily gain as a percentage. For each group (vaccinated and
together with them. After S weeks, test all the piglets for the controls), ca1culate the average of the average daily gains.
presence of antibodies against Aujeszky' s disease virus. The test is invalíd unless all the control pigs display signs of
The test is not valid if any vaccinated piglet does not show Aujeszky' s disease and the average of their average daily
an antibody response. The vaccine virus complíes with the gains is less than -O.S kg. The vaccine complies with the test
test if no antibodies against Aujeszky' s disease virus are if:
detected in any group of contact controls and if all the - all the vaccinated pigs survive and the difference between
vaccinated piglets show an antibody response. the average s of the average daily gains for the 2 groups is
2-3-4 Increase in virulence not less than 1.5 kg;
Carry out the test according to general chapter 5.2.6 using - the geometrical mean titres and the duration of excretion
piglets 3-S days old and that do not have antibodies against of the challenge virus are significantly lower in vaccinates
Aujeszky's disease virus. If the properties of the vaccine virus than in controls.
allow sequential passage through S groups via natural 2-3-5-2 Vaccines intended for passive protection If the vaccine is
spreading, this method may be used, otherwise passage as intended for use in sows for tl1e passive protection of piglets,
described below is carried out. the suitabilíty of the vaccine virus for this purpose may be
Administer to each piglet of the 1st group by the intranasal demonstrated by the following method.
route a quantity of the vaccine virus that will allow recovery U se for the test not fewer than 12 sows that do not have
of virus for the passages described below. After 3-S days, antibodies against Aujeszky's disease virus. Vaccinate not
prepare a suspension from the brain, lung, tonsils and local fewer than 8 sows, according to the schedule to be
lymph glands of each piglet and pool the samples. Administer recommended. Maintain not fewer than 4 sows as controls.
1 mL of the suspension of pooled samples by the intranasal At 6-10 days of age, challenge the piglets from the sows with
route to each piglet of the next group. Carry out this passage a sufficient quantity of virulent Aujeszky' s disease virus.
operation not fewer than 4 times, verify the presence of the Observe the piglets at least daily for 21 days.
virus at each passage. If the virus is not found at a passage The test is not valíd if the average number of piglets per lítter
level, repeat the passage by administration to a group of for each group is les s than 6.
10 animals. The vaccine complies with the test if not les s than
If the sth group of animals shows no evidence of an increase 80 per cent protection against mortalíty is found in the
in virulence indicative of reversion during the observation piglets from the vaccinated sows compared to those from the
period, further testing is not required. Othelwise, carry out control sows.
an additional safety test and compare the clinical signs and
2-4 MANUFACTURER'S TESTS
any relevant parameters in a group of at least 8 animals
receiving the material used for the 1sr passage and another 2-4-1 Batch potency test
It is not necessary to carry out the Potency test (section 3-6)
similar group receiving the virus at the final passage level.
for each batch of the vaccine if it has been carried out on a
The vaccine virus complies with the test if no indication of representative batch using a vaccinating dos e containing not
increased virulence of the virus recovered for the final more than the minimum virus titre stated on the label.
passage compared with the material used for the 1st passage The test described under Potency is carried out for a given
is observed. If virus is not recovered after an initial passage in vaccine, on one or more occasions, as decided by or with the
2 animals and a subsequent repeat passage in 10 animals, the agreement of the competent authority. Where this test is not
vaccine virus also complies with the test. carried out, an alternative validated method is used, the
2-3-5 Irnmunogenicity criteria for acceptance being set with reference to a batch of
A test is carried out for each route and method of vaccine that has given satisfactory results in tl1e test described
administration to be recommended for vaccination. under Potency.
The quantity of vaccine virus to be administered to each pig
3 BATCH TESTS
is not greater than the minimum virus titre to be stated on
3-1 Identification
the label and the virus is at the most attenuated passage level
The vaccine virus is identified by a suitable method,
that will be present in a batch of vaccine.
e.g. when mixed with a monospecific antiserum, the vaccine
2-3-5-1 Vaccines intended for active i11111lUnisation Use for the virus is no longer able to infect susceptible cell cultures into
test not fewer than lS fattening pigs of the age to be which it is inoculated.
recommended and that do not have antibodies against
Aujeszky's disease virus. The body mass of none of the pigs 3-2 Bacteria and fungi
differs from the average body mass of the group by more The vaccine, including where applicable, the diluent supplied
than 20 per cent. Vaccinate not fewer than 10 pigs, for reconstitution, complíes with the test for sterility
according to the schedule to be recornmended. Maintain not prescribed in the monograph Vaccines for veteril1aJY use
fewer than S pigs as controls. At the end of the fattening (0062).
period (80-90 kg), weigh and challenge each pig by the 3-3 Mycoplasmas (2.6.7)
intranasal route with a sufficient quantity of virulent The vaccine complies with the test for mycoplasmas.
Aujeszky's disease virus (challenge with at least 10 6 CCID so 3-4 Extraneous agents
of a virulent strain having undergone not more than Neutralise the vaccine virus with a suitable monospecific
3 passages and administered in not less than 4 mL of diluent antise:rum or monoclonal antibodies against Aujeszky's
has been found to be satisfactory). Determine the titre of disease virus and inoculate into cell cultures known for their
virus in swabs taken from the nasal cavity of each pig daily susceptibility to viruses pathogenic for pigs and to
from the day before challenge until virus is no longer pestiviruses. Carry out at least 1 passage and maintain the
detected. Weigh each pig 7 days after challenge or at the cultures for 14 days. The vaccine complíes with the test if no
time of death if this occurs earlier and ca1culate the average cytopathic effect develops and there is no sign of the
Vet-208 Veterinary Vaccines 2016

presence of haemadsorbing agents. Carry out a specific test 2-3 SEED LOTS
for pestiviruses. 2-3-1 Extraneous agents
3-5 Virus titre The master seed lot complies with the test for extraneous
Titrate the vaccine virus in suitable ceH cultures. The vaccine agents in seed lots (2.6.24). In these tests on the master seed
complies with the test if 1 dose contains not les s than the lot, the organisms used are not more than 5 passages from
minimum virus titre stated on the label. the master seed lot at the start of the test.
3-6 Potency 2-4 CHOICE OF VACCINE COMPOSITION
The vaccine complies with the requirements of the test The vaccine is shown to be satisfactory with respect to safety
described below when administered by a recommended route (5.2.6) and efficacy (5.2.7) for the birds for which it is
and method. intended.
Use not fewer than 10 pigs weighing 15-35 kg and that do The following tests for safety (section 2-4-1) and
not have antibodies against Aujeszky's disease virus. immunogenicity (section 2-4-2) may be used during the
The body mas s of none of the pigs differs from the average demonstration of safety and efficacy.
body mass of the group by more than 25 per cent. Vaccinate 2-4-1 Safety
not fewer than 5 pigs with 1 dose of the vaccine. Maintain The test is carried out for each route of administration to be
not fewer than 5 pigs as controls. After 3 weeks, weigh each recommended for vaccination and for each avian species for
pig, then chaHenge them by the intranasal route with a which the vaccine is intended. U se a batch of vaccine
sufficient quantity of virulent Aujeszky' s disease virus. Weigh containing not les s than the maximum potency that may be
each pig 7 days after chaHenge or at the time of death if this expected in a batch of vaccine.
occurs earlier and calculate the average daily gain as a
F 01' each test, use not fewer than 8 birds not older than the
percentage. For each group (vaccinated and controls),
minimum age to be recommended for vaccination. In the
calculate the average of the average dai1y gains.
case of chickens, use chickens from a flock free from
The test is invalid unless aH the control pigs display signs of specified pathogens (SPF) (5.2.2) and if the vaccine is used
Aujeszky's disease and the average of their average daily for species other than chickens, they have not been
gains is less than -0.5 kg. The vaccine complies with the test vaccinated and do not have antibodies against avian
if aH the vaccinated pigs survive and the difference between infectious bronchitis virus. Administer by a route to be
the average s of the average daily gains for the 2 groups is not recommended and method to each bird 1 dos e of the
less than 1.6 kg. vaccine. Observe the birds for at least daily for at least
_____________________________________________ PhE~
14 days after the administration of the vaccine.
The test is not valid if non-specific mortality occurS.
The vaccine complies with the test if no bird shows abnormal
signs of disease or dies from causes attributable to the
Avian Infectious Bronchitis *** vaccine.
** ** 2-4-2 Irnmunogenicity
Vaccine (Inactivated) **** * A test is carried out for each route and method of
(Ph. Eur. monograph 0959) administration to be recommended, using in each case
CAUTION Accidental injection of oily vaccine can cause chickens from an SPF flock (5.2.2) and for each serotype in
serious local reactions in mano Expert medical advice should the vaccine. The vaccine administered to each chicken is of
be sought immediately and the doctor should be informed minimum potency.
that the vaccine is an oil emulsiono Use for the test 4 groups of not fewer than 30 chickens
PhE~ _____________________________________________ treated as foHows:
-- group A: unvaccinated control s;
1 DEFINITION
-:- group B: vaccinated with inactivated avian infectious
Avian infectious bronchitis vaccine (inactivated) is a bronchitis vaccine;
preparation of one or more suitable strains of one or more -- group C: vaccinated with live avian infectious bronchitis
serotypes of avian infectious bronchitis virus, inactivated vaccine and inactivated avian infectious bronchitis vaccine
while maintaining adequate immunogenic properties. This according to the schedule to be recommended;
monograph applies to vaccines intended to protect birds -- group D: vaccinated with live avian infectious bronchitis
against a drop in egg production or quality; for vaccines also vaccine.
intended for protection against respiratory signs, a
Monitor egg production and quality in aH chickens from
demonstration of efficacy additional to that described under
point of lay until at least 4 weeks after challenge. At the peak
Potency is required.
of lay, challenge all groups with a quantity of virulent aviim
2 PRODUCTION infectious bronchitis virus sufficient to cause a drop in egg
2-1 PREPARATION OF THE VACCINE production or quality over 3 consecutive weeks during the
The vaccine virus is propagated in fertilised hens' eggs or in 4 weeks following challenge. The test is invalid unless there is
ceH cultures. The vaccine may be adjuvanted. a drop in egg production in group A compared to the normal
2-2 SUBSTRATE FOR VIRUS PROPAGATION level noted before challenge of at least 35 per cent where
challenge has been made with a Massachusetts-type strain;
2-2-1 Embryonated hens' eggs
where it is necessary to carry out a challenge with a strain of
If the vaccine virus is grown in embryonated hens' eggs, they
another serotype for which there is documented evidence that
are obtained from healthy flocks.
the strain willnotcause a 35 per cent arop in egg
2-2-2 Cell cultures production, the challenge must produce a drop in egg
If the vaccine is grown in ceH cultures, they comply with the production commensurate with the documented evidence
requirements for ceH cultures for production of veterinary and in any case not less than 15 per cent. The vaccine
vaccines (5.2.4).
2016 Veterinary Vaccines Vet-2 O9

complies with the test if egg production or quality is into the allantoic cavity of each of ten 9- to 11-day-old
significantly better in group C than in group D and embryonated hens' eggs from an SPF fiod: (5.2.2) 0.2 mL of
significantly better in group B than in group A. tlle pooled aHantoic liquid from the live embryos and into
2-5 MANUFACTURER'S TESTS each of 10 similar eggs 0.2 mL of the pooled liquid from the
dead erilbryos and incubate for 5-6 days. Examine for
2-5-1 Residuallive virus
abnormalities all embryos which die after 24 h of injection or
An amplification test for residuallive avían infectious
which survive 5-6 days. If more than 20 per cent of the
bronchitis virus is carried out on each batch of antigen
embryos die at either stage repeat the test from that stage.
immediately after inactivation and on tlle final bulk vaccine
The vaccine complies with the test if there is no death or
or, if the vaccine contains an adjuvant, on tlle bulk antigen or
abnormality attributable to the vaccine virus.
mixture of bulk antigens immediately before the addition of
adjuvant; the test is carried out in embryonated hen eggs B. For vaccine prepared with cell-culture-adapted strains of
from SPF fiocks (5.2.2) or in suitable cell cultures (5.2.4), virus, inoculate 10 doses of the vaccine into suitable cell
whichever is the most sensitive for the vaccine strain. cultures. If the vaccine contains an oil adjuvant, eliminate it
The quantity of inactivated virus harvest used in the test is by suitable means. Incubate at 38 ± 1 oC for 7 days. Make
equivalent to not les s than 10 doses of vaccine. The vaccine a passage on another set of cell cultures and incubate
complies with the test if no live virus is detected. at 38 ± 1 oC for 7 days. The vaccine complies with the test
if none of the cultures show signs of infection.
2-5-2 Batch potency test
It is not necessary to carry out the potency test (section 3-5) 3-4 Specified extraneous agents
for each batch of vaccine if it has been carried out using a Use 10 chickens, 14-28 days old, from an SPF fiod: (5.2.2).
batch of vaccine with a minimum potency. Where the test is Vaccinate each chicken by a recommended route with a
not carried out, an alternative validated method is used, the double dose of the vaccine. After 3 weeks, administer 1 dose
criteria for acceptance being set with reference to a batch of by the same route. Collect serum samples from each chicken
vaccine that has given satisfactory results in tlle test described 2 weeks later and carry out tests for antibodies against the
under Potency. The following test may be used. following agents by the metllods prescribed in general
chapter 5.2.2. Chicken flocks free fr0111 specified pathogens for the
Administer 1 dose of v:accine by the intramuscular route to
production and qualit)! control of vaccines: avían
each of not fewer than 10 chickens, between 2 weeks of age
encephalomyelitis virus, avían leucosis virus es, egg-drop
and the minimum age stated for vaccination and from an
syndrome virus, avían infectious bursal disease virus, avían
SPF fiock (5.2.2), and maintain 5 hatch mates as
infectious laryngotracheitis virus, influenza A virus, Marek's
unvaccinated controls. Collect serum samples from each
disease virus, N ewcastle disease virus.
chicken just before administration of the vaccine and after
the period defined when testing the reference vaccine; The vaccine complies with the test if it does not stimulate tlle
determine the antibody titre of each serum, for each serotype formation of antibodies against these agents.
in the vaccine, by a suitable serological method, for example, 3-5 Potency
serum neutralisation. The test is invalid unless the sera The vaccine complies with the requirements of the test
collected from the unvaccinated controls and from tlle mentioned under Immunogenicity (section 2-4-2) when
chickens just before the administration of the vaccine are free administered by a recommended route and method.
from detectable specific antibody. The vaccine complies with
4 LABELLING
the test if tlle antibody levels are not significantly less than
The label states whether the strain in the vaccine is embryo-
those obtained with a batch that has given satisfactory results
adapted or cell-culture-adapted.
in the test described under Potency.
_____________________________________________ PhE~

3 BATCH TESTS
3-1 Identification
When injected into chickens that do not have antibodies
against each of the virus serotypes in the vaccine, the vaccine
Avian Infectious Bronchitis ***
*** ***
stimulates the production of such antibodies, detectable by
virus neutralisation.
" Vaccine, Living ***
3-2 Bacteria and fungi
(Avian Infectious Bmnchitis Vaccine (Live)J
The vaccine, including where applicable the diluent supplied
Ph Bur monograph 0442)
for reconstitution, complies with tlle test for sterility
~E~ _____________________________________________
prescribed in the monograph Vaccines for veterinaJY
use (0062). 1 DEFINITION
3-3 Residuallive virus Avian infectious bronchitis vaccine (live) is a preparation of
A test for residual live virus is carried out to confirm one or more suitable strains of different types of avían
inactivation of avian infectious bronchitis virus. infectious bronchitis virus. This monograph applies to
A. For vaccine prepared Witll embryo-adapted strains of vaccines intended for administration to chickens for active
virus, inject 2/5 of adose into the allantoic cavity of ten 9- to immunisation against respiratory disease caused by avían
11-day-old embryonated hens' eggs from an SPF fiock infectious bronchitis virus.
(5.2.2) and incubate. Observe for 5-6 days and pool 2 PRODUCTION
separately the allantoic liquid from eggs containing live 2-1 PREPARATION OF THE VACClNE
embryos and that from eggs containing dead embryos, The vaccine virus is grown in embryonated hens' eggs or in
excluding those that die within the first 24 h after injection. cell cultures.
Examine for abnormalities aH embryos which die after 24 h
of injection or which survive 5-6 days. No death or
abnormality attributable to the vaccine virus occurs. Inject
Vet-210 Veterinary Vaccines 2016

2-2 SUBSTRATE FOR VIRUS PROPAGATION The following test may be carried out: use not fewer than
2-2-1 Embryonated hens' eggs 40 female chickens from an SPF flock (5.2.2) that are not
If the vaccine virus is grown in embryonated hens' eggs, they older than the minimum age to be recommended for
are obtained from flocks free from specified pathogens (SPF) vaccination; use the vaccine virus at the least attenuated
(5.2.2). passage level that will be present between the master seed lot
2-2-2 Cell cultures and a batch of the vaccine; administer to each chicken by a
If the vaccine virus is grown in cell cultures, they comply route to be recommended a quantity of virus equivalent to
with the requirements for cell cultures for production of not less than the maximum titre likely to be present in 1 dose
veterinary vaccines (5.2.4). of vaccine; at least 10 weeks after administration of the
vaccine virus, euthanise the chickens and carry out a
2-3 SEED LOTS macroscopic examination of the oviducts. The vaccine virus
2-3-1 Extraneous agents complies with the test if abnOlTIlalities are present in not
The master seed lot complies with the tests for extraneous more than 5 per cent of the oviducts.
agents in seed lots (2.6.24). In these tests on the master seed
2-4-2 Increase in virulence
lot, the organisms used are not more that 5 passages from
Carry out the test according to general chapter 5.2.6 using
the master seed lot at the start of the test.
2-week-old SPF chickens (5.2.2). If the properties of the
2-4 CHOICE OF VACCINE VIRUS vaccine virus allow sequential passage through 5 groups via
The vaccine virus shall be shown to be satisfactory with natural spreading, this method may be used, otherwise,
respect to safety (5.2.6) and efficacy (5.2.7) for the chickens passage as described below is carried out.
for which it is intended. Administer to each chicken of the 1st group by eye-drop a
The following tests for safety (section 2-4-1), increase in quantity of the vaccine virus that will allow recovery of virus
virulence (section 2-4-2) and immunogenicity (section 2-4-3) for the passages described below. 2-4 days after
may be used during the demonstration of safety and efficacy. administration of the vaccine virus, prepare a suspension
2-4-1 Safety from the mucosa of the trachea of each chicken and pool
2-4-1-1 Safety for the respirat01y tract and kidneys. Carry out these samples. Administer 0.05 mL of the pooled samples by
the test in chickens not older than the minimum age to be eye-drop to each chicken of the next group. Carry out this
recommended for vaccination. Use vaccine virus at the least passage operation not fewer than 4 times; verify the presence
attenuated passage level that will be present between the of the virus at each passage. If the virus is not found at a
master seed lot and a batch of the vaccine. passage level, repeat the passage by administration to a group
of 10 chickens. Carry out the test for safety for the
U se not fewer than 15 chickens of the same origin and from
respiratory tract and kidneys (section 2-4-1-1) and, where
an SPF flock (5.2.2). Administer to each chicken by the
applicable, the test for safety for the reproductive tract
oculonasal route a quantity of the vaccine virus equivalent to
(section 2-4-1-2) using the matelial used for the 1st passage
not less than 10 times the maximum virus titre likely to be
and the virus at the final passage level. Administer the virus
contained in 1 dose of the vaccine. On each of days 5, 7
by the route to be recommended for vaccination that is likely
and 10 after administration of the virus, euthanise not fewer
to be the least safe.
than 5 of the chickens and take samples of trachea and
kidney. Fix kidney samples for histological examination. The vaccine virus complies with the test if no indication of
Remove the tracheas and prepare 3 transverse sections from an increase in virulence of the virus recovered for the final
the upper part, 4 from the middle part and 3 from the lower passage compared with the material used for the 1sr passage
part of the trachea of each chicken; examine all tracheal is observed. If virus is not recovered after an initial passage in
explants as soon as possible and at the latest 2 h after 5 animals and a subsequent repeat passage in 10 animals, the
sampling by low-magnification microscopy for ciliary activity. vaccine virus also complies with the test.
S core for ciliostasis on a scale from O (100 per cent ciliary 2-4-3 Immunogenicity
activity) to 4 (no activity, complete ciliostasis); calculate the Immunogenicity is demonstrated for each strain of virus to
mean ciliostasis score (the maximum for each trachea be inc1uded in the vaccine. A test is carried out for each
being 40) for the 5 chickens euthanised on each of days 5, 7 route and method of administration to be recommended
and 10. using in each case chickens from an SPF flock (5.2.2) that
The test is not valid if more than 10 per cent of the chickens are not older than the· minimum age to be recommended for
die from causes not attributable to the vaccine virus. vaccination. The quantity of the vaccine virus administered
to each chicken is not greater than the minimum virus titre
The vaccine virus complies with the test if:
to be stated on the label and the virus is at the most
- no chicken shows notable clinical signs of avian infectious
attenuated passage level that will be present in a batch of the
bronchitis or dies from causes attributable to the vaccine
vaccine.
virus;
- any inflammatory lesions seen during the kidney Either or both of the tests below may be used during the
histological examination are, at most, moderate. demonstration of immunogenicity.
A risk/benefit analysis is carried out, taking into account the 2-4-3-1 Cilia/y activity of tracheal explams. U se not fewer than
average ciliostasis scores obtained and the benefits expected 25 chickens of the same origin' and from an SPF flock
from the use of the vaccine. (5.2.2). Vaccinate by a route to berecommended not fewer
than 20 chickens. Maintain not fewer than 5 chickens as
2-4-1-2 S afety for the I:eproductive tracto If the
controls. Challertge each chicken after 21 days by eye-drop
recommendations for use state or imply that the vaccine may
with a sufficient quantity of virulent avian imectious
be used in females less than 3 weeks old that are
bronchitis virus of the same type as the vaccine virus to be
subsequently kept to sexual maturity, it shall be
tested. Euthanise the chickens 4-7 days after challenge and
demonstrated that there is no damage to the development of
prepare 3 transverse sections from the upper part, 4 from the
the reproductive tract when the vaccine is given to chickens
middle part, and 3 from the lower part of the trachea of each
of the minimum age to be recommended for vaccination.
2016 Veterinary Vaccines Vet-211

chicken. Examine all tracheal explants as soon as possible into which it is inoculated, whereas after further admixture
and at the latest 2 h after sampling by low-magnification with type-specific antiserum against the strain to be identified
microscopy for ciliary activity. For a given tracheal section, it no longer produces such infection.
ciliary activity is considered as normal when at least 3-2 Bacteria and fungi
50 per cent of the intemal ring shows vigorous ciliary Vaccines intended for administration by injection comply
movement. A chicken is considered not affected if not fewer with the test for sterility prescribed in the monograph
than 9 out of 10 rings show normal ciliary activity. Vaccines for veterinaJY use (0062).
The test is not valid if: Frozen or freeze-dried vaccines produced in embryonated
-- fewer than 80 per cent of the control chickens show eggs and not intended for administration by injection comply
cessation or extreme loss of vigour of ciliary activity; eithet with the test for sterility prescribed in the monograph
-- and/or during the period between the vaccination and Vaccines for veteri12aJY use (0062) or with the following test:
challenge, more than 10 per cent of vaccinated or control carry out a quantitative test for bacterial and fungal
chickens show abnormal clinical signs or die from causes contamination; carry out identification tests for micro-
not attributable to the vaccine. organisms detected in the vaccine; the vaccine does not
The vaccine virus complies with the test if not fewer than contain pathogenic micro-organisms and contains not more
80 per cent of the vaccinated chickens show normal ciliary than 1 non-pathogenic micro-organism per dose.
activity. Any diluent supplied for reconstitution of the vaccine
2-4-3-2 Virus recove¡y fmm tracheal szuabs. Use not fewer than complies with the test for sterility prescribed in the
30 chickens of the same origin and from an SPF flock monograph Vaccines for veteri12aJY use (0062).
(5.2.2). Vaccinate by a route to be recommended not fewer 3-3 Mycoplasmas
than 20 chickens. Maintain not fewer than 10 chickens as The vaccine complies with the test for mycoplasmas (2.6.7).
controls. Challenge each chicken after 21 days by eye-drop
with a sufficient quantity of virulent avian infectious 3-4 Extraneous agents
bronchitis virus of the same type as the vaccine virus to be The vaccine complies with the tests for extraneous agents in
tested. Euthanise the chickens 4-7 days after challenge and batches of finished product (2.6.25).
prepare a suspension from swabs of the tracheal mucosa of 3-5 Virus titre
each chicken. Inoculate 0.2 mL of the suspension into the Titrate the vaccine virus by inoculation into embryonated
allantoic cavity of each of 5 embryonated hens' eggs, hens' eggs from an SPF flock (5.2.2) or into suitable cell
9-11 days old, from an SPF flock (5.2.2). Incubate the eggs cultures (5.2.4). If the vaccine contains more than 1 strain of
for 6-8 days after inoculation. Eggs that after 1 day of virus, titrate each strain after having neutralised the others
incubation do not contain a live embryo are eliminated and with type-specific avían infectious bronchitis antisera.
considered as non-specific deaths. Record the other eggs The vaccine complies with the test if 1 dose contains for each
containing a dead embryo and after 6-8 days' incubation vaccine virus not less than the minimum titre stated on the
examine each egg containing a live embryo for lesions label.
characteristic of avian infectious bronchitis. Make 3-6 Potency
successively 3 such passages. If 1 embryo of a series of eggs The vaccine complies with the requirements of 1 of the tests
dies or shows characteristic lesions, the inoculum is prescribed under Irnmunogenicity (section 2-4-3) when
considered to be a carrier of avian infectious bronchitis virus. administered according to the recommended schedule by a
The examination of a series of eggs is considered to be recommended route and method. It is not necessary to carry
definitely negative if no inoculum concemed is a carrier. out the potency test for each batch of the vaccine if it has
The test is not valid if: been carried out on a representative batch using a vaccinating
-- the challenge virus is re-isolated from fewer than dose containing not more than the minimum virus titre
80 per cent of the control chickens; stated on the label.
-- and/or during the period between vaccination and _____________________________________________ PhE~

challenge, more than 10 per cent of the vaccinated or


control chickens show abnormal clinical signs or die from
causes not attributable to the vaccine;
-- and/or more than 1 egg in any group is eliminated
because of non-specific embryo death. Avian Paramyxovirus 3 Vaccine
The vaccine virus complies with the test if the challenge virus
is re-isolated from not more than 20 per cent of the
tor Turkeys, Inactivated
(A vian Paramyxovirus 3- Vaccine (Inactivated) for
vaccinated chickens.
Turkeys) Ph Eur monograph 1392)
3 BATCH TESTS PhE~ _____________________________________________
3-1 Identification
3-1-1 Vaccines containing one type of virus. The vaccine, 1 DEFINITION
diluted if necessary and mixed with avian infectious Avian paramyxovirus 3 vaccine (inactivated) for turkeys is a
bronchitis virus antiserum specific for the virus type, no preparation of a suítable strain of avian paramyxovirus 3,
longer infects embryonated hens' eggs from an SPF flock inactivated while maintaining adequate irnmunogenic
(5.2.2) or susceptible cell cultures (5.2.4) into which it is properties. This monograph applies to vaccines intended for
inoculated. . protection of turkeys against a drop in egg production and
3-1-2 Vaccines containing more than one type ofvirus. loss of egg quality.
The vaccine, diluted if necessary and mixed with type- 2 PRODUCTION
specific antisera against each strain present in the vaccine 2-1 PREPARATION OF THE VACCINE
except that to be identified, infects embryonated hens' eggs The vaccine virus is propagated in embryonated eggs or in
from an SPF flock (5.2.2) or susceptible cell cultures (5.2.4) cell cultures. The vaccine may be adjuvanted.
Vet-212 Veterinary Vaccines 2016

2-2 SUBSTRATE FOR VIRUS PROPAGATION 2-5 MANUFACTURER'S TESTS


2-2-1 Embryonated eggs 2-5-1 Residuallive virus
If the vaccine virus is grown in emblyonated eggs, they are The test for residuallive virus is carried out in emblyonated
obtained from healthy flocks. eggs or suitable cell cultures (5.2.4), whichever is the most
2-2-2 Cell cultures sensitive for the vaccine strain. The quantity of inactivated
If the vaccine virus is grown in cell cultures, they comply virus harvest used in the test is equivalent to not less than
with the requirements for cell cultures for production of 10 dos es of vaccine. The vaccine complies with the test if no
veterinary vaccines (5.2.4). live virus is detected.

2-3 SEED LOTS 2-5-2 Batch potency test


It is not necessary to cany out the potency test (section 3-5)
2-3-1 Extraneous agents
for each batch of vaccine if it has been carried out using a
The master seed lot complies with the tests for extraneous
batch of vaccine with a minimum potency. Where the test is
agents in seed lots (2.6.24). In these tests on the master seed
not carried out, an alternative validated method is used, the
lot, the organisms used are not more than 5 passages from
criteria for acceptance being set with reference to a batch of
the master seed lot at the start of the test.
vaccine that has given satisfactory results in the test described
2-4 CHOICE OF VACCINE COMPOSITION under Potency.
The vaccine is shown to be satisfactory with respect to safety
3 BATCH TESTS
(5.2.6) and efficacy (5.2.7) for each category of turkeys for
3-1 Identification
which it is intended.
When injected into animal s that do not have antibodies
The following tests for safety (section 2-4-1) and against avian paramyxovirus 3, the vaccine stimulates the
irnmunogenicity (section 2-4-2) may be used during the production of such antibodies.
demonstration of safety and efficacy.
3-2 Bacteria and fungi
2-4-1 Safety The vaccine, including where applicable the diluent supplied
The test is carried out for each route of administration to be for reconstitution, complies with the test for sterility
recommended for vaccination. Use a batch of vaccine prescribed in the monograph Vaccines for veterinalY
containing not less than the maximum potency that may be use (0062).
expected in a batch of vaccine.
3-3 Residuallive virus
For each test, use not fewer than 8 turkeys not older than the
A test for residuallive virus is carried out to confirm
minimum age to be recommended for vaccination, that have
inactivation of avian paramyxovirus 3.
not been vaccinated and that do not have antibodies against
avian paramyxovirus 3. Administer by a recommended route Inject 2/5 of adose into the allantoic cavity of each of not
and method to each turkey 1 dose of the vaccine. If the fewer than 10 embryonated hen eggs 9-11 days old, from
schedule to be recommended requires a 2nd dose, administer flocks free from specified pathogens (SPF) (5.2.2) and
1 dose to each turkey after an interval of at least 14 days. incubate. Observe for 6 days and pool separately the allantoic
Observe the turkeys at least dai1y for at least 14 days after the fluid from eggs containing live emblyos, and that from eggs
last administration of the vaccine. containing dead emblyos, excluding those dying within 24 h
of the injection. Examine embryos that die within 24 h of
The test is not valid if non-specific mortality occurs.
injection for the presence of avian paramyxovirus 3.
The vaccine complies with the test if no turkey shows
abnormal signs of disease 01' dies from causes attributable to The vaccine does not comply with the test if avian
the vaccine. paramyxovirus 3 is found.
Inject into the allantoic cavity of each of not fewer than ten
2-4-2 Irnmunogenicity
9- to 11-day-old fertilised hen eggs from an SPF flock
A test is carried out for each route and method of
(5.2.2), 0.2 mL of the pooled allantoic fluid from the live
administration to be recommended, using in each case
embryos and, into each of 10 similar eggs, 0.2 mL ofthe
turkeys of the minimum age to be recommended for
pooled fluid from the dead embryos, and incubate
vaccination. The vaccine administered to each turkey is of
for 5-6 days. Test the allantoic fluid from each egg for the
minimum potency.
presence of haemagglutinins using chicken elythrocytes.
Use for the test 2 groups each of not fewer than 20 turkeys
The vaccine complies with the test if there is no evidenée of
of the same origin and of the same age, that do not have
haemagglutinating activity and if not more than 20 per cent
antibodies against avian paramyxovirus 3. Vaccinate
of the embryos die at either stage. If more than 20 per cent
one group in accordance with the recommendations stated
of the emblyos die at one of the stages, repeat that stage;
on the label. Maintain the other group as controls.
the vaccine complies with the test if there is no evidence of
The test is not valid if serological tests carried out on serum haemagglutinating activity and not more than 20 per cent of
samples obtained at the time of first vaccination show the the embryos die at that stage.
presence of antibodies against avian paramyxovirus 3 in
Antibiotics may be used in the test to control extraneous
either vaccinates or control s 01' if tests carried out at the time
bacterial infection.
of challenge show such antibodies in controls.
At the egg-production peak, challenge the 2 groups by the 3-4 Specified extraneous ag~nts
oculonasal route with a sufficient quantity of a virulent strain Use 10 chickens, 14-28 days old, from an SPF flod:: (5.2.2).
of avian paramyxovirus 3. For not less than 6 weeks after Vaccinate each chicleen by a recommended route with a
challenge, record the number of eggs laid weekly for each double dose of th'e vaccine. After 3 weeks, administer 1 dose
group, distinguishing between normal and abnormal eggs. by the same route .. Collect serum samples from each chicken
The vaccine complies with the test if egg production and 2 weeks later and carry out tests for antibodies against the
quality are significantly better in the vaccinated group than in following agents by the methods prescribed in general
the control group. chapter 5.2.2. Chicken flocks free from specified pathogens for the
production and quality control of vaccines: avian
2016 Veterinary Vaccines Vet-213

encephalomyelitis virus, avian infectious bronchitis virus, observation period (as a basis for comparison in the test for
avian leucosis viruses, egg-drop syndrome virus, avian increase in virulence).
infectious bursal disease virus, avian infectious The test is not valid if more than 10 per cent of the chickens
laryngotracheitis virus, influenza A virus, Marek' s disease younger than 3 weeks of age show abnormal signs of disease
VIrus. or die from causes not attributable to the vaccine virus.
The vaccine complies with the test if it does not stimulate the For chickens older than 3 weeks of age, the test is not val id if
formation of antibodies against these agents. non-specific mortality occurs.
3-5 Potency The vaccine virus complies with the test if no chicken shows
The vaccine complies with the requirements of the test abnormal signs of disease or die s from causes attributable to
mentioned under Immunogenicity (section 2-4-2) when the vaccine.
administered by a recommended route and method. 2-4-2 Increase in virulence
_____________________________________________ PhEw Can-y out the test according to general chapter 5.2.6 using
1-day-old chickens from an SPF fiock (5.2.2). Ifthe
properties of the vaccine virus allow sequential passage
through 5 groups via natural spreading, this method may be
used, otherwise passage as described below is carried out.
Avian Viral Tenosynovitis Vaccine ***** Administer to each chicken of the 1st group by a suitable
** **
(Live) *** route a quantity of the vaccine virus that will allow recovery
(Ph Bur monograph 1956) of virus for the passages described below. Euthanise the
_____________________________________________ ~Ew
chickens at the moment when the virus concentration in the
most suitable material (for example, tendons, tendon sheaths
1 DEFINITION and liquid exudates from the hock joints, spleen) is sufficient.
Avian viral tenosynovitis vaccine (live) is a preparation of a Prepare a suspension from this material from each chicken
suitable strain of avian tenosynovitis virus (avian and pool these samples. Administer 0.1 mL of the pooled
orthoreovirus). This monograph applies to vaccines intended samples by the route of administration most likely to lead to
for administration to chickens for active immunisation. increase in virulence to each chicken of the next group. Carry
out this passage operation not fewer than 4 times; verify the
2 PRODUCTION
presence of the virus at each passage. If the virus is not
2-1 PREPARATION OF THE VACCINE found at a passage level, repeat the passage by administration
The vaccine virus is grown in cell cultures. to a group of 10 chickens.
2-2 SUBSTRATE FOR VIRUS PROPAGATION If the 5th group of chickens shows no evidence of an increase
2-2-1 Cell cultures in virulence indicative of reversion dming the observation
Cell cultures comply with the requirements for cell cultures period, further testing is not required. Otherwise, carry out
for production of veterinary vaccines (5.2.4). an additional safety test and compare the clinical signs and
2-3 SEED LOTS any relevant parameters in a group of at least 10 chickens
2-3-1 Extraneous agents receiving the material used for the 1st passage and another
The master seed lot complies with the tests for extraneous similar group receiving the virus at the final passage leve!.
agents in seed lots (2.6.24). In these tests on the master seed The vaccine virus complies with the test if no indication of
lot, the organisms used are not more than 5 passages from an increase in virulence of the virus at the final passage level
the master seed lot at the start of the tests. compared with the material used for the 1st passage is
observed. If the virus is not recovered after an initial passage
2-4 CHOICE OF VACCINE VIRUS in 5 chickens and a subsequent repeat passage in
The vaccine virus shall be shown to be satisfactory with 10 chickens, the vaccine virus also complies with the test.
respect to safety (5.2.6) and efficacy (5.2.7) for the chickens
for which it is intended. 2-4-3 Immunogenicity
A test is canied out for each route and medl0d of
The following tests for safety (section 2-4-1), increase in administration to be recommended for vaccination using in_
virulence (section 2-4-2) and immunogenicity (section 2-4-3) each case chickens not older than the minimum age to be
may be used during the demonsu"ation of safety and efficacy. recommended for vaccination. The quantity of the vaccine
2-4-1 Safety virus administered to each chicken is not greater than the
Carry out the test for each route and method of minimum virus titre to be stated on the label and the virus is
administration to be recommended for vaccination using in at the most attenuated passage level that will be present in a
each case chickens not older than the minimum age to be batch of the vaccine. Use not fewer than 30 chickens of the
recommended for vaccination and from a fiod: free from same origin and from an SPF fiock (5.2.2). Administer the
specified pathogens (SPF) (5.2.2). Use vaccine virus at the vaccine by a route to be recommended to not fewer than
least attenuated passage level that will be present in a batch 20 chickens. Maintain not fewer than 10 chickens as
of the vaccin~. controls. Challenge each chicken after 21 days by a suitable
For each test performed in chickens younger than 3 weeks of route witha sufficient quantity of virulent avian tenosynovitis
age, use not fewer than 10 chickens. For each test performed virus. Observe the chickens at least daily for 21 days after
in chickens older than 3 weeks of age, use not fewer than challenge. Record the deaths and the surviving chickens that
8 chickens. Administer to each chicken a quantity of the show dinical signs of disease. If the challenge is administered
vaccine virus equivalent to not less than 10 times the by the foot pad, any transient swelling of the foot pad during
maximum virus titre li1zely to be contained in 1 dose of the the first 5 days after challenge may be considered non-
vaccine. Observe the chickens at least dai1y for at least specific. At the end of the observation period, euthanise all
21 days. Carry out histological examination of the joints and the surviving chickens and carry out macroscopic and/or
tendon sheaths of the legs and feet at the end of the
V et- 214 Veterinary Vaccines 2016

microscopic examination for lesions of the joints and tendon


Bordetella Bronchiseptica Vaccine *****
sheaths of the legs and feet, e.g. exudate and swelling. ** **
The test is not valid if: (live) for 009S ***
- during the observation period after challenge fewer than (Ph. Eur. 1110nograph 2525)
80 per cent of the control chickens die or show severe PhE~ __________________ ~ _ ____
clinical signs of avian viral tenosynovitis or show
macroscopical and/or microscopicallesions in the joints 1 DEFINITION
and tendon sheaths of the legs and feet, Bordetella bronchiseptica Vaccine (live) for dogs is a
- or if during the period between vaccination and challenge preparation of a suitable strain of Bordetella bronchiseptica.
more than 10 per cent of the control or vaccinated This monograph applies to vaccines intended for the active
chickens show abnormal clinical signs or die from causes immunisation of dogs against respiratory disease caused by
not attributable to the vaccine. B. bronchiseptica.
The vaccine virus complies with the test if during the 2 PRODUCTION
observation period after challenge not fewer than 90 per cent 2-1 PREPARATION OF THE VACCINE
of the vaccinated chickens survive and show no notable The vaccine strain is grown in a suitable medium.
clinical signs of disease or show macroscopical and/or
2-2 CHOICE OF VACCINE COMPOSITION
microscopicallesions in the joints and tendon sheaths of the
legs and feet. The vaccine strain is shown to be satisfactory with respect to
safety (5.2.6) and efficacy (5.2.7) for the dogs for which it is
3 BATCH TESTS intended.
3-1 Identification The following tests for safety (section 2-2-1), excretion and
Carry out an irnmunostaining test in cell cultures to identify transmission of the vaccine strain (section 2-2-2), increase in
the vaccine virus. virulence (section 2-2-3) and irnmunogenicity (section 2-2-4)
3-2 Bacteria and fungi may be used during the demonstration of safety and efficacy.
Vaccines intended for administration by injection comply 2-2-1 Safety
with the test for sterility prescribed in the monograph Carry out the test for each route and method of
Vaccines for veterina1Y use (0062). administration to be recommended for vaccination using in
Frozen or freeze-dried vaccines produced in embryonated each case dogs not older than the minimum age to be
eggs and not intended for administration by injection either recommended for vaccination. The vaccine strain to be
comply with the test for sterility prescribed in the monograph administered is at the least attenuated passage level that will
Vaccines for veterina1Y use (0062) or with the following test: be present in a batch of the vaccine.
carry out a quantitative test for bacterial and fungal For each test, use not fewer than 8 dogs, shown to be free
contamination; carry out identification tests for from B. bronchiseptica and that do not have antibodies against
microorganisms detected in the vaccine; the vaccine does not B. bronchiseptica. Administer to each dog a quantity of the
contain pathogenic microorganisms and contains not more vaccine strain equivalent to not less than 10 times d1e
than 1 non-pathogenic microorganism per dose. maximum number of live bacteria lil<::ely to be contained in
Any diluent supplied for reconstitution of the vaccine 1 dose of the vaccine. Observe the dogs at least daily for at
complies with the test for sterility prescribed in the least 14 days.
monograph Vaccines for veterina1Y use (0062). The vaccine strain complies with the test if no dog shows
3-3 Mycoplasmas abnormal local or systemic reactions 01' dies from causes
The vaccine complies with the test for mycoplasmas (2.6.7). attributable to the vaccine strain.
3-4 Extraneous agents 2-2-2 Excretion and transmission of the vaccine strain
The vaccine complies with the tests for extraneous agents in Use dogs not older than 10 weeks of age. Administer the
batches of finished product (2.6.25). strain by the route to be recommended for vaccination most
3-5 Virus titre likely to lead to excretion. The vaccine strain to be
Titrate the vaccine virus by inoculation into suitable cell administered is at the least attenuated passage level that will
cultures (5.2.4). The vaccine complies with the test if 1 dos e be present in a batch of the vaccine.
contains not les s than the minimum virus titre stated on the For each test, use not fewer than 8 dogs, shown to be free
label. from B. bronchiseptica and that do not have antibodies against
3-6 Potency B. bronchiseptica. Administer to not fewer than 4 dogs a
The vaccine complies with the requirements of the test quantity of the vaccine st:rain equivalent to not les s than the
prescribed under Irnmunogenicity (section 2-4-3) when maximum number of live bacteria likely to be contained in
administered by a recommended route and method. It is not 1 dose of the vaccine. 2 days after vaccination add 4 dogs to
neceSSa1y to carry out the potency test for each batch of the the group of vaccinated dogs. Observe the animals for
vaccine if it has been carried out on a representative batch 70 days. Collect nasal swabs 01' washings from each dog at
using a vaccinating dose containing not more than the weekly intervals. Verify the presence of the excreted vaccine
minimum virus titre stated on the label. strain with a suitable method ..
_ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ Ph Eur The vaccine strain complies with the test if no dog shows
abnormal local 01' systemic reactions or dies from causes
attributable to die vaccine strain.
The results are noted and used to formulate the label
statement (whether the vaccinated strain is excreted, the
period over which there is excretion and whether or not the
vaccine strain spreads to in-contact dogs).
2016 Veterinary Vaccines Vet-215

2-2-3 Increase in virulence 3 BATCH TESTS


Carry out the test according to general chapter 5.2.6 using 3-1 Identification
dogs not older than 10 weeks of age, which are free from The vaccine strain is identified by suitable methods.
B. bronchiseptica and that do not have antibodies against 3-2 Bacteria and fungi
B. bronchiseptica. If the properties of the vaccine strain allow Carry out the test by inoculation of suitable media.
sequential passage through 5 groups via natural spreading, The vaccine complies with the test if it does not contain
this method may be used, otherwise passage as described extraneous micro-organisms. Any diluent supplied for
below is carried out. reconstitution of the vaccine complies with the test for
Administer to each dog by a route to be recommended a sterility prescribed in the monograph Vaccines for veterinary
quantity of the vaccine strain that will allow recovery of use (0062).
bacteria for the passages described below. Administer the
3-3 Live bacteria
strain by the route to be recommended for vaccination most
Make a count of live bacteria on a solid medium suitable for
likely to lead to reversion to virulence. On one occasion
the culture of B. bronchiseptica. The vaccine complies with
between 4 and 6 days after administration, collect nasal
the test if 1 dos e contains not less than the minimum
swabs or washings from each dog, verify the presence of
number of live B. bronchiseptica stated on the label.
bacteria and pool positive samples. Administer 1 mL of the
pooled samples by a suitable route (for example, the 3-4 Potency
intranasal route) to each dog of the next group. Carry out The vaccine complies with the requirements of the test
this passage operation not fewer than 4 times; verify the prescribed under Immunogenicity (section 2-2-4) when
presence of the bacteria at each passage. If the bacteria are administered by a recommended route and method. It is not
not found at a passage level, repeat the passage by necessary to carry out the potency test for each batch of the
administration to a group of 10 animaIs: vaccine if it has been carried out on a representative batch
using a vaccinating dose containing not more than the
If the 5th group of animals shows no evidence of an increase
minimum number of live B. bronchiseptica stated on the label.
in virulence indicative of reversion during the observation
period, further testing is not required. Othelwise, carry out 4 LABELLING
an additional safety test and compare the clinical signs and The label states:
any relevant parameters in a group of at least 8 animals -- where applicable, the period after vaccination during
receiving the material used for the 1st passage and another which the vaccine is excreted;
similar group receiving the bacteria at the final passage level. -- where applicable, that the vaccine strain may be
The vaccine strain complies with the test if no indication of transmitted to other dogs.
increased virulence of the bacteria recovered for the final _____________________________________________ PhE~

passage compared with the material used for the 1st passage
is observed. If bacteria are not recovered after an initial
passage in 2 animals and a subsequent repeat passage in
10 animals, the vaccine strain also complies with the test.
Bovine Parainfluenza Virus ***
2-2-4 Immunogenicity *** ***
A test is carried out for each route and method of Vaccine, Living ***
administration to be recommended for vaccination using in (Bovine ParaiJ1fluenza Virus Vaccine (Live))
each case dogs of the minimum age to be recommended. Ph Bur monograph 1176)
The quantity of vaccine strain to be administered to each dog PhE~ _____________________________________________
is not greater than the minimum number of live bacteria to
be stated on the label and the strain is at the most attenuated 1 DEFINITION
passage level that will be present in a batch of vaccine. Eovine parainfluenza virus vaccine (live) is a preparation of a
U se for the test not fewer than 15 dogs which are free from suitable su"ain of bovine parainfluenza 3 virus. This
B. bronchiseptica and that do not have antibodies against monograph applies to vaccines intended for the active
B. bronchiseptica. Vaccinate not fewer than 10 dogs, according irnmunisation of cattle against infection with bovine
to the schedule to be recommended. Maintain not fewer than parainfluenza virus.
5 dogs as controls. Challenge each dog after 20-22 days by 2 PRODUCTION
the intranasal route with a quantity of a suspension of 2-1 PREPARATION OF THE VACCINE
virulent B. bronchiseptica sufficient to cause typical signs of The vaccine virus is grown in cell cultures.
respiratory disease in a dog that does not have antibodies
against B. bronchiseptica. Observe the dogs at least daily for 2-2 SUBSTRATE FOR VIRUS PROPAGATION
14 days after challenge. Collect nasal swabs or washings from 2-2-1 Cell cultures
each dog daily from day 2 to 14 after challenge and The cell cultures comply with the requirements for cell
determine the number of excreted B. bronchiseptica in each cultures for production of veterinary vaccines (5.2.4).
sample. U se a scoring system to record the signs of 2-3 CHOICE OF VACCINE VIRUS
respiratory disease in each dogo The vaccine virus is shown to be satisfactory Witl1 respect to
The test is invalid if more than 20 per cent of the controls safety (5.2.6) and efficacy (5.2.7) for the cattle for which it is
show no typical signs of the disease. intended.
The vaccine complies with the test if there is a significant The following tests for safety (section 2-3-1), increase in
decrease in the score for respiratory signs and in the number virulence (section 2-3-2) and irnmunogenicity (2-3-3) may be
of B. bronchiseptica excreted in vaccinates compared to used during the demonsu"atión of safety and efficacy.
controls. 2-3-1 Safety
Carry out the test for each route and method of
administration to be recommended for vaccination.
Vet-216 Veterinary Vaccines 2016

Use vaccine virus at the least attenuated passage leve! that demonstrated that valid results are obtained in these
will be present in a batch of the vaccine. conditions. Collect sera from the calves before vaccination,
For each test, use not fewer than 5 calves of the minimum 7 days and 14 days after the time of vaccination and just
age to be recommended for vaccination and preferably that before challenge. Vaccinate not fewer than 5 calves,
do not have antibodies against bovine parainfluenza 3 virus according to the schedule to be recommended. Maintain not
or, where justified, use calves with a very low leve! of such fewer than 5 calves as controls. Challenge each calf after
antibodies as long as they have not been vaccinated against 20-22 days by a respiratory tract route with a sufficient
bovine parainfluenza virus and administration of the vaccine quantity of a suspension of a low-passage virulent bovine
do es not cause an anamnestic response. Administer to each parainfluenza 3 virus. Observe the calves at least daily for
calf a quantity of the vaccine virus equivalent to not les s than 14 days after challenge and monitor each of them for signs,
10 times the maximum virus titre likely to be contained in in particular respiratOly signs and virus shedding (by nasal
1 dose of the vaccine. Observe the calves at least daily for at swabs or tracheobronchial washing).
least 14 days. Measure the body temperature of each calf on The test is not valid if tests for antibodies against bovine
the day before vaccination, at the time of vaccination and for parainfluenza 3 virus on the sera indicate that there was
the 4 subsequent days. intercurrent infection with the virus during the test or if,
The vaccine virus complíes with the test if no abnormal effect during the observation period after challenge, more than 2 of
on body temperature occurs and if no calf shows abnormal, the 5 control calves show no excretion of the challenge virus,
local or systemic reactions or dies from causes attributable to as shown by nasal swabs or samples harvested by
the vaccine virus. tracheobronchial washing.
2-3-2 Increase in virulence The vaccine virus complies with the test if, during the
Carry out the test according to chapter 5.2.6. Evaluation of observation period after challenge, in vaccinated calves
safety of veterinar.y vaccines and immunosem, using calves that compared to control s there is a significant reduction in mean
do not have antibodies against bovine parainfluenza 3 virus. titre and in mean duration of virus excretion, and a notable
If the properties of the vaccine virus allow sequential passage reduction in general and local signs (if the challenge virus
through 5 groups via natural spreading, this method may be used produces such signs).
used, otherwise passage as described below is carried out. 3 BATCH TESTS
Administer to each calf of the 1st group by the intranasal 3-1 Identification
route a quantity of the vaccine virus that will allow recovely Carry out an irnmunostaining test in suitable cell cultures,
of virus for the passages described below. On each of days using a monospecific antiserum.
3 to 7 after administration of the virus, take nasal swabs from 3-2 Bacteria and fungi
each calf and collect in not more than 5 mL of a suitable The vaccine, including where applicable the diluent supplied
medium, which is then used to inoculate cell cultures to for reconstitution, complies with the test for sterility
verify the presence of virus. Administer about 1 mL of the prescribed in the monograph Vaccines for veterúwIJ' use
suspension from the swabs that contain the maximum (0062).
amount of virus, as indicated by the titration of cell cultures,
3-3 Mycoplasmas (2.6.7)
to each calf of the next group. Cany out this passage
The vaccine complies with the test for mycoplasmas.
operation not fewer than 4 times; verify the presence of the
virus at each passage. If the virus is not found at a passage 3-4 Extraneous agents
leve!, repeat the passage by administration to a group of Neutralise the vaccine virus with a suitable monospecific
10 animals. antiserum against bovine parainfluenza 3 virus and inoculate
If the 5th group of animal s shows no evidence of an increase into cell cultures known for their susceptibility to virus es
in virulence indicative of reversion during the observation pathogenic for cattle. Carry out at least 1 passage and
period, further testing is not required. Otherwise, carry out maintain the cultures for 14 days.
an additional safety test and compare the clinical signs and The vaccine complies with the test if no cytopathic effect
any relevant parameters in a group of at least 8 animal s develops and there is no sign of the presence of
receiving the material used for the 1st passage and another haemadsorbing agents. Carry out a specific test for
similar group receiving the virus at the final passage. pestiviruses.
The vaccine virus complies with the test if no indication of 3-5 Virus titre
increased virulence of the virus recovered for the final Titrate the vaccine virus in suitable cell cultures. The vaccine
passage compared with the material used for the 1st passage complies with the test if 1 dose contains not less than the
is observed; account is taken of the titre of excreted virus in minimum virus titre stated on the labe!'
the nasal swabs. If virus is not recovered after an initial 3-6 Potency
passage in 2 animals and a subsequent repeated passage in The vaccine complíes with the requirements of the test'
10 animals, the vaccine virus also complies with the test. prescribed under Irnmunogenicity (section 2-3-3) when
2-3-3 Immunogenicity administered by a recommended route and method. It is not
A test is carried out for each route and method of necessaty to carry out the potency test for each batch of the
administration to be recommended for vaccination using in vaccine if it has been carriedout on a representative batch
each case calves of the minimum age to be recommended. using a vaccinating dose containing not more than the
The quantity of vaccine to be administered to each calf is not minimum virus titre stated on the labe!'
greater than the minimum virus titre to be stated on the label ________~-----------------------------------~Ew
and the virus is at the most attenuated passage leve! that will
be present in a batch of vaccine.
U se for the test not fewer than 10 calves that do not have
antibodies against bovine parainfluenza 3 virus; calves having
low levels of such antibodies may be used if it has been
2016 Veterinary Vaccines Vet-217

3 to 7 after administration of the virus, take nasal swabs from


Bovine Respiratory Syncytial Virus each calf and collect in not more than 5 mL of a suitable
Vaccine, living medium, which is then used to inoculate cell cultures to
(Bovine Respirato1)' Syncytial Virus Vaccine (Live) J verify the presence of virus. Administer about 1 mL of the
Ph Eur monograph 1177) suspension from the swabs that contain tlle maximum
PhE~ _____________________________________________ amount of virus, as indicated by the titratíon of cell cultures,
to each calf of the next group. Carry out this passage
1 DEFINITION operation not fewer than 4 times; verify the presence of tlle
Bovine respiratory syncytial virus vaccine Oive) is a virus at each passage. If the virus is not found at a passage
preparation of a suitable strain of bovine respiratory syncytial level, repeat the passage by administration to a group of
virus. This monograph applies to vaccines intended for the 10 animals.
active immunisation of cattle against infection with bovine If the 5 th group of calves shows no evidence of an increase in
respiratory syncytial virus. virulence indicative of reversion during the observation
2 PRODUCTION period, further testing is not required. Othelwise, cany out
2-1 PREPARATION OF THE VACCINE an additional safety test and compare the clinical signs and
The vaccine virus is grown in cell cultures. any relevant parameters in a group of at least 8 animals
receiving the material used for the 1sr passage and another
2-2 SUBSTRATE FOR VIRUS PROPAGATION similar group receiving the virus at the final passage level.
2-2-1 Cell cultures The vaccine virus complies with the test if no calf shows
The cell cultures comply with the requirements for cell
signs attributable to the vaccine virus and no indication of
cultures for production of veterinary vaccines (5.2.4).
increased virulence of the virus recovered for the final
2-3 CHOICE OF VACCINE VIRUS passage compared with the material used for the 1sr passage
The vaccine virus is shown to be satisfactory with respect to is observed; account is taken of the titre of excreted virus in
safety (5.2.6) and efficacy (5.2.7) for the cattle for which it is the nasal swabs. If virus is not recovered after an initial
intended. passage in 2 animals and a subsequent repeated passage in
The following tests for safety (section 2-3-1), increase in 10 animals, the vaccine virus also complies with the test.
virulence (section 2-3-2) and immunogenicity (section 2-3-3) 2-3-3 Irnmunogenicity
may be used during the demonstration of safety and efficacy. A test is canied out for each route and metllod of
2-3-1 Safety administration to be recommended for vaccination using in
Carry out the test for each route and method of each case calves of the minimum age to be recommended.
administration to be recommended for vaccination, using in The quantity of vaccine to be administered to each calf is not
each case calves of the minimum age to be recommended for greater than tlle minimum virus titre to be stated on the label
vaccination. Use vaccine virus at the least attenuated passage and the virus is at the most attenuated passage level that will
level tllat will be present in a batch of the vaccine. be present in a batch of vaccine.
2-3-1-1 LaboratOlY test For each test, use not fewer than U se for the test not fewer tllan 10 calves that do not have
5 calves that do not have antibodies against bovine antibodies against bovine respirat01Y syncytial virus. Collect
respiratory syncytial virus. Administer to each calf a quantity sera from the calves before the time of vaccination, 7 and
of the vaccine virus equivalent to not less than 10 times the 14 days after the time of vaccination and just before
maximum virus titre likely to be contained in 1 dose of tlle challenge. Vaccinate not fewer than 5 calves, according to tlle
vaccine. Observe the calves at least daily for at least 14 days. schedule to be recommended. Maintain not fewer tllan
Measure the body temperature of each calf on the day before 5 calves as controls. Challenge each calf after 20-22 days by
vaccination, at the time of vaccination and daily for the a respirat01y tract route with a sufficient quantity of a
following 7 days. suspension of a low-passage virulent bovine respiratory
syncytial virus. Observe tlle calves at least daily for 14 days
The vaccine virus complies with the test if no abnormal effect
after challenge and monitor each of them for signs, in
on body temperature occurs and if no calf shows abnormal
particular respiratory signs and virus shedding (by nasal
local or systemic reactions or dies from causes attributable to
swabs or tracheobronchial washing).
the vaccine virus.
The test is not valid if antibodies against bovine respiratory
2-3-1-2 Field studies The calves used for the field trials are
syncytial virus are detected in any sample from control calves
also used to evaluate the incidence of hypersensitivity
before challenge or if more than 2 of tlle 5 control calves
reactions in vaccinated calves following subsequent exposure
show no excretion of the challenge virus, as shown by nasal
to the vaccine or to wild virus. The vaccine complies with the
swabs or samples harvested by tracheobronchial washing.
test if it is not associated with an abnormal incidence of
immediate hypersensitivity reactions. The vaccine virus complies with tlle test if, dming the
observation period after challenge, there is a significant
2-3-2 Increase in virulence
reduction in mean titre and in mean duration of virus
Carry out the test according to chapter 5.2.6. Evaluation of
excretion in vaccinates compared to controls, and a notable
safety of veterinaJY vaccines and immunosera, using calves that
reduction in general and local signs in vaccinated calves (if
do not have antibodies against bovine respiratory syncytial
the challenge virus used produces such signs).
virus. If the properties of the vaccine virus allow sequential
passage through 5 groups via natural spreading, this method 3 BATCH TESTS
may be used, otherwise passage as described below is carried 3-1 Identification
out. Identify the vaccine by an inununostaining test in suitable
Administer to each calf of the 1sr group by the intranasal cell cultures using a monospecific antiserum.
route a quantity of the vaccine virus that will allow recovery
of virus for the passages described below. On each of days
Vet-218 Veterinary Vaccines 2016

3-2 Bacteria and fungi The following tests for safety (section 2-3-1) and
The vaccine, inc1uding where applicable the diluent supplied immunogenicity (section 2-3-2) may be used during the
for reconstitution, complies with the test for sterility demonstration of safety and efficacy.
prescribed in the monograph Vaccines for veterina1Y 2-3-1 Safety
use (0062). Carry out the test for each route and method of
3-3 Mycoplasmas (2.6.7) administration to be recommended for vaccination and in
The vaccine complies with the test for mycoplasmas. each categOly of cattle for which the vaccine is intended.
3-4 Extraneous agents U se a batch of vaccine containing not less than the
Neutralise the vaccine virus with a suitable monospecific maximum potency that may be expected in a batch of
antiserum against bovine respiratory syncytial virus and vaccine.
inoculate into cell cultures known for their susceptibility to 2-3-1-1 General safety. For each test, use not fewer than
viruses pathogenic for cattle. Carry out at least one passage 8 cattle of the minimum age to be recommended for
and maintain the cultures for 14 days. vaccination and that do not have bovine diarrhoea virus or
The vaccine complies with the test if no cytopathic effect antibodies against the virus. Administer to each animal
develops and there is no sign of the presence of 1 dose of tlle vaccine. If the schedule to be recommended
haemadsorbing agents. Carry out a specific test for requires a 2nd dose, administer another dose after an interval
pestiviruses. of at least 14 days. Observe the cattle at least daily for at
least 14 days.
3-5 Virus titre
Titrate the vaccine virus in suitable cell cultures. The vaccine The vaccine complies with the test if no animal shows
complies with the test if 1 dose contains not less than the abnormal local or systemic reactions or dies from causes
minimum virus titre stated on the label. attributable to the vaccine.
2-3-1-2 Safety in pregnant cattle. If the vaccine is intended for
3-6 Potency
use in pregnant cattle, use not fewer than 8 cattle at tlle
The vaccine complies with tlle requirements of the test
beginning of each semester for which use is not
prescribed under Immunogenicity (section 2-3-3) when
contraindicated. Administer to each animal 1 dose of the
administered by a recommended route and method. It is not
vaccine. If the schedule to be recommended requires a
neceSSaly to cany out the potency test for each batch of the
2 nd dose, administer another dose after an interval of at least
vaccine if it has been carried out on a representative batch
14 days. Observe the cattle at least daily until calving.
using a vaccinating dose containing not more than the
minimum virus titre stated on the label. The vaccine complies with the test if no animal shows
_____________________________________________ ~E~
abnOlmallocal or systemic reactions or dies from causes
attributable to the vaccine and if no adverse effects on
gestation or the offspring are noted.
2-3-1-3 Exa711ination of reproductive pe¡formance. If the vaccine
is intended for administration shortly before or at
Bovine Viral Diarrhoea Vaccine ***** insemination, absence of undesirable effects on conception
** ** rate must be demonstrated.
(Inactivated) *** 2-3-2 Irnmunogenicity
(Ph. Eur. monograph 1952)
The following test is suitable to demonstrate the
~E~ _____________________________________________
immunogenicity of the vaccine with respect to bovine
1 DEFINITION diarrhoea virus of genotype 1; if protection against bovine
Bovine viral diarrhoea vaccine (inactivated) is a preparation diarrhoea virus of genotype 2 is c1aimed, an additional test,
of one or more suitable strains of bovine dialThoea virus similar to that described below, but using bovine diarrhoea
inactivated while maintaining adequate immunogenic virus of genotype 2 for challenge, is carried out.
properties. This monograph applies to vaccines intended for A test is carried out for each route and metll0d of
the active immunisation of heifers and cows for protection of administration to be recommended. The vaccine
their progeny against transplacental infection. administered to each heifer is of minimum potency.
2 PRODUCTION Use for the test not fewer than 20 heifers free from bovine
2-1 PREPARATION OF THE VACCINE diarrhoea virus and that do not have antibodies against
bovine diarrhoea virus. Vaccinate not fewer than 13 heifers
The vaccine virus is grown in cell cultures. The viral
according to the schedule to be recommended. Maintain not
suspensions of each vaccine virus are harvested separately
fewer tllan 7 heifers as controls. Keep all the animals as one
and inactivated by a method that maintains immunogenicity.
group. Inseminate the heifers. Take a blood sample from
The viral suspensions may be purified and concentrated.
non-vaccinated heifers shortly before challenge. The test is
The vaccine may be adjuvanted.
discontinued if fewer than 10 vaccinated heifers 01' 5 non-
2-2 SUBSTRATE FOR VIRUS PROPAGATION vaccinated heifers are pregnant at the time ofchallenge.
2-2-1 Cell cultures Challenge each heifer between the 60 th and 90th days of
The cell cultures comply with the requirements for cell gestation. For both test models described (observation until
cultures for production of veterinalY vaccines (5.2.4). calving and harvest of foetuses at 28 days), challenge may be
2-3 CHOICE OF VACCINE COMPOSITION made by the intranasal route with a sufficient quantity of a
The vaccine is shown to be satisfactory with respect to non-cytopathic strain of bovine diarrhoea virus 01'
safety (5.2.6) and efficacy (5.2.7) for the cattle for which it is altematively, where the heifers are observed until calving,
intended. challenge may be made by contact with a persistently
viraemic animal. Observe the heifers clinically at least daily
from challenge either until the end of gestation or until
2016 Veterinary Vaccines Vet-219

harvest of foetuses after 28 days. If abortion occurs, examine 7 days and observe the 2nd culture for not less than 7 days.
the aborted foetus for bovine diarrhoea virus by suitable The vaccine complies with the test if no live virus is detected.
methods. If cattle are observed until calving, irnmediately If the vaccine contains an adjuvant, separate the adjuvant if
after birth and prior to ingestion of colostrum, examine all possible from the liquid phase by a method that does not
calves for viraemia and antibodies against bovine diarrhoea interfere with the detection of possible live virus.
virus. If foetuses are harvested 28 days after challenge, 3-4 Potency
examine the foetuses for bovine diarrhoea virus by suitable The vaccine complies with the requirements of the test
methods. Transplacental infection is considered to have prescribed under Irnmunogenicity (section 2-3-2) when
occurred if virus is detected in foetal organs or in the blood administered by a recommended route and method.
of newborn calves or if antibodies are detected in precolostral _____________________________________________ PhEw
sera of calves.
The test is not valid if any of the control heifers have
neutralising antibody before challenge or if transplacental
infection fails to occur in more than 10 per cent of the calves
from the control heifers. The vaccine complies with the test if Brucella Melitensis (Strain Rev. 1) *****
at least 90 per cent of the calves from the vaccinated heifers ** **
are protected from transplacental infection. Vaccine, Living ***
(Brucellosis VacGÍne (Live) (Brucella Melitensis Rev. 1
2-4 MANUFACTURER'S TESTS Strain)) for Veterina1Y Use) Ph. Eur. 1110nograph 0793)
2-4-1 Residuallive virus ~Ew _____________________________________________
The test for residual live virus is carried out using a quantity
of inactivated virus harvest equivalent to not less than 1 DEFINITION
25 dos es of vaccine in cells of the same type as those used Brucellosis vaccine (live) (Brucella melitensis Rev. 1 strain)
for production of the vaccine or cells shown to be at least as for veterinary use is a suspension of live Bl'ucella melitensis
sensitive; the cells are passaged after 7 days and observed for Rev. 1 strain. The vaccine contains not fewer than
a total of not les s than 14 days. The inactivated virus harvest 0.5 x 10 9 and not more than 4 x 10 9 live bacteria per dose.
complies with the test if no live virus is detected. This monograph applies to vaccines intended for the active
2-4-2 Batch potency test immunisation of sheep and goats against disease caused by
It is not necessary to carry out the potency test (section 3-4) B. melitensis.
for each batch of vaccine if it has been carried out using a 2 PRODUCTION
batch of vaccine with a minimum potency. Where the test is 2-1 PREPARATION OF THE VACCINE
not carried out, an alternative validated method is used, the B. melitensis Rev. 1 strain is cultured in a suitable medium.
criteria for acceptance being set with reference to a batch of The method of culture is such as to avoid bacterial
vaccine that has given satisfactory results in the test described dissociation and thus maintain tl1e smooth characteristic of
under Potency. The following test may be used. the culture. The bacteria are suspended in a buffer solution
Use for the test 7 suitable laboratory animals or calves that that may contain a suitable stabiliser. The suspension is
do not have antibodies against bovine diarrhoea virus. distributed into containers.
Administer by the subcutaneous route to 5 animals a suitable
2-2 CHOICE OF VACCINE STRAIN
dose of the vaccine. Maintain 2 animals as controls.
The vaccine strain is shown to be satisfactory with respect to
A 2nd dose of vaccine may be administered after a suitable
safety (5.2.6) and efficacy (5. 2.7) for the sheep and goats for
interval if this has been shown to provide a suitably
discriminating test system. Collect blood samples before the which it is intended.
1st vaccination and at a given interval between 14 and The following tests for safety (section 2-2-1), residual
21 days after the last vaccination. Determine the antibody virulence (section 2-2-2), determination of dissociation phase
titres against bovine diarrhoea virus by seroneutralisation on of master seed lot (section 2-2-3) and irnmunogenicity in
suitable cell cultures. mice (section 2-2-4) may be used during the demonstration
of safety and efficacy.
The test is not valid if the control animal s show antibodies
against bovine diarrhoea virus. The vaccine complies with the 2-2-1 Safety
test if the level of antibodies in the vaccinates is not lower U se 8 sheep, 4-6 months old, that do not have antibodies
than that found for a batch of vaccine that has given against B. melitensis. Administer to each sheep by a route to
satisfactory results in the test described under Potency. be recommended 3 doses of the vaccine. Observe the sheep
at least daily for at least 14 days.
3 BATCH TESTS
3-1 Identification The vaccine complies with the test if no sheep shows notable
When administered to animal s tl1at do not have specific signs of disease or dies from causes attributable to the
neutralising antibodies against bovine diarrhoea virus, the vaccme.
vaccine stimulates the production of such antibodies. 2-2-2 Residual virulence
3-2 Bacteria and fungi The test is carried out on the master seed lot and on a
The vaccine, including where applicable the diluent supplied representative batchof vaccine. If the quantity of the master
for reconstitution, complies with the test for sterility seed sufficient for pelforming the test is not available, the
prescribed in the monograph VacGÍnes for veterina1Y lowest passage seed used for the production that is available
use (0062). in sufficient quantity may be used.
Use 32 female CD1 mice, 5,,6 weeks old. Vaccinate each
3-3 Residuallive virus
mouse by the subcutaneous route with a suspension
Carry out a test for residual live bovine diarrhoea virus by
(0.1 mL) containing 10 8 live bacteria. Euthanise the mice in
inoculating not less than 10 doses onto cells known to be
groups of 8, selected at random, 3, 6, 9 and 12 weeks latero
sensitive to bovine diarrhoea virus; passage the cells after
Remove the spleens and homogenise individually and
Vet-220 Veterinary Vaccines 2016

aseptically in 1 mL of phosphate buffered saline pH 6.8 R. 3-2 Determination of dissociation phase


Spread the entire suspension on plates containing a suitable Examine not fewer than 200 colonies by a suitable technique.
culture medium (lower limit of detection: 1 bacterium per The culture of the vaccine strain is seen to be in the
spleen). Carry out in parallel a similar test using Brucella smooth (S) phase.
711elitensis Rev. 1 strain BRP (reference strain). Calculate the The vaccine complies with the test if not fewer than
50 per cent persistence time by the usual statistical methods 95 per cent of the colonies are of the smooth type.
(5.3) for probit analysis.
3-3 Bacteria and fungi
The product complies with the test if the 50 per cent The vaccine complies with the test if it does not contain
persistence time for the vaccine strain does not differ extraneous micro-organisms. Veri:fy the absence of micro-
significantly from that of tl1e reference strain. organisms other than B. melitensis Rev. 1 strain as described
2-2-3 Determination of dissociation phase of the in the test for sterility prescribed in tl1e monograph Vaccines
master seed 10t for veterina7Ji use (0062).
Examine not fewer than 200 colonies by a suitable technique. 3-4 Live bacteria
The culture of the vaccine strain is seen to be in the Make a count of live bacteria on a solid medium suitable for
smooth (S) phase. the culture of B. melitensis Rev. 1 strain.
The seed lot complies with the test if not fewer than The vaccine complies with the test if it contains not fewer
99 per cent of the colonies are of the smooth type. than 0.5 x 10 9 and not more than 4 x 10 9 live bacteria
2-2-4 Irnmunogenicity in mice per dose.
The test is carried out on tl1e master seed lot and on a
4LABELLING
representative batch of vaccine. If the quantity of the master
The label states:
seed sufficient for pelforming the test is not available, the
- that the vaccine may be dangerous for man;
lowest passage seed used for the production that is available
- that the vaccine is not to be used in pregnant animals;
in sufficient quantity may be used.
- that the vaccine may be dangerous for cattle and that they
Use for the test healthy CD1 female mice, 5-7 weeks old and are not to be kept in contact with sheep 01' goats
from the same stock. Distribute the mice into 3 groups of vaccinated less than 24 h previously.
6 mice. Dilute tl1e vaccine strain and the Brucella melitensis ___________________________________________ ~Ew

Rev. 1 strain BRP (reference strain) to a concentration of


10 6 CFU/mL.
Vaccinate bytl1e subcutaneous route the mice of tl1e 1sr
group with 0.1 mL of the diluted vaccine strain and the mice
of the 2nd group with 0.1 mL of the diluted reference strain; Calf Coronavirus Diarrhoea ***
keep tl1e 3rd group as the unvaccinated control. After *** ***
30 days, challenge all the mice with 2 x 10 5 bacteria of Vaccine (Inactivated) ***
B. abo11Zls strain 544 (COTdependent). Euthanise the mice (Ph. ElIr. 771onograph 1953)
15 days later and remove the spleen for B. abO/tus isolation. PhEw ______________________________________
Record the number of B. abO/tus per spleen (X) and
transfonn this value to obtain Y = 10gIO (X/log IO X). Then 1 DEFINITION
calculate the mean and standard deviation of each group. Calf coronavirus diarrhoea va.ccine (inactivated) is a
The test is valid if: preparation of one 01' more suitable strains of bovine
- tl1e mean of tl1e unvaccinated control group is at least 4.5 coronavirus, inactivated while maintaining adequate
(mean ofY); immunogenic properties. This monograph applies to vaccines
- tl1e mean of the group receiving the reference strain is intended for the active immunisation of dams for passive
lower than 2.5 (mean of Y); and protection of their progeny against coronavirus diarrhoea
- the standard deviation of each group is lower than 0.8. during tl1e first few weeks of life.
Carry out a statistical comparison of the irnmunogenicity 2 PRODUCTION
values of the 3 groups using the least significant differences 2-1 PREPARATION OF THE VACCINE
test. The vaccine strain complies with the test if: Each vaccine virus is grown separately in cell cultures.
- the irnmunogenicity value of tl1e group receiving the The viral suspensions of each vaccine virus are harvested
vaccine strain is significantly lower tl1an the separately and inactivated by a method that maintains
immunogenicity value of the control group; and immunogenicity. The viral suspensions may be purified and
- tl1e irnmunogenicity value of the group receiving the concentrated. The vaccine may be adjuvanted.
vaccine strain is not significantly different from tl1e 2-2 SUBSTRATE FOR VIRUS PROPAGATION
immunogenicity value of the group receiving the reference
2-2-1 Cell cultures
strain.
The cell cultures comply Witl1 the requirements for cell
3 BATCH TESTS cultures for production of veterinary vaccines (5.2.4).
3-1 Identification 2-3 CHOICE OF VACCINE COMPOSITION
B. melitensis present in the vaccine is identified by suitable
The vaccine is shown to be satisfactory with respect to safety
morphological, serological and biochemical tests and by
(5.2.6) and efficacy (5.2.7) for the pregnant cows for which it
culture: Rev. 1 strain is inhibited by addition to the suitable
is intended.
culture medium of eitl1er benzylpenicillin sodium (3 ~lg/mL),
thionin (20 ~lg/mL) or basic fuchsin (20 ~lg/mL); the strain The following tests for safety (section2-3-1) and
grows on agar containing 2.5 ~lg of streptomycin per immunogenicity (section 2-3-2) may be used during the
millilitre. demonstration of safety and efficacy.
2016 Veterinary Vaccines Vet-221

2-3-1 Safety in pregnant cows vaccine that has given satisfactory results in the test described
Carry out the test for each route and method of under Potency. The following test may be used.
administration to be recommended for vaccination, using in To obtain a valid assay, it may be necessary to cany out a
each case pregnant cows that have not been vaccinated test using several groups of animals, each receiving a different
against bovine coronavirus. Use a batch of vaccine containing dose. For each dose required, carry out the test as follows.
not less than the maximum potency that may be expected in U se for the test not fewer dlan 7 animals of a suitable species
a batch of vaccine. and that do not have specific antibodies agail1st bovine
For each test, use not fewer than 8 cows per group at the coronavirus. Vaccinate not fewer than 5 animals using
stage or at different stages of pregnancy according to the 1 injection of a suitable dose. Maintain not fewer than
schedule to be recommended. Administer to each pregnant 2 animals as controls. Where dle recommended schedule
animal 1 dose of the vaccine. If the schedule to be requires a booster injection to be given, a booster vaccination
recommended requires a 2nd dose, administer another dose may also be given in this test provided it has been
after an interval of at least 14 days. After each injection, demonstrated that this will still provide a suitably sensitive
measure the body temperature on the day of the injection test system. At a given interval not less than 14 days after the
and on the 4 following days. Observe the pregnant cows at last injection, collect blood from each animal and prepare
least daily until calving. serum samples. Use a suitable validated test to measure the
The vaccine complies with the test if no pregnant cow shows antibody response. The vaccine complies widl the test if the
abnormal local or systemic reactions or die s from causes antibody level in the vaccinates is not significandy less than
attributable to the vaccine and if no adverse effects on that obtained with a batch that has given satisfactory results
gestation or the offspring are noted. in the test described under Potency and there is no
significant increase in antibody titre in the controls.
2-3-2 Immunogenicity
A test is carried out for each route and method of 3 BATCH TESTS
administration to be recommended. The vaccine 3-1 Identification
administered to each cow is of minimum potency. Injected into animal s that do not have specific antibodies
Use for the test not fewer than 15 pregnant cows, preferably against bovine coronavirus, the vaccine stimulates dle
that do not have antibodies against bovine coronavirus. formation of such antibodies.
Where such cows are not available, use cows that: have not 3-2 Bacteria and fungi
been vaccinated against bovine coronavirus; come from a The vaccine, including where applicable dle diluent supplied
farm where there is no recent history of infection with bovine for reconstitution, complies with the test for sterility
coronavirus; and have a low level of antibodies against bovine prescribed in the monograph Vaccines for veterinarji use
coronavirus, the levels being comparable in all cows. (0062).
Vaccinate not fewer than 10 pregnant cows according to the 3-3 Residuallive virus
schedule to be recommended. Maintain not fewer than 5 Carry out a test for residual live virus using 10 doses of
pregnant cows as controls. Starting at calving, take the vaccine and 2 passages in cell cultures of the same type as
colostrum and dlen mil k from each cow and keep it in dl0se used for production of dle vaccine or odler cell cultures
suitable conditions. Determine individually dle protective of suitable sensitivity. The vaccine complies with the test if
activity of the colostrum and milk from each cow using calves no live virus is detected. If dle vaccine contains an adjuvant
born from healthy cows, and which may be born by that intetferes with dle test, separate it if possible from the
Caesarean section, and maintained in an environment where liquid phase of the vaccine by a medlod that does not
they are not exposed to infection by bovine coronavirus. inactivate virus nor interfere in any other way widl detection
Feed colostrum and then milk to each calf every 6 h or of live virus es.
according to the schedule to be recommended. At 5-7 days
after birth, challenge each calf by the oral route with a 3-4 Specified extraneous agents
sufficient quantity of a virulent strain of bovine coronavirus. Use 2 cattle not less dlan 6 months old and that do not have
Observe the calves at least daily for 7 days. Note dle antibodies against bovine herpesvirus 1 (BHVl), bovine
incidence, severity and duration of diarrhoea and the leukaemia virus (BLV) and bovine viral diarrhoea virus
duration and quantity of virus excretion. (BVDV). Administer to each animal by a recommended
route a double dose of the vaccine, dlen another dose after
The vaccine complies with the test if there is a significant
14 days. Observe the catde at least daily until 14 days after
reduction in diarrhoea and virus excretion in calves given
the last administration. Take a blood sample at the end of
colostrum and milk from vaccinated cows compared to those
the observation periodo The vaccine complies with the test if
given colostrum and milk from controls.
it does not stimulate the formation of antibodies against
2-4 MANUFACTURER'S TESTS bovine herpesvirus 1 (BHV1), bovine leukaemia virus (BLV)
2-4-1 Residuallive virus and bovine viral diarrhoea virus (BVDV).
The test for residual live virus is carried out using 2 passages 3-5 Potency
in cell cultures of the same type as those used for production The vaccine complies with the requirements of the test
or in cells shown to be at least as sensitive. The quantity of prescribed under Irnmunogenicity (section 2-3-2) when
inactivated virus harvest used in the test is equivalent to not administered by a recommended route and method.
les s than 10 dos es of vaccine. The inactivated virus harvest
complies with the test if no live virus is detected. 4 LABELLING
The label sta tes the recommended schedule for administering
2-4-2 Batch potency test
colostrum and milk, post-part~ml.
It is not necessary to carry out the potency test (section 3-5)
_____________________________________________ PhE~

for each batch of vaccine if it has been carried out usinga


batch of vaccine with a minimum potency. Where the test is
not carried out, an alternative validated method is used, the
criteria for acceptance being set with reference to a batch of
Vet-222 Veterinary Vaccines 2016

Calf Rotavirus Diarrhoea Vaccine *** recommended. Maintain not fewer than 5 pregnant cows as
*** *** controls. Starting at calving, take the colostrum and then
(Inactivated) *** milk from each cow and keep it in suitable conditions.
(Ph. Eur. 111onograph 1954) Determine individually the protective activity of the
PhE~ _____________________________________________ colostrum and milk from each cow using calves born from
healthy cows, and which may be born by Caesarean section,
1 DEFINITION and maintained in an environment where they are not
Calf rotavirus diarrhoea vaccine (inactivated) is a preparation exposed to infection by bovine rotavirus. Feed colostrum and
of one or more suitable strains of bovine rotavirus, then milk to each calf every 6 h or according to the schedule
inactivated while maintaining adequate irnmunogenic to be recommended. At 5-7 days after birth, challenge each
properties. This monograph applies to vaccines intended for calf by the oral route with a sufficient quantity of a virulent
the active irnmunisation of dams for passive protection of strain of bovine rotavirus. Observe the calves at least daily for
their progeny against rotavirus diarrhoea during the first 7 days. Note the incidence, severity and duration of
few weeks of life. diarrhoea and the duration and quantity of virus excretion.
2 PRODUCTION The vaccine complies with the test if there is a significant
2-1 PREPARATION OF THE VACCINE reduction in diarrhoea and virus excretion in calves given
Each vaccine virus is grown separately in cell cultures. colostrum and milk from vaccinated cows compared to those
The viral suspensions of each vaccine virus are harvested given colostrum and milk from controls.
separately and inactivated by a method that maintains 2-4 MANUFACTURER'S TESTS
irnmunogenicity. The viral suspensions may be purified and 2-4-1 Residuallive virus
concentrated. The vaccine may be adjuvanted. The test for residual live virus is carried out using 2 passages
2-2 SUBSTRATE FOR VIRUS PROPAGATION in cell cultures of the same type as those used for production
2-2-1 Cell cultures or in cells shown to be at least as sensitive. The quantity of
The cell cultures comply with the requirements for cell inactivated virus harvest used in the test is equivalent to not
cultures for production of veterinary vaccines (5.2.4). less than 100 doses of vaccine. The inactivated viral harvest
complies with the test if no live virus is detected.
2-3 CHOICE OF VACCINE COMPOSITION
2-4-2 Batch potency test
The vaccine is shown to be satisfactory with respect to safety
It is not necessary to carry out the potency test (section 3-5)
(5.2.6) and efficacy (5.2.7) for the pregnant cows for which it
for each batch of vaccine if it has been carried out using a
is intended.
batch of vaccine with a minimum potency. Where the test is
The following tests for safety (section 2-3-1) and not carried out, an alternative validated method is used, the
irnmunogenicity (section 2-3-2) may be used during the criteria for acceptance being set with reference to a batch of
demonstration of safety and efficacy. vaccine that has given satisfactory results in the test described
2-3-1 Safety in pregnant cows under Potency. The following test may be used.
Carry out the test for each route and method of To obtain a valid assay, it may be necessary to carry out a
administration to be recommended for vaccination, using in test using several groups of animals, each receiving a different
each case pregnant cows that have not been vaccinated dose. For each dose required, carry out the test as follows.
against bovine rotavirus. Use a batch of vaccine containing Use for the test not fewer than 7 animals of a suitable species
not les s than the maximum potency that may be expected in and that do not have antibodies against bovine rotavirus.
a batch of vaccine. Vaccinate not fewer than 5 animals using 1 injection of a
For each test, use not fewer than 8 cows per group at the suitable dose. Maintain not fewer than 2 animals as controls.
stage or at different stages of pregnancy according to the Where the recommended schedule requires a booster
schedule to be recommended. Administer to each pregnant injection to be given, a booster vaccination may also be given
animal 1 dos e of the vacciné. If the schedule to be in this test provided it has been demonstrated that this will
recommended requires a 2nd dose, administer another dose still provide a suitably sensitive test system. At a given
after an interval of at least 14 days. After each injection, interval not less than 14 days after the last injection, collect
measure the body temperature on the day of the injection blood from each animal and prepare serum samples. Use a
and on the 4 following days. Observe the pregnant cows at suitable validated test to measure the antibody response.
least daily until calving. The vaccine complies with the test if the antibody level in the
The vaccine complies with the test if no pregnant cow shows vaccinates is not significantly lessthan that obtained with a
abnormal local or systemic reactions or dies from causes batch that has given satisfactory results in the test described
attributable to the vaccine and if no adverse effects on under Potency and there is no significant increase in antibody
gestation or the offspring are noted. titre in the controls.
2-3-2 Inununogenicity 3 BATCH TESTS
A test is carried out for each route and method of 3-1 Identification
administration to be recommended. The vaccine Injected into animals that do not have specific antibodies
administered to each cow is of minimum potency. against bovine rotavirus, the vaccine stimulates the formation
Use for the test not fewer than 15 pregnant cows, preferably of such antibodies.
that do not have antibodies against bovine rotavirus. Where 3-2 Bacteria and fungi
such cows are not available, use cows that: have not been The vaccine~ including where applicable the diluent supplied
vaccinated against bovine rotavirus; come from a farm where for reconstitution, complies with the test for sterility
there is no recent history of infection with bovine rotavirus; prescribed in the monograph Vaccines for veterina1Y
and have a low level of antibodies against bovine rotavirus, use (0062).
the levels being comparable in all cows. Vaccinate not fewer
than 10 pregnant cows according to the schedule to be
2016 Veterinary Vaccines Vet-223

3-3 Residuallive virus 2-3-1 Safety


Carry out a test for residual live virus using 10 doses of Carry out the test for each route and method of
vaccine and 2 passages in cell cultures of the same type as administration to be recommended for vaccination. Use a
those used for production of the vaccine or other cell cultures batch of vaccine containing not less than the maximum
of suitable sensitivity. The vaccine complies with the test if potency that may be expected in a batch of vaccine.
no live virus is detected. If the vaccine contains an adjuvant For each test, use not fewer than 8 dogs of the minimum age
that intelferes with the test, separate it if possible from the to be recommended and that do not have antibodies against
liquid phase of the vaccine by a method that does not canine adenovirus 1 or 2. Administer to each dog 1 dose of
inactivate virus nor intelfere in any other way with detection the vaccine. If the schedule to be recommended requires a
of live viruses. 2nd dose, administer 1 dose after an intel"val of at least
3-4 Specified extraneous agents 14 days. Observe the dogs at least daily for at least 14 days
Use 2 cattle not less than 6 months old and that do not have after the last administration.
antibodies against bovine herpesvirus 1 (BHV1), bovine The vaccine complies with the test if no dog shows abnormal
leukaemia virus (BLV) and bovine viral diarrhoea virus local or systemic reactions or dies from causes attributable to
(BVDV). Administer to each animal by a recommended tlle vaccine.
route a double dose of the vaccine, then another dose after 2-3-2 Immunogenicity
14 days. Observe the cattle at least daily until 14 days after For vaccines intended to protect against hepatitis, the test
the last administration. Take a blood sample at the end of
described below is suitable for demonstration of
the observation periodo The vaccine complies with the test if immunogenicity. If the vaccine is indicated for protection
it does not stimulate the formation of antibodies against against respiratory signs, a furthel" test to demonstrate
bovine herpesvirus 1 (BHV 1), bovine leukaemia virus (BLV)
immunogenicity for this indication is also necessary.
and bovine viral dialThoea virus (BVDV).
A test is carried out fol" each l"oute and method of
3-5 Potency administration to be recommended for vaccination, using in
The vaccine complies with the requirements of the test each case dogs of the minimum age to be recommended.
prescribed under Immunogenicity (section 2-3-2) when The vaccine administered to each dog is of minimum
administered by a recommended route and method. potency.
4 LABELLING Use for the test not fewer than 7 dogs that do not have
The label sta tes the recommended schedule for administering antibodies against canine adenovirus. Vaccinate not fewer
colostrum and milk, post-pa11Lnn. than 5 dogs, according to the schedule to be recommended.
_____________________________________________ PhEw Maintain not fewer than 2 dogs as controls. Challenge each
dog aftel" 20-22 days by the intravenous route with a
sufficient quantity of a suspension of patllogenic canine
adenovirus. Observe the dogs at least daily for 21 days after
*** challenge. Dogs displaying typical signs of serious infection
Canine Adenovirus Vaccine,
*** *** with canine adenovirus are eutllanised to avoid unnecessary
Inactivated *** suffering.
(Canine Adenovirlls Vaccine (Inactivated)) The test is not valid if, during the observation period after
Ph Bu!' 111onograph 1298) challenge, fewer than 100 per cent of the control dogs die
PhEw _____________________________________________ from or show typical signs of serious infection with canine
adenovirus. The vaccine complies with tlle test if, during the
1 DEFINITION observation period, all the vaccinated dogs survive and show
Canine adenovirus vaccine (inactivated) is a preparation of no signs of disease.
one or more suitable strains of canine adenovirus 1 (canine
2-4 MANUFACTURER'S TESTS
contagious hepatitis virus) and/or canine adenovirus 2,
2-4-1 Residuallive virus
inactivated while maintaining adequate immunogenic
The test for residual live virus is carried out using a quantity
properties. This monograph applies to vaccines intended for
of inactivated virus harvest equivalent to at least 10 dos es of
the active immunisation of dogs against canine contagious
vaccine with 2 passages in cell cultures of the same type as
hepatitis and/or respiratory disease caused by canine
those used for production or in cell cultures shown to be at
adenovirus.
least as sensitive. The inactivated viral harvest complies with
2 PRODUCTION the test if no live virus is detected.
2-1 PREPARATION OF THE VACCINE 2-4-2 Batch potency
The vaccine virus is grown in cell cultures. The virus harvest It is not necessary to carry out the Potency test (section 3-4)
is inactivated. The vaccine may be adjuvanted. for each batch of vaccine if it has been carried out using a
2-2 SUBSTRATE FOR VIRUS PROPAGATION batch of vaccine with a minimum potency. Where the test is
2-2-1 Cell cultures not carried out, an alternative validated method is used, the
The cell cultures comply with the requirements for cell criteria for acceptance being set with reference to a batch of
cultures for production of veterinary vaccines (5.2.4). vaccine that has given satisfactory results in the test described
under Potency.
2-3 CHOICE OF VACCINE COMPOSITION
The vaccine is shown to be satisfactory with respect to safety 3 BATCH TESTS
(5.2.6) and efficacy (5.2.7) for the dogs for which it is 3-1 Identification
intended. When injected into animals that do not have specific
The following tests for safety (section 2-3-1) and antibodies against the type or types of canine adenovirus
immunogenicity (section 2-3-2) may be used during the stated on the label, the vaccine stimulates the formation of
demonstration of safety and efficacy. such antibodies.
Vet-224 Veterinary Vaccines 2016

3-2 Bacteria and fungi 2-3-2 Increase in virulence


The vaccine, including where applicable the diluent supplied Can")' out the test according to general chapter 5.2.6 using
for reconstitution, complies with the test for sterility dogs 5-7 weeks old, that do not have antibodies against
prescribed in the monograph Vaccines for veterinaJ]' canine adenoviruses. lf the properties of the vaccine virus
use (0062). allow sequential passage through 5 groups via natural
3-3 Residuallive virus spreading, this method may be used, otherwise passage as
Carry out a test for residual canine adenovirus using 10 doses described below is carried out.
of vaccine by inoculation into sensitive cell cultures; make a Administer to each dog of the 1st group by a route to be
passage after 6-8 days and maintain the cultures for 14 days. recommended a quantity of the vaccine virus that will allow
The vaccine complies with the test if no live virus is detected. recovery of virus for the passages described below.
If the vaccine contains an adjuvant, separate the adjuvant Administer the virus by the route to be recommended for
from the liquid phase by a method that does not inactivate or vaccination most likely to lead to reversion of virulence. After
otherwise interfere with the detection of live virus. 4-6 days, prepare a suspension from the nasal and pharyngeal
mucosa, tonsils, lung, spleen and if they are likely to contain
3-4 Potency
The vaccine complies with the requirements of the test virus, liver and kidney of each dog and pool the samples.
mentioned under lmmunogenicity (section 2-3-2) when Administer 1 mL of the pooled samples by a suitable route -
for example, the intranasal route - to each dog of the next
administered by a recommended route and method.
_____________________________________________ PhE~
group. Carry out this passage operation not fewer than
4 times; verify the presence of the virus at each passage.
lf the virus is not found at a passage level, repeat the passage
by administration to a group of 10 animals.
If the 5th group of animals shows no evidence of an increase
Canine Adenovirus Vaccine, in virulence indicative of reversion during the observation
period, further testing is not required. Otherwise, carry out
Living an additional safety test and compare the clinical signs and
(Canine Adenovirus Vaccine (Live) any relevant parameters in a group of at least 8 animals
Ph Bur monograph 1951) receiving the material used for the 1st passage and another
PhE~ _____________________________________________ similar group receiving the virus at the final passage level.
The vaccine virus complies with the test if no indication of
1 DEFINITION
increased virulence of the virus recovered for the final
Canine adenovirus vaccine (live) is a preparation of a suitable
passage compared with the material used for the 1st passage
strain of canine adenovirus 2. This monograph applies to
is observed. If virus is not recovered after an initial passage in
vaccines intended for the active immunisation of dogs against
2 animals and a subsequent repeat passage in 10 animals, the
canine contagious hepatitis and/or respiratory disease caused
vaccine virus also complies with the test.
by canine adenovirus.
2-3-3 Irnmunogenicity
2 PRODUCTION A test is carried out for each route and method of
2-1 PREPARATION OF THE VACCINE administration to be recommended for vaccination using in
The vaccine virus is grown in cell cultures. each case dogs of the minimum age to be recommended.
2-2 SUBSTRATE FOR VIRUS PROPAGATION The quantity of vaccine virus to be administered to each dog
2-2-1 Cell cultures is not greater than the minimum virus titre to be stated on
The cell cultures comply with the requirements for cell the label and the virus is at the most attenuated passage level
cultures for production of veterinary vaccines (5.2.4). that will be present in a batch of vaccine.
2-3-3-1 Vaccines intended to protect against hepatitis. Use for
2-3 CHOICE OF VACCINE VIRUS
the test not fewer than 7 dogs that do not have antibodies
The vaccine virus is shown to be satisfactory with respect to
against canine adenoviruses. Vaccinate not fewer than 5 dogs,
safety (5.2.6) and efficacy (5.2.7) for the dogs for which it is
according to the schedule to be recommended. Maintain not
intended.
fewer than 2 dogs as controls. Challenge each dog after
The following tests for safety (section 2-3-1), increase in 20-22 days by the inu'avenous route with a sufficient quantity
virulence (section 2-3-2) and immunogenicity (section 2-3-3) of a suspension of virulent canine adenovirus 1 (canine
may be used during the demonstration of safety and efficacy. contagious hepatitis virus). Observe the dogs at least daily for
2-3-1 Safety 21 days after challenge. Dogs displaying typical signs of
Carry out the test for each route and method of serious infection with canine adenovirus are euthanised to
administration to be recommended for vaccination. avoid unnecessary suffering.
Use vaccine virus at the least attenuated passage level that The test is not val id if during the observation period after
will be present in a batch of the vaccine. challenge, fewer than 100 per cent of the control dogs die or
For each test, use not fewer than 5 dogs of the minimum age show notable signs of canine adenovirosis.
to be recommended for vaccination and that do not have The vaccine virus complies with the test if during the
antibodies against canine adenoviruses. Administer to each observation period after challenge, all the vaccinated dogs
dog a quantity of the vaccine virus equivalent to not less than survive and show no signs of disease except for a possible
10 times the maximum virus titre likely to be contained in transient elevated rectal temperature.
1 dose of the vaccine. Observe the dogs at least daily for at
2-3-3-2 Vaccine intended to protect against respirat01y signs.
least 14 days.
U se for the test not fewer than 20 dogs that do not have
The vaccine virus complies with the test if no dog shows antibodies against canine adenoviruses. Vaccinate not fewer
abnonnal local 01' systemic reactions, signs of disease 01' dies than 10 dogs, according to the schedule to be recommended.
from causes attributable to the vaccine virus. Maintain not fewer than 10 dogs as controls. Challenge each
2016 Veterinary Vaccines Vet-225

dog after 20-22 days by the intranasal route with a quantity 2 PRODUCTION
of a suspension of virulent canine adenovirus 2 sufficient to 2-1 PREPARATION OF THE VACCINE
cause typical signs of respiratory disease in a dog that does The vaccine virus is grown in emblyonated hens' eggs or in
not have antibodies against canine adenoviruses. Observe the cell cultures.
dogs at least daily for 10 days after challenge. Record the
2-2 SUBSTRATE FOR VIRUS PROPAGATION
incidence of signs of respiratory and general disease in each
dog (for example, sneezing, coughing, nasal and lachrymal 2-2-1 Embryonated hens' eggs
If the vaccine virus is grown in emblyonated hens' eggs, they
discharge, loss of appetite). Collect nasal swabs or washings
from each dog daily from days 2 to 10 after challenge and are obtained from fiocks free from specified pathogens (SPF)
test these samples to determine the presence and titre of (5.2.2).
excreted virus. 2-2-2 Cell cultures
The vaccine complies with the test if there is a notable If the vaccine virus is grown in cell cultures, they comply
decrease in the incidence and severity of signs and in virus with the requirements for cell cultures for production of
excretion in vaccinates compared to controls. veterinary vaccines (5.2.4).

3 BATCH TESTS 2-3 CHOICE OF VACCINE VIRUS


3-1 Identification The vaccine virus is shown to be satisfactory with respect to
The vaccine mixed with monospecific antiserum against safety (5.2.6) and efficacy (5. 2.7) for the dogs for which it is
canine adenovirus 2 no longer infects susceptible cell intended.
cultures. The following tests for safety (section 2-3-1), increase in
3-2 Bacteria and fungi virulence (section 2-3-2) and irnmunogenicity (section 2-3-3)
The vaccine, including where applicable the diluent supplied may be used during the demonstration of safety and efficacy.
for reconstitution, complies with the test for sterility 2-3-1 Safety
prescribed in the monograph Vaccines for veterinaJY Cany out the test for each route and method of
use (0062). administration to be recommended for vaccination.
3-3 Mycop1asmas (2.6.7) U se vaccine virus at the least attenuated passage level that
The vaccine complies with the test for mycoplasmas. will be present in a batch of the vaccine.
For each test, use not fewer than 5 dogs of the minimum age
3-4 Extraneous agents
to be recommended for vaccination and that do not have
Neutralise the vaccine virus with a suitable monospecific
antibodies against canine distemper virus. Administer to each
antiserum against canine adenovirus 2 and inoculate into cell
dog a quantity of the vaccine virus equivalent to not les s than
cultures known for their susceptibility to virus es pathogenic
10 times the maximum virus titre likely to be contained in
for the dogo Carry out a passage after 6-8 days and maintain
1 dose of the vaccine. Observe the dogs at least daily for
the cultures for a total of 14 days.
42 days.
The vaccine complies with the test if no cytopathic effect
The vaccine virus complies with the test if no dog shows
develops and there is no sign of the presence of
abnormal local or systemic reactions, signs of disease or dies
haemadsorbing agents.
from causes attributable to the vaccine virus.
3-5 Virus titre
2-3-2 Increase in virulence
Titrate the vaccine virus in suitable cell cultures. The vaccine
Cany out the test according to general chapter 5. 2. 6 using
complies with the test if one dose contains not les s than the
dogs 5-7 weeks old, that do not have antibodies against
minimum virus titre stated on the label.
canine distemper virus. If the properties of the vaccine virus
3-6 Potency allow sequential passage through 5 groups via natural
The vaccine complies with the requirements of one or both spreading, this method may be used, otherwise passage as
of the tests prescribed under Immunogenicity (section 2-3-3) described below is carried out.
when administered by a recommended route and method. Administer to each dog of the 1st group by a route to be
It is not necessary to carry out the potency test for each
recommended a quantity of the vaccine virus that will allow
batch of the vaccine if it has been carried out on a
recovery of virus for the passages described below.
representative batch using a vaccinating dose containing not
Administer the virus by the route to be recommended for
more than the minimum virus titre stated on the label.
vaccination most likely to lead to reversion to virulence. After
_____________________________________________ PhEw
5-10 days, prepare a suspension from the nasal mucosa,
tonsils, thymus, spleen and the lungs and their local lymph
nodes of each dog and pool the samples. Administer 1 mL of
the pooled samples by the intranasal route to each dog of the

*****
next group. Cany out this passage operation not fewer than
Canine Distemper Vaccine, Living 4 times; verify the presence of the virus at each passage.
** ** If the virus is not found at a passage level, repeat the passage
(Canine Diste7l1per Vaccine (Live) J *** by administration to a group of 10 animals.
Ph Bur 1110nograph 0448)
~Ew _____________________________________________ If the 5th group of animals shows no evidence of an increase
in virulence indicative of reversion during the observation
1 DEFINITION period~ further testing is not required. Otherwise, carry out
Canine distemper vaccine (live) is a preparation of a suitable an additional safety test and compare the clinical signs and
strain of distemper virus. This monograph applies to vaccines any relevant parameters in a- group of at least 8 animals
intended for the active irnmunisation of dogs against canine receiving the material used for the 1st passage and another
distemper. similar group receiving the virus at the final passage level.
Vet-226 Veterinary Vaccines 2016

The vaccine virus complies with the test if no indication of


Canine Parainfluenza Virus ***
increased virulence of the virus recovered for the final ** **
passage compared with the material used for the 1st passage Vaccine (Live) *****
is observed. If virus is not recovered after an initial passage in (Ph. Eur. 1110nograph 1955)
2 animals and a subsequent repeat passage in 10 animals-, the ~E~ _____________________________________________
vaccine virus also complies with the test.
2-3-3 Immunogenicity 1 DEFINITION
A test is carried out for each route and method of Canine parainfiuenza virus vaccine (live) is a preparation of a
administration to be recommended for vaccination using in suitable strain of parainfiuenza virus of canine origino This
each case dogs 8-16 weeks old. The quantity of vaccine virus monograph applies to vaccines intended for the active
to be administered to each dog is not greater than the irnrnunisation of dogs against respiratory signs of infection
minimum virus titre to be stated on the label and the virus is with parainfiuenza virus of canine origino
at the most attenuated passage level that will be present in a 2 PRODUCTION
batch of vaccine. 2-1 PREPARATION OF THE VACCINE
U se for the test not fewer than 7 dogs that do not have The vaccine virus is grown in cell cultures.
antibodies against canine distemper virus. Vaccinate not
2-2 SUBSTRATE FOR VIRUS PROPAGATION
fewer than 5 dogs according to the schedule to be
2-2-1 Cell cultures
recommended. Maintain not fewer than 2 dogs as controls.
The cell cultures comply with the requirements for cell
Challenge each dog after 20-22 days by the intravenous route
cultures for production of veterinary vaccines (5.2.4).
with a sufficient quantity of a suspension of virulent canine
distemper virus. Observe the dogs at least daily for 21 days 2-3 CHOICE OF VACCINE VIRUS
after challenge. Dogs displaying typical signs of serious The vaccine virus is shown to be satisfactory with respect to
infection with canine distemper virus are euthanised to avoid safety (5.2.6) and efficacy (5.2.7) for the dogs for which it is
unnecessary suffering. intended.
The test is not valid if during the observation period after The following tests for safety (section 2-3-1)-, increase in
challenge-, fewer than 100 per cent of the control dogs die or virulence (section 2-3-2) and irnmunogenicity (section 2-3-3)
show notable signs of canine distemper. may be used during the demonstration of safety and efficacy.
The vaccine virus complies with the test if during the 2-3-1 Safety
observation period after challenge-, all the vaccinated dogs Carry out the test for each route and method of
survive and show no signs of disease. administration to be recommended for vaccination.
3 BATCH TESTS Use vaccine virus at the least attenuated passage level that
3-1 Identification will be present in a batch of the vaccine.
The vaccine mixed with a monospecific distemper antiserum For each test-, use not fewer than 5 dogs of the minimurn age
against canine distemper virus no longer provokes cytopathic to be recommended for vaccination and that do not have
effects in susceptible cell cultures. antibodies against parainfiuenza virus of canine origino
3-2 Bacteria and fungi Adrninister to each dog a quantity of the vaccine virus
equivalent to not less than 10 times the maximurn virus titre
The vaccine-, including where applicable the diluent supplied
for reconstitution-, complies with the test for sterility likely to be contained in 1 dose of the vaccine. Observe the
prescribed in the monograph Vaccines f01" veterina¡y dogs at least daily for at least 14 days.
use (0062). The vaccine virus complies with the test if no dog shows
3-3 Mycop1asmas (2.6.7) abnorrnal local or systernic reactions-, signs of disease or dies
The vaccine complies with the test for mycoplasmas. from causes attributable to the vaccine virus.
3-4 Extraneous agents 2-3-2 Increase in virulence
Neutralise the vaccine virus with a suitable monospecific Can)' out the test according to general chapter 5.2.6 using
antiserum against canine distemper virus and inoculate into dogs 5-7 weeks old-, that do not have antibodies against
cell cultures known for their susceptibility to viruses parainfiuenza virus of canine origino If the properties of the
pathogenic for the dogo Carry out a passage after 6-8 days vaccine virus allow sequential passage through 5 groups via
and maintain the cultures for 14 days. natural spreading-, this method may be used-, otherwise
passage as described below is carried out.
Thevaccine complies with the test if no cytopathic effect
develops and there is no sign of the presence of Administer to each dog of the 1st group by the intranasal
haemadsorbing agents. route and by a route to be recommended a quantity of the
vaccine virus that will allow recovery of virus for the passages
3-5 Virus titre
described below. Administer the virus by the route to be
Titrate the vaccine virus in suitable cell cultures. The vaccine
recommended for vaccination most likely to lead to reversion
complies with the test if one dose contains not less than the
to virulence. After 3-10 days-, prepare a suspension from
minimum virus titre stated on the label.
nasal swabs of each dogo Adrninister 1 mL of the suspension
3-6 Potency from the swabs that contain the maximurn amount of virus
The vaccine complies with the requirements of the test by the intranasal route to each dog of the next group. Carry
prescribed under Irnmunogenicity (section 2-3-3) when out this passage operation not fewer than 4 times; verify the
administered by a recommended route and method. It is not presence of the virus at each passage. If the virus is not
necessary to carry out the potency test for each batch of the found at a passage level-, repeat the passage by administration
vaccine if it has been carried out on a representative batch to a group of 1O animals.
using a vaccinating dose containing not more than the
If the 5 th group of animals shows no evidence of an increase
minimum virus titre stated on the label.
in virulence indicative of reversion during the observation
_____________________________________________ PhE~
2016 Veterinary Vaccines Vet-227

period, further testing is not required. Otherwise, carry out administered by a recommended route and method. It is not
an additional safety test and compare the clinical signs and necessary to carry out the potency test for each batch of the
any relevant parameters in a group of at least 8 animal s vaccine if it has been carried out on a representative batch
receiving the material used for the 1st passage and another using a vaccinating dose containing not more than the
similar group receiving the virus at the final passage level. mínimum virus titre stated on the label.
The vaccine virus complies with the test if no indication of _____________________________________________ PhE~

increased virulence of the virus recovered for the final


passage compared with the material used for the 1st passage
is observed. If virus is not recovered after an initial passage in
2 animal s and a subsequent repeat passage in 10 animals, the
vaccine virus also complies with the test.
Canine Parvovirus Vaccine,
2-3-3 Immunogenicity Inactivated
A test is carried out for each route and method of (Canine Parvovirosis Vaccine (Inactivated))
administ:ration to be recommended for vaccination, using in Ph Bur monograph 0795)
each case dogs of the mínimum age to be recommended. PhE~ _____________________________________________
The quantity of vaccine virus to be admínistered to each dog
1 DEFlNITION
is not greater than the mínimum virus titre to be stated on
Canine parvovirosis vaccine (inactivated) is a preparation of a
the label and the virus is at the most attenuated passage level
suitable st:rain of canine parvovirus inactivated while
that will be present in a batch of vaccine.
maintaining adequate irnmunogenic properties. This
U se for the test not fewer than 15 dogs that do not have monograph applies to vaccines intended for the active
antibodies against parainfluenza virus of canine origino irnmunisation of dogs against canine parvovirosis.
Vaccinate not fewer than 10 dogs according to the schedule
to be recommended. Maintain not fewer than 5 dogs as 2 PRODUCTION
controls. Challenge each dog after not les s than 20-22 days 2-1 PREPARATION OF THE VACCINE
by the intratracheal or intranasal route with a sufficient The vaccine virus is grown in cell cultures. The virus harvest
quantity of a suspension of virulent parainfiuenza virus of is inactivated. The vaccine may be adjuvanted.
canine origino Observe the dogs at least daily for 14 days 2-2 SUBSTRATE FOR VIRUS PROPAGATION
after challenge. Collect nasal swabs 01' washings from each 2-2-1 Cell cultures
dog daily from day 2 to 10 after challenge and test these The cell cultures comply with the requirements for cell
samples for the presence of excreted virus. Use a scoring cultures for production of veterinary vaccines (5.2.4).
system to record the incidence of coughing in each dogo
2-3 CHOICE OF VACCINE COMPOSITION
The test is not valid if more than 1 of the control dogs shows
neither coughing nor the excretion of the challenge virus. The vaccine is shown to be satisfactory with respect to safety
(5.2.6) and efficacy (5.2.7) for the dogs for which it is
The vaccine complies with the test if the scores for coughing
intended.
or virus excretion for the vaccinated dogs are significantly
lower than in the controls. The following tests for safety (section 2-3-1) and
irnmunogenicity (section 2-3-2) may be used during the
3 BATCH TESTS demonstration of safety and efficacy.
3-1 Identification
2-3-1 Safety
Carry out an irnmunofiuorescence test in suitable cell
Carry out the test for each route and method of
cultures, using a monospecific antiserum.
administration to be recommended for vaccination. U se a
3-2 Bacteria and fungi batch of vaccine containing not les s than the maximum
The vaccine, including where applicable the diluent supplied potency that may be expected in a batch of vaccine.
for reconstitution, complies with the test for sterility For each test, use not fewer than 8 dogs not older than the
prescribed in the monograph Vaccines for vetel'ina1y mínimum age to be recommended for vaccination and that
use (0062). do not have antibodies against canine parvovirus. Administer
3-3 Mycoplasmas (2.6.7) to each dog 1 dose of the vaccine. If the schedule to be
The vaccine complies with the test for mycoplasmas. recommended requires a 2nd dose, administer 1 dose after an
3-4 Extraneous agents interval of at least 14 days. Observe the dogs at least daily for
Neutralise the vaccine virus with a suitable monospecific at least 14 days after the last administration.
antiserum against parainfiuenza virus of canine origin and The vaccine complies with the test if no dog shows abnormal
inoculate into cell cultures known for their susceptibility to local or systemic reactions or dies from causes attributable to
viruses pathogenic for the dogo Carry out a passage after the vaccine.
6-8 days and maintain the cultures for a total of 14 days. 2-3-2 Immunogenicity
The vaccine complies with the test if no cytopathic effect A test is carried out for each route and method of
develops and there is no sign of the presence of administration to be recommended for vaccination using in
haemadsorbing agents. each case dogs of the minimum age to be recommended.
3-5 Virus titre The vaccine administered to each dog is of minimum
Titrate the vaccine virus in suitable cell cultures. The vaccine potency.
complies with the test if one dose contains not less than the Use for the test not fewer than 7 dogs that do not have
minimum virus titre stated on the label. antibodies against canine parvovirus. Vaccinate not fewer
3-6 Potency than 5 dogs according to the schedule to be recommended.
The vaccine complies with the requirements of the test Maintain not fewer than 2 dogs as controls. Challenge each
prescribed under Irnmunogenicity (section 2-3-3) when dog after 20-22 days by the oronasal route with a sufficient
quantity of a suspension of pathogenic canine parvovirus.
Vet-228 Veterinary Vaccines 2016

Observe the dogs at least daily for 14 days after challenge. antibody titres less than 4 ND so per 0.1 mL of serum,
At the end of the observation period, carry out measured by the method described be1ow. Vaccinate each
haemagglutination tests for and titration of the virus in the dog according to the recommended schedule. 14 days after
faeces. vaccination, examine the serum of each dog as follows. Heat
The test is not valid if fewer than 100 per cent of the control the serum at 56 oC for 30 min and prepare serial dilutions
dogs show notable signs of the disease 01" leucopenia and using a medium suitable for canine cells. Add to each
excretion of the virus. The vaccine complies with the test if dilution an equal volume of a virus suspension containing an
all the vaccinated dogs survive and show no signs of disease amount of virus such that when the volume of serum-virus
nor leucopenia and if the maximum titre of virus excreted in mixture appropriate for the assay system is inoculated into
the faeces is less than 1/100 of the geometric mean of the cell cultures, each culture receives approximately
maximum titres found in the controls. 104 CCID so . Incubate the mixtures at 37 oC for 1 h and
inoculate 4 canine ceIl cultures with a suitable volume of
2-4 MANUFACTURER'S TESTS each mixture. Incubate the cell cultures at 37 oC for 7 days,
2-4-1 Residuallive virus passage and incubate for a further 7 days. Examine the
A test for residuallive virus is carried out on the bulk harvest cultures for evidence of specific cytopathic effects and
of each batch to confirm inactivation of the canine ca1culate the antibody titre. The vaccine complies with the
parvovirus. The quantity of inactivated virus harvest used in test ifthe mean titre is not less than 32 ND so per 0.1 mL of
the test is equivalent to not less than 100 doses of the serum. If one dog fails to respond, repeat the test using
vaccine. The inactivated viral harvest is inoculated into 2 more dogs and calculate the result as the mean of the titres
suitable non-confiuent cells; after incubation for 8 days, a obtained from all of the 3 dogs that have responded.
sub culture is made using trypsinised cells. After incubation _____________________________________________ ~E~

for a further 8 days, the cultures are examined for residual


live parvovirus by an immunofiuorescence test.
The immunofiuorescence test may be supplemented by a
haemagglutination test 01" other suitable tests on the
supernatant of the cell cultures. The inactivated viral harvest
complies with the test if no live virus is detected.
Canine Parvovirus Vaccine, Living
(Canine Parvovirosis Vaccine (Live) ,
3 BATCH TESTS
Ph Bur monograph 0964)
3-1 Identification
PhE~ _____________________________________________
When injected into animals that do not have antibodies
against canine parvovirus, the vaccine stimulates the 1 DEFINITION
production of such antibodies. Canine parvovirosis vaccine (live) is a preparation of a
3-2 Bacteria and fungi suitable strain of canine parvovirus. This monograph applies
The vaccine, including where applicable the diluent supplied to vaccines intended for the active immunisation of dogs
for reconstitution, complies with the test for stelility against canine parvovirosis.
prescribed in the monograph Vaccines for veteriJZalY 2 PRODUCTION
use (0062).
2-1 PREPARATION OF THE VACCINE
3-3 Potency The vaccine virus is grown in cell cultures.
Carry out test 3-3-1 ol" test 3-3-2.
2-2 SUBSTRATE FOR VIRUS PROPAGATION
3-3-1 Test in guinea-pigs for haemagglutination-inhibiting
2-2-1 Cell cultures
antibodies. Use for the test not fewer than 5 guinea-pigs that
The cell cultures comply with the requirements fOl" cell
do not have specific antibodies. Administer to each guinea-
cultures for production of veterinary vaccines (5.2.4).
pig by the subcutaneous route half of the dose to be
recommended. After 14 days, inject again half of the dose to 2-3 CHOICE OF VACCINE VIRUS
be recommended. 14 days later, collect blood samples and The vaccine virus is shown to be satisfactory with respect to
separate the serum. Inactivate each serum by heating at safety (5.2.6) and efficacy (5.2.7) for the dogs for which it is
56 oC for 30 mino To 1 volume of each serum add intended.
9 volumes of a 200 gIL suspension of light kaolin R in The following tests for safety (section 2-3-1), increase in
phosphate buffered saline pH 7.4 R. Shake each mixture for virulence (section 2-3-2) and immunogenicity (section 2-3-3)
20 mino Centrifuge, collect the supernatant and mix with may be used during the demonstl"ation of safety and efficacy.
1 volume of a concentrated suspension of pig erythrocytes.
2-3-1 Safety
Allow to stand at 4 oC for 60 min and centlifuge.
Carry out the test for each route and method of
The dilution ofthe serum obtained is 1:10. Using each
administration to be recommended for vaccination, using in
serum, prepare a series of twofold dilutions. To 0.025 mL of
each case dogs of the minimum age to be recommended for
each of the latter dilutions add 0.025 mL of a suspension of
vaccination. Use vaccine virus at the least attenuated passage
canine parvo virus antigen containing
level that will be present in a batch of the vaccine.
4 haemagglutinating units. Allow to stand at 37 oC for
30 min and add 0.05 mL of a suspension of pig erythrocytes 2-3-1-1 General safet:y. For each test, use not fewer than
containing 30 x 10 6 cells per millilitre. Allow to stand at 5 dogs that do not have haemagglutination-inhibiting
4 oC for 90 min and note the last dilution of serum that still antibodies against canine parvovirus. A count of white blood
completely inhibits haemagglutination. The vaccine complies ceIls in circulating blood is made on days 4, 2 and O before
with the test if the median antibody titre of the sera collected injection of the vaccine strain. Administer to each dog a
after the second vaccination is not les s than 1/80. quantity of the vaccine virus equivalent to not less than
10 times the maximum virus titre likely to be contained in
3-3-2 Test in dogs for virus-neutralising antibodies. Use for the
1 dose of the vaccine. Observe the dogs at least daily for at
test not fewer than 2 healthy dogs, 8-12 weeks old, that have
2016 Veterinary Vaccines Vet-229

least 14 days. A count of white blood cells in circulating is not greater than the minimum virus titre to be stated on
blood is made on days 3, 5, 7 and 10 after the injection. the label and the virus is at the most attenuated passage level
The test is not valid if a diminution in the number of that will be present in a batch of vaccine.
circulating white blood cells greater than 50 per cent of the U se for the test not fewer than 7 dogs that do not have
initial number - determined as the average of the 3 values haemagglutination-inhibiting antibodies against canine
found before injection of the vaccine strain - is noted. parvovirus. Vaccinate not fewer than 5 dogs. Maintain not
The vaccine virus complies with the test if no dog shows fewer than 2 dogs as controls. Challenge each dog after
abnormal local or systemic reactions, signs of disease or die s 20-22 days by the oronasal route with a sufficient quantity of
from causes attributable to the vaccine virus and if, for each a suspension of virulent canine parvovirus. Observe the dogs
dog and each blood count, after vaccination, the number of at least daily for 14 days after challenge. At the end of the
leucocytes is not less than 50 per cent of the initial value. observation period, carry out a haemagglutination test for
2-3-1-2 Effects on the thymus. For each test, use not fewer and titration of the virus in the faeces.
than 8 dogs that do not have haemagglutination-inhibiting The test is not valid if fewer than 100 per cent of the control
antibodies against canine parvovirus. Administer to each of dogs show typical signs of the disease andlor leucopenia and
not fewer than 4 dogs a quantity of the vaccine virus excretion of the virus.
equivalent to not less than 10 times the maximum virus titre The vaccine virus complies with the test if all the vaccinated
likely to be contained in 1 dose of the vaccine. Maintain not dogs survive and show no sign of disease nor leucopenia and
fewer than 4 dogs as controls. Observe the dogs at least daily. if the maximum titre of virus excreted in the faeces is less
After 14 days, euthanise 2 dogs from each group and after than 1/100 of the geometric mean of the maximum titres
21 days, the remaining dogs from each group. Carry out found in the controls.
histological examination of the thymus of each dogo
3 BATCH TESTS
The vaccine virus complies with the test if there is no more
3-1 Identification
than slight hypoplasia of the thymus after 14 days and no
The vaccine is grown in a susceptible cell line in a substrate
damage is evident after 21 days.
suitable for presenting for fiuorescent antibody or
2-3-2 Increase in virulence irnmunoperoxidase tests. Suitable controls are included.
Carry out the test according to general chapter 5.2.6 using A proportion of the cells is tested with a monoc1onal
dogs of the minimum age to be recommended for antibody specific for canine parvovirus and a propOltÍon of
vaccination, that do not have haemagglutination-inhibiting the cells tested with a monoc1onal antibody specific for feline
antibodies against canine parvovirus. If the properties of the parvovirus. Canine parvovirus antigen is detected but no
vaccine virus allow sequential passage through 5 groups via feline parvovirus is detected in the cells inoculated with the
natural spreading, this method may be used, otherwise vaccine.
passage as described below is carried out.
3-2 Bacteria and fungi
Administer to each dog of the 1st group by a route to be The vaccine, inc1uding where applicable the diluent supplied
recommended a quantity of the vaccine virus that will allow for reconstitution, complies with the test for sterility
recovery of virus for the passages described below. Collect prescribed in the monograph Vaccines for veterinaJY
the faeces from each dog from the 2nd to the 10th day after use (0062).
administration of the virus, check them for the presence of
3-3 Mycoplasmas (2.6.7)
the virus and pool the faeces containing virus. Administer
The vaccine complies with the test for mycoplasmas.
1 mL of the suspension of pooled faeces by the oro nasal
route to each dog of the next group. Carry out this passage 3-4 Extraneous agents
operation not fewer than 4 times; veri:fy the presence of the Neutralise the vaccine virus with a suitable monospecific
virus at each passage. If the virus is not found at a passage antiserum against canine parvovirus and inoculate into ceH
level, repeat the passage by administration to a group of cultures known for their susceptibility to virus es pathogenic
10 animals. for the dogo
If the 5th group of animals shows no evidence of an increase The vaccine complies with the test if no cytopathic effect
in virulence indicative of reversion during the observation develops and there is no sign of haemagglutinating or
period, further testing is not required. Otherwise, carry out haemadsorbing agents and no other sign of the presence of
an additional safety test and compare the clinical signs and extraneous viruses.
any relevant parameters in a group of at least 8 animal s 3-5 Virus titre
receiving the material used for the 1st passage and another Tiu"ate the vaccine virus by inoculation into suitable cell
similar group receiving the virus at the final passage level. cultures. The vaccine complies with the test if one dose
The vaccine virus complies with the test if no indication of contains not less than the minimum virus tiu"e stated on the
increased virulence of the virus recovered for the final label.
passage compared with the material used for the 1st passage 3-6 Potency
is observed; account is taken, notably, of the count of white The vaccine complies widl the requirements of the test
blood cells, of results of histological examination of the prescribed under Irnmunogenicity (section 2-3-3) when
thymus and of the titre of excreted virus. If virus is not administered by a recommended route and method. It is not
recovered after an initial passage in 2 animals and a necessary to carry out the potency test for each batch of the
subsequent repeat passage in 10 animals, the vaccine virus vaccine if it has been carried out on a representative batch
also complies with the test. using avaccinating dose containing not more than the
2-3-3 ItmTIunogenicity minimum virus titre stated 01.1 the label.
A test is carried out for each route and method of _____________________________________________ PhEw
administration to be recommended using in each case dogs
of the minimum age to be recommended for vaccination.
The quantity of vaccine virus to be administered to each dog
Vet-230 Veterinary Vaccines 2016

*** 3-1 Identification


Clostridium Botulinum Vaccine
********* When injected into a healthy animal free from antibodies
Botulinum Vaccine against the type or types of C. botulinum from which the
(Clostridiu711 Botulinum Vaccine for Veterina/Y Use) vaccine was prepared, the vaccine stimulates the production
Ph Bu¡" monograph 0360) of such antibodies.
When Clostridium Botulinum Vaccine or Botulinum Vaccine 3-2 Bacteria and fungi
is prescribed or demanded and the types to be present are The vaccine, inc1uding where applicable the diluent supplied
not stated, Clostridium Botulinum Vaccine prepared from for reconstitution, complies with the test for sterility
types C and D shall be dispensed or supplied. prescribed in the monograph Vaccines for veterina/Y
PhE~ _____________________________________________ use (0062).
3-3 Residual toxicity
1 DEFINITION Inject 0.5 mL of the vaccine by the subcutaneous route into
Clostridium botulinum vaccine for veterinary use is prepared each of 5 mice, each weighing 17-22 g. Observe the animals
from liquid cultures of suitable strains of Closuidium at least daily for 7 days.
botulinum type C or type D or a mixture of these types.
The vaccine complies with the test if no animal shows
The whole culture or its filtrate or a mixture of the two is
notable signs of disease or dies from causes attributable to
inactivated to eliminate its toxicity while maintaining
the vaccine.
adequate irnmunogenic properties. This monograph applies
to vaccines intended for active irnmunisation of animals 3-4 Potency
against botulismo Use for the test healthy white mice from a unifOlm stock,
each weighing 18-20 g. Use as challenge dose a quantity of a
2 PRODUCTION toxin of C. botulinum of the same type as that used in the
2-1 PREPARATION OF THE VACCINE preparation of the vaccine corresponding to 25 times the
C. botulinum used for production is grown in an appropriate paralytic dose 50 per cent, a paralytic dose 50 per cent being
liquid medium. the quantity of toxin that, when injected by the
The preparation may be adsorbed, precipitated or intraperitoneal route into mice, causes paralysis in
concentrated. It may be treated with a suitable adjuvant and 50 per cent of the animals within an observation period of
may be freeze-dried. 7 days. If 2 types of C. botulinu111 have been used in the
2-2 CHOICE OF VACCINE COMPOSITION preparation of the vaccine, carry out the potency
determination for each. Dilute the vaccine to be examined
The vaccine is shown to be satisfactory with respect to safety
8-fold using a 9 giL solution of sodium chloride R. Inject
(5.2.6) and efficacy (5.2.7) for the animals for which it is
0.2 mL of the dilution subcutaneously into each of 20 mice.
intended.
After 21 days, inject the challenge dose by dle intraperitoneal
The following test for safety (section 2-2-1) may be used route into each of dle vaccinated mice and into each of 10
during the demonstration of safety. control mice. Observe the mice for 7 days and record the
2-2-1 Safety number of animals that show signs of botulismo
Carry out the tests for each route and method of The test is not valid unless all the control mice show signs of
administration to be recommended for vaccination and where botulism during the observation periodo The vaccine
applicable, in animals of each category for which the vaccine complies with the test if not fewer than 80 per cent of the
is intended, using in each case animals not older than the vaccinated mice are protected.
minimum age to be recommended for vaccination. Use a
batch of vaccine containing not less than the maximum 4 LABELLING
potency that may be expected in a batch of vaccine. The label states:
-- the type or types of C. botulinum from which the vaccine
For each test, use not fewer than 8 animal s that do not have
has been prepared;
antibodies against C. botulinum. Administer to each animal
-- whether the preparation is a toxoid or a vaccine prepared
1 dose of the vaccine. If the schedule to be recommended
from a whole inactivated culture or a mixture of the two.
requires a 2nd dose, administer another dose after an interval
_____________________________________________ PhE~

of at least 14 days. Observe the animals at least daily until


14 days after the last administration.
The vaccine complies with the test if no animal shows
abnormal local or systemic reactions or dies from causes
attributable to the vaccine. ***
Clostridium Chauvoei Vaccine
2-3 MANUFACTURER'S TESTS *** ***
2-3-1 Batch potency test Blackleg Vaccine ***
It is not necessary to carry out the potency test (section 3-4) (Closuidium Chauvoei Vaccine for Vete¡ina¡y Use)
for each batch of the vaccine if it has been carried out using Ph Bu/" monograph 0361)
a batch of vaccine with a minimum potency. Where the test PhE~ ____________~-------------------------------
is not carried out, an altemative validated method is used,
the criteria for acceptance being set with reference to a batch 1 DEFINITION
of vaccine that has given satisfactory results in the test Clostridium chauvoei vaccine for veterinary use is prepared
described under Potency. from liquid cultúres of one or more suitable strains of
Clostlidiu111 chauvoei. The whole culture is inactivated to
3 BATCH TESTS eliminate its toxicity while maintaining adequate
The identification) the tests and the dete17nination of potency apply irnmunogenic properties. This monograph applies to vaccines
to the liquid preparation and to the freeze-dried preparation intended for active immunisation of animal s against disease
reconstituted as stated on the label. caused by C. chauvoei.
2016 Veterinary Vaccines Vet-231

2 PRODUCTION euthanised and are then considered to have died from


2-1 PREPARATION OF THE VACCINE C. chauvoei infection.
C chauvoei Used for production is grown in an appropriate _____________________________________________ PhE~

liquid medium. Inactivated cultures may be treated with a


suitable adjuvant.
2-2 CHOICE OF VACCINE COMPOSITION
The vaccine is shown 10 be satisfactory with respect to safety
Clostridium Novyi Type B Vaccine *****
(5.2.6) and efficacy (5.2.7) for the animals for which it is ** **
intended. Black Disease Vaccine ***
The following test for safety (section 2-2-1) may be used (Clostrz'dium Novyi (Type B) Vaccine fo1' Veterina7Y Use,
during the demonstration of safety. Ph Bur 11Zo11ograph 0362)
2-2-1 Safety PhE~ _____________________________________________
Carry out the tests for each route and method of
1 DEFINITION
administration to be recommended for vaccination and where
Clostridium novyi (type B) vaccine for veterinary use is
applicable, in animals of each category for which the vaccine
prepared from a liquid culture of a suitable strain of
is intended, using in each case animal s not older than the
Clostridium 110vyi (type B).
minimum age 10 be recommended for vaccination. U se a
batch of vaccine containing not less than the maximum The whole culture 01' its filtrate 01' a mixture of the two is
potency that may be expected in a batch of vaccine. inactivated to eliminate its toxicity while maintaining
adequate irnmunogenic properties. This monograph applies
For each test, use not fewer than 8 animal s that do not have
to vaccines intended for active irnmunisation of animal s
antibodies against C. chauvoei. Administer to each animal
and/or to protect passively their progeny against disease
1 dose of the vaccine. If the schedule to be recornmended
caused by C. 110vyi (type B).
requires a 2nd dose, administer another dose after an interval
of at least 14 days. Observe the animals at least daily until 2 PRODUCTION
14 days after the last administration. 2-1 PREPARATION OF THE VACCINE
The vaccine complies with the test if no animal shows C 110vyi (type B) used for production is grown in an
abnormal local or systemic reactions 01" dies from causes appropriate liquid medium. Toxoids and/ol' inactivated
attributable to the vaccine. cultures may be treated with a suitable adjuvant, after
concentration if necessary.
3 BATCH TESTS
3-1 Identification 2-2 CHOICE OF VACCINE COMPOSITION
The vaccine protects susceptible animals against infection The vaccine is shown to be satisfac10ry with respect to safety
with C. challvoei. The potency test may also serve for (5.2.6) and efficacy (5.2.7) for the animals for which it is
identification. intended. For the latter, it shall be demonstrated that for
3-2 Bacteria and fungi each target species the vaccine, when administered according
The vaccine, including where applicable the diluent supplied 10 the schedule to be recommended, stimulates an immune
for reconstitution, complies with the test for sterility response (for example, induction of antibodies) consistent
prescribed in the monograph Vaccines for veterina7JI use with the clairns made fol' the producto
(0062). The following test fol' safety (section 2-2-1) may be used
during the demonstration of safety.
3-3 Potency
Use for the test notfewer than 10 healthy guinea-pigs, each 2-2-1 Safety
weighing 350-450 g. Administer to each guinea-pig by the Carry out the tests fol' each route and method of
subcutaneous route a quantity of the vaccine not greater than administration to be recommended for vaccination and where
the minimum dose stated on the label as the 1st dose. After applicable, in animals of each category for which the vaccine
28 days, administer to the same animals a quantity of the is intended, using in each case animals not older than the
vaccine not greater than the minimum dose stated on the minimum age to be recommended fol' vaccination. U se a
label as the 2nd dose. 14 days after the 2nd vaccination, batch of vaccine containing not les s than the maximum
inoculate by the intramuscular route each of the vaccinated potency that may be expected in a batch of vaccine.
guinea-pigs and each of 5 control animals with a suitable For each test, use not fewer than 8 animal s that do not have
quantity of a virulent culture, 01' of a spore suspension, of antibodies against C. novyi (type B). Administer to each
C. chauvoei, activated if necessary with an activating agent animal 1 dose of the vaccine. If the schedule 10 be
such as ca1cium chloride. recommended requil'es a 2nd dose, administer anothel' dose
The vaccine complies with the test if not more than after an interval of at least 14 days. Observe the animals at
10 per cent of the vaccinated guinea-pigs die from least daily until 14 days after the last administration.
C. chauvoei infection within 5 days and all the control The vaccine complies with dle test if no animal shows
animals die from C. chauvoei infection within 48 h of abnormal local 01' systemic reactions 01' dies from causes
challenge 01' within 72 h if a spore suspension was used for attributable to the vaccine. If dle test is carried out in
the challenge. If more than 10 per cent but not more than pregnant animals, no adverse effects on gestation 01' the
20 per cent of the vaccinated animals die, repeat the test. offspring are noted.
The vaccine complies with the test if not more than
2-3 MANUFACTURER'S TESTS
10 per cent of the 2nd group of vaccinated animals die within
2-3-1 Residual toxicity
5 days and all of the 2nd group of control animals die within
A test for detoxification is carried out immediately after the
48 h of challenge or within 72 h if a spore suspension was
de1Oxification process and, when there is risk of revel'sion, a
used for the challenge. To avoid unnecessary suffering
2nd test is can"Íed out at as late a stage as possible during the
following virulent challenge, moribund animals are
Vet-232 Veterinary Vaccines 2016

production process. The test for residual toxicity mice or other suitable animals against the toxic effects of a
(section 3-3) may be omitted by the manufacturer. fixed dose of C. novyi alpha toxin with the quantity of a
2-3-2 Batch potency test reference preparation of Clostridium novyi alpha antitoxin,
It is not necessary to cany out the potency test (section 3-4) calibrated in International Units, necessaty to give the same
for each batch of vaccine if it has been carried out using a protection. For this comparison, a suitable preparation of
batch of vaccine with a minimum potency. C. novyi alpha toxin for use as a test toxin is required.
The dose of the test toxin is determined in relation to the
Where the test is not carried out, an alternative validated
reference preparation; the potency of the serum to be
method is used, the criteria for acceptance being set with
examined is detelmined in relatíon to the reference
reference to a batch of vaccine that has given satisfact01Y
preparation using the test toxin.
results in the test described under Potency and that has been
shown to be satisfactory with respect to immunogenicity in Clostridia (multicomponent) rabbit antiserum BRP is suitable for
the target species. The following test may be used after a use as a reference serum.
satisfactory correlation with the test under Potency 3-4-1 Preparation of test toxin. Prepare the test toxin from a
(section 3-4) has been established. sterile filtrate of an approximately 5-day culture in liquid
Vaccinate rabbits as described under Potency and prepare medium of C. novyi type B and dry by a suitable method.
sera. Determine the level of antibodies against the alpha toxin Select the test toxin by determining for mice the L+/10 dose
of C. novyi in the individual sera by a suitable method such and the LD so , the observation period being 72 h.
as an immunochemical method (2.7.1) or neutralisation in A suitable alpha toxin contains not les s than 1 L+/10 dose in
cell cultures. Use a homologous reference serum calibrated in 0.05 mg and not less than 10 LDso in each L+/10 dose.
International Units of C. novyi alpha antitoxin. Clostridia 3-4-2 Determination of test dose of toxin. Prepare a solution of
(multicomponent) rabbit antiserum BRP is suitable for use as a the reference preparation in a suitable liquid so that it
reference serum. contains 1 IU/mL. Prepare a solution of the test toxin in a
The vaccine complies with the test if the level of antibodies is suítable liquid so that 1 mL contains a precisely known
not less than that found for a batch of vaccine that has given amount such as 1 mg. Prepare mixtures of the solution of the
satisfactory results in the test described under Potency and reference preparation and the solution of the test toxin such
that has been shown to be satisfactory with respect to . that each mixture contains 1.0 mL of the solution of the
immunogenicity in the target species. reference preparation (1 IV), one of a series of graded
volumes of the solution of the test toxin and sufficient of a
3 BATCH TESTS
suitable liquid to bring the total volume to 2.0 mL. Allow the
3-1 Identification
mixtures to stand at room temperature for 60 min. Using not
When injected into animals that do not have novyi alpha
fewer dlan 2 mice, each weighing 17-22 g, for each mixture,
antitoxin, the vaccine stimulates the formation of such
inject adose of 0.2 mL by the intramuscular or the
antitoxins.
subcutaneous route into each mouse. Observe the mice for
3-2 Bacteria and fungi 72 h. If aH dle mice die, the amount of toxin present in
The vaccine, including where applicable the diluent supplied 0.2 mL of the mixture is in excess of the test dose. If none of
for reconstitution, complies with the test for sterility the mice die, the amount of toxin present in 0.2 mL of the
prescribed in the monograph Vaccines for veterina/)I mixture is less than the test dose. Prepare fresh mixtures
use (0062). such that 2.0 mL of each mixture contains 1.0 mL of the
3-3 Residual toxicity solution of the reference preparatíon (1 IU) and one of a
Administer 0.5 mL of the vaccine by the subcutaneous route series of graded volumes of the solution of the test toxin
into each of 5 mice, each weighing 17-22 g. Observe the separated from each other by steps of not more than
animals at least daily for 7 days. 20 per cent and covering the expected end-point. AHow the
The vaccine complies with the test if no animal shows mixtures to stand at room temperature for 60 min. Using not
notable signs of disease or dies from causes attributable to fewer dlan 2 mice for each mixture, inject adose of 0.2 mL
the vaccine. by the intramuscular or the subcutaneous route into each
mouse. Observe the mice for 72 h. Repeat the determination
3-4 Potency
at least once and combine the results of the separate tests
Use for the test not fewer than 10 healthy rabbits, that have been made with mixtures of the same composition
3-6 months old. Administer to each rabbit by the so that a series of total s ís obtained, each total representing
subcutaneous route a quantity of vaccine not greater than the the mortality due to a mixture of a given composition.
minimum dose stated on the label as dle 1st dose. After
21-28 days, administer to the same animals a quantity of the The test dose of toxin is the amount present in 0.2 mL of
vaccine not greater than the minimum dose stated on the that mixture whích causes the death of one half of the total
number of mice injected with it.
label as the 2nd dose. 10-14 days after the 2nd injection, bleed
the rabbits and pool the sera. 3-4-3 Determination of the potency of the seru71l obtained fmm
rabbits
The vaccine complies with the test if the potency of the
pooled sera is not less than 3.5 IU/mL. PreliminaJ)I test Dissolve a quantíty of the test toxin in a
The International Unit is the specific neutralising activity for suitable liquid so that 1.0 mL contains 10 times the test dose
C. novyi alpha toxin contained in a stated amount of the (solution of the test toxin). Prepare a series of mixtures of the
solution of the test toxin and of the serum to be examÍned
International Standard, which consists of a quantity of dried
immune horse serum. The equivalence in International Units such that each mixture contains 1.0 mL of the solutíon of the
of the International Standard is stated by the World Health test toxin, o:ne of a series of graded volumes of the serum to
Organization. be examined and sufficient of a suitable liquid to bring the
final volume to 2.0 mL. Allow the mixtures to stand at room
The potency of the pooled sera obtained from dle rabbits is temperature for 60 min. Using not fewer than 2 mice for
determined by comparing the quantity necessary to protect each mixture, inject adose of 0.2 mL by the intramuscular
2016 Veterinary Vaccines Vet-233

or the subcutaneous route into each mouse. Observe the to vaccines intended for active irnmunisation of animals
mice for 72 h. If none of the mice die, 0.2 mL of the mixture and/or to protect passively their progeny against disease
contains more than 0.1 IV. If aH the mice die, 0.2 mL of the caused by C. perfringens.
mixture contains less than 0.1 IU.
2 PRODUCTION
Final test Prepare a series of mixtures of the solution of the 2-1 PREPARATION OF THE VACCINE
test toxin and the serum to be examined such that 2.0 mL of C pelfringens Used for production is grown in an appropriate
each mixture contains 1.0 mL of the solution of the test liquid medium. Toxoids andlor inactivated cultures may be
toxin and one of a series of graded volumes of the serum to treated with a suitable adjuvant.
be examined, separated from each other by steps of not more
than 20 per cent and covering the expected end-point as 2-2 CHOICE OF VACCINE COMPOSITION
determined by the preliminary test. Prepare further mixtures The vaccine is shown to be satisfactory with respect to safety
of the solution of the test toxin and of the solution of the (5.2.6) and efficacy (5.2.7) for the animals for which it is
reference preparation such that 2.0 mL of each mixture intended. For the latter, it shaH be demonstrated tllat for
contains 1.0 mL of the solution of the test toxin and one of a each target species the vaccine, when administered according
series of graded volumes of the solution of the reference to the schedule to be recommended, stimulates an immune
preparation, in order to confirm the test dose of the toxin. response (for example, induction of antibodies) consistent
Allow the mixtures to stand at room temperature for 60 min. with the claims made for the product.
U sing not fewer than 2 mice for each mixture, proceed as The following test for safety (section 2-2-1) may be used
described in the preliminary test. during the demonstration of safety.
The test mixture that contains 0.1 IU in 0.2 mL is that 2-2-1 Safety
mixture which kills the same or almost the same number of Carry out the tests for each route and method of
mice as the reference mixture containing 0.1 IU in 0.2 mL. administration to be recommended for vaccination and where
Repeat the determination at least once and calculate the applicable, in animals of each category for which the vaccine
average of aH valid estimates. The test is valid only if the is intended, using in each case animals not older than tlle
reference preparation gives a result within 20 per cent of the minimum age to be recommended for vaccination. Use a
expected value. batch of vaccine containing not less than the maximum
The confidence limits (P = 0.95) have been estimated to be: potency that may be expected in a batch of vaccine.
-- 85 per cent and 114 per cent when 2 animals per dose For each test, use not fewer tllan 8 animal s that do not have
are used; antibodies against C. perfiingens. Administer to each animal
-- 91.5 per cent and 109 per cent when 4 animals per dose 1 dose of the vaccine. If the schedule to be recornmended
are used; requires a 2nd dose, administer another dose after an interval
-- 93 per cent and 108 per cent when 6 animal s per dose of at least 14 days. Observe the animal s at least daily until
are used. 14 days after the last administration.
4 LABELLING The vaccine complies with tlle test if no animal shows
The label states: abnormal local or systemic reactions 01' dies from causes
-- whether the product is a toxoid, a vaccine prepared from attributable to the vaccine. If the test is carried out in
a whole inactivated culture or a mixture of the two; pregnant animals, no adverse effects on gestation 01' tlle
-- for each target species, the irnmunising effect produced offspring are noted.
(for example, antibody production, protection against 2-3 MANUFACTURER'S TESTS
signs of disease or infection). 2-3-1 Residual toxicity
_____________________________________________ PhEw A test for detoxification is carried out immediately after tlle
detoxification process and, when tllere is risk of reversion, a
2nd test is carried out at as late a stage as possible during the
production process. The test for residual toxicity
(section 3-3) may be omitted by the manufacturero
Clostridium Perfringens Vaccines 2-3-2 Batch potency test
(Clostridium Perfiingens Vaccine for Veterinary Use) It is not necessary to carry out tlle potency test (section 3-4)
Ph Bur monogmph 0363) for each batch of vaccine if it has been carried out using a
batch of vaccine with a minimum potency. Where the test is
The foHowing titles may be used for appropriate vaccines:
not carried out, an alteni.ative validated method is used, the
Clostridium Perfringens Type B Vaccine; Lamb Dysentery criteria for acceptance being set Witll reference to a batch of
Vaccine; vaccine that has given satisfactory results in tlle test described
Clostridium Perfringens Type C Vaccine; Struck Vaccine; under Potency and that has been shown to be satisfactory
Clostridium Perfringens Type D Vaccine; Pulpy Kidney with respect to irnmunogenicity in the target species.
Vaccine The following test may be used after a satisfactory correlation
~Ew _____________________________________________ with the test under Potency (section 3-4) has been
established.
1 DEFINITION Vaccinate rabbits as described under Potency and prepare
Clostridium perfringens vaccine for veterinary use is prepared sera. Determine the leve! of antibodies against the beta
from liquid cultures of suitable strains of Clostlidium and/or 'epsilon toxins of C. perfringens in the individual sera
perfiingens type B, C. pelfringens type C or C. perfringens by a suitable metllod such as an immunochemical method
type D or a mixture of these types. (2.7.1) 01' neutralisation in cell cultures. Use a homologous
The whole cultures or their filtrates or a mixture of the two reference serum calibrated in International Units of
are inactivated to eliminate their toxicity while maintaining C. perfiingens beta andlor epsilon antitoxin. Clostridia
adequate irnmunogenic properties. This monograph applies
Vet-234 Veterinary Vaccines 2016

(multic0111ponem) rabbit antiserum BRP is suitable for use as a Units of the Intemational Standard, is stated by the World
reference serum. Health Organization.
The vaccine complies with the test if the level or levels of The potency of the pooled sera obtained from the rabbits is
antibodies are not less than that found for a batch of vaccine detelmined by comparing the quantity necessary to protect
that has given satisfactory results in the test described under mice 01' other suitable animals against the toxic effects of a
Potency and that has been shown to be satisfactory with fixed dos e of C. perfringens beta toxin or C. perfringens epsilon
respect to immunogenicity in the target species. toxin with the quantity of a reference preparation of
3 BATCH TESTS Clostridium perfringens beta antitoxin 01' Clostridium
perfringens epsilon antitoxin, as appropriate, calibrated in
3-1 Identification
Intemational Units, necessary to give the same protection.
Type B When injected into animals that do not have beta and
For this comparison, a suitable preparation of C. perfringens
epsilon antitoxins, the vaccine stimulates the formation of
beta 01' epsilon toxin for use as a test toxin is required.
such antitoxins.
The dose of the test toxin is determined in relation to the
Type C When injected into animals that do not have beta appropriate reference preparation; the potency of the serum
antitoxin, the vaccine stimulates the formation of such to be examined is determined in relation to the appropriate
antitoxin. reference preparation using the appropriate test toxin.
Type D When injected into animals that do not have epsilon Clostridia (multicomponent) rabbit antiserum BRP is suitable for
antitoxin, the vaccine stimulates the formation of such use as a reference serum.
antitoxin.
3-4-3 Preparation of test toxin. Prepare the test toxin from a
3-2 Bacteria and fungi sterile filtrate of an early culture in liquid medium of
The vaccine, including where applicable the diluent supplied C. perfringens type B, type C 01' type D as appropriate and
for reconstitution, complies with the test for sterility dry by a suitable method. Use a beta 01' epsilon toxin as
prescribed in the monograph Vaccines for veterinaiY use appropriate. Select the test toxin by detetmining for mice the
(0062). L+ and the LDso for the beta toxin and the L+/10 dose and
3-3 Residual toxicity the LDso for the epsilon toxin, the observation period being
Administer 0.5 mL of the vaccine by the subcutaneous route 72 h.
to each of 5 mice, each weighing 17-22 g. Observe the mice A suitable beta toxin contains not less than 1 L+ in 0.2 mg
at least daily for 7 days. and not les s than 25 LDso in 1 L+ dose. A suitable epsilon
The vaccine complies with the test if no animal shows toxin contains not less than 1 L+/10 dose in 0.005 mg and
notable signs of disease or dies from causes attributable to not less than 20 LDso in 1 L+11 O dose.
the vaccine. 3-4-4 Determination of test dose of toxin. Prepare a solution of
3-4 Potency the reference preparation in a suitable liquid so that it
Use for the test not fewer than 10 healthy rabbits, contains 5 IU/rnL for Clostridium perfringens beta antitoxin
3-6 months old. Administer to each rabbit by the and 0.5 IU/mL for Clostridium perfringens epsilon antitoxin.
subcutaneous route a quantity of vaccine not greater than the Prepare a solution of the test toxin in a suitable liquid so that
minimum dose stated on the label as the 1st dose. After 1 mL contains a precisely known amount such as 10 mg for
21-28 days, administer to the same animals a quantity of the beta toxin and 1 mg for epsilon toxin. Prepare mixtures of
vaccine not gn;ater than the minimum dose stated on the the solution of the reference preparation and the solution of
label as the 2nd dose. 10-14 days after the 2nd injection, bleed the test toxin such that each contains 2.0 mL of the solution
the rabbits and pool the sera. of the reference preparation, one of a series of graded
Type B The vaccine complies with the test if the potency of volumes of the solution of the test toxin and sufficient of a
the pooled sera is not less than 10 IU of beta antitoxin and suitable liquid to bring the total volume to 5.0 mL. Allow the
not less than 5 IU of epsilon antitoxin per millilitre. mixtures to stand at room temperature for 30 mino Using not
fewer than 2 mice, each weighing 17-22 g, for each mixture,
Type C The vaccine complies with the test if the potency of
inject adose of 0.5 mL by the intravenous 01' the
the pooled sera is not less than 10 IU of beta antitoxin per
inu'aperitoneal route into each mouse. Observe the mice for
milliliu·e.
72 h. If all the mice die, the amount of toxin present in
Type D The vaccine complies with the test if the potency of 0.5 mL of the mixture is in excess of the test dose. If none of
the pooled sera is not less than 5 IU of epsilon antitoxin per the mice die the amount of toxin present in 0.5 mL of the
millilitre. mixture is less than the test dose. Prepare fresh mixtures
3-4-1 Interna tia n al standard for Clostridium perfringens beta such that 5. O mL of each mixture contains 2. O mL of the
antitoxin solution of the reference preparation and one of a series of
The Intemational Unit is the specific neutralising activity for graded volumes of the solution of the test toxin separated
C. pe¡jringens beta toxin contained in a stated amount of the from each other by steps of not more than 20 per cent and
Intemational Standard, which consists of a quantity of dried covering the expected end-point. Allow the mixtures to stand
irnmune horse serum. The equivalence in Intemational Units at room temperature for 30 mino U sing not fewer than
of the Intemational Standard is stated by the World Health 2 mice for each mixture, inj~ct adose of 0.5 mL by the
Organization. intravenous 01' the intraperitoneal route into each mouse.
3-4-2 International standard for Clostridiu111 perfringens epsilon Observe the mice for 72 h. Repeat the determination at least
antitoxin once and add ~ogether the results of the separate tests that
have been made with mixtures of the same composition so
The Intemational Unit is the specific neutralising activity for
that a series of total s is obtained, each total representing the
C. perfringens epsilon toxin contained in a stated amount of
mortality due to a mixture of given composition.
the Intemational Standard, which consists of a quantity of
dried immune horse serum. The equivalence in Intemational
2016 Veterinary Vaccines Vet-235

The test dose of toxin is the amount present in 0.5 mL of


Clostridium Septicum Vaccine ***
that mixture which causes the death of one half of the total *** ***
number of mice injected with it. Braxy Vaccine ***
3-4-5 Determination of the potency of the senmz obtained fr0111 (Clostridiu111 Septicum Vaccine for VeterinalY Use,
rabbíts Ph Bur monograph 0364)
PreliI1linaly test Dissolve a quantity of the test toxin in a PhE~ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ ____
suitable liquid so that 2.0 mL contains 10 times the test dose
(solution of the test toxin). Prepare a series of mixtures of the 1 DEFINITION
solution of the test toxin and of the serum to be examined Clostridium septicum vaccine for veterinary use is prepared
such that each contains 2.0 mL of the solution of the test from a liquid culture of a suitable strain of Clostridium
toxin, one of a series of graded volumes of the serum to be septicum.
examined and sufficient of a suitable liquid to bring the final The whole culture or its filtra te or a mixture of the two is
volume to 5.0 mL. A1low the mixtures to stand at room inactivated to eliminate its toxicity while maintaining
temperature for 30 min. Vsing not fewer than 2 mice for adequate immunogenic properties. This monograph applies
each mixture, inject adose of 0.5 mL by the intravenous or to vaccines intended for active immunisation of animal s
the intraperitoneal route into each mouse. Observe the mice and/or to protect passively their progeny against disease
for 72 h. If none of the mice die, 0.5 mL of the mixture caused by C. septicu11l.
contains more than 1 IV of beta antitoxin or 0.1 IV of 2 PRODUCTION
epsilon antitoxin. If all the mice die, 0.5 mL of the mixture 2-1 PREPARATION OF THE VACCINE
contains less than 1 IV ofbeta antitoxinor 0.1 IV of epsilon
C septicuJ11 used for production is grown in an appropriate
antitoxin.
liquid medium. Toxoid andlor inactivated cultures may be
Final test Prepare a series of mixtures of the solution of the treated with a suitable adjuvant.
test toxin and the serum to be examined such that 5.0 mL of
each mixture contains 2.0 mL of the solution of the test 2-2 CHOICE OF VACCINE COMPOSITION
t~xin and one of a series of graded volumes of the serum to The vaccine is shown to be satisfactory with respect to safety
be examined separated from each ot1ler by steps of not more (5.2.6) and efficacy (5.2.7) for the animals for which it is
than 20 per cent and covering the expected end-point as intended. For the latter, it shall be demonstrated that for
determined by the preliminary test. Prepare further mixtures each target species the vaccine, when administered according
of the solution of the test toxin and of the solution of the to the schedule to be recommended, stimulates an immune
reference preparation such that 5.0 mL of each mixture response (for example, induction of antibodies) consistent
contains 2.0 mL of the solution of the test toxin and one of a with the claims made for the product.
series of graded volumes of the solution of the reference The following test for safety (section 2-2-1) may be used
preparation, in order to confirm the test das e of the toxin. during the demonstration of safety.
Allow the mixtures to stand at room temperature for 30 min. 2-2-1 Safety
U sing not fewer than 2 mice for each mixture proceed as Carry out the tests for each route and method of
described in the preliminary test. administration to be recommended for vaccination and where
Beta antitoxin The test mixture that contains 1 IU in 0.5 mL applicable, in animals of each category for which the vaccine
is that mixture which kills the same or almost the same is intended, using in each case animals not older than the
number of mice as the reference mixture containing 1 IU in minimum age to be recommended for vaccination. U se a
0.5 mL. batch of vaccine containing not less than the maximum
Epsilon antitoxin The test mixture that contains 0.1 IU in potency that may be expected in a batch of vaccine.
0.5 mL is that mixture which IdUs the same or almost the For each test, use not fewer than 8 animals that do not have
same number of mice as the reference mixture containing antibodies against C. septicum. Administer to each animal
0.1 ID in 0.5 mL. Repeat the determination at least once 1 dose of the vaccine. If the schedule to be recommended
and calculate the average of all valid estimates. The test is requires a 2nd dose, administer another dose after an interval
valid only if the reference preparation gives a result within of at least 14 days. Observe the animals at least daily until
20 per cent of the expected value. 14 days after the last administration.
The confidence limits (P = 0.95) have been estimated to be: The vaccine complies with the test if no animal shows
- 85 per cent and 114 per cent when 2 animals per dose abnormallocal or systemic reactions or dies from causes
are used; attributable to the vaccine. If the test is carried out in
91.5 per cent and 109 per cent when 4 animals per dose pregnant animals, no adverse effets on gestation or the
are used; offspring are noted.
93 per cent and 108 per cent when 6 animals per dose 2-3 MANUFACTURER'S TESTS
are used.
2-3-1 Residual toxicity
4 LABELLING A test for detoxification is carried out immediately after the
The label states: detoxification process and, when there is risk of reversion, a
- whether the preparation is a toxoid or a vaccine prepared 2nd test is· carried out at as late a stage as possible during the
from a whole inactivated culture or a mixture of the two; production process. The test for residual toxicity
- for each target species, the immunising effect produced (section 3-3) may be omitted by the manufacturer.
(for example, antibody production, protection against 2-3-2 Batch potency test
signs of disease or infection). It is not necessary to carry out the potency test (section 3-4)
_ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ PhEur for each batch of vaccine if it has been carried out using a
batch of vaccine with a minimum potency. Where the test is
not carried out, an alternative validated method is used, the
Vet-236 Veterinary Vaccines 2016

criteria for acceptance being set with reference to a batch of Clostridia (multícomponent) rabbít antiserum BRP is suitable for
vaccine that has given satisfactory results in the test described use as a reference serum.
under Potency and that has been shown to be satisfactory 3-4-1 Preparatíon of test toxin. Prepare the test toxin from a
with respect to immunogenicity in the target species. stelile filtrate of a 1- to 3-day culture of C. septicum in liquid
The following test may be used after a satisfactory correlation medium and dry by a suitable method. Select the test toxin
with the test under Potency (section 3-4) has been by detelmining for mice the L+/5 dose and the LD so , the
established. observation period being 72 h.
Vaccinate rabbits as described under Potency and prepare A suitable toxin contains not less than 1 L+/5 dose in 1.0 mg
sera. Determine the level of antibodies against the toxin of and not les s than 10 LD so in each L+/5 dose.
C. septícu71l in the individual sera by a suitable method such
3-4-2 Determinatíon of test dose of toxil1. Prepare a solution of
as an immunochemical method (2.7.1) or neutralisation in
d1e reference preparation in a suitable liquid so that it
cell cultures. Use a homologous reference serum calibrated in
contains 1.0 IU/mL. Prepare a solution of the test toxin in a
Intemational Units of C. septicw1Z antitoxin. Clostridia
suitable liquid so that 1 mL contains a precisely known
(multícomponent) rabbít antíserum BRP is suitable for use as a
amount, such as 4 mg. Prepare mixtures of the solution of
reference serUffi. The vaccine complies with the test if the
the reference preparation and the solution of the test toxin
level of antibodies is not les s than that found for a batch of
such that each mixture contains 2.0 mL of the solution of the
vaccine that has given satisfactory results in the test described
reference preparation (2 IU), one of a series of graded
under Potency and that has been shown to be satisfactory
volumes of the solution of the test toxin and sufficient of a
with respect to immunogenicity in the target species.
suitable liquid to bring the total volume to 5.0 mL. Allow the
3 BATCH TESTS mixtures to stand at room temperature for 60 mino Using not
3-1 Identification fewer than 2 mice, each weighing 17-22 g, for each mixture,
When injected into animals that do not have C. septicum inject adose of 0.5 mL by the intravenous 01' the
antitoxin, the vaccine stimulates the formation of-such intraperitoneal route into each mouse. Observe the mice for
antitoxin. 72 h. If all the mice die, the amount of toxin present in
3-2 Bactetia and fungi 0.5 mL of the mixture is in excess of d1e test dose. If none of
The vaccine, including where applicable !he diluent supplied the mice die, the amount of toxin present in 0.5 mL of the
for reconstitution, complies with the test for sterility mixture is less than the test dose. Prepare fresh mixtures
prescribed in the monograph VacGÍnes for veterinary use such that 5. O mL of each mixture contains 2. O mL of the
(0062). reference preparation (2 IV) and one of a series of graded
volumes of the solution of d1e test toxin separated from each
3-3 Residual toxicity other by steps of not more than 20 per cent and covering d1e
Inject 0.5 mL of the vaccine by the subcutaneous route into expected end-point. Allow the mixtures to stand at room
each of 5 mice, each weighing 17-22 g. Observe the mice at temperature for 60 mino Using not fewer than 2 mice for
least daily for 7 days. each mixture, inject adose of 0.5 mL by d1e intravenous 01'
The vaccine complies with the test if no animal shows the intraperitoneal route into each mouse. Observe the mice
notable signs of disease or dies from causes attributable to for 72 h. Repeat d1e detelmination at least once and add
the vaccine. together d1e results of the separate tests that have been made
3-4 Potency with mixtures of the same composition so that a series of
Use for the test not fewer than 10 healthy rabbits, totals is obtained, each total representing the mortality due to
3-6 months old. Administer to each rabbit by the a mixture of a given composition.
subcutaneous route a quantity of vaccine not greater than the The test dose of toxin is the amount present in 0.5 mL of
minimum dos e stated on the label as the 1sr dose. After that mixture which causes the death of one half of the total
21-28 days, administer to the same animal s a quantity of the number of mice injected wid1 it.
vaccine not greater than the minimum dose stated on the 3-4-3 Determínatíon of the potencJl of the serum obtained fmm
label as the 2nd dose. 10-14 days after the 2nd injection, bleed rabbits
the rabbits and pool the sera.
Preliminmy test Dissolve a quantity of the test toxin in a
The vaccine complies with the test if the potency of the suitable liquid so that 2.0 mL contains 10 times the test dose
pooled sera is not less than 2.5 IU/mL. (solution of the test toxin). Prepare a series of mixtures of the
The Intemational Unit is the specific neutralising activity for solution of the test toxin -and of the serum to be examined
C. septícum toxin contained in a stated amount of the such that each contains 2.0 mL of the solution of the test
Intemational Standard, which consists of a quantity of dried toxin, one of a series of graded volumes of the serum to be
immune horse serum. The equivalence in Intemational Units examined and sufficient of a suitable liquid to bring the final
of the Intemational Standard is stated by the World Health volume to 5.0 mL. Allow the mixtures to stand at room
Organization. temperature for 60 mino Using not fewer than 2 mice for
The potency of the pooled sera obtained from the rabbits is each mixture, inject adose of 0.5 mL by the intravenous 01'
determined by comparing the quantity necessary to protect the intraperitoneal route into each mouse. Observe d1e mice
mice or other suitable animals against the toxic effects of a for 72 h. If none of the mice die, 0.5 mL of the mixture
dos e of C. septicul1l toxin with the quantity of a reference contains more than 0.2 IV. If aH the mice die, 0.5 mL of the
preparation of Clostlidium septicum antitoxin, calibrated in mixture contains less than 0.2 IU.
Intemational Units, necessary to give the same protection. Final test Prepare a series of mixtures of the solution of the
For this compmison, a suitable preparation of C. septicu17l test toxin and of the serum to be examined such that 5.0 mL
toxin for use as a test toxin is required. The dos e of the test of each mixture contains 2.0 mL of the solution of the test
toxin is determined in relation to the reference preparation; toxin and one of a series of graded volumes of the serum to
the potency of the serum to be examined is determined in be examined, separated from each other by steps of not more
relation to d1e reference preparation using the test toxin. than 20 per cent and covering the expected end-point as
2016 Veterinary Vaccines Vet-237

detennined by the preliminalY test. Prepare further mixtures (section 2-2-3) may be used during demonstration of safety
of the solution of the test toxin and of the solution of the and efficacy.
reference preparation such that 5.0 mL of each mixture The C. tetani strain used in the preparation of d1e vaccine is
contains 2.0 mL of the solution of the test toxin and one of a shown to be satisfactOlY with respect to the production of the
series of graded volumes of the solution of the reference neurotoxin.
preparation to confirm the test dose of the toxin. Allow the
2-2-1 Production of antigens
mixtures to stand at room temperature for 60 mino Vsing not
The production of the neurotoxin of C. tetani is verified by a
fewer than 2 mice for each mixture proceed as described in
suitable immunochemical method (2.7.1) carried out on the
the preliminary test. The test mixture which contains 0.2 IV
neurotoxin obtained from the vaccine strain under the
in 0.5 mL is that mixture which ldlls the same or almost the
conditions used for the production of the vaccine.
same number of mice as the reference mixture containing
0.2 IV in 0.5 mL. Repeat the detennination at least once 2-2-2 Safety
and calculate the average of all valid estimates. The test is Carry out the test for each route and method of
valid only if the reference preparation gives a result within administration to be recommended for vaccination and where
20 per cent of the expected value. applicable, in animals of each category for which the vaccine
is intended, using in each case animal s not older than the
The confidence limits (P = 0.95) have been estimated to be:
minimum age to be recommended for vaccination and of the
85 per cent and 114 per cent when 2 animals per dose
most sensitive category for the species. Vse a batch of
are used;
vaccine containing not less than the maximum potency that
91.5 per cent and 109 per cent when 4 animals per dose
may be expected in a batch of vaccine.
are used;
-- 93 per cent and 108 per cent when 6 animals per dose For each test use not fewer d1an 8 animals, free from
are used. antitoxic antibodies. Administer to each animal 1 dose of
vacCine. If the schedule to be recommended requires a 2 nd
4 LABELLING dose, administer another dose after an interval of at least
The .label states: 14 days. Observe the animals at least daily until at least
-- whether the preparation is a toxoid or a vaccine prepared 14 days after d1e last administration.
from a whole inactivated culture, or a mixture of the two;
The vaccine complies with the test if no animal shows
-- for each target species, the irnmunising effect produced
abnormal local or systemic reactions or dies from causes
(for example, antibody production, protection against
attributable to the vaccine. If d1e test is carried out in
signs of disease or infection).
pregnant animals, no adverse effects on gestation or d1e
_____________________________________________ ~E~

offspring are noted.


2-2-3 Irnmunogenicity
2-2-3-1 1717 J1nm oge nicity test in the target species
It shall be demonstrated for each target species that the
Clostridium Tetani Vaccines ***** vaccine, when administered according to d1e schedule to be
** ** recommended and by the route to be recommended,
Tetanus Toxoids (Veterinary) *** stimulates an immune response (for example, induction of
(Tetanus Vaccine for VeterinaJY Use, Ph Bur 71l0nograph 0697) antitoxic antibodies 01' induction of protective levels of
The name Clostridium Tetani Vaccine for Equidae (Tetanus antitoxic antibodies) consistent wid1 the claims made for the
Toxoid for Equidae) may be used for vaccines with an producto
appropriate potency. 2-2-3-2 1m71lunogenicity test in guinea-pigs
When Tetanus Toxoid is demanded for veterinary use, Administer 1 dos e of vaccine by the subcutaneous route to
Clostridium Tetani Vaccine shall be supplied. each of at least 5 guinea-pigs that do not have antibodies
PhE~ _____________________________________________ against the neurotoxin of C. tetani. After 28 days, administer
again to each guinea-pig 1 dose by d1e subcutaneous route.
1 DEFINITION 14 days after the 2nd dose, collect blood from each guinea-
Tetanus vaccine for veterinary use is a preparation of d1e pig and prepare semm samples. Detelmine for each serum
neurotoxin of Clostridium tetani inactivated to eliminate its the titre of antibodies against d1e neurotoxin of C. tetani
toxicity while maintaining adequate immunogenic properties. using a suitable immunochemical method (2.7.1) such as a
The vaccine may be used to induce active and/or passive toxin-binding-inl1ibition test (ToBI test) and a homologous
irnmunity. reference serum. Detennine the average antibody titre of the
2 PRODUCTION serum samples.
2-1 PREPARATION OF THE VACCINE Clostrzdium tetani guinea-pig antiserum for vaccines for veterinalY
C. tetani used for production is grown in an appropriate use BRP is suitable for use as a reference serum.
liquid medium. The toxin is purified and then detoxified or it Tetanus vaccine intended for use in animals other than
may be detoxified before purification. The antigenic purity is hOl"SeS complies with the test if the average antibody titre is
detennined in Lf units of tetanus toxoid per milligram of not less than 7.5 IV/rnL.
protein and shown to be not less than the value approved for Tetanus vaccine intended for use in hOl"SeS complies with the
the particular producto test if ,the average antibody titre is not less than 30 IV/mL.
2-2 CHOICE OF VACCINE COMPOSITION For tetanus vaccine presented as a combined vaccine for use
The vaccine is shown to be satisfactOlY wid1 respect to safety in animals other than horses, d1e above test may be carried
(5.2.6) and efficacy (5.2.7) for the animals for which it is out in susceptible rabbits instead of guinea-pigs. The vaccine
intended. The following tests for production of antigens complies with the test if the average antibody titre of the sera
(section 2-2-1), safety (section 2-2-2) and immunogenicity of d1e vaccinated rabbits is not less than 2.5 IV/rnL.
Vet-238 Veterinary Vaccines 2016

Clostridia (multicomponent) rabbit antiseruJ1Z BRP and


Clostridium tetani rabbit antiserll1n BRP are suitable for use as
Coccidiosis Vaccine (live) for
reference sera. Chickens
2-3 MANUFACTURER'S TESTS (Ph. Eur. monograph 2326)
2 -3 -1 Absence of toxin and irreversibility of toxoid PhE~ _____________________________________________
Carry out a test for reversion to toxicity on the detoxified
1 DEFINITION
harvest using 2 groups of 5 guinea-pigs, each weighing
350-450 g; if the vaccine is adsorbed, carry out the test with Coccidiosis vaccine (live) for chickens is a preparation of
the shortest practical time interval before adsorption. Prepare sporulated oocysts of a suitable line or lines of species of
a dilution of the detoxified harvest so that the guinea-pigs coccidial parasites (Eimeria spp.). This monograph applies to
each receive 10 times the amount of toxoid (measured in vaccines intended for administration to chickens for active
Lf units) that will be present in adose of vaccine. Divide the irnmunisation.
dilution into 2 equal parts. Keep 1 part at 5 ± 3 oC and the 2 PRODUCTION
other at 37 oC for 6 weeks. Attribute each dilution to a 2-1 PREPARATION OF THE VACCINE
separate group of guinea-pigs and inject into each guinea-pig Oocysts are produced in chickens from a flock free from
the dilution attributed to its group. Observe the animal s at specified pathogens (SPF) (5.2.2) or in embryonated hens'
least daily for 21 days. The toxoid complies with the test if eggs from an SPF flock (5.2.2). The eggs must be subject to
no guinea-pig shows signs of disease or dies from causes disinfection and/or incubation conditions validated to ensure
attributable to the neurotoxin of C. tetani. the inactivation of any Eimeria that may be on the shel1s.
2-3-2 Batch potency test The hatched chickens must then be reared in disinfected
It is not necessary to carry out the potency test (section 3-4) premises, in isolation conditions that ensure no infection with
for each batch of vaccine if it has been carried out using a Eimeria. The chickens must not have been treated with
batch of vaccine with a minimum potency. Where the test is coccidiostats. Oocysts are collected from faeces or contents of
not carried out~ an alternative validated method is used, the the intestinal tract of infected chickens during the patent
criteria for acceptance being set with reference to a batch of periodo Oocysts of different Eimeria lines are produced
vaccine that has given satisfactory results in the test described separately. Oocysts are isolated, purified, disinfected,
under Potency. sporulated and counted. The vaccine is produced by
Where the test described under Potency is used as the batch blending defined numbers of sporulated oocysts of each line
potency test, the vaccine complies with the test if the in a suitable medium.
antibody titre in International Units is not les s than that 2-2 SEED LOTS
found for a batch of vaccine shown to be satisfactory with 2-2-1 Identification
respect to irnmunogenicity in the target species. The identity of each Eimeria master seed is established from
3 BATCH TESTS the characteristics of the coccidia produced from it, based on
3-1 Identification an appropriate selection of the following characteristics: size
1f the nature of the adjuvam allozvs it, can)! out test A. Otherzvise and shape of the oocyst; localisation of the developmental
carry out test B. stages in the chicken intestine; pathognomonic lesions
A. Dissolve in the vaccine sufficient sodium citrate R to give a (E. ten ella, E. acervulina, E. necatlix, E. maxima and
100 giL solution. Maintain the solution at 37 oC for about E. brunettz) and lack of macroscopic lesions (E. praecox and
16 h and centrifuge until a clear supernatant is obtained. E. 111itis); size of schizonts in the intestinal mucosa; size of
The supernatant reacts with a suitable tetanus antitoxin, gametocytes in the mucosa; differences in the electrophoretic
giving a precipita te. mobilities of certain isoenzymes, e.g. lactate dehydrogenase
B. When injected into animal s that do not have antibodies and glucose phosphate isomerase; and by the use of
against the neurotoxin of C. tetani, the vaccine stimulates the molecular biology techniques. Artificially attenuated lines
production of such antibodies. may be distinguished from the parent strains by studying
parameters appropriate to the method of attenuation.
3-2 Bacteria and fungi
The vaccine, including where applicable the diluent supplied 2-2-2 Extraneous agents
for reconstitution, complies with the test for sterility Carry out tests 1-6 of chapter 2.6.24. Avian viral vaccines:
prescribed in the monograph Vaccines for veterinalY tests for extraneous agents in seed lots. General provisions a-d, f
use (0062). and h and section 7 of chapter 2.6.24 are also applicable.
3-3 Residual toxicity In these tests on the master seed lot, use organisms that are
Administer 5 mL of the vaccine by the subcutaneous route, not more than 5 passages from the master seed lot at the
as 2 equal divided doses at separate sites, into each of start of the tests. Each master seed lot complies with the
5 healthy guinea-pigs, each weighing 350-450 g, that have requirements of each test.
not previously been treated with any material that will 2-3 CHOICE OF VACCINE COMPOSITION
interfere with the test. The vaccine complies with the test if Only coccidial lines shown to be satisfactory with respect to
no animal shows notable signs of disease or dies from causes residual pathogenicity and increase in virulence may be used
attributable to the vaccine. If within 21 days of the injection in the preparation of the vaccine, and the tests described
any of the animal s shows signs of or dies from tetanus, the below (sections 2-3-2 and 2-3-3) may be used to
vaccine do es not comply with the test. If more than 1 animal demonstrate this. The vaccine shall be shown to be
dies from non-specific causes, repeat the test. If any animal satisfactory with respect to safety (5.2.6) and efficacy (5.2.7)
dies in the 2nd test, the vaccine does not comply with the test. for the chickens· for which it is intended. The following tests
3-4 Potency under Specific test for the safety of the vaccine composition
The vaccine complies with the requirements of the test (section 2-3-1) and Immunogenicity (section 2-3-4) may be
mentioned under Irnmunogenicity (section 2-2-3-2). used during the demonstration of safety and efficacy.
_____________________________________________ ~E~
2016 Veterinary Vaccines Vet-239

2-3-1 Specific test for the safety ofthe vaccine 2 Lesions are much closer together, but not coalescent;
composition lesions may extend as far posterior as 20 cm below the
Carry out the test with a preparation containing oocysts of duodenum in 3-week-old birds. The intestinal walls show
each species at the least attenuated passage level that will be no thickening. Digestive tract contents are nOlmal.
present in a batch of vaccine. U se not fewer than 10 chickens 3 Lesions are numerous enough to cause coalescence with
from an SPF fiod: (5.2.2). The chickens must be hatched reduction in lesion size and give the intestine a coated
and reared as described in section 2-1 and must not have appearance. The intestinal wall is thickened and the
been treated with coccidiostats. Use chickens of the category contents are wately. Lesions may extend as far posterior
that is expected to be the most sensitive, i.e. 14-day-old as the yolk sac diverticulum.
chickens. During the test, chickens are housed in suitable 4 The mucosal wall is greyish with completely coalescent
conditions with the use of fioor pens or cages with solid colonies. Congestion may be confined to small petechiae
fioors to favour reinfection with oocysts. Administer by or, in extremely heavy infections, the entire mucosa may
gavage or another suitable route to each chicken a quantity of be bright red in colour. Individuallesions may be
vaccinal oocysts consisting of the equivalent of not less than indistinguishable in the upper intestine. Typical ladder-
10 times the maximum quantity of oocysts of each coccidial like lesions appear in the middle part of the intestine.
species likely to be contained in 1 dose of the vaccine. The intestinal wall is vely much thickened and the
Observe the chickens at least daily for at least 14 days. intestine is filled with a creamy exudate, which may bear
The test is not val id if more than 10 per cent of the large numbers of oocysts. Birds dying of coccidiosis are
vaccinated chickens die from causes not attributable to the scored as 4.
vaccinal oocysts. Eimeria bnl11etti
The vaccine complies with the test if no vaccinated chicken O No gross lesions.
shows abnOlmal signs of disease or dies from causes No gross lesions. In the absence of distinct lesions, the
attributable to the vaccine. presence of parasites may go undetected unless scrapings
from suspicious areas are examined microscopically.
2-3-2 Test for residual pathogenicity
2 The intestinal wall may appear grey in colour. The lower
Cany out a separate test with each coccidial species and line
portion may be thickened and fiecks of pink material
to be included in the vaccine. Use in each case a preparation
sloughed from the intestine are presento
containing oocysts at the least attenuated passage level that
3 The intestinal wall is thickened and a blood-tinged
will be present between the master seed lot and a batch of
catarrhal exudate is presento Transverse red streaks may
the vaccine. For each test use not fewer than 20 chickens
be present in the lower rectum and lesions occur in the
from an SPF fiod:: (5.2.2). The chickens must be hatched
cae cal tonsils. Soft mucous plugs may be present in this
and reared as described in section \2-1 and must not have
latter area.
been treated with coccidiostats. Use chickens of the categOly
4 Extensive coagulation necrosis of the mucosal surface of
that is expected to be the most sensitive, i.e. 14-day-old
the lower intestine may be presento In some birds a dry
chickens. During the test, the chickens are placed in cages
necrotic membrane may line the intestine and caseous
(01' any other suitable accomodation that prevents reinfection
cores may plug the entrance to the caeca. Lesions may
and allows collection of faeces). Administer by gavage or
extend into the middle or upper intestine. Birds dying of
another suitable route to each chicken the equivalent of not
coccidiosis are scored as 4.
less than 10 times the maximum quantity of the vaccinal
Ei771eria maxima
oocysts likely to be contained in 1 dose of the vaccine.
O No gross lesions.
Observe the chickens at least daily for 14 days. The test is
Small red petechiae may appear on the serosal side of the
not valid if more than 10 per cent of the chickens die from
mid-intestine. There is no ballooning or thickening of the
causes not attributable to the vaccinal oocysts. Collect faeces
intestine, though small amounts of orange mucous may
and detelmine oocyst production daily from day 3 until
be presento
day 14. On one day between days 4 and 8, depending on the
2 The serosal surface may be speckled with nun1erous red
length of the pre-patent period, when lesions are expected to
petechiae. The intestine may be filled with orange
be maximal, and on day 14, euthanise not fewer than
mucous, but there is little 01' no ballooning of the
9 chickens and examine the intestinal tract for specific lesions
intestine. There is thickening of the wall.
indicative of infection with the coccidial species or, for
3 The intestinal wall is ballooned and thickened. The
species known not to induce macroscopic lesions (E. mitis
mucosal surface is roughened. Intestinal contents are
and E. praecox), microscopic evidence of infection such as
composed of pinpoint blood clots and mucous.
demonstration of oocysts or developing oocysts in the
4 The intestinal wall may be ballooned for most of its
intestinal contents or scrapings of the intenstinal wall.
length. It contains numerous blood clots and digested red
For species that have the potential to produce relevant
blood cells giving a characteristic colour and putrid
macroscopic pathological changes if not attenuated, a scoring
odour. The wall is greatly thickened. Birds dying of
system with a scale from O to 4 is used to record the
coccidiosis are scored as 4.
species-specific lesions visible in the intestine as follows.
Eimeria acervulina Eimeria necatrix
O No gross lesions. O No gross lesions:
Scattered, white, plaque-like lesions containing Small scattered petechiae and white spots are easily seen
developing oocysts are confined to the duodenum. These on the serosal surface. There is little, if any, damage
lesions are elongated with the longer axis transversely apparent on the mucosal surface.
oriented on the intestinal walls like the rungs of a ladder. 2 Numerous petechiae are _seerí on the serosal surface.
They may be seen on either the serosal or mucosal Slight ballooning confined to the midgut area may be
intestinal surfaces. There may be up to a maximum of 5 presento
lesions per square centimetre.
Vet-240 Veterinary Vaccines 2016

3 There is extensive haemonhage into the lumen of the The line complies with the test if no indication of an increase
intestine. The serosal surface is covered with red in virulence of the maximally passaged oocysts compared
petechiae and/or white plaques, and is rough and with the unpassaged oocysts is observed.
thickened with many pinpoint haemorrhages. Normal The test is not valid if oocysts are not recovered at any
intestinal contents are lacking. Ballooning extends over passage level.
the lower half of the small intestine.
2-3-4 Immunogenicity
4 Extensive haemorrhage gives the intestine a dark colour,
The efficacy of each coccidial species and line included in the
and the intestinal contents consist of red 01' brown
vaccine is detelmined in a separate study with an appropriate
mucous. Ballooning may extend throughout much of the
challenge strain. For each component, a test is carried out
length of the intestine. Birds dying of coccidiosis are
with vaccine administered by each route and method of
scored as 4.
administration to be recommended, using in each case
Eimeria ten ella chickens not older than the minimum age to be
O No gross lesions. recommended for vaccination. The quantity of each of the
Very few scattered petechiae are seen on the caecal wall, components in the batch of vaccine administered to each
and there is no thickening of the caecal walls. Normal chicken is not greater than the minimum number of oocysts
caecal contents are presento to be stated on the label and the oocysts are at the most
2 Lesions are more numerous with noticeable blood in the attenuated passage level that will be present in a batch of
caecal contents, and the caecal wall is somewhat vaccine. Use for the test not fewer than 40 chickens from an
thickened. Normal caecal contents are presento SPF flock (5.2.2). The chickens must be hatched and reared
3 Large amounts of blood or caecal cores are present, and as described in section 2-1 and must not have been treated
the caecal walls are greatly thickened. There is little if any with coccidiostats. Vaccinate not fewer than 20 chickens and
normal faecal content in the caeca. maintain not fewer than 20 chickens as controls. For the
4 The cae cal wall is greatly distended with blood 01' large evaluation of weight gain with Eimeria strains showing a low
caseous cores. Faecal debris is lacking 01' included in the pathogenicity, the number of chickens used may be higher.
cores. Birds dying of coccidiosis are scored as 4. The test may require different challenge doses for different
The species and line comply with the test for attenuation if test parameters and so may be assessed as separate challenge
no more than mild coccidiallesions 01' limited signs of groups. For example, a lower challenge dose may be needed
infection are observed; where the scoring system described to determine the effect on oocyst output than the dose
above is appropriate, the average lesion score on the day of needed to determine the effect on weight gain and lesion
sampling between days 4 and 8 and on day 14 is not greater scoring. After vaccination, the chickens are housed in suitable
than 1.5 points and no individual score is greater than conditions with the use of floor pens or cages with solid
3 points. The quantity and time of oocyst production is floors to favour reinfection with oocysts. On a suitable day
determined. between days 14 and 21 after vaccination, weigh each
2-3-3 Increase in virulence chicken, move them to cages (01' any other suitable
Carry out a separate test according to general chapter 5.2.6 accomodation that prevents reinfection and allows collection
with each coccidial species and line to be included in the of faeces) and challenge each chicken by gavage or another
vaccine. Use a preparation containing oocysts at the master suitable route with a sufficient quantity of virulent coccidia to
seed lot level. If the quantity of tlle master seed sufficient for induce in the unvaccinated controls signs of disease
performing the test is not available, the lowest passage seed characteristic of the Eimeria challenge species. Observe the
used for tlle production that is available in sufficient quantity chickens at least daily until the end of the test. Record deaths
may be used. For each test use 14-day-old chickens from an and the number of surviving chickens that show clinical signs
SPF flock (5.2.2). If tlle properties of the vaccinal oocysts of disease. Collect faeces and determine oocyst production
allow sequential passage through 5 groups via natural from day 3 after challenge until the end of the test. On an
spreading, this method may be used, otherwise, passage as appropriate day between days 4 and 8 after challenge,
described below is carried out. The chickens must be depending on the length of the pre-patent period of the
hatched and reared as described in section 2-1 and must not challenge species, weigh each chicken. Euthanise 10 chickens
have bee!1 treated with coccidiostats. During the test, the from each group and examine them for lesions in the
chickens are placed in cages (or any other suitable intestinal tracto Where appropriate, record the specific lesions
accomodation that prevents reinfection and allows collection indicative of the coccidial challenge species (using the scoring
of faeces). Administer by gavage 01' another suitable route to system described in section 2-3-2). For species known notto
each chicken of the 1st group a quantity of oocysts that will induce macroscopic lesions (E. niitis and E. praecox), examine
allow recovery of the oocysts for the passages described the chickens for microscopic evidence of infection such as
below. Collect faeces daily from day 2 to day 14 after demonstration of oocysts 01' developing oocysts in the
infection and prepare a pooled suspension of sporulated intestinal contents or scrapings of the intestinal wall.
oocysts from tlle 5 chickens. Administer a suitable quantity On day 14 after challenge, weigh each of the remaining
by gavage 01' another suitable route to each chicken of the chickens.
next group. Carry out this passage operation not fewer than The test is not valid if:
4 times, verifying the presence of oocysts at each passage. - during the period between vaccination and challenge,
If the vaccinal oocysts are not found at a passage level, repeat more than 10 per cent of the vaccinated or control
the passage by administration to a group of 10 chickens. chickens show abnormal clinical signs or die from causes
Carry out the test for residual pathogenicity (section 2-3-2), not attributable to the vaccine;
using the material used for the 1st passage and the oocysts - for challenges with E. tenella, E. acel''Vulina, E. necatlix,
that have been recovered in the final passage. Compare the E. maxima 01' E. bnmetti, fewer than 80 per cent of the
results obtained for signs of lesions 01' infection in the control chickens euthanised between days 4 and 8 have
intestinal tract and oocyst output from administration of marked characteristic lesions of the challenge infection in
passaged and unpassaged oocysts.
2016 Veterinary Vaccines Vet-241

the intestine at post-mortem examination (e.g. lesion 3-3 Mycoplasmas


scores not less than 2); The vaccine complies with the test for mycoplasmas (2.6.7).
- for challenges with E. mitis or E. praecox, fewer than 3-4 Extraneous agents
80 per cent of the control chickens euthanised Carry out tests 1-6 of chapter 2.6.25. Avian live virus
between days 4 and 8 are infected. vaccines: tests for extraneous agents in batches of finished producto
The vaccine complies with the test if: General provisions a-d, g and h are also applicable.
- for all the Eimeria challenge species, the production of the The vaccine complies with the requirements of each test.
oocysts is significantly decreased in vaccinates compared 3-5 Sporulated oocyst count
with controls; The sporulated oocysts content per dose is determined by
- for all the Eimeria challenge species, no vaccinated counting the sporulated oocysts in a suitable counting
chicken die s due to the challenge infection; chamber, under the microscope. The contents are not less
- for challenge with E. ten ella, E. aceruulina, E. necatrix, than the minimum and not more than the maximum content
E. maxima or E. brunettz~ at least 80 per cent of the of sporulated oocysts stated on the label.
vaccinates show no more than mild signs of disease and
these are less marked than those in the control s; 3-6 Potency
- for challenges with E. ten ella, E. acervulina, E. necatrix, The vaccine complies with the requirements of the test
E. 111axi111a or E. brunetti, at least 80 per cent of the prescribed under Irnmunogenicity (section 2-3-4) using
vaccinates have no or minimal lesions in the intestine 1 dose of the vaccine administered by a recommended route.
(e.g. mean lesion scores not greater than 1) and no bird 4 LABELLING
has a les ion score of 4; The label states the minimum and maximum number of
- for challenges with E. ten ella, E. acervulina, E. necatrix, sporulated oocysts per dose.
E. 111axima, E. brunetti, E. mitis, 01' E. praecox, the growth ____________________________________________ PhE~

rate in the vaccinates is significantly greater than in the


controls.
2-4 MANUFACTURER' S TESTS
2-4-1 In-process test for sporulation rate and oocyst
count Contagious Pustular Dermatitis Vaccine,
A sample of each oocyst bulk is examined microscopically Living
after the sporulation step and before blending to detelmine
OlfVaccine
the percentage of sporulated oocysts and the oocyst count.
The values obtained are within the limits shown to allow DEFINITION
preparation of a satisfactory vaccine. Contagious Pustular Dermatitis Vaccine, Living is a
2-4-2 Batch potency test for each Eimeria species in preparation of a suitable strain of orf virus (contagious
the vaccine pustular dennatitis virus) for administration to sheep, by skin
It is not necessary to carry out the potency test (section 3-6) scarification.
for each batch of the vaccine if it has been canied out using PRODUCTION
a batch or batches of vaccine with minimum potency and The vaccinal organisms are obtained from the vesiculo-
sporulated oocyst contento Where the test is not carried out, pustular lesions produced on the skin of lambs inoculated
an alternative validated method is used, the criteria for with the seed virus 01' prepared in suitable cell cultures
acceptance for each component being set with reference to a (Appendix XV J(Vet) 1). If lambs are used for production,
batch of vaccine that has given satisfactory results in the test they are obtained from a flock known to be free from the
described under Potency. natural disease, are healthy and have not been exposed
2-4-3 Freedom from extraneous agents previously to orf virus. Both the flock and the lambs are
The disinfection method applied during the preparation of monitored for freedom from infectious diseases, the range of
the final product from the harvested oocysts may be validated diseases in each case being agreed with the competent
to shm.y effective inactivation of certain potential extraneous authority.
agents. Where relevant validation data is available and where The harvested material may be freeze-dried. The finished
justified and authorised, sorne 01' all of the tests indicated preparation administered to the animals may contain glycerol
under Extraneous agents (section 3-4) may be omitted as and may contain an approved dye.
routine tests on each batch. CHOICE OF VACCINE STRAIN
3 BATCH TESTS The vaccine strain is shown to be satisfactory with respect to
3-1 Identification safety and irnmunogenicity for the animals for which the
3-1-1 Microscopical examination is used to confinn the vaccine is intended. The following tests may be used during
presence of coccidial oocysts in the batch of vaccine. the demonstration of safety, Appendix XV K(Vet) 1,
3-1-2 The potency test (or batch potency test) is used to reversion to virulence and irnmunogenicity,
confirm the presence of oocysts of each of the Eimeria species Appendix XV K(Vet) 2.
stated on the label. Safety Carry out á test in each category of animal for which
the vaccine is to be recommended. Vaccinate at least five
3-2 Bacteria and fungi
animal s that do not have antibodies to orf virus. Administer
The vaccine, including where applicable the diluent supplied
to each, by skin scarification of one site 01' a small number of
for reconstitution, complies with the test for sterility
contiguous sites, a quantity_ of virus containing not les s than
prescribed in the monograph Vaccines for veterinary use (0062)
ten times the maximum virus titre likely to be contained in a
and comply with the test with a medium selective for
dos e of the vaccine. Observe the animals for 4 weeks.
Campylobacter spp.
No systemic or local reactions occur except for mild localised
lesions of orf which resolve in les s than three weeks after
Vet-242 Veterinary Vaccines 2016

vaccination to leave no more than a small area of scar tissue. Mycoplasmas


If the vaccine is for use or may be used in pregnant animals, Complies with the test for absence of mycoplasmas,
for the test in this category, administer the vaccine at the Appendix XVI B(Vet) 3.
relevant stage or stages of pregnancy, pro long the observation Safety
period up to the time of parturition and note any effects on Administer by skin scarification of one site 01' a small number
gestation or the offspring. of contiguous sites, ten doses of the vaccine to each of two
Reversíon to virulence Admínister by skin scarification a lambs of the minimum age to be recommended for
quantity of virus containing not less than one dose of the vaccination and that are free from antibodies to orf virus.
vaccine to each of two lambs of the minimum age to be Observe the lambs for two weeks. No systemic or local
recommended for vaccination and that do not have reactions occur except for mild local lesions of Oli.
antibodies to orf virus. Six to eight days later, anaesthetise Extraneous viruses
the lambs and remove, pool and homogenise the Oli lesions Neutralise the vaccine if necessmy. Inoculate the vaccine
in buffer contaíning suitable antibiotics and inoculate into onto suitable cell cultures that are susceptible to ovine
two more lambs. Repeat this passage operation four more viruses, make at least one passage and maintain the cultures
times. If the virus disappears (no lesions develop) a second for at least 14 days. No cytopathic effect develops. Conduct a
series of passages is carried out. Observe the lambs given the test for freedom from haemadsorbing agents. The cell
last passage material for four weeks. The vaccine complies cultures show no signs of viral contamination.
with the test if the virus from the last passage produces no
systemic reactions and the local reactions observed are no POTENCY
more severe than those seen in the lambs given the Vaccinate each of no fewer than two healthy susceptible
unpassaged material. sheep, 6 to 12 months old, with serial dilutions of the
Immunogenicity Carry out a test in each category of animal vaccine applied to the scarified skin. Characteristic lesions of
for which the vaccine is to be recommended. Vaccinate, by contagious pustular dermatitis appearing on the fourth to
skin scarification, at least five animals that do not have eighth day after inoculation are recorded. The vaccine
antibodies to Oli virus, with a quantity of virus containing the contains not less than 100 MID in the dose stated on the
mínimum virus titre likely to be contained in adose of label.
vaccine. Maintain separately at least two animals of the same STORAGE
age and from the same source as unvaccinated controls. When stored in the prescribed conditions the vaccine may be
Eighteen to twenty four days later, challenge all the animals expected to retain its potency for at least ayear.
by skin scarification with a sufficient quantity of virulent
strain of orf virus to cause disease in the controls. Observe
the animals for 21 days and monitor and score the skin
lesions. The test is not valid unless there are marked lesions
Duck Plague Vaccine (Live) ***
*** ***
of orf in both the control animals after challenge.
The vaccine complies with the test if, after challenge, there
are no more than mild transient lesions of Oli in the
(Ph Eur m01Zograph 1938) ***
_____________________________________________ PhE~

vaccinates.
BATCH TESTING 1 DEFINITION
Extraneous bacteria and fungi: Duck plague vaccine (live) is a preparation of a suitable
A. Vaccines produced in lambs For each batch, the number of strain of duck plague virus (anatid herpesvirus 1). This
non-pathogenic organisms per dos e shall be within the limits monograph applies to vaccines intended for the active
set for the product and shown to be safe. immunisation of ducks.
B. Vaccines produced in cell cultures Each batch complies with 2 PRODUCTION
the test described under Veterinary Vaccines. 2-1 PREPARATION OF THE VACCINE
Virus titre Where the nature of the vaccine strain allows a The vaccine virus is grown in emblyonated hens' eggs or in
satisfactory test to be conducted in vitra, the test for Potency cell cultures. The vaccine may be freeze-dried.
may be omitted as a routine control on each batch of 2-2 SUBSTRATE FOR VIRUS PROPAGATION
vaccine. Where the Potency test is not carried out, an 2-2-1 Embryonated hens' eggs
alternative validated in vitro test is conducted on each batch If the vaccine virus is grown in emblyonated hens' eggs, they
to measure the virus contento The batch complies with the are obtained from flocks free from specified pathogens (SPF)
tests if the titre of the batch is not less than the minimum (5.2.2).
titre stated on the label.
2-2-2 Cell cultures
The vaccine complíes with the requirements stated under
If the vaccine virus is grown in cell cultures, they comply
Veterina1J1 Vaccines with the following modifications.
with the requirements for cell cultures for production of
IDENTIFICATION veterinary vaccines (5.2.4).
Produces the characteristic lesions of contagious pustular
2-3 SEED LOTS
dermatitis when applied to a scarified area of the skin of
2-3-1 Extraneous agents
lambs.
The master seed lot complies with the test Jor extraneous
TESTS agents in seed lots (2.6.24). In these tests on the master seed
Extraneous bacteria and fungí lot, the organisms used are not more than 5 passages from
The vaccine is shown by appropriate methods to be free from the master seed lot at the start of the tésts.
pathogenic organisms.
2016 Veterinary Vaccines Vet-243

2-4 CHOICE OF VACCINE VIRUS 2-4-3 Irnmunogenicity


The vaccine virus shall be shown to be satisfactory with A test is carried out for each route and method of
respect to safety (5.2.6) and efficacy (5. 2.7) for the ducks for administration to be recommended for vaccination, using in
which the vaccine is intended. each case domestic ducks not older than the minimum age to
The following tests for safety (section 2-4-1), increase in be recommended for vaccination. The quantity of the vaccine
virulence (section 2-4-2) and irnmunogenicity (section 2-4-3) virus administered to each duele is not greater than the
may be used during demonstration of safety and efficacy. minimum virus titre to be stated on the label and the virus is
at the most attenuated passage level that will be present in a
2-4-1 Safety
batch of the vaccine. For each test, use not fewer than
Carry out the test for each route and method of
30 ducks of the same origin and that do not have antibodies
administration to be recommended for vaccination, using in
against duele plague virus. Vaccinate by a route to be
each case ducks from a species considered to be the most
recommended not fewer than 20 dueles. Maintain not fewer
susceptible among the species to be recommended for
than 10 dueles as controls. After 5 days, challenge each duck
vacciriation, not older than the minimum age to be
by a suitable route with a sufficient quantity of virulent duele
recommended for vaccination and that do not have
plague virus. Observe the dueles at least daily for 14 days
antibodies against duck plague virus. Use vaccine virus at the
after challenge. Record the deaths and the number of
least attenuated passage level that will be present in a batch
surviving dueles that show elinical signs of disease.
of the vaccine.
The test is not valid if during the observation period after
For each test performed in dueles younger than 3 weeks of
challenge fewer than 80 per cent of the control dueles die or
age, use not fewer than 10 susceptible ducks. For each test
show typical signs of duele plague andlor if during the period
performed in dueles older than 3 weeks of age, use not fewer
between the vaccination and challenge more than 10 per cent
than 8 susceptible dueles. Administer to each duck a quantity
of control or vaccinated dueles show abnormal clinical signs
of vaccine virus equivalent to not less than 10 times the
of disease or die from causes not attributable to the vaccine.
maximum virus titre likely to be contained in 1 dose of the
vaccine. Observe the dueles at least daily for at least 14 days. The vaccine virus complies with the test if during the
observation period after challenge not fewer than 80 per cent
The test is not valid if more than 10 per cent of the dueles
of the vaccinated ducks survive and show no notable elinical
younger than 3 weeks of age show abnormal signs of disease
signs of duele plague.
01' die from causes not attributable to the vaccine. For ducks
older than 3 weeks of age, the test is not valid if non-specific 3 BATCH TESTS
mortality occurs. 3-1 Identification
The vaccine virus complies with the test if no duele shows The vaccine, diluted if necessary and mixed with a
abnormal signs of disease or dies from causes attributable to monospecific duck plague virus antiserum, no longer infects
the vaccine virus. embryonated hens' eggs from an SPF fiock (5.2.2) or
susceptible cell cultures (5.2.4) into which it is inoculated.
2-4-2 Increase in virulence
Carry out the test according to general chapter 5.2.6 using 3-2 Bacteria and fungi
domestic ducks that do not have antibodies against duele The vaccine, ineluding where applicable the diluent supplied
plague virus and of an age suitable for the multiplication of for reconstitution, complies with the test for sterility
the virus. If the properties of the vaccine virus allow prescribed in the monograph Vaccines for vetetina/J' use
sequential passage through 5 groups via natural spreading, (0062).
this method may be used, otherwise passage as described 3-3 Mycoplasmas
below is carried out. Administer to each duele of the 1st The vaccine complies with the test for mycoplasmas (2.6.7).
group by a route to be recommended a quantity of the 3-4 Extraneous agents
vaccine virus that will allow recovery of virus for the passages The vaccine complies with the tests for extraneous agents in
described below. 2 to 4 days later, take samples of liver and batches of finished product (2.6.25).
spleen from each duele and pool all samples. Administer
0.1 mL of the pooled suspension by the oro-nasal or a 3-5 Virus titre
Titrate the vaccine virus by inoculation into embryonated
parenteral route to each duele of the next group. Carry out
hens' eggs from an SPF fiock (5.2.2) or into suitable cell
this passage operation not fewer than 4 times; verify the
cultures (5.2.4). The vaccine complies with the test if 1 dose
presence of the virus at each passage. If the virus is not
contains not less than the minimum virus titre stated on the
found at a passage level, repeat the passage by administration
label.
to a group of 10 ducks.
If the 5th group of dueles shows no evidence of an increase in 3-6 Potency
virulence indicative of reversion during the observation The vaccine complies with the test prescribed under
period, further testing is not required. Otherwise, carry out immunogenicity (section 2-4-3), when administered by a
an additional safety test and compare the elinical signs and recommended route and method. It is not necessary to carry
any relevant parameters in a group of at least 10 dueles out the potency test for each batch of the vaccine if it has
receiving the material used for the 1st passage and another been carried out on a representative batch using a vaccinating
similar group receiving the virus at the final passage level. dos e containing not more than the minimum virus titre
stated on the label.
The vaccine virus complies with the test if no indication of
_____________________________________________ PhE~

an increase in virulence of the virus at the final passage level


compared with the material used for the 1st passage is
observed. If virus is not recovered after an initial passage in
5 dueles and a subsequent repeat passage in 10 ducks, the
vaccine virus also complies with the test.
Vet-244 Veterinary Vaccines 2016

2-4-2 Increase in virulence


Duck Viral Hepatitis Type I Carry out the test according to general chapter 5.2.6 using
Vaccine (Live) 1-day-old domestic ducldings that do not have antibodies
(Ph. Eur. 1110nograph 1315) against duck hepatitis virus type I. If the properties of the
~E~ _____________________________________________ vaccine virus allow sequential passage through 5 groups vía
natural spreading, this method may be used, otherwise
1 DEFINITION passage as described below is carried out.
Duck viral hepatitis type I vaccine (live) is a preparation of a Administer to each duckling of the 1sr group by the oro-nasal
suitable strain of duck hepatitis virus type I. Tms monograph route a quantity of vaccine virus that will allow recovery of
applies to vaccines intended for the active irnmunisation of virus for the passages described below. 2 ro 4 days later, take
breeder ducks in order ro protect passively their progeny samples of liver from each duclding and pool the samples.
and/or for the active irnmunisation of ducklings. Administer 1 mL of the pooled liver suspension by the oro-
2 PRODUCTION nasal route to each duckling of the next group. Carry out this
2-1 PREPARATION OF THE VACCINE operation 4 times. Verify the presence of the virus at each
The vaccine virus is grown in embryonated hens I eggs or in passage. If the virus is not found at a passage level, repeat the
cell cultures. passage by administration to a group of 10 ducldings.
Observe the ducldings given the last passage at least daily for
2-2 SUBSTRATE FOR VIRUS PROPAGATION 21 days.
2-2-1 Embryonated hens I eggs If the 5th group of ducldings shows no evidence of an
If the vaccine virus is grown in embryonated hens' eggs, they increase in virulence indicative of reversion during the
are obtained from fiocks free from specified pathogens (SPF) observation period, further testing is not required. Otherwíse,
(5.2.2). carry out an additional safety test and compare the clinical
2-2-2 Cell cultures signs and any relevant parameters in a group of at least
If the vaccine virus is grown in cell cultures, they comply 10 ducldings receiving the material used for the 1sr passage
with the requirements for cell cultures for production of and another similar group receiving the virus at the final
veterinary vaccines (5.2.4). passage level.
2-3 SEED LOTS The vaccine virus complies with the test if no indicatíon of
2-3-1 Extraneous agents an increase in virulence of the virus at the final passage level
The master seed lot complies with the tests for extraneous compared with the material used for the 1sr passage is
agents in seed lots (2.6.24). In these tests on the master seed observed. If the virus is not recovered after an initial passage
lot, the organisms used are not more than 5 passages from in 5 ducklings and a subsequent repeat passage in
the master seed lot at the start of the tests. 10 ducldings, the vaccine virus also complies with the test.
2-4 CHOICE OF VACCINE VIRUS 2-4-3 Irnmunogenicity
The vaccine virus shall be shown to be satisfactory with A test is carried out for each route and method of
respect to safety (5.2.6) and efficacy (5. 2.7) for the ducks for administration to be recommended for vaccination, using in
wmch it is intended. each case domestic ducks not older than the minimum age to
be recommended for vaccination. The quantity of the vaccine
The following tests for safety (section 2-4-1), increase in
virus administered to each bird is not greater than the
virulence (section 2-4-2) and immunogenicity (section 2-4-3)
minimum virus titre to be stated on the label and the virus is
may be used during demonstration of safety and efficacy.
at the most attenuated passage level that will be present in a
2-4-1 Safety batch of the vaccine.
Carry out the test for each route and method of 2-4-3-1 Vaccines for passive immunisation of ducklings. Use for
administration to be recommended for vaccination using in the test not fewer than 15 laying ducks or ducks intended for
each case susceptible domes tic ducks (Anas platyrhynchos) not laying, as appropriate, of the same origin and that do not
older than the minimum age to be recommended for have antibodies against duck hepatitis virus type I. Vaccinate
vaccination and that do not have antibodies against duck by a route to be recommended not fewer than 10 ducks
hepatitis virus type I. Use vaccine virus at the least using the schedule to be recommended. Maintain not fewer
attenuated passage level that will be present in a batch of the than 5 ducks as controls. Starting from 4 weeks after onset of
vaccine. lay, collect embryonated eggs from vaccinated and control
For each test performed in ducklings younger than 3 weeks ducks and incubate them. Challenge not fewer than twenty
of age, use not fewer than 10 ducklings. For each test l-week-old ducldings representative of the vaccinated group
performed in ducldings older than 3 weeks of age, use not and not fewer than 10 from the control group by the oro-
fewer than 8 ducldings. Administer to each duckling a nasal route with a sufficient quantity of virulent duck
quantity of vaccine virus equivalent ro not less than 10 times hepatitis virus type I. Observe the ducldings at least daily for
the maximum virus titre likely to be contained in 1 dose of 14 days after challenge. Record the deaths and the number
vaccine. Observe the ducldings at least daily for at least of surviving ducklings that show clinical signs of disease.
14 days. The test is not valid if:
The test is not valid if more than 10 per cent of the during the observation period after challenge fewer than
ducldings younger than 3 weeks of age show abnormal signs 70 per cent of the challenged ducklings from the control
of disease or die from causes not attributable to the vaccine. ducks die or ·show typical signs of the disease,
For ducklings older than 3 weeks of age, the test is not valid and/or dilring the period berween vaccination and
if non-specific mortality occurs. collection' of the eggs more than 10 per cent of the
The vaccine virus complies with the test if no duclding shows control or vaccinated ducks show abnormal clinical signs
abnormal signs of disease or dies from causes attributable to or die from causes not attributable to the vaccine.
the vaccine virus.
2016 Veterinary Vaccines Vet-245

The vaccine virus complies with the test if during the contain pathogenic micro-organisms and contains not more
observation period after challenge the percentage re1ative than 1 non-padlogenic micro-organism per dose.
protection ca1culated using the following expression is not Any di1uent supp1ied for reconstitution of dle vaccine
1ess than 80 per cent: comp1ies with the test for sterility prescribed in the
monograph Vaccines for veterinaJY use (0062).
v-o 3-3 Mycoplasmas
100 _ e x 100
The vaccine complies with the test for mycop1asmas (2.6.7).
3-4 Extraneous agents
V percentage of challenged duck1ings from vaccinated The vaccine complies with the tests for extraneous agents in
ducks that survive to the end of the observation batches of finished product (2.6.25).
period without clinica1 signs of the disease;
e percentage of challenged duck1ings from 3-5 Virus titre
unvaccinated control ducks that survive to the end Titrate the vaccine virus by inocu1ation into embryonated
of the observation period without clinica1 signs of hens' eggs from an SPF flock (5.2.2) or into suitab1e cell
the disease. cultures (5.2.4). The vaccine comp1ies with the test if 1 dose
contains not 1ess than the minimum virus titre stated on rhe
2-4-3-2 Vaccines for active immunisation of ducklings. Use for 1abel.
the test not fewer than 30 ducldings of the same origin and
3-6 Potency
that do not have antibodies against duck hepatitis virus
Depending on the indications, the vaccine comp1ies with 1 or
type 1. Vaccinate by a route to be recornmended not fewer
both of the tests prescribed under Irnmunogenicity (section
than 20 ducldings. Maintain not fewer than 10 ducldings as
2-4-3), when administered by a recommended route and
contro1s. Challenge each duclding after at 1east 5 days by the
method. It is not necessary to carry out the potency test for
oro-nasal route with a sufficient quantity of viru1ent duck
each batch of the vaccine if it has been carried out on a
hepatitis virus type 1. Observe the ducldings at 1east dai1y for
representative batch using a vaccinating dose containing not
14 days after challenge. Record the deaths and the number
more than the minimum virus tit:re stated on the 1abel.
of surviving ducldings that show clinica1 signs of disease.
The test is not va1id if: 4 LABELLING
-- during the observation period after challenge fewer than If it has been found that the vaccine may show reversion to
70 per cent of the control ducldings die or show typica1 viru1ence, the 1abe1 indicates the precautions necessary to
signs of the disease; avoid transmission of viru1ent virus to unvaccinated
-- and/or during the period between vaccination and duck1ings.
challenge more than 10 per cent of the control or _____________________________________________ PhE~

vaccinated ducldings show abnorma1 clinica1 signs or die


fram causes not attributab1e to the vaccine.
The vaccine virus complies with the test if during the
observation period after challenge the percentage re1ative
Egg Drop Syndrome 76 ***
protection ca1cu1ated using the following expression is not
*** ***
1ess than 80 per cent: (Adenovirus) Vaccine ***
(Bgg Drop S)!7ldrome '76 Vaccine (Inactivated),
v-o Ph Bur 11l0nograph 1202)
100 _ e x 100
CAUTION Accidental injection of oily vaccine can cause se¡ious local
reactions in mano Expert medical advice should be sought
V percentage of challenged vaccinated duck1ings that immediately and the doctor should be il1j01"med that the vaccine is
survive to the end of the observation period without an oil emulsiono
clinica1 signs of the disease; PhE~ _____________________________________________
e percentage of challenged unvaccinated control
duck1ings that survive to the end of dle observation 1 DEFINITION
period without clinica1 signs of the disease. Egg drop syndrome '76 vaccine (inactivated) is a preparation
of a suitab1e strain of egg drop syndrome '76 virus
3 BATCH TESTS (haemagg1utinating avian adenovirus), inactivated whi1e
3-1 Identification maintaining adequate irnmunogenic properties. This
The vaccine, di1uted if necessary and mixed with a monograph applies to vaccines intended for protection of
monospecific duck hepatitis virus type I antiserum, no 10nger 1aying birds against a drop in egg production and/or for
infects embryonated hens' eggs from an SPF flock (5.2.2) or prevention of los s of egg qua1ity.
susceptible cell cultures (5.2.4) into wb,ich it is inocu1ated.
2 PRODUCTION
3-2 Bacteria and fungi
2-1 PREPARATION OF THE VACCINE
Vaccines intended for administration by injection comp1y
The vaccine strain is propagated in embryonated hens' or
with the test for steri1ity prescribed in the monograph
ducks' eggs or in cell cultures. The vaccine may be
Vaccines for veterinaJY use (0062).
adjuvanted.
Frozen or freeze-dried vaccines produced in embryonated
eggs and not intended for administration by injection either 2-2 SUBSTRATE FOR VIRUS PROPAGATION
comp1y with the test for steri1ity prescribed in the monograph 2-2-1 Embryonated hens' or ducks' eggs
Vaccines for veterinary use (0062) or with the following test: If the vaccine virus is grown -in embryonated hens' or ducks'
carry out a quantitative test for bacteria1 and funga1 eggs, they are obtained from hea1thy flocks.
contamination; carry out identification tests for micro-
organisms detected in the vaccine; the vaccine does not
Vet-246 Veterinary Vaccines 2016

2-2-2 Cell cultures 2-5 MANUFACTURER'S TESTS


If the vaccine virus is grown in cell cultures, they comply 2-5-1 Residuallive virus
with the requirements for cell cultures for production of The test for residual live virus is carried out in embryonated
veterinary vaccines (5.2.4). ducks' eggs from a fiod: free from egg drop syndrome '76
2-3 SEED LOTS virus infection, or in embryonated hens' eggs from an SPF
2-3-1 Extraneous agents fiock (5.2.2), or in suitable cell cultures, whichever is the
The master seed lot complies with the test for extraneous most sensitive for the vaccine strain. The quantity of
agents in seed lots (2.6.24). In these tests on the master seed inactivated virus harvest used in the test is equivalent to not
lot, the organisms used are not more than 5 passages from less than 10 doses of the vaccine. The inactivated virus
the master seed lot at the start of the test. harvest complies with the test if no live virus is detected.
2-5-2 Batch potency test
2-4 CHOICE OF VACCINE COMPOSITION
It is not necessary to cany out the potency test (section 3-5)
The vaccine is shown to be satisfactory with respect to safety
for each batch of vaccine if it has been carried out using a
(5.2.6) and efficacy (5.2.7) for the birds for which it is
batch of vaccine with a minimum potency. Where the test is
intended.
not carried out, an alternative validated metil0d is used, the
The following tests for safety (section 2-4-1) and criteria for acceptance being set with reference to a batch of
immunogenicity (section 2-4-2) may be used during the vaccine that has given satisfact01Y results in the test described
demonstration of safety and efficacy. under Potency. The following test may be used.
2-4-1 Safety Vaccinate not fewer than ten 14- to 28-day-old chickens
The test is carried out for each route of administration to be from an SPF fiock (5.2.2) with 1 dose of vaccine by one of
recommended for vaccination. Use a batch of vaccine the recommended routes. 4 weeks later, collect serum
containing not les s than the maximum potency that may be samples from each bird and from 5 unvaccinated control
expected in a batch of vaccine. birds of the same age and from the same source. Measure
For each test, use not fewer than 8 hens not older than the the antibody response in a haemagglutination (HA) inhibition
minimum age to be recommended for vaccination and from test on each serum using 4 HA units of antigen and chicken
a fiock free from specified pathogens (SPF) (5.2.2). erythrocytes. The test is not valid if there are specific
Administer by a route and method to be recommended to antibodies in the sera of the unvaccinated birds. The vaccine
each hen 1 dose of the vaccine. Observe the hens at least complies with the test if the mean tiu"e of the vaccinated
daily for at least 14 days after the administration of the group is not less than that found previously for a batch of
vaccine. vaccine tilat has given satisfactory results in the test described
The test is not valid if non-specific mortality occurs. under Potency.
The vaccine complies with the test if no hen shows abnormal 3 BATCH TESTS
signs of disease or dies from causes attributable to the 3-1 Identification
vaccme. When injected into chickens that do not have antibodies
2-4-2 Immunogenicity against egg drop syndrome '76 virus, the vaccine stimulates
A test is carried out for each route and method of the production of such antibodies.
adminisu"ation to be recommended, using in each case hens 3-2 Bacteria and fungi
from an SPF fiod: (5.2.2) and of the age at which The vaccine, inc1uding where applicable the diluent supplied
vaccination is recommended. The vaccine administered to for reconstitution, complies with tile test for sterility
each hen is of minimum potency. prescribed in the monograph Vaccines fOI" veterina1Y use
Vaccinate each of 2 groups of 30 hens. Maintain 2 control (0062).
groups one of 10 hens and the other of 30 hens, of the same 3-3 Residuallive virus
age and from the same source as the vaccinates. Maintain A test for residual live virus is carried out to confirm
individual egg production records from point of lay until inactivation of egg drop syndrome '76 virus.
4 weeks after challenge. At 30 weeks of age, challenge each
A. For a vaccine prepared in eggs, carry out the test in
hen from 1 group of 30 vaccinates and the group of 10
embryonated ducks' eggs from a fiod: free from egg drop
control hens with a quantity of egg drop syndrome '76 virus
syndrome '76 virus infection or, if it is known to provide a
sufficient to cause a well marked drop in egg production
more sensitive test system, in hens' eggs from an SPF fiock
and/or quality. The test is invalid unless there is a well
(5.2.2). Inject 2/5 of adose into the allantoic cavity of each
marked drop in egg production andlor quality in the control
of ten 10- to 14-day-old embryomited eggs that are free from
hens. The vaccine complies with the test if the vaccinated
parental antibodies to egg drop syndrome '76 virus. Incubate
hens show no marked drop in egg production andlor quality.
the eggs and observe for 8 days. Pool separately the allantoic
When the second group of vaccinated hens and the group of fiuid from eggs containing live embryos, and that from eggs
30 conu"ol hens are nearing the end of lay, challenge these containing dead embryos, exc1uding those that die from non-
hens, as before. The test is invalid unless there is a well specific causes within 24 h of the injection. Inject into the
marked drop in egg production andlor quality in the control allantoic cavity of each of ten 10- to 14-day-old embtyonated
hens. The vaccine complies with the test if the vaccinated eggs that do not have parentalantibodies to egg drop
hens show no marked drop in egg production andlor quality. syndrome '76 virus, 0.2 mL of the pooled allantoic fiuid
Carry out serological tests on serum samples obtained at the from the live embryos and into each of 10 similar eggs,
time of vaccination, 4 weeks later and just prior to challenge. 0.2 mL of the po'oled allantoic fluid from the dead embryos
The test is not valid if antibodies against egg drop syndrome and incubate fo1' 8 days. Examine the aJlantoic fiuid from
'76 virus are detected in any sample from control hens. each egg for the presence of haemagglutinating activity using
chicken erythrocytes. If more than 20 per cent of the
embryos die at either stage, repeat that stage. The vaccine
complies with the test if there is no evidence of
2016 Veterinary Vaccines Vet-247

haemagglutinating activity and if, in any repeat test, not more concentrated and are inactivated; they may be treated to
than 20 per cent of the embryos die from non-specific fragment the virus and the viral fragments may be purified
causes. and concentrated. The vaccine may be adjuvanted.
Antibiotics may be used in the test to control extraneous 2-2 SUBSTRATE FOR VIRUS PROPAGATION
bacterial infection. 2-2-1 Cell cultures
E. For a vaccine adapted to growth in cell cultures, inoculate The cell cultures comply with the requirements for cell
10 doses into suitable cell cultures. If the vaccine contains an cultures for production of veterinary vaccines (5.2.4).
oi1y adjuvant, eliminate it by suitable means. Incubate the
2-3 CHOICE OF VACCINE COMPOSITION
cultures at 38 ± 1 oC for 7 days. Make a passage on another
The vaccine is shown to be satisfactory with respect to safety
set of cell cultures and incubate at 38 ± 1 oC for 7 days.
(5.2.6) and efficacy (5.2.7) for the horses for which it is
Examine the cultures regular1y and at the end of the
intended. Where a particular breed of horse is known to be
incubation period examine the supernatant for the presence
especially sensitive to the vaccine, horses from that breed are
of haemagglutinating activity. The vaccine complies with the
included in the test for safety.
test if the cell cultures show no sign of infection and if there
is no haemagglutinating activity in the supernatant. The following tests for safety (section 2-3-1) and
irnmunogenicity (section 2-3-2) may be used during the
3-4 Specified extraneous agents
demonsu"ation of safety and efficacy.
Use 10 chickens, 14-28 days old, from an SPF fiod: (5.2.2).
Vaccinate each chicken by a recommended route with a 2-3-1 Safety
double dose of the vaccine. After 3 weeks, administer 1 dose Carry out the test for each route and method of
by the same route. Collect serum samples from each chicleen administration to be recommended for vaccination and in
2 weeks later and carry out tests for antibodies against the horses of each category for which the vaccine is intended.
following agents by the methods prescribed in general U se a batch of vaccine containing not less than the
chapter 5.2.2. Chicken flocks free fmm specified pathogens for the maximum potency that may be expected in a batch of
productio7Z and quality control of vaccines: avian vaccine.
encephalomyelitis virus, avian leucosis viruses, infectious U se for the test not fewer than 8 horses that have not been
bronchitis virus, avian infectious bursal disease virus, avian previously vaccinated with an equine herpesvirus vaccine,
infe.ctious laryngotracheitis virus, infiuenza A virus, Marek's that have at most a low antibody titre not indicative of recent
disease virus, N ewcastle disease virus and, for vaccine infection and that do not excrete equid herpesvirus.
produced in duele eggs, Chlamydia (by a complement-fixation Administer to each horse 1 dose of the vaccine, then another
test 01' agar gel precipitation test), duele hepatitis virus type I dose after 14 days. Observe the horses at least daily until at
(by a fiuorescent-antibody test or serum-neuu"alisation test) least 14 days after the last adminisu"ation.
and Derzsy' s disease virus (by a serum-neutralisation test). The vaccine complies with the test if no horse shows
The vaccine complies with the test if it does not stimulate the abnormallocal or systemic reactions or dies from causes
formation of antibodies against tl1ese agents. attributable to the vaccine during the 28 days of the test.
3-5 Potency 2-3-2 Immunogenicity
The vaccine complies Witl1 tl1e requirements of the test The type of irnmunogenicity test depends on the claims for
mentioned under ImmlU10genicity (section 2-4-2) when the producto For vaccines intended to protect against the
administered by a recommended route and method. disease of the respiratory tract, carry out test 2-3-2-1, using
equid herpesvirus 1 and/or equid herpesvirus 4 depending on
4 LABELLING
the claims for protection. For vaccines intended to protect
The label states whether the su"ain in the vaccine is duele- 01'
against abmtion carry out test 2-3-2-2.
hen-embryo-adapted 01' cell-culture-adapted.
A test is carried out for each route and method of
_____________________________________________ PhE~
adminisu"ation to be recommended, using in each case horses
that have not been vaccinated with an equine herpesvirus
vaccine, that have at most a low antibody titre not indicative
of recent infection, and that do not excrete equid herpesvirus.
Equine Herpesvirus Vaccine, *** To demonstrate that no recent infection occurs, immediately
*** *** before vaccination: draw a blood sample from each horse and
Inactivated *** test individually for antibodies against equid herpesviruses 1
(Bquine Herpesvinls Vaccine (In activa ted) J and 4; collect 10 mL of heparinised blood and test the
Ph Bur monograph 1613) washed leucocytes for equid herpesviruses 1 and 4; collect a
~E~ _____________________________________________ nasopharyngeal swab and test for equid herpesviruses 1 and
4. There is no indication of an active infection. Immediately
1 DEFINITION before challenge collect a nasopharyngeal swab and test for
Equine herpesvirus vaccine (inactivated) is a preparation of equid herpesviruses 1 and 4. If there is an indication of virus
one 01' more suitable strains of equid herpesvirus 1 and/or excretion remove the horse from the test. Keep the horses in
equid herpesvirus 4, inactivated while maintaining adequate strict isolation. The vaccine administered to each horse is of
irnmunogenic propelties or a suspension of an inactivated minimum potency.
fraction of the virus. This monograph applies to vaccines 2-3-2-1 Vaccines intendedfor protection against disease ofthe
intended for the active irnmunisation of horses against disease respirat01J' tracto U se for the test not fewer than 10 hm"ses, not
caused by equid herpesvirus 1 andlor equid herpesvirus 4. less than 6 months old. Vaccinate not fewer than 6 horses
2 PRODUCTION according to the schedule to"be recommended. Maintain not
2-1 PREPARATION OF THE VACCINE fewer than 4 hm"ses as controls. At least 2 weeks after the last
Each strain of vaccine virus is grown separately in cell vaccination, challenge each hm"se by nasal instillation with a
cultures. The viral suspensions may be purified and quantity of equid herpesvirus 1 or 4, sufficient to produce in
Vet-248 Veterinary Vaccines 2016

a susceptible horse characteristic signs of the disease such as virus es, the vaccine stimulates the production of specific
pyrexia and virus excretion (and possibly nasal discharge and antibodies against the virus type or types inc1uded in the
coughing). Observe the horses at least daily for 14 days. producto The method used must distinguish between
Collect nasopharyngeal swabs daily from each individual antibodies against equid herpesviruses 1 and 4.
horse to isolate the virus. 3-2 Bacteria and fungi
The vaccine complies with the test if the vaccÍnated horses The vaccine, inc1uding where applicable the diluent supplied
show no more than slight signs; the signs in vaccinates are for reconstitution, complies with the test for sterility
less severe than in controls. The average number of days on prescribed in the monograph Vaccines for veterinary use
which virus is excreted and the respective virus titres are (0062).
significantly lower in vaccinated horses than in controls. 3-3 Residuallive virus
2-3-2-2 Vaccines intended for protection against abol1ion. Carry out a test for residual live virus using not less than
Use not fewer than 10 pregnant horses. In addition to the 25 doses of vaccine by inoculating cell cultures sensitive to
testing described aboye, 6, 4, 3, 2 and 1 month before the equid herpesviruses 1 and 4; make a passage after 5-7 days
first vaccination draw a blood sample from each horse and and maintain the cultures for 14 days. The vaccine complies
test individually for antibodies against equid herpesvirues 1 with the test if no live virus is detected. If the vaccÍne
and 4. There is no evidence of recent infection or virus contains an adjuvant, separate the adjuvant from the liquid
excretion. Vaccinate not fewer than 6 horses according to the phase, by a method that do es not inactivate or otherwise
schedule to be recommended. Maintain not fewer than interfere with the detection of live virus, or carry out a test
4 horses as controls. Between day 260 and day 290 of for inactivation on the mixture of bulk antigens before
pregnancy but not earlier than 3 weeks after the last addition of the adjuvant.
vaccination, challenge each horse, by nasal instillation, with a
3-4 Potency
quantity of equid herpesvirus 1 sufficient to produce abortion
The vaccine éomplies with the requirements of the test
in susceptible horses. Observe the horses at least daily up to
mentioned under Irnmunogenicity (section 2-3-2) when
foaling or abortion. Collect samples of fetal lung and liver
administered by a recommended route and method.
tissues from aborted fetuses and carry out tests for virus in
_____________________________________________ ~Ew

cell cultures.
The test is not valid if more than one control horse gives
birth to a healthy foal and if the challenge virus is not
isolated from the aborted fetuses. The vaccine complies with
the test if not more than one vaccinated horse aborts. Equine Influenza Vaccine,
2-4 MANUFACTURER'S TESTS Inactivated
2-4-1 Residuallive virus
(Bquine Influenza Vaccine (In activa ted) ,
The test for residual live virus is carried out using 2 passages
Ph Bur 7110nograph 0249)
in the same type of cell culture as that used in the
PhEw _____________________________________________
production or in cell cultures shown to be at least as
sensitive. The quantity of inactivated virus harvest used in 1 DEFINITION
the test is equivalent to not less than 25 doses of the vaccine. Equine influenza vaccine (inactivated) is a preparation of one
The inactivated virus harvest complies with the test if no live or more suitable strains of equine influenza virus, inactivated
virus is detected. while maintaining adequate irnmunogenic properties. Suitable
2-4-2 Batch potency test strains contain both haemagglutinin and neuraminidase. This
It is not necessary to carry out the potency test (section 3-4) monograph applies to vaccines intended for the active
for each batch of vaccine if it has been carried out using a irnmunisation of horses against equine influenza.
batch of vaccine with a minimum potency. Where the test is 2 PRODUCTION
not carried out, an alternative validated method is used, the
2-1 PREPARATION OF THE VACCINE
criteria for acceptance being set with reference to a batch of
Each strain of virus is grown separately in embryonated hens'
vaccine that has given satisfactory results in the test described
eggs or in cell cultures. The viral suspensions may be purified
under Potency. The following test may be used.
and concentrated. The antigen content of the vaccine is
VaccÍnate not fewer than 5 rabbits, guinea-pigs or mice with based on the haemagglutinin content of the viral suspensions
a single injection of a suitable dose. Where the recommended determined as described under Manufacturer's tests;
schedule requires a 2nd injection to be given, the the amount of haemagglutinin fot each strain is not les s than
recommended schedule may be used in laboratory animal s that in the vaccine shown to be satisfactory in the test for
provided it has been demonstrated that this will still provide potency. The vaccine may be adjuvanted.
a suitably sensitive test system. At a given interval within the
range of 14-21 days after the last injection, collect blood 2-2 SUBSTRATE FOR VIRUS PROPAGATION
from each animal and prepare serum samples. Use a suitable 2-2-1 Embryonated hens' eggs
validated test such as an enzyme-linked immunosorbent assay If the vaccine virus is grown in embryonated hens' eggs, they
to measure the response to each of the antigens stated on the are obtained from a healthy fiock.
label. The vaccine complies with the test if the antibody 2-2-2 Cell cultures
levels are not significantly less than those obtained with a If the vaccine virus is grown in cell cultures, they comply
batch that has given satisfactory results in the test described with the requirements for cell cultures for production of
under Potency. veterinary váccines (5.2.4).
3 BATCH TESTS 2-3 CHOICE OF VACCINE COMPOSITION
3-1 Idenfication The choice of strains used in the vaccine is based on
In animals that do not have antibodies against equid epidemiological data. The World Organisation for Animal
herpesvirus 1 and equid herpesvirus 4 or a fraction of the Health (OIE, formerly the Office international des épizooties)
2016 Veterinary Vaccines Vet-249

reviews the epidemiological data periodically and if necessary suitable for use as reference sera for the single radial
recommends new strains corresponding to prevailing haemolysis test.
epidemiological evidence. Such strains are used in The c1aims for the product reflect the type of
accordance with the regulations in force in the signatory irnmunogenicity demonstrated (protection against challenge
states of the Convention on the Elaboration of a European or antibody production).
Pharmacopoeia.
2-3-2-1 Protection from signs of disease and reduction of virus
The vaccine is shown to be satisfactory with respect to safety excretion. Carry out the irnmunogenicity test using a challenge
(5.2.6) and efficacy (5.2.7) for the horses for which it is strain against which the vaccine is stated to provide
intended. Where a particular breed of horse is known to be protection. U se where possible a recent iso late.
especially sensitive to the vaccine, horses from that breed are
A test is carried out for each route and method of
inc1uded in the tests for safety.
administration to be recommended, using in each case horses
The following tests for safety (section 2-3-1) and not less than 6 months old. The vaccine administered to
irnmunogenicity (section 2-3-2) may be used during the each horse is of minimum potency.
demonstration of safety and efficacy. Use for the test not fewer than 10 horses that do not have
2-3-1 Safety antibodies against equine influenza virus. Draw a blood
Carry out the test for each route and method of sample from each horse and test individually for antibodies
administration to be recommended for vaccination and in against equine influenza virus 10 determine seronegativity.
horses of each category for which the vaccine is intended. Vaccinate not fewer than 6 horses according to the schedule
Use a batch of vaccine containing not less than the to be recommended. Maintain not fewer than 4 horses as
maximum potency that may be expected in a batch of controls. Draw a second blood sample from each vaccinated
vaccine. horse 7 days after the first vaccination and test individually
Use for the test not fewer than 8 horses that preferably do for antibodies against equine influenza virus, to detect an
not have antibodies against equine influenza virus or, where anamnestic sero-response. Horses showing sero-conversion at
justified, use horses with a low level of such antibodies as this stage are exc1uded from the test. At least 2 weeks after
long as they have not been vaccinated against equine the last vaccination, challenge each horse by aerosol with a
influenza and administration of the vaccine does not cause an quantity of equine influenza virus sufficient to produce
anamnestic response. Administer to each horse 1 dose of the characteristic signs of disease such as fever, nasal discharge
vaccine, then another dose after at least 14 days. Observe the and coughing in a susceptible horse. Observe the horses at
horses at least daily until at least 14 days after the last least daily for 14 days. Collect nasal swabs daily from each
administration. individual horse to isolate the virus.
The vaccine complies with the test if no horse shows The vaccine complies with the test if the vaccinated horses
abnormallocal or systemic reactions or dies from causes show no more than slight signs; the controls show
attributable to the vaccine during the 28 days of the test. characteristic signs. The average number of days on which
2-3-2 Immunogenicity virus is excreted and the respective virus titres are
The test described under 2-3-2-1 is suitable to demonstrate significantIy lower in vaccinated horses tI1an in control
the irnmunogenicity of the strains present in the vaccine. horses.
A test with virulent challenge is carried out for at least one 2-3-2-2 Presence of antibodies afier vaccination
vaccine strain (see test under 2-3-2-1). For other strains in 2-3-2-2-1 Single radial haemolysis. Heat each serum at 56 oC
the vaccine, demonstration of irnmunogenicity may, where for 30 min. Perfonn tests on each serum using respectively
justified, be based on the serological response induced in the antigen or antigens prepared from the strain(s) used in
horses by the vaccine (see tests under 2-3-2-2); justification the production of the vaccine. Mix 1 mL of sheep
for protection against these strains may be based on erythrocyte suspension in barbital buffer solution (1 volume
published data on the correlation of the antibody titre with of erythrocytes in 10 volumes of final suspension) with 1 mL
protection against antigenically related strains. of a suitable dilution of the influenza virus strain in barbital
Where serology is used, the test is carried out as described buffer solution and incubate the mixture at 4 oC for 30 min.
under 2-3-2-1 but instead of virulent challenge, a blood To 2 mL of the virus/erythrocyte mixture, add 1 mL of a
sample is drawn 2 weeks after the last vaccination and the 3 giL solution of chromiu71l(JJI) trichloride hexahydrate R, mix
antibody titre of each serum is determined by a suitable and allow to stand for 10 mino Heat the sensitised
immunochemical method (2. 7.1), such as the single radial erythrocytes to 47 oC in a water-bath. Mix 15 mL of a
haemolysis test or the haemagglutination-inhibition test 10 gIL solution of agarose for electrophoresis R in barbital
shown below; a reference serum is used to validate the test. buffer solution, 0.7 mL of sensitised erythrocyte suspension
The acceptance criteria depend on the strain and are based and the appropriate amount of diluted guinea-pig
on available data; for A/equine-2 virus, vaccines have usually complement in barbital buffer solution at 47 oC. Pour the
been found satisfactory if the antibody titre of each serum is mixture into Petri dishes and allow the agar to set. Punch
not less than 85 mm 2 where the single radial haemolysis test holes in the agar layer and place in each hole 5 ~lL of the
is used, or not less than 1:64 (before mixture with the undiluted serum to be tested or the control serum. Incubate
suspension of antigen and erythrocytes) where the the Petri dishes at 37 oC for 18 h. Measure the diameter of
haemagglutination-inhibition test is used. the haemolysis zone and calculate its are a, which expresses
the antibody titre, in square millimetres.
Equine influenza subtype 1 strain A/eq/Nezu111arket/77 horse
antiserum BRP, equine influenza subtype 2 European-like Equine influenza subtype 1 strain A/eq/Nezu1JZarket/77 horse
strain A/eq/Nezumarket/2/93 horse antiseru11l BRP, equine antiserum BRP, equine influenza subtype 2 European-like
influenza subtype 2 Amencan-like strain A/eq/NezuJ11arket/1 /93 strain A/eq/Nezu111arketl2/93 horse antiserum BRP, equine
horse antiseru11l BRP and equine influenza subtype 2 Amerz'can- influenza subtype 2 American-like strain A/eq/Nezu111arket/1 /93
like strain A/eq/South Africa/4/03 hm'se antiserum BRP are horse antiserum BRP and equine influenza subtype 2 Ametican-
like stI'ain A/eq/South Africa/4/03 Izorse antisetum BRP are
Vet-250 Veterinary Vaccines 2016

suitable for use as reference sera for the single radial The vaccine complies with the test if the antibody titres are
haemolysis test. not significantly lower than those obtained in guinea-pigs
2-3-2-2-2 Haemagglutination-inhibition test. Inactivate each with a reference batch of vaccine shown to have satisfactory
serum by heating at 56 oC for 30 mino To 1 volume of each potency in horses.
serum add 3 volumes of phosphate buffered saline pH 7.4 R 2-4-3 Bacteria! endotoxins
and 4 volumes of a 250 gIL suspension of light kaolin R in For vaccines produced in eggs, the content of bacterial
the same buffer solution. Shake each mixture for 10 mino endotoxins is detetmined on the virus hat\lest to monitor
Centrifuge, collect the supernatant and mix with a production.
concentrated suspension of chicken erythrocytes. Allow to 2-4-4 Haemagglutinin content
stand at 37 oC for 60 min and centrifuge. The dilution of the The content of haemagglutinin in the inactivated virus
serum obtained is 1:8. Perform tests on each serum using suspension, after purification and concentration where
each antigen prepared from the strains used in the applicable, is determined by a suitable irnmunochemical
production of the vaccine. Using each diluted serum, prepare method (2.7.1), such as single radial irnmunodiffusion, using
a series of 2-fold dilutions. To 0.025 mL of each of the latter a suitable haemagglutinin reference preparation;
dilutions add 0.025 mL of a suspension of antigen treated the inactivated virus suspension complies with the test if the
with ether R and containing 4 haemagglutinating units. Allow content is shown to be within the limits shown to allow
the mixture to stand for 30 min and add 0.05 mL of a preparation of a satisfactory vaccine.
suspension of chicken etythrocytes containing
2 x 107 erythrocytes/rnL. Allow to stand for 1 h and note 3 BATCH TESTS
the last dilution of serum that still completely inhibits 3-1 Identification
haemagglutination. In animals that do not have specific antibodies against equine
influenza virus, the vaccine stimulates the production of such
2-4 MANUFACTURER'S TESTS
antibodies.
2-4-1 Residuallive virus
The test for residual live virus is carried out using 3-2 Bacteria and fungi
method 2-4-1-1 or method 2-4-1-2, whichever is more The vaccine, including where applicable the diluent supplied
sensitive. The quantity of inactivated virus harvest used is for reconstitution, complies with the test for sterility
equivalent to not les s than 10 dos es of vaccine. prescribed in the monograph Vaccines for veterina1Y use
(0062).
2-4-1-1 Test in cel! cultures. Inoculate the vaccine into suitable
cells; after incubation for 8 days, make a subculture. 3-3 Residuallive virus
Incubate for a further 6-8 days. Hat\lest about 0.1 mL of the Inoculate 0.2 mL of the vaccine into the allantoic cavity of
supernatant and examine for live virus by a each of 10 embryonated eggs and incubate at 33-37 oC for
haemagglutination test. If haemagglutination is found, cany 3-4 days. The test is invalid unless at least 8 of the 10
out a further passage in cell culture and test for embryos sUl""Vive. Hat\lest 0.5 mL of the allantoic fluid from
haemagglutination; the inactivated virus hat\lest complies each sUl\living embtyo and pool the fluids. Inoculate 0.2 mL
with the test if no haemagglutination occurs. of the pooled fluid into a further 10 embtyonated eggs and
incubate at 33-37 c C for 3-4 days. The test is invalid unless
2-4-1-2 Test in e11lbl}l07zated eggs. Inoculate 0.2 mL into the
not fewer than 8 of tlle 10 embtyos sUl\live. Hat\lest about
allantoic cavity of each of 10 embtyonated eggs and incubate
0.1 mL of the allantoic fluid from each surviving embtyo and
at 33-37 oC for 3-4 days. The test is invalid unless not fewer
examine each individual hat\lest for live virus by a
than 8 of the 10 embryos sUl\live. Hat\lest 0.5 mL of the
haemagglutination test. If haemagglutination is found for any
allantoic fluid from each surviving embtyo and pool the
of the fluids, carry out for that fluid a furdler passage in eggs
fluids. Inoculate 0.2 mL of the pooled fluid into a further
and test for haemagglutination; the vaccine complies with the
10 embtyonated eggs and incubate at 33-37 oC for 3-4 days.
test if no haemagglutination occurs.
The test is invalid unless at least 8 of the 10 embryonated
embryos sUl""Vive. Hat\lest about 0.1 mL of the allantoic fluid 3-4 Potency
from each sUl\living embryo and examine each individual The vaccine complies with the requirements of the testes)
hat\lest for live virus by a haemagglutination test. mentioned under Immunogenicity (section 2-3-2) when
If haemagglutination is found for any of the fluids, cany out administered by a recommended route and method.
a further passage of that fluid in eggs and test for _____________________________________________ PhE~

haemagglutination; the inactivated virus hat\lest complies


with the test if no haemagglutination occurs.
2-4-2 Batch potency test
It is not necessaty to carry out the potency test (section 3-4) Feline Calicivirus Vaccine,
for each batch of vaccine if it has been carried out using a
batch of vaccine with a minimUlTI potency. Where the test is Inactivated
not carried out, an alternative validated method is used, the (Feline Calicivirosis Vaccine (In activa ted) )
criteria for acceptance being set with reference to a batch of Ph Bur monograph 1101)
vaccine that has given satisfactory results in the testes) PhE~ __________ ~ _________________________________
described under Potency. The following test may be used.
1 DEFINITION
Use 5 guinea-pigs that do not have specific antibodies. Feline calicivir~sis vaccine (inactivated) is a preparation of
Vaccinate each guinea-pig by the subcutaneous route with one or more suitable strains of feline calicivirus inactivated
one dose of vaccine. 21 days later, collect blood samples and while maintaining adequate immunogenic properties or of
separate the serum. Cany out tests on the serum for specific fractions of one or more strains of feline calicivirus with
antibodies by a suitable immunochemical method (2.7.1) adequate immunogenic properties. This monograph applies
such as single radial haemolysis or haemagglutination- to vaccines intended for the active immunisation of cats
inhibition, using reference sera to validate the test. against feline calicivirosis.
2016 Veterinary Vaccines Vet-251

2 PRODUCTION - copious 2
2-1 PREPARATION OF THE VACCINE Ocular discharge
The vaccine virus is grown in cell cultures. The virus harvest Weight loss 2
is inactivated; the virus may be disrupted and the fractions Virus excretion (total number of days):
purified and concentrated. The vaccine may be adjuvanted. ::; 4 days
5-7 days 2
2-2 SUBSTRATE FOR VIRUS PROPAGATION
> 7 days 3
2-2-1 Cell cultures
The cell cultures comply with the requirements for cell 2-4 MANUFACTURER'S TESTS
cultures for production of veterinary vaccines (5.2.4). 2-4-1 Residuallive virus
2-3 CHOICE OF VACCINE COMPOSITION The test for residual live calicivirus is carried out using 2
passages in cell cultures of the same type as those used for
The vaccine is shown to be satisfactory with respect to safety
preparation of the vaccine or in cell cultures shown to be at
(5.2.6) and efficacy (5.2.7) for the cats for which it is
least as sensitive; the quantity of inactivated virus harvest
intended.
used in the test is equivalent to not less than 25 doses of
The following tests for safety (section 2-3-1) and vaccine. The inactivated viral harvest complies with the test if
irnmunogenicity (section 2-3-2) may be used during the no live virus is detected.
demonstration of safety and efficacy.
2-4-2 Batch potency test
2-3-1 Safety It is not necessary to carry out the Potency test (section 3-4)
Carry out the test for each route and method of for each batch of vaccine if it has been carried out using a
administration to be recommended for vaccination. U se a batch of vaccine with a minimum potency. Where the test is
batch of vaccine containing not les s than the maximum not carried out, an alternative validated method is used, the
potency that may be expected in a batch of vaccine. criteria for acceptance being set with reference to a batch of
For each test, use not fewer than 8 cats of the minimum age vaccine that has given satisfactory results in the test described
to be recommended for vaccination and that do not have under Potency. The following test may be used.
antibodies against feline calicivirus. Administer to each cat Use for the test groups of 15 seronegative mice. Administer
1 dose of the vaccine. If the schedule to be recommended to each mouse half adose of the vaccine and 7 days later,
requires a 2nd dose, administer 1 dose after an interval of at repeat the administration. After 21 days following the first
least 14 days after the last administration. Observe the cats at injection, take blood samples and detelmine dle level of
least daily for at least 14 days after the last administration. antibodies against feline calicivirus by an
The vaccine complies with the test if no cat shows abnOlmal irnmunofiuorescence technique using pools of serum from
local or systemic reactions or dies from causes attributable to groups of 3 mice. The vaccine complies with the test if the
the vaccine. antibody levels are not significandy lower than those obtained
2-3-2 Immunogenicity with a batch of vaccine that has given satisfactory results in
A test is carried out for each strain of feline calicivirus in the the test described under Potency.
vaccine and for each route and method of administration to 3 BATCH TESTS
be recommended for vaccination, using in each case cats 3-1 Identification
8-12 weeks old. The vaccine administered to each cat is of When injected into animals that do not have specific
minimum potency. antibodies against feline calicivirus, the vaccine stimulates the
Use for the test not fewer than 20 cats dlat do not have formation of such antibodies.
antibodies against feline calicivirus. Vaccinate not fewer than 3-2 Bacteria and fungi
10 cats, according to the schedule to be recommended. The vaccine, including where applicable the diluent supplied
Maintain not fewer than 10 cats as controls. Challenge each
for reconstitution, complies with the test for sterility
cat after 4 weeks by the intranasal route with a sufficient prescribed in the monograph Vaccines for veteril1a1Y
quantity of a suspension of virulent feline calicivirus. Observe use (0062).
the cats at least daily for 14 days after challenge. Collect
nasal washings daily on days 2 to 14 to test for virus 3-3 Residuallive virus
excretion. Note daily the body temperature and signs of Carry out a test for residual live calicivirus using 10 doses of
disease using the scoring system shown below. vaccine and 2 passages in cell cultures of the same type as
those used for preparation of the vaccine or in cell cultures
The test is not valid if during the observation period after
shown to be at least as sensitive. The vaccine complies with
challenge, fewer than 80 per cent of the control cats show
the test if no live virus is detected. If the vaccine contains an
notable signs of feline calicivirosis (hyperthermia, buccal
adjuvant that would interfere with the test, where possible
ulcers, respiratory signs). The vaccine complies with the test
separate the adjuvant from the liquid phase by a medlod that
if during the observation period after challenge, the score for
does not inactivate or otherwise intelfere with detection of
the vaccinated cats is significandy lower than that for the
live virus.
controls.
Observed signs Score 3-4 Potency
Death 10 The vaccine complies with the requirements of the test
Depressed state 2 prescribed under Irnmunogenicity (section 2-3-2) when
Temperature 2': 39.5 oC administered by a recommended route and method.
Temperature ::; 37 oC 2 ~~ _________________________________________ PhE~

Ulcer (nasal or oral)


- small and few in number 1
- large and numerous 3
Nasal discharge
- slight
Vet-252 Veterinary Vaccines 2016

Feline Calicivirus Vaccine, Living *** any relevant parameters in a group of at least 8 animals
*** *** receiving the material used for the first passage and another
(Feline Calicivirosis Vaccine (Live), *** similar group receiving the virus at the final passage level.
Ph Bur monograph 1102) The vaccine virus complies with the test if no indication of
PhE~ _____________________________________________ increased virulence of the virus recovered for the final
passage compared with the material used for the first passage
1 DEFINITION is observed. If virus is not recovered after an initial passage in
Feline calicivirosis vaccine (live) is a preparation of one 01' 2 animal s and a subsequent repeat passage in 10 animals, the
more suitable strains of feline calicivirus. This monograph vaccine virus also complies with the test.
applies to vaccines intended for the active immunisation of
cats against feline calicivirosis.
2-3-3 Immunogenicity
A test is carried out for each strain of feline calicivirus in the
2 PRODUCTION vaccine, for each route and method of administration to be
2-1 PREPARATION OF THE VACCINE recommended for vaccination. The quantity of vaccine virus
The vaccine virus is grown in cell cultures. to be administered to each cat is not greater than the
2-2 SUBSTRATE FOR VIRUS PROPAGATION minimum virus titre to be stated on the label and the virus is
at the most attenuated passage level that will be present in a
2-2-1 Cell cultures
The cell cultures comply with the requirements for cell batch of vaccine.
cultures for production of veterinary vaccines (5.2.4). Use for the test not fewer than 20 cats, 8-12 weeks old, that
do not have antibodies against feline calicivirus. Vaccinate
2-3 CHOICE OF VACCINE VIRUS not fewer than 10 cats, according to the schedule to be
The vaccine virus is shown to be satisfactory with respect to recommended. Maintain not fewer than 10 cats as controls.
safety (5.2.6) and efficacy (5.2.7) for the cats for which it is Challenge each cat after 4 weeks by the intranasal route with
intended. a sufficient quantity of a suspension of virulent feline
The following tests for safety (section 2-3-1), increase in calicivirus virus. Observe the cats at least daily for 14 days
virulence (section 2-3-2) and immunogenicity (section 2-3-3) after challenge. Collect nasal washings daily on days 2 to
may be used during the demonstration of safety and efficacy. 14 to test for virus excretion. Note daily the body
2-3-1 Safety tempeniture and signs of disease using the scoring system
Can)' out the test for each route and method of shown below.
administration to be recommended for vaccination. The test is not valid if during the observation period after
U se vaccine virus at the least attenuated passage level that challenge, fewer than 80 per cent of the control cats show
will be present in a batch of the vaccine. notable signs of feline calicivirosis (hypelthermia, buccal
For each test, use not fewer than 8 cats of the minimum age ulcers, respiratol)' signs).
to be recommended for vaccination and that do not have The vaccine virus complies with dle test if during the
antibodies against feline calicivirus. Administer to each cat a observation period after challenge, the score for the
quantity of the vaccine virus equivalent to not less than vaccinated cats is significantly lower than dlat for the
10 times the maximum virus titre likely to be contained in controls.
1 dose of the vaccine. Observe the cats at least daily for at Observed signs Score
least 14 days. Death 10
The vaccine virus complies with the test if no cat shows Depressed state 2
abnormal local 01' systemic reactions, 01' dies from causes Temperature ~ 39.5 oC
attributable to the vaccine virus. Temperature :::; 37 oC 2
Ulcer (nasal 01' oral)
2-3-2 Increase in virulence
- small and few in number 1
Carry out the test according to general chapter 5.2.6 using
- large and numerous 3
cats that do not have antibodies against feline calicivirus.
Nasal discharge
If the properties of the vaccine virus allow sequential passage
- slight
through 5 groups via natural spreading, this method may be
- copious 2
used, otherwise passage as described below is carried out.
Ocular discharge 1
Administer to each cat of the 1sr group by a route to be Weight loss 2
recommended a quantity of the vaccine virus that will allow Virus excretion (total number of days):
recovery of virus for the passages described below. :::; 4 days
Administer the virus by the route to be recommended for 5-7 days 2
vaccination most likely to lead to reversion of virulence. After > 7 days 3
5 days, remove the nasal mucus, tonsils and trachea of each
cato Mix, homogenise in 10 mL of buffered saline and allow 3 BATCH TESTS
the solids to settle. Administer the supernatant by the 3-1 Identification
intranasal route to each cat of the next group. Carry out this When neutralised by one or more monospecific antisera, the
passage operation 4 times; verify the presence of the virus at vaccine no longer infects susceptible cell cultures into which
each passage. If the virus is not found at a passage level, it is inoculated.
repeat the passage by administration to a group of 3-2 Bacteria an4 fungi
10 animals. The vaccine,including where applicable the diluent supplied
If the 5th group of animal s shows no evidence of an increase for reconstitution, complies with the test for sterility
in virulence indicative of reversion during the observation prescribed in the monograph Vaccines for veterina¡y use
period, further testing is not required. Otherwise, carry out (0062).
an additional safety test and compare the clinical signs and
2016 Veterinary Vaccines Vet-253

3-3 Mycoplasmas (2.6.7) antibodies against C. felis. Administer to each cat 1 dose of
The vaccine complies with the test for mycoplasmas. the vaccine. If the schedule to be recommended requires a
3-4 Extraneous agents 2 nd dose, administer 1 dose after an interval of at least
Neutralise the vaccine virus with one or more suitable 14 days.
monospecific antisera against feline calicivirus and inoculate Observe the cats at least dai1y for at least 14 days after the
into cell cultures known for their susceptibility to viruses last administration.
pathogenic for the cato Carry out at least 1 passage and The vaccine complies with the test if no cat shows abnormal
maintain the cultures for 14 days. The vaccine complies with local 01' systemic reactions or dies from causes attributable to
the test if no cytopathic effect develops and there is no sign the vaccine.
of the presence of haemadsorbing agents.
2-2-2 Immunogenicity
3-5 Virus titre Carry out the test for each route and method of
Titrate the vaccine virus in suitable cell cultures at a administration to be recommended for vaccination, using in
temperature favourable to replication of the virus. each case cats not older dlan the minirnum age to be
The vaccine complies with the test if one dose contains not recommended for vaccination. The vaccine to be
les s than the minimum virus tiu"e stated on the label. administered to each cat is of minirnum potency.
3-6 Potency Vaccinate 10 cats that are free from antibodies against C. felis
The vaccine complies with the requirements of the test and keep 10 cats as controls. Not later than 4 weeks after the
prescribed under Irnmunogenicity (section 2-3-3) when last adminisu"ation of vaccine, administer by a suitable route
administered by a recommended route and method. It is not to each cat a quantity of a virulent strain of C. felis sufficient
necessary to carry out the potency test for each batch of the to produce in susceptible cats typical signs of disease such as
vaccine if it has been carried out on a representative batch conjunctivitis and nasal discharge. Observe the cats for
using a vaccinating dose containing not more than the 28 days. Where reduction of ch1amydophila excretion is to be
minimum virus titre stated on the label. c1aimed, collect nasal washings and/or conjunctival swabs
_____________________________________________ PhE~ on days 7, 14, 17, 21, 24 and 28 after challenge to test for
ch1amydophila excretion. The duration of excretion for the
vaccinated animals is significandy lower than for the controls.
Note daily the body temperature and signs of disease using a
suitable scoring system. The vaccine complies with the test if
Feline Chlamydiosis Vaccine ***** the score for the vaccinated cats is significantly lower than
** **
(Inactivated) ***
that for the controls.
(Feline Chlamydiosis Vaccine (In activa ted) J Ph Bur 2-3 MANUFACTURER'S TESTS
monograph 2324) 2-3-1 Batch potency test
~E~ _____________________________________________ It is not necessary to carry out the potency test (section 3-4)
for each batch of the vaccine if it has been canied out using
1 DEFINITION a batch of vaccine widl a minimum potency. Where the test
Feline chlamydiosis vaccine (inactivated) is a preparation of is not carried out on a batch, an alternative validated medl0d
one or more suitable su"ains of Chlamydophila felis, which is used, the criteria for acceptance being set with reference to
have been inactivated by a suitable method. This monograph a batch of vaccine that has given satisfactory results in the
applies to vaccines intended for administration to cats for potency test (section 3-4). The following test may be used.
active irnmunisation. Inject a suitable dose by a suitable route into each of
2 PRODUCTION 5 seronegative cats 01' another suitable species. Where the
2-1 PREPARATION OF THE VACCINE schedule stated on the label requires a booster injection to be
The seed material is cultured in embryonated hens' eggs given, a booster vaccination may also be given in this test
from a healthy fiod: or in suitable cell cultures (5.2.4). If the provided it has been demonsu"ated that this will still provide
vaccine contains more than one strain of bacterium, the a suitably sensitive test system. Before the vaccination and at
different strains are grown and harvested separately. a given interval usually within the range of 14-21 days after
The bacterial harvests are inactivated using suitable and the last injection, collect blood from each animal and prepare
validated methods. The suspensions may be treated to serum samples. Determine individually for each serum the
fragment the micro-organisms and the fragments may be titre of antibodies against each strain stated on the label,
purified and concentrated. The vaccine may contain using a suitable test such as enzyme-linked immunosorbent
adjuvants. assay (2.7.1). The vaccine complies with the test if the
antibody levels are not significandy lower than those obtained
2-2 CHOICE OF VACCINE COMPOSITION for a batch that has given satisfactory results in the potency
The vaccine is shown to be satisfactory with respect to safety test (section 3-4).
(5.2.6) and efficacy (5.2.7) in cats for which it is intended.
2-3-2 Bacterial endotoxins
The following tests for safety (section 2-2-1) and
A test for bacterial endotoxins (2.6.14) is carried out on the
immunogenicity (section 2-2-2) may be used during the
finallot 01', where the nature of the adjuvant prevents
demonstration of safety and efficacy.
perfOlmance of a satisfactory test, on the bulk antigen or the
2-2-1 Safety mixture of bulk antigens irnmediately before addition of the
Carry out the test for each route and method of adjuvant. The maximum acceptable amount of bacterial
administration to be recommended for vaccination. U se a endotoxins is that found for' a batch of vaccine that has been
batch of vaccine containing not less than the maximum shown to be satisfactory in dle safety test (section 2-2-1).
potency that may be expected in a batch of vaccine. The method chosen for determining the maxirnum
For each test, use not fewer than 8 cats of the minirnum age acceptable amount of bacterial endotoxins is subsequendy
to be recommended for vaccination and that do not have used for the testing of each batch.
Vet-254 Veterinary Vaccines 2016

3 BATCH TESTS The vaccine complies with the test if no cat shows abnormal
3-1 Identification local or systemic reactions or dies from causes attributable to
When injected into seronegative animals, the vaccine the vaccine.
stimulates the production of antibodies against each of the 2-3-2 Irnmunogenicity
strains of C. felis present in the vaccine. A test is carried out for each route and method of
3-2 Residuallive chlarnydophila administration to be recommended for vaccination, using in
The vaccine complies with a suitable test for residual live each case cats 8-12 weeks old. The vaccine administered to
chlamydophila. each cat is of minimum potency.
3-3 Bacteria and fungi U se for the test not fewer than 10 cats that do not have
The vaccine, including where applicable the diluent supplied antibodies against feline panleucopenia virus and canine
for reconstitution, complies with the test for sterility parvovirus. Vaccinate not fewer than 5 cats, according to the
prescribed in the monograph Vaccines for veterina1Y use schedule to be recommended. Maintain not fewer than 5 cats
(0062). as controls. Carry out leucocyte counts 8 days and 4 days
before challenge and calculate the mean of the 2 counts to
3-4 Potency
serve as the initial value. Challenge each cat after 20-22 days
The vaccine complies with the test for immunogenicity
by the intraperitoneal route with a sufficient quantity of a
(section 2-2-2).
suspension of virulent feline panleucopenia virus. Observe the
_____________________________________________ ~E~
cats at least daily for 14 days after challenge. Carry out
leucocyte counts on the 4 th, 6th, 8th and 10d1 days after
chaUenge.
The test is not valid if during the observation períod after
Feline Infectious Enteritis Vaccine, challenge, fewer than 100 per cent of the control cats show
on not fewer than one occasion a diminution in the number
Inactivated of leucocytes of at least 75 per cent of the initial value or die
Feline Panleucopenia Vaccine, Inactivated from panleucopenia. The vaccine complies with the test if
(Feline Infectious Enteritis (Feline Panleucopenia) Vaccine during the observation period after challenge, aU the
(In activa ted) J Ph Eur monograph 0794) vaccinated cats survive and show no signs of disease nor
~E~ _____________________________________________ leucopenia; that is to say, the diminution in the number of
leucocytes does not exceed, in any of the 4 counts,
1 DEFINITION 50 per cent of the initial value.
Feline infectious enteritis (feline panleucopenia) vaccine
2-4 MANUFACTURER'S TESTS
(inactivated) is a preparation of a suitable strain of feline
2-4-1 Residuallive virus
panleucopenia virus or canine parvovirus inactivated while
The test for residual live virus is carried out using a quantity
maintaining adequate immunogenic properties. This
of inactivated virus harvest equivalent to not less than
monograph applies to vaccines intended for the active
100 doses of the vaccine by a validated method such as the
immunisation of cats against feline infectious enteritis (feline
following: inoculate into suitable non-confiuent ceUs and after
panleucopenia) .
incubation for 8 days, make a subculture using trypsinised
2 PRODUCTION cells. After incubation for a further 8 days, examine the
2-1 PREPARATION OF THE VACCINE cultures for residual live parvovirus by an
The vaccine virus is grown in ceU cultures. The virus harvest immunofiuorescence test. The immunofiuorescence test may
is inactivated. The vaccine may be adjuvanted. be supplemented by a haemagglutination test or other
suitable tests on the supematant of the cell cultures.
2-2 SUBSTRATE FOR VIRUS PROPAGATION
The inactivated viral harvest complies with the test if no live
2-2-1 Cell cultures
virus is detected.
The ceU cultures comply with the requirements for ceU
cultures for production of veterinary vaccines (5.2.4). 2-4-2 Batch potency test
For routine testing of batches of vaccine, a test based on
2-3 CHOICE OF VACCINE COMPOSITION production of haemagglutination-inhibiting antibodies in
The vaccine is shown to be satisfactory with respect to safety guinea-pigs may be used instead of test 3-3-1 or 3-3-2
(5.2.6) and efficacy (5.2.7) for the cats for which it is described under Potency if a satisfactory correlation with the
intended. test for immunogenicity has been established.
The foUowing tests for safety (section 2-3-1) and
3 BATCH TESTS
immunogenicity (section 2-3-2) may be used during the
demonstration of safety and efficacy. 3-1 Identification
When injected into animals, the vaccine stimulates the
2-3-1 Safety production of antibodies against the parvovirus present in the
Carry out the test for each route and method of vaccíne.
administration to be recommended for vaccination. U se a
batch of vaccine containing not less than the maximum 3-2 Bacteria and fungi
potency that may be expected in a batch of vaccine. The vaccine, including where applicable the diluent supplied
for reconstitution, complies with the test for sterility
For each test, use not fewer than 8 cats of the minimum age prescribed in the monograph Vaccines for veterina1Y use
to be recommended for vaccination and that do not have
(0062).
antibodies against feline panleucopenia virus. Administer to
each cat 1 dose of the vaccine. If the schedule to be 3-3 Potency
recommended requires a 2nd dose, administer 1 dose after an Carry out test 3-3-1 or test 3-3-2.
ínterval of at least 14 days. Observe the cats at least daíly for
at least 14 days after the last administratíon.
2016 Veterinary Vaccines Vet-255

3-3-1 Test in cats for haemagglutination-inhibiting active immunisation of cats against feline infectious enteritis
antibodies ' (feline panleucopenia).
U se for the test not fewer than 4 cats, 8-12 weeks old, that
2 PRODUCTION
do not have antibodies against feline panleucopenia virus and
2-1 PREPARATION OF THE VACCINE
canine parvovirus. Vaccinate not fewer than 2 cats with
The vaccine virus is grown in cell cultures.
1 dose of the vaccine. Maintain not fewer than 2 cats as
controls. After 21 days, draw a blood sample from each cat 2-2 SUBSTRATE FOR VIRUS PROPAGATION
and separate the serum from each sample. Inactivate each 2-2-1 Cel1 cultures
serum by heating at 56 oC for 30 mino To 1 volume of each The cell cultures comply with the requirements for cell
serum add 9 volumes of a 200 gIL suspension of light cultures for production of veterinary vaccines (5.2.4).
kaolin R in phosphate buffered saline pH 7.4 R. Shake each
2-3 CHOICE OF VACCINE vmus
mixture for 20 mino Centrifuge, collect the supernatant and
The vaccine virus is shown to be satisfactory with respect to
mix with 1 volume of a concentrated suspension of pig
safety (5.2.6) and efficacy (5.2.7) for the cats for which it is
erythrocytes. Allow to stand a:t 4 oC for 60 min and
intended, including safety for pregnant queens if the vaccine
centrifuge. The dilution ofthe serum obtained is 1:10. Using
may be used in such queens. If the virus is excreted in the
each serum, prepare a series of twofold dilutions.
faeces, the effect in pregnant queens must be documented.
To 0.025 mL of each of the latter dilutions add 0.025 mL of
a suspension of canine parvovirus or feline panleucopenia The following tests for safety (section 2-3-1), increase in
virus antigen containing 4 haemagglutinating units. Allow to virulence (section 2-3-2) and immunogenicity (section 2-3-3)
stand at 37 oC for 30 min and add 0.05 mL of a suspension may be used during the demonstration of safety and efficacy.
of pig erythrocytes containing 30 x 10 6 cells per millilitre. 2-3-1 Safety
Allow to stand at 4 oC for 90 min and note the last dilution Carry out the test for each route and method of
of serum that still completely inhibits haemagglutination. administration to be recommended for vaccination.
The test is not valíd if either control cat develops antibodies Use vaccine virus at the least attenuated passage level that
against canine parvovirus or feline panleucopenia virus. will be present in a batch of the vaccine.
The vaccine complíes with the test if both vaccinated cats 2-3-1-1 General safety. For each test, use not fewer than
have developed titres of at least 1:20. 5 cats of the mínimum age to be recommended for
3-3-2 Test in cats for virus-neutralising antibodies vaccination and that do not have antibodies against feline
Use for the test not fewer than 2 cats, 8-12 weeks old, that panleucopenia virus and canine parvovirus. Make counts of
have antibody titres less than 4ND so per 0.1 mL of serum leucocytes in circulating blood on days 8 and 4 before
measured by the method described below. Vaccinate each cat injection of the vaccine virus and calculate the mean of the
according to the recommended schedule. 14 days after 2 counts to serve as the initial value. Administer to each cat a
vaccination, examine the serum of each cat as follows. Heat quantity of the vaccine virus equivalent to not less than
the serum at 56 2C for 30 min and prepare serial dilutions 10 times the maximum virus titre likely to be contained in
using a medium suitable for felíne cells. Add to each dilution 1 dose of dle vaccine. Observe dle cats at least daily for at
an equal volume of a virus suspension containing an amount least 14 days. Make leucocyte counts on dle 4 th , 6 th, 8th and
of virus such that when the volume of serum-virus mixture 10th days after inoculation.
appropriate for the assay system is inoculated into cell The vaccine virus complies widl the test if no cat shows
cultures, each culture receives approximately 104 CCID so . abnormallocal or systemic reactions, signs of disease or dies
Incubate the mixtures at 37 oC for 1 h and inoculate 4 feline from causes attributable to dle vaccine virus and if, for each
cell cultures with a suitable volume of each mixture. Incubate cat and each blood count, the number of leucocytes is not
the cell cultures at 37 oC for 7 days, passage and incubate for less than 50 per cent of the initial value.
a further 7 days. Examine the cultures for evidence of 2-3-1-2 Safety in pregnant queens. If the vaccine is intended
specific cytopathic effects and calculate the antibody titre. for use or may be used in pregnant queens, use not fewer
The vaccine complíes with the test if the mean titre is not than 5 queens per group, at the stage of pregnancy to be
less than 32 ND so per 0.1 mL of serum. If one cat fails to recommended 01' at a range of stages of pregnancy according
respond, repeat the test using 2 more cats and calculate the to the schedule to be recommended. Administer to each
result as the mean of the titres obtained from all of the 3 cats queen a quantity of vaccine virus at least equivalent to the
that have responded. maximum virus titre likely to be contained in 1 dose of the
_____________________________________________ PhE~
vaccine. Observe the queens at least daily until 1 day after
parturition and observe their kittens until at least dle age of
3 weeks.
The vaccine virus complíes with dle test if no queen shows
abnormal local 01' systemic reactions, signs of disease 01' dies
Feline Infectious Enteritis Vaccine, ***** from causes attributable to the vaccine virus and if no
** ** adverse effects on the pregnancy or the offspring, such as
living *** foetal resorption 01' ataxia in the kittens, are noted.
Feline Panleucopenia Vaccine, Living
2-3-2 Increase in virulence
(Feline Infectious Entel1tis (Feline Panleucopenia) Vaccine
Canyr out the test according to general chapter 5.2.6 using
(Live), Ph Eur 11lonograph 0251)
cats ofthe minimum age to be recommended for vaccination
PhE~ _____________________________________________
and that do not have antibodies agaínst feline panleucopenia
1 DEFINITION virus and canine parvovirus. If the properties of the vaccine
Felíne infectious enteritis (felíne panleucopenia) vaccine (live) virus allow sequential passage through 5 groups via natural
is a preparation of a suitable strain of feline panleucopenia spreading, this method may be used, otherwise passage as
virus. This monograph applies to vaccines intended for the described below is carried out.
Vet-256 Veterinary Vaccines 2016

Administer to each cat of the 1sr group by a route to be of the cells with monoclonal antibodies specific for feline
recommended a quantity of the vaccine virus that will allow panleucopenia virus and a proportion with monoclonal
recovery of virus for the passages described below. Collect antibodies specific for canine parvovirus. Feline
the faeces from each cat from the 2nd to the 10th day after panleucopenia virus is detected but no canine parvovirus is
administration of the virus J check them for the presence of detected in the cells inoculated with the vaccine.
the virus and pool the faeces containing virus. Administer 3-2 Bacteria and fungi
1 mL of the suspension of pooled faeces by either the oral or The vaccine J including where applicable the diluent supplied
the intranasal route to each cat of the next group. Carry out for reconstitutionJ complies with the test for sterility
this passage operation not fewer than 4 times; verify the prescribed in the monograph Vaccines for veterinary
presence of the virus at each passage. If the virus is not use (0062).
found at a passage levelJ repeat the passage by administration
to a group of 10 animals. 3-3 Mycoplasmas (2.6.7)
The vaccine complies with the test for mycoplasmas.
If the 5th group of animals shows no evidence of an increase
in virulence indicative of reversion during the observation 3-4 Extraneous agents
periodJ further testing is not required. Otherwise J carry out Neutralise the vaccine virus with a suitable monospecific
an additional safety test and compare the clinical signs and antiserum against feline panleucopenia virus and inoculate
any relevant parameters (count of white blood cells J results of into cell cultures known for their susceptibility to viruses
histological examination of the thymus and titre of excreted pathogenic for the cato Carry out at least 1 passage and
virus) in a group of at least 8 animals receiving the material maintain the cultures for 14 days. The vaccine complies with
used for the 1sr passage and another similar group receiving the test if no cytopathic effect develops and there is no sign
the virus at the final passage level. of the presence of haemadsorbing agents.
The vaccine virus complies with the test if no cat dies 01' 3-5 Virus titre
shows signs attributable to the vaccine virus and no Titrate the vaccine virus in suitable cell cultures. The vaccine
indication of increasing virulence of the virus recovered for complies with the test if one dose contains not les s than the
the final passage compared with the material used for the minimum virus titre stated on the label.
1sr passage is observed. Account is taken J notably, of the 3-6 Potency
count of white blood cells, of results of histological The vaccine complies with the requirements of the test
examination of the thymus and of the titre of excreted virus. prescribed under Irnmunogenicity (section 2-3-3) when
If virus is not recovered after an initial passage in 2 animals administered by a recommended route and method. It is not
and a subsequent repeat passage in 10 animals, the vaccine necessary to carry out the potency test for each batch of the
virus also complies with the test. vaccine if it has been carried out on a representative batch
2-3-3 Irnmunogenicity using a vaccinating dose containing not more than the
A test is carried out for each route and method of minimum virus titre stated on the label.
administration to be recommended for vaccination. _____________________________________________ PhE~

The quantity of vaccine virus to be administered to each cat


is not greater than the minimum virus titre to be stated on
the label and the virus is at the most attenuated passage level
that will be present in a batch of vaccine.
Feline Leukaemia Vaccine, ***
Use for the test not fewer than 10 cats, 8-12 weeks old, that ** **
do not have antibodies against feline panleucopenia virus and Inactivated *****
canine parvovirus. Vaccinate not fewer than 5 cats, according (Feline Leukaemia Vaccine (Inactivated) J
to the schedule to be recommended. Maintain not fewer than Ph Bur monogmph 1321)
5 cats as controls. Carry out leucocyte counts 8 days and PhE~ _____________________________________________
4 days before challenge and calculate the mean of the 2
counts to serve as the initial value. Challenge each cat after 1 DEFINITION
20-22 days by the intraperitoneal route with a sufficient Feline leukaemia vaccine (inactivated) is a preparation of
quantity of a suspension of virulent feline panleucopenia irnmunogens from a suitable strain of feline leukaemia virus.
virus. Observe the cats at least daily for 14 days after This monograph applies to vaccines intended for the active
challenge. Carry out leucocyte counts on the 4 th, 6th, 8th and irnmunisation of cats against feline leukaemia.
10 d, days after challenge.
2 PRODUCTION
The test is not valid if during the observation period after 2-1 PREPARATION OF THE VACCINE
challenge, fewer than 100 per cent of the control cats show,
The irnmunogens consist either of a suitable strain of feline
on fewer than one occasion, a diminution in the number of
leukaemia virus inactivated while maintaining adequate
leucocytes of at least 75 per cent of the initial value 01' die
irnmunogenic properties or of a fraction of the virus with
from feline panleucopenia. The vaccine virus complies with
adequate irnmunogenic properties; the irnmunogenic fraction
the test if during the observation period after challengeJ all
may be produced by recombinant DNA technology.
the vaccinated cats survive and show no signs of disease nor
The vaccine may be adjuvanted.
leucopenia; that is to say, the diminution in the number of
leucocytes does not exceed, in any of the 4 counts, 2-2 CHOICE OF VACCINE COMPOSITION
50 per cent of the initial value. The vaccine is shown to be satisfactory with respect to safety
(5.2.6) andefficacy (5. 2.7) for the cats for which it is
3 BATCH TESTS
intended.
3-1 Identification
Carry out replication of the vaccine virus in a susceptible cell The following tests for safety (section 2-2-1) and
line in a substrate suitable for a fiuorescent antibody test 01' irnmunogenicity (section 2-2-2) may be used during the
peroxidase test. Prepare suitable controls. Test a proportion demonstration of safety and efficacy.
2016 Veterinary Vaccines Vet-257

2-2-1 Safety 2-4 MANUFACTURER'S TESTS


Carry out the test for each route and method of 2-4-1 Residuallive virus
administration to be recommended for vaccination. Use a Where applicab1e, the test for residual live virus is carried out
batch of vaccine containing not less than the maximum using a quantity of inactivated virus harvest equivalent to not
potency that may be expected in a batch of vaccine. less than 25 doses of vaccine and 2 passages in the same type
2-2-1-1 General safety and immunosuppression. Use for the test of cell cultures as used for the production of the vaccine or in
not fewer than 15 cats of the minimum age to be cell cultures shown to be at least as sensitive. The inactivated
recommended and that do not have antibodies against gp 70 viral harvest complies with the test if no live virus is detected.
antigen of feline leukaemia virus nor display viraemia or 2-4-2 Batch potency test
antigenaemia at the time of the test; absence of antibodies It is not necessary to carry out the Potency test (section 3-4)
and antigen is demonstrated by enzyme-linked for each batch of vaccine if it has been carried out using a
immunosorbent assay (2. 7.1). Administer to each of not batch of vaccine with a minimum potency. Where the test is
fewer than 10 cats 1 dose of the vaccine. If the schedule to not carried out, an alternative validated method is used, the
be recommended requires a 2nd dose, administer 1 dos e after criteria for acceptance being set with reference to a batch of
an interval of at least 14 days. Maintain not fewer than 5 cats vaccine that has given satisfactory results in the test described
as controls. Record the body temperature of each cat on under Potency.
the day before each vaccination, at the time of vaccination,
2-4-3 Bacterial endotoxins
4 h and 8 h later, and once per day during the
For vaccines produced by recombinant DNA technology with
4 following days. Observe the cats at least daily for not less
a bacterial host cell such as Bscherichia coli, a test for bacterial
than 4 weeks after the last vaccination. 1, 2 and 4 weeks after
endotoxins (2.6.14) is carried out on each final lot or, where
the last vaccination, submit the cats to suitable tests for
the nature of the adjuvant prevents performance of a
evidence of an immunosuppressive effect.
satisfactory test, on the antigen immediately before addition
The vaccine complies with the test if no cat shows abnormal of the adjuvant. The value found is within the limit approved
local or systemic reactions or dies from causes attributable to for the particular vaccine and which has been shown to be
the vaccine and if no significant difference is observed in safe for cats.
vaccinated cats compared with controls regarding
immunosuppressive effect. 3 BATCH TESTS
3-1 Identification
2-2-2 Immunogenicity When injected into healthy cats that do not have specific
A test is carried out for each route and method of antibodies against the antigen or antigens stated on the label,
administration to be recommended, using in each case cats of the vaccine stimulates the production of such antibodies.
the minimum age to be recommended for vaccination.
The vaccine administered to each cat is of minimum 3-2 Bacteria and fungi
potency. The vaccine, including where applicable the diluent supplied
for reconstitution, complies with the test for sterility
Use for the test not fewer than 25 cats that do not have
prescribed in the monograph Vaccines for veterinary use
antibodies against the antigens of feline leukaemia virus and
(0062).
against the feline oncogene membrane antigen (anti-FOCMA
antibodies), and showing no viraemia or antigenaemia at the 3-3 Residuallive virus
time of the test. Vaccinate not fewer than 15 cats according If the vaccine contains inactivated virus, carry out a test for
to the schedule to be recommended. Maintain not fewer than residual live feline leukaemia virus by making 2 passages on
10 cats as controls. Challenge each cat after 14 days by the susceptible cell cultures. The vaccine complies with the test if
peritoneal or oronasal route, on one or several occasions, no virus is detected. If the vaccine contains an adjuvant, if
with a sufficient quantity of suspension of an possible separate the adjuvant from the liquid phase by a
epidemiologically relevant virulent strain of feline leukaemia method that does not inactivate the virus nor interfere in any
virus, consisting predominantly of type A virus. Observe the other way with the detection of virus.
cats at least daily for 15 weeks and, from the 3rd week 3-4 Potency
onwards, test each week for viraemia or antigenaemia The vaccine complies with the requirements of the test
(p27 protein) by suitable methods such as prescribed under Immunogenicity (section 2-2-2) when
immunofluorescence on circulating leucocytes or enzyme- administered by a recommended route and method.
linked immunosorbent assay. A cat is considered persistently _____________________________________________ PhE~

infected if it shows positive viraemia or antigenaemia for


3 consecutive weeks or on 5 occasions, consecutively or not,
between the 3rd and the 15 ü1 week.
The test is not valid if during the observation period after
Feline Viral Rhinotracheitis ***
** **
challenge, fewer than 80 per cent of the control cats show
persistant viraemia or antigenaemia. The vaccine complies
with the test if during the observation period after challenge,
Vaccine, Inactivated *****
not fewer than 80 per cent of the vaccinated cats show no (Feline Viral Rhinotracheitis Vaccine (In activa ted) J
persistent infection. Ph Bur monograph 1207)
PhE~ _____________________________________________
2-3 IN-PROCESS CONTROL TESTS
During production, suitable immunochemical tests are 1 DEFINITION
carried out for the evaluation of the quality and purity of the Feline viral rhinotracheitis vaccrn.e (inactivated) is a
viral antigens included in the vaccine composition. preparation of a suitable strain of feline rhinotracheitis virus
The values found are within the limits approved for the (feline herpesvirus 1), inactivated while maintaining adequate
particular vaccine. immunogenic properties, or of an inactivated fraction of the
virus having adequate immunogenic properties. This
Vet-258 Veterinary Vaccines 2016

monograph applies to vaccines intended for the active Nasal discharge, slight
immunisation of cats against feline viral rhinotracheitis. Nasal discharge, copious 2
2 PRODUCTION Cough 2
2-1 PREPARATION OF THE VACCINE Sneezing 1
Sneezing, paroxysmal 2
The vaccine virus is grown in cell cultures. The virus harvest
Ocular discharge, slight 1
is inactivated; the virus may be disrupted and the fractions
Ocular discharge, serious 2
purified and concentrated. The vaccine may be adjuvanted.
Conjunctivitis 2
2-2 SUBSTRATE FOR VIRUS PROPAGATION Weight loss ::: 5.0 per cent 5
2-2-1 Cell cultures Virus excretion (total number of days):
The cell cultures comply with the requirements for cell ~ 4 days
cultures for production of veterinary vaccines (5.2.4). 5-7 days 2
2-3 CHOICE OF VACCINE COMPOSITION > 7 days 3
The vaccine is shown to be satisfactory with respect to safety 2-4 MANUFACTURER'S TESTS
(5.2.6) and efficacy (5.2.7) for the cats for which ir is 2-4-1 Residuallive virus
intended. The test for residual live virus is carried out using 2 passages
The following tests for safety (section 2-3-1) and in celI cultures of the same type as those used for preparation
immunogenicity (section 2-3-2) may be used during the of the vaccine or in celI cultures shown to be at least as
demonstration of safety and efficacy. sensitive; the quantity of inactivated virus harvest used in the
2-3-1 Safety test is equivalent to not less than 25 doses of vaccine.
Carry out the test for each route and method of The inactivated viral harvest complies with the test if no live
administration to be recoÍnmended for vaccination. U se a virus is detected.
batch of vaccine containing not less than the maximum 2-4-2 Batch potency test
potency that may be expected in a batch of vaccine. It is not necessary to carry out the Potency test (section 3-4)
For each test, use not fewer than 8 cats of the minimum age for each batch of vaccine if it has been canied out using a
to be recommended for vaccination and that do not have batch of vaccine ·with a miriimum potency. Where the test is
antibodies against feline herpesvirus 1 or against a fraction of not carried out, an alternative validated method is used, the
the virus. Administer to each cat 1 dose of the vaccine. criteria for acceptance being set with reference to a batch of
If the schedule to be recommended requires a 2nd dose, vaccine that has given satisfactory results in the test described
administer 1 dos e after an interval of at least 14 days after under Potency. The following test may be used.
the last administration. Observe the cats at least daily for at Use for the test a group of 15 seronegative mice. Administer
least 14 days after the last administration. The vaccine to each mouse half adose of the vaccine and, 7 days later,
complies with the test if no cat shows abnormal local or repeat the admi.l1istration. 21 days after d1e first injection,
systemic reactions or dies from causes attributable to the take blood samples and determine the level of antibodies
vaccine. against feline herpesvirus 1 by a suitable immunochemical
2-3-2 Irnmunogenicity method (2.7.1), such as an immunofiuorescence technique
A test is carried out for each route and method of using pools of serum from groups of 3 mice. The vaccine
administration to be recommended for vaccination, using in complies with the test if the antibody leve1s are not
each case cats 8-12 weeks old. The vaccine administered to significandy lower than those obtained with a batch of
each cat is of minimum potency. vaccine that has given satisfactory results in the test described
under Potency.
Use for the test not fewer than 20 cats that do not have
antibodies against feline herpesvirus 1 or against a fraction of 3 BATCH TESTS
the virus. Vaccinate not fewer than 10 cats, according to the 3-1 Identification
schedule to be recommended. Maintain not fewer than When administered to animals that do not have specific
10 cats as controls. Challenge each cat after 4 weeks by the antibodies against feline herpesvirus 1 or against the fraction
intranasal route with a quantity of a suspension of virulent of the virus used to produce the vaccine, the vaccine
feline herpesvirus 1 sufficient to produce typical signs of the stimulates the production of such antibodies.
disease such as fever, nasal discharge and cough in a cat that 3-2 Bacteria and fungi
does not have antibodies against feline herpesvirus 1 or a The vaccine, inc1uding where applicable the diluent supplied
fraction of the virus. Observe the cats at least daily for for reconstitution, complies with the test for sterility
14 days after challenge. Collect nasal washings daily on days prescribed in the monograph Vaccines for veterina1Y
2 to 14 after challenge to test for virus excretion. Note daily use (0062).
the body temperature and signs of disease using the scoring
3-3 Residuallive virus
system shown below.
Carry out a test for residual live feline herpesvirus 1 using
The vaccine complies wid1 the test if the score for the 10 doses of vaccine and 2 passages in cell cultures of the
vaccinated cats is significandy lower than that for the same type as those used for preparation of the vaccine, or in
controls. other suitably sensitive celI cultures. The vaccine complies
Sign Score with the test if no live virus is detected. If the vaccine
Death 10 contains an adjuvant that interferes with the test, where
Depressed state 2 possible separate the adjuvant from the liquid phase by a
Temperature: method that does not inactivate or othetwise interfere with
39.5 oC - 40.0 oC 1 detection of live virus.
2': 40.0 oC 2
~ 37.0 oC 3
Glossitis 3
2016 Veterinary Vaccines Vet-259

3-4 Potency to settle. Administer 1 mL of the supernatant by the


The vaccine complies with the requirements of the test intranasal route to each cat of the next group. Carry out this
prescribed under Immunogenicity (section 2-3-2) when passage operation not fewer than 4 times; verify the presence
administered by a recommended route and method. of the virus at each passage. If the virus is not found at a
_ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ PhEur passage level, repeat the passage by administration to a group
of 10 animals.
If the 5th group of animals shows no evidence of an increase
in virulence indicative of reversion during the observation
Feline Viral Rhinotracheitis period, further testing is not required. Otherwise, carry out
an additional safety test and compare the c1inical signs and
Vaccine, Living any relevant parameters in a group of at least 8 animal s
(Feline Viral Rhinotracheitis Vaccine (Live)) receiving the material used for the first passage and another
Ph Bur 111onograph 1206) similar group receiving the virus at the final passage level.
~E~ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ __ The vaccine virus complies with the test if no indication of
increased virulence of the virus recovered for the final
1 DEFINITION passage compared with the material used for the first passage
Feline viral rhinotracheitis vaccine (live) is a preparation of a is observed. If virus is not recovered after an initial passage in
suitable strain of feline rhinotracheitis virus (feline 2 animals and a subsequent repeat passage in 10 animals, the
herpesvirus 1). This monograph applies to vaccines intended vaccine virus also complies with the test.
for the active immunisation of cats against feline viral
rhinotracheitis. 2-3-3 Irnmunogenicity
A test is carried out for each route and method of
2 PRODUCTION administration to be recommended for vaccination.
2-1 PREPARATION OF THE VACCINE The quantity of vaccine virus to be administered to each cat
The vaccine virus is grown in cell cultures. is not greater than the minimum virus titre to be stated on
2-2 SUBSTRATE FOR VIRUS PROPAGATION the label and the virus is at the most attenuated passage level
2-2-1 Cell cultures that will,be present in a batch of vaccine.
The cell cultures comply with the requirements for cell Use for the test not fewer than 20 cats, 8-12 weeks old, that
cultures for production of veterinary vaccines (5.2.4). do not have antibodies against feline herpesvirus 1. Vaccinate
not fewer than 10 cats, according to the schedule to be
2-3 CHOICE OF VACCINE VIRUS recommended. Maintain not fewer than 10 cats as controls.
The vaccine virus is shown to be satisfactory with respect to Challenge each cat after 4 weeks by the intranasal route with
safety (5.2.6) and efficacy (5.2.7) for the cats for which it is a quantity of a suspension of virulent feline herpesvirus 1
intended. sufficient to cause typical signs of disease such as fever, nasal
The following tests for safety (section 2-3-1), increase in discharge and cough. Observe the cats at least daily for
virulence (section 2-3-2) and immunogenicity (section 2-3-3) 14 days after challenge. Collect nasal washings daily on days
may be used during the demonstration of safety and efficacy. 2 to 14 after challenge to test for virus excretion. Note daily
2-3-1 Safety the body temperature and signs of disease using the scoring
Carry out the test for each route and method of system shown below.
administration to be recommended for vaccination. The vaccine virus complies with the test if, during the
U se vaccine virus at the least attenuated passage level that observation period after challenge, the score for the
will be present in a batch of the vaccine. vaccinated cats is significantly lower than that for the
For each test, use not fewer than 8 cats of the minimum age controls.
to be recommended for vaccination and that do not have Sign Score
antibodies against feline herpesvirus 1. Administer to each cat Death 10
a quantity of the vaccine virus equivalent to not less than Depressed state 2
10 times the maximum virus titre likely to be contained in Temperature:
1 dose of the vaccine. Observe the cats at least daily for at 39.5 oC - 40.0 oC
least 14 days. 2': 40.0 oC 2
:::; 37.0 oC 3
The vaccine virus complies with the test if no cat shows
abnormal local 01' systemic reactions 01' signs of disease, 01' Glossitis 3
dies from causes attributable to the vaccine virus. Nasal discharge, slight 1
Nasal discharge, copious 2
2-3-2 Increase in virulence Cough 2
Carry out the test according to general chapter 5.2.6 using Sneezing 1
cats that do not have antibodies against feline herpesvirus 1. Sneezing, paroxysmal 2
If the properties of the vaccine virus allow sequential passage Ocular discharge, slight
through 5 groups via natural spreading, this method may be Ocular discharge, serious 2
used, otherwise passage as described below is carried out. Conjunctivitis 2
Administer to each cat of the 1st group by a route to be Weight loss 2': 5.0 per cent 5
recommended a quantity of the vaccine virus that will allow Virus excretion (total number of days):
recovery of virus for the passages described below. :::; 4 days
Administer the virus by the route to be recommended for 5-7 days 2
vaccination most likely to lead to reversion of virulence. After > 7 days 3
2-4 days, remove the nasal mucus, tonsils and local
lymphatic ganglia and the trachea of each cat. Mix,
homogenise in 10 mL of buffered saline and allow the solids
Vet-260 Veterinary Vaccines 2016

3 BATCH TESTS 2-3 CHOICE OF VACCINE VIRUS


3-1 Identification The vaccine virus is shown to be satisfactory with respect to
When mixed with a monospecific antiserum, the vaccine no safety (5.2.6) and efficacy (5.2.7) for the ferrets, or for the
longer infects susceptible cell cultures into which it is ferrets and minks for which it is intended.
inoculated. The following tests for safety (section 2-3-1) and
3-2 Bacteria and fungi immunogenicity (section 2-3-3) may be used during the
The vaccine, including where applicable the diluent supplied demonstration of safety and efficacy. The tests are performed
for reconstitution, complies with the test for sterility in each species for which the vaccine is intended.
prescribed in the monograph Vaccines for veterz'na¡y use 2-3-1 Safety
(0062). Carry out the test for each route and method of
3-3 Mycoplasmas (2.6.7) administration to be recommended for vaccination.
The vaccine complies with the test for mycoplasmas. U se vaccine virus at the least attenuated passage level that
will be present in a batch of the vaccine.
3-4 Extraneous agents
Neutralise the vaccine virus with a suitable monospecific For each test, use not fewer than 5 ferrets ami/or minks of
antiserum against feline herpesvirus 1 and inoculate into cell the minimum age to be recommended for vaccination and
cultures known for their susceptibility to viruses pathogenic that do not have antibodies against distemper virus.
for the cato Cany out at least 1 passage and maintain the Administer to each ferret anci/or mink a quantity of the
cultures for 14 days. The vaccine complies with the test if no vaccine virus equivalent to not less than 10 times the
cytopathic effect develops and there is no sign of the maximum virus titre likely to be contained in 1 dose of the
presence of haemadsorbing agents. vaccine. Observe the animal s at least daily for 42 days.
3-5 Virus titre The vaccine complies with the test if no animal shows
Titrate the vaccine virus in suitable cell cultures and at a abnormallocal or systematic reactions, signs of disease or
temperature favourable to replication of the virus. die s from causes attributable to the vaccine.
The vaccine complies with the test if 1 dose contains not less 2-3-2 Increase in virulence
than the minimum virus titre stated on the label. Carry out the test according to general chapter 5.2.6 using
animals of the most susceptible target species. Use animals
3-6 Potency
that do not have antibodies against distemper virus. If the
The vaccine complies with the requirements of the test
properties of the vaccine virus allow sequential passage
prescribed under Immunogenicity (section 2-3-3) when
through 5 groups via natural spreading, this method may be
administered by a recommended route and method. It is not
used, otherwise passage as described below is carried out.
necessaly to carry out the potency test for each batch of the
vaccine if it has been carried out on a representative batch Administer to each animal of the 1st group by a route to be
using a vaccinating dose containing not more than the recommended a quantity of the vaccine virus that will allow
minimum virus titre stated on the label. recovery of virus for the passages described below.
_____________________________________________ PhE~
Administer the virus by the route to be recommended for
vaccination most likely to lead to reversion to virulence. After
5-10 days, prepare a suspension from, for example, the nasal
mucosa, tonsils, thymus, spleen and the lungs and their local
lymph nodes of each animal and pool the samples.
Ferret and Mink Distemper
*
****** Administer 1 mL of the pooled samples by the intranasal
route to each animal of the next group. Carry out this
Vaccine, living ***** passage operation not fewer than 4 times; verify the presence
(Distemper Vaccine (Live) for Mustelids) of the virus at each passage. If the virus is not found at a
Ph Bur monograph 0449) passage level, repeat the passage by administration to a group
PhE~ _____________________________________________ of 10 animals.
If the 5 th group of animals shows no evidence of an increase
1 DEFINITION
in virulence indicative of reversion during the observation
Distemper vaccine (live) for mustelids is a preparation of a
period, further testing is not required. Otherwise, carry out
suitable strain of distemper virus that is attenuated for ferrets,
an additional safety test and compare the clinical signs and
or for ferrets and minks. This monograph applies to vaccines
any relevant parameters in a group of at least 8 animals
intended for the active immunisation of ferrets, or ferrets and
receiving the material used for the 1st passage and another
minks, against disease caused by distemper virus.
similar group receiving the virus at the final passage level.
2 PRODUCTION The vaccine virus complies with the test if no indication of
2-1 PREPARATION OF THE VACCINE an increased virulence of the virus recovered for the final
The vaccine virus is grown in embryonated hens' eggs or in passage compared with the material used for the 1st passage
cell cultures. is observed. If virus is not recovered after an initial passage in
2-2 SUBSTRATE FOR VIRUS PROPAGATION 2 animals and a subsequent repeat passage in 10 animals, the
2-2-1 Embryonated hens' eggs vaccine virus also complies witÍ1 the test.
If the vaccine virus is grown in embryonated hens' eggs, they 2-3-3 Irnmunogenicity
are obtained from flocks free from specified pathogens (SPF) A test is carried 9ut for each route and method of
(5.2.2). administration to be recommended for vaccination using
animal s of the target species (ferrets and/or minks) for which
2-2-2 Cell cultures
the vaccine is intended. Use animal s not older than the
If the vaccine virus is grown in cell cultures, they comply
minimum age to be recommended for vaccination.
with the requirements for cell cultures for production of
The quantity of the vaccine virus administered to each
veterinary vaccines (5.2.4).
animal is not greater than the minimum virus titre to be
2016 Veterinary Vaccines Vet-261

stated on the label and the virus is at dle most attenuated 2 PRODUCTION
passage level that will be present in a batch of vaccine. 2-1 PREPARATION OF THE VACCINE
Use for the test not fewer than 7 ferrets and/or minks that do The vaccine virus is grown in cell cultures and then
not have antibodies against distemper virus. Vaccinate not separated from cellular material by filtration 01" other suitable
fewer than 5 animals, according to the schedule to be procedures. The harvested virus is inactivated in suitable
recommended. Maintain not fewer than 2 animals as conditions and may be concentrated and purified. It is used
controls. Challenge each animal after 20-22 days by the for the preparation of vaccine irnmediately 01" after storage at
intramuscular route with a quantity of a suspension of a temperature shown to be consistent with antigen stability.
virulent distemper virus sufficient to cause the death of a The vaccine is prepared from inactivated virus by blending
ferret andlor a mink. Observe the animals at least daily for with one or more adjuvants. For a given strain, the quantity
21 days after challenge. Animals displaying typical signs of of 146S antigen blended in each batch of vaccine is not lower
serious infection with distemper virus are euthanised to avoid than that of a batch of vaccine that has been found
unecessary suffering. satisfactory with respect to Immunogenicity.
The test is not valid if 1 or both of the control animals do 2-2 SUBSTRATE FOR VIRUS PROPAGATION
not die of distemper. The vaccine virus complies with the 2-2-1 Cell cultures
test if the vaccinated animals remain in normal health. The cell cultures comply with the requirements for cell
3 BATCH TESTS cultures for production of veterinary vaccines (5.2.4).
3-1 Identification
2-3 VALIDATION OF THE INACTIVATION
The vaccine mixed with a specific distemper antiserum no
PROCEDURE
longer provokes cytopathic effects in susceptible cell cultures
During inactivation, the virus titre is monitored by a sensitive
or lesions on the chorio-allantoic membranes of fertilised hen
and reproducible technique. The inactivation procedure is
eggs 9-11 days old.
not satisfactory unless the decrease in virus utre, plotted
3-2 Bacteria and fungi logarithmically, is linear and extrapolation indicates that there
The vaccine, including where applicable the diluent supplied is less than 1 infectious virus unit per 104 litres of liquid
for reconstitution, complies with the test for sterility preparation at the end of inactivation.
prescribed in the monograph Vaccines for veterinalY use
(0062). 2-4 CHOICE OF VACCINE COMPOSITION
The vaccine is shown to be satisfactory widl respect to safety
3-3 Mycoplasmas (2.6.7)
(5.2.6) and efficacy (5.2.7) for each species for which it is
The vaccine complies with the test for mycoplasmas.
intended.
3-4 Extraneous agents
The following tests for safety (section 2-4-1) and
Neutralise the vaccine virus with a suitable monospecific
irnmunogenicity (section 2-4-2) may be used during the
antiserum against distemper virus and inoculate into
demonstration of safety and efficacy.
susceptible cell cultures. The vaccine complies with the test if
no cytopathic effect develops and there is no sign of the 2-4-1 Safety
presence of haemagglutinating 01" haemadsorbing agents. Can)' out dle test for each route and medlod of
3-5 Virus titre administration to be recommended for vaccination and in
Titrate the vaccine virus in suitable cell cultures 01" fertilised each categOl)' of each species for which dle vaccine is
hens I eggs 9-11 days old. The vaccine complies with the test intended, using in each case animals of the minimum age to
if one dose contains not less than the minimum virus titre be recommended. Use a representative batch of vaccine
stated on dle label. containing not less than the maximum antigen content that
may be expected in a batch of vaccine.
3-6 Potency
The vaccine complies with the requirements of the test For each test, use not fewer than 8 animals that do not have
prescribed under Irnmunogenicity (section 2-3-3) when antibodies against foot-and-mouth disease virus. Administer
administered by a recommended route and method. It is not to each animal 1 dose of the vaccine. If the schedule to be
necessary to carry out the potency test for each batch of the recommended requires a 2nd dose, administer 1 dose after an
vaccine if it has been carried out on a representative batch interval of at least 14 days. Observe the animals at least daily
using a vaccinating dose containing not more than the for at least 14 days after the last administration.
minimum virus titre stated on the label. The vaccine complies with the test if no animal shows
_____________________________________________ PhEw abnormallocal or systemic reactions, 01" dies from causes
attributable to the vaccine.
2-4-2 Irnmunogenicity
Carry out an irnmunogenicity test for each strain of foot-and-
Foot and Mouth Disease ***
*** *** mouth disease virus that may be included in dle vaccine.
(Ruminants) Vaccine *** Each test is carried out for each route and method of
(Foot-and-Mouth Disease (Ruminants) Vaccine administration to be recommended for vaccination, using in
(Inactivated) J Ph Bur monogmph 0063) each case cattle not less than 6 months old. The vaccine
PhEw _____________________________________________ administered to each catde is of minimum antigen contento
Either of the following 2 tests is suitable to demonstrate
1 DEFINITION immun,ogenicity of the vaccine for cattle.
Foot-and-mouth disease (ruminants) vaccine (inactivated) is 2-4-2-1 PDso challenge test. The potency of the vaccine is
a preparation containing one or more suitable strains of foot-
expressed as the number of 50 per cent catde protective
and-mouth disease virus inactivated while maintaining
doses (PD so ) contained in the dose stated on the label.
adequate irnmunogenic properties. This monograph applies
The PD so is determined in cattle given primal)' vaccination
to vaccines intended for active irnmunisation of ruminants and challenged by the inoculation of 10 000 IDso of virulent
against foot-and-mouth disease.
Vet-262 Veterinary Vaccines 2016

bovine virus of the same strain as that used in the The test is not valid if both control cattle do not show lesions
preparation of the vaccine in the conditions described below. on at least 3 feet. From the number of protected cattle in the
The vaccine virus may be used for challenge. vaccinated group, calculate the percentage of protected cattle.
Use for the test not fewer than 17 cattle obtained from areas The vaccine complies with the test if the potency is not les s
free from foot-and-mouth disease, that have never been than that to be stated on the label; the minimum potency to
vaccinated against foot-and-mouth disease and do not have be stated on the label is not less than 75 per cent.
antibodies neutralising the different strains of foot-and-mouth 2-5 MANUFACTURER'S TESTS
disease virus. Vaccinate not fewer than 3 groups of not fewer 2-5-1 Identification
than 5 cattle per group, using a different dose of the vaccine The bulk inactivated antigen is identified by a suitable
for each group. Administer the different doses by injecting irnmunochemical method (2.7.1).
different volumes of the vaccine and not by dilution of the
2-5-2 Residual1ive virus
vaccine. Maintain 2 cattle as controls. For example, if the
The limit of detection of the cell cultures to be used with
label states that the injection of 2 mL corresponds to the
respect to the virus to be tested is established by detetmining
administration of 1 dose of vaccine, a 1/4 dose of vaccine
would be obtained by injecting 0.5 mL, and a 1/10 dose the number of CCID so and the 146S antigen content of a
sample of live virus. The cells are not suitable if an amount
would be obtained by injecting 0.2 mL. Challenge all the
cattle after 20-22 days by the intradetmal route, into at least of virus corresponding to 1 ~lg of 146S antigen has less than
10 6 CCID so . A proportion of each batch of bulk inactivated
2 sites on the upper surface of the tongue (0.1 mL per site),
antigen representing at least 200 doses is tested for freedom
with adose equivalent to approximately 10 000 IDso of a
from live virus by inoculation into suitable cell cultures.
suspension of a fully virulent virus, obtained from cattle and
A passage is made during culture of the cells. For mis
of the same strain as that used in the preparation of the
purpose, the sample of the inactivated antigen may be
vaccine. Observe the cattle at least daily for 8 days. In the
interest of animal welfare, individual animals may be concentrated to allow testing of such large samples in cell
cultures. It must be shown that the selected concentration
euthanised before the end of the observation period and
and assay systems are not detrimental to detection of
considered as unprotected if a vaccinated animal shows
lesions of foot-and-mouth disease on at least 1 foot or if a infectious virus within the test sample and that tl1e
control animal shows lesions of foot-and-mouth disease on at concentrated inactivated antigen does not interfere with virus
replication or cause toxic changes. A positive control is
least 3 feet. Unprotected cattle show lesions at sites other
than the tongue. Protected cattle may display lingual lesions. inc1uded in each test.
2-5-3 Antigen content
The test is not valid if both control cattle do not show lesions
The 146S antigen content of each batch of bulk inactivated
on at least 3 feet. From the number of protected cattle in
antigen is determined by an in vitra method (for example, by
each group, calculate the PD so content of the vaccine.
sucrose density gradient centrifugation and ultraviolet
The vaccine complies with the test if the potency is not les s spectrophotometry at 259 mn).
than that to be stated on the label; the minimum potency to
2-5-4 Batch potency test
be stated on the label is not less than 3 PD so per dos e for
It is not necessary to carry out the potency test (section 3-3)
cattle.
for each batch of vaccine if it has been carried out using a
2-4-2-2 PPG test. The following test could also be used to batch of vaccine with a minimum antigen contento Where the
demonstrate immunogenicity of the vaccine for cattle test is not carried out, an alternative validated method is
(referred to as the 'Percentage of protection against used, tl1e criteria for acceptance being set with reference to a
generalised foot infection' (PPG test). batch of vaccine that has given satisfactory results in the test
The potency of the vaccine is expressed as the percentage of described under Potency and has been shown to be
cattle that do not show lesions on any feet. The PPG is satisfactory with respect to immunogenicity in the target
determined in cattle given primary vaccination and species.
challenged by the inoculation of 10 000 IDso of virulent virus The following test may be used after a satisfactory pass level
of the same strain as that used in the preparation of the for a given su"ain has been established. Once a pass level has
vaccine under the conditions described below. The vaccine been established for a given strain, the same level of antigen
virus may be used for challenge. Use for the test not fewer may be used when this strain is formulated in combination
than 18 cattle obtained from areas free from foot-and-mouth with any other antigen provided that the formulation of the
disease, that have never been vaccinated against foot-and- vaccine differs only in the strains inc1uded.
moutl1 disease and do not have antibodies neutralising the
2-5-4-1 Vaccines for use in cattle. Use cattle of the minimum
different strains of foot-and-mouth disease virus. Vaccinate
age recommended for vaccination obtained from areas free
not fewer than 16 cattle with 1 full dose. Maintain 2 cattle as
from foot-and-mouth disease, that have never been
controls. Challenge all the cattle after 20-22 days by the
vaccinated against foot-and-mouth disease and do not have
intradermal route, into at least 2 sites on the upper surface of
antibodies neutralising the different strains of foot-and-mouth
the tongue (0.1 mL per site), with adose equivalent to
disease virus. Vaccinate not fewer than 5 cattle by a
approximately 10 000 IDso of a suspension of a fully virulent
recommended route. Use a suitable dose of the vaccine for
virus of the same strain as that used in the preparation of the
each animal. After a defined period, not greater than 28 days
vaccine. Observe the cattle at least daily for 8 days. In the
following vaccination, draw a blood sample and detetmine
interest of animal welfare, individual animal s may be
individually in each serum the level of antibodies against each
euthanised before the end of the observation period and
strain used in the preparabon of the vaccine by a validated
considered as unprotected if a vaccinated animal shows
technique (e.g. sero-neutralisation test, ELISA). The vaccine
lesions of foot-and-mouth disease on at least 1 foot or if a
complies with the test if the geometric-mean of the antibody
control animal shows lesions of foot-and-mouth disease on at
titre in cattle is not significantly lower than the pass level.
least 3 feet. Unprotected cattle show lesions at sites other
than the tongue. Protected cattle may display linguallesions. 2-5-4-2 Vaccines for use in other ruminants. The potency of
each batch shall be demonstrated in a suitable, validated test.
2016 Veterinary Vaccines Vet-263

EMERGENCY USE: in situations of extreme urgency and age to be recommended for vaccination. For each test
subject to agreement by the competent authority, a batch of perfonned in birds older than 3 weeks of age, use not fewer
vaccine may be released before completion of the tests and than 8 birds not older than the minimum age to be
the detennination of potency if a test for sterility has been recommended for vaccination. In the case of chickens, use
canied out on the bulk inactivated antigen and all other chickens from a fiod: free from specified pathogens (SPF)
components of the vaccine and if the test for safety and the (5.2.2) and in the case of turkeys, ducks or geese, use birds
determination of potency have been canied out on a that have not been vaccinated and that do not have
representative batch of vaccine prepared from the same bulk antibodies against P. 11Zultocida. Administer by a route and
inactivated antigen. In this context, a batch is not considered method to be recommended to each bird 1 dos e of vaccine.
to be representative unless it has been prepared with not If the schedule to be recommended requires a 2nd dose,
more than the amount of antigen or antigens and with the administer 1 dose to each bird after an interval of at least
same formulation as the batch to be released. 14 days. Observe the birds at least daily for at least 14 days
for the last administration of the vaccine.
3 BATCH TESTS
3-1 Identification The test is not valid if more than 10 per cent of the birds
The serum of an animal that did not have antibodies against younger than 3 weeks of age show abnormal signs of disease
foot-and-mouth disease virus prior to being immunised with or die from causes not attributable to the vaccine. For birds
the vaccine neutralises the st:rains of the virus used to prepare older than 3 weeks of age, the test is not valid if non-specific
the vaccine, when tested by a suitably sensitive method. mortality occurs.
3-2 Bacteria and fungi The vaccine complies with dle test if no bird shows abnonnal
The vaccine and, where applicable, the liquid supplied with signs of disease or dies from causes atttibutable to the
it, comply with the test for sterility presClibed in the vaccme.
monograph Vaccines for veterinary use (0062). 2;.2-2 Immunogenicity
3-3 Potency The test is carried out for each route and method of
The vaccine complies with the requirements of the test administration to be recommended for vaccination, for each
mentioned under Immunogenicity (section 2-4-2) when avian species for which the vaccine is intended and for each
administered by a recommended route and method. serovar of P. multocida against which protection is claimed.
_____________________________________________ PhEw
Use for each test not fewer than 30 birds not older than the
minimum age to be recommended for vaccination. U se birds
that have not been vaccinated and that are free from
antibodies against P. multocida. For each test, administer to
each of not fewer than 20 birds a quantity of the vaccine not
Fowl Cholera Vaccine (Inactivated) ***** greater than 1 dose. If re-vaccination is recommended, repeat
** ** this operation after the recommended interval. Maintain not
(Ph. Eur. 71Zo71ograph 1945) *** fewer than 10 birds as controls. Challenge each of the birds
~Ew _____________________________________________ of both groups 21 days after dle last administration by dle
intramuscular route with a sufficient quantity of virulent
1 DEFINITION
P. multocida. Observe the birds for 14 days after challenge.
Fowl cholera vaccine (inactivated) is a preparation of one or
more suitable strains of one or more serovars of Pasteurella The test is not valid if during the observation period after
71Iultocida, inactivated while maintaining adequate challenge, fewer than 70 per cent of dle control birds die or
immunogenic properties. This monograph applies to vaccines show signs of infection (such as either clinical signs or
intended for the active immunisation of chickens, turkeys, bacterial re-isolation in organs) or if dUling the period before
ducks and geese against acute fowl cholera. challenge, more than 10 per cent of the birds from the
conu'ol group or from the vaccinated group show abnormal
2 PRODUCTION signs of disease or die from causes not atttibutable to the
2-1 PREPARATION OF THE VACCINE vaccine.
The seed matelial is cultured in a suitable medium. If the The vaccine complies widl the test if, at the end of the
vaccine contains more than one strain of bactelium, the observation period after challenge, not fewer than 70 per cent
different strains are grown and harvested separately. of the birds from the vaccinated group survive and show no
The bactelial harvests are inactivated. The vaccine may be signs of disease. Mild signs that do not persist beyond dle
adjuvanted. observation period may be tolerated.
2-2 CHOICE OF VACCINE COMPOSITION 2-3 MANUFACTURER'S TESTS
The vaccine is shown to be satisfactory with respect to safety 2-3-1 Batch potency test
(5.2.6) and efficacy (5.2.7) for the species for which it is It is not necessary to carry out the potency test (section 3-3)
intended. for each batch of vaccine if it has been carried out using a
The following tests for safety (section 2-2-1) and batch of vaccine with minimum potency. Where the test is
immunogenicity (section 2-2-2) may be used during the not carried out, an alternative validated method is used, the
demonstration of safety and efficacy. criteria for acceptance being set with reference to a batch of
2-2-1 Safety vaccine that has given satisfactory results in the test described
The test is carried out for each route of administration to be under Potency. The following test may be used.
recommended for vaccination and for each avian species for Use no! fewer than 15 SPF chickens (5.2.2), 3-4 weeks old.
which the vaccine is intended. Use a batch of vaccine Collect serum samples from _each vaccinated and control
containing not les s than the maximum potency that may be chicken just before vaccination and check for the absence of
expected in a batch of vaccine. antibodies against each serovar of P. l1lultocida in the vaccine.
For each test perfonned in birds younger than 3 weeks of Administer to each of 10 chickens 1 dose of the vaccine by
age, use not fewer than 10 birds not older than the minimum the subcutaneous route. Maintain 5 chickens as controls.
Vet-264 Veterinary Vaccines 2016

Collect serum samples 5 weeks after vaccination from each 2-2-2 Cell cultures
vaccinated and control chicken. Measure the titres of serum If the vaccine virus is grown in cen cultures, they comply
antibodies against each serovar of P. 11Zultocida stated on the with the requirements for cen cultures for production of
label using a suitable validated serological method. Calculate veterinary vaccines (5.2.4).
the mean titres for the group of vaccinates.
2-3 SEED LOTS
The test is not valid if specific P. 11Zultocida antibodies are 2-3-1 Extraneous agents
found: before vaccination in 1 or more sera from chickens to The master seed lot complies with the tests for extraneous
be vaccinated or from controls; in 1 or more sera from agents in seed lots (2.6.24). In these tests on the master seed
control chickens 5 weeks after the time of administration of lot, the organisms used are not more than 5 passages from
the vaccine. the master seed lot at the start of the tests.
The vaccine complies with the test if the mean antibody
titres of the group of vaccinates are equal to or greater than 2-4 CHOICE OF VACCINE VIRUS
the titres obtained with a batch that has given satisfactory The vaccine virus shall be shown to be satisfactory with
results in the test described under Potency. respect to safety (5.2.6) and efficacy (5.2.7) for the chickens
for which it is intended.
2-3-2 Bacteria! endotoxins
A test for bacterial endotoxins (2.6.14) is carried out on the The following tests for safety (section 2-4-1), increase in
final lot or, where the nature of the adjuvant prevents virulence (section 2":4-2) and irnmunogenicity (section 2-4-3)
performance of a satisfactory test, on the bulk antigen or the may be used during demonstration of safety and efficacy.
mixture of bulk antigens irnmediately before addition of the 2-4-1 Safety
adjuvant. The maximum acceptable amount of bacterial Carry out the test for each route and method of
endotoxins is that found for a batch of vaccine that has been administration to be recommended for vaccination using in
shown satisfactory in safety test (section 2-2-1). The method each case chickens not older than the minimum age to be
chosen for determining the maximum acceptable amount of recommended for vaccination from an SPF fiod: (5.2.2).
bacterial endotoxins is used subsequently for testing each U se vaccine virus at the least attenuated passage level that
batch. will be present in a batch of the vaccine.
3 BATCH TESTS F or each test performed in chickens younger than 3 weeks of
3-1 Identification age, use not fewer than 10 chickens. For each test performed
When injected into SPF chickens (5.2.2), the vaccine in chickens older than 3 weeks of age, use not fewer than
stimulates the production of antibodies against each of the 8 chickens. Administer to each chicken a quantity of the
serovars of P. multocida in the vaccine. vaccine virus equivalent to not les s than 10 times the
3-2 Bacteria and fungi maximum virus titre likely to be contained in 1 dose of the
The vaccine, inc1uding where applicable the diluent supplied vaccine. Observe the chickens at least daily for at least
for reconstitution, complies with the test for sterility 14 days.
prescribed in the monograph Vaccines for veterinalY The test is not valid if more than 10 per cent of the chickens
use (0062). younger than 3 weeks of age show abnormal signs of disease
3-3 Potency or die from causes not attributable to the vaccine.
The vaccine complies with the requirements of the test For chickens older than 3 weeks of age, the test is not valid if
mentioned under Immunogenicity (section 2-2-2) when non-specific mortality occurs.
administered by a recommended route and method. The vaccine virus complies with the test if no chicken shows
abnonnal signs of disease or dies from causes attributable to
LABELLING
the vaccine virus.
The label states:
- the serovar(s) used to prepare the vaccine; 2-4-2 Increase in virulence
- the serovar(s) against which protection is c1aimed. Carry out the test according to general chapter 5.2.6 using
_ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ PhEur chickens not older than the minimum age to be
recommended for vaccination and from an SPF flock (5.2.2).
If the properties of the vaccine virus allow sequential passage
through 5 groups via natural spreading, this method may be
Fowl POX Vaccine, Living used, otherwise, passage as described below is carried out.
Administer to each chicken óf the 1st group by a suitable
(Fo'lul-pox Vaccine (Live)J Ph Bur mOllograph 0649) route a quantity of the vaccine virus that will allow recovery
_ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ __
PhE~
of virus for the passages described below. Prepare 4-7 days
1 DEFINITION after administration a suspension from the induced skin
Fowl-pox vaccine (live) is a preparation of a suitable strain of lesions of each chicken and pool these samples. Administer
avian pox virus. This monograph applies to vaccines intended 0.2 mL of the pooled samples by cutaneous scarification of
for administration to chickens for active irnmunisation. the comb or other unfeathered part of the body, or by
another suitable method to each chicken of the next group.
2 PRODUCTION Carry out this passageoperation- not fewer than 4 times;
2-1 PREPARATION OF THE VACCINE verify the presence of the virus at each passage. If the virus is
The vaccine virus is grown in embryonated hens' eggs or in not found at a passage level, repeat the passage by
cen cultures. administration to a group of 10 chickens.
2-2 SUBSTRATE FOR VIRUS PROPAGATION If the 5th group of chickens shows no evidence of an increase
2-2-1 Embryonated hens' eggs in virulence indicative of reversion during the observation
If the vaccine virus is grown in embryonated hens' eggs, they period, further testing is not required. Otherwise, carry out
are obtained from fiocks free from specified pathogens (SPF) an additional safety test and compare the clínical signs and
(5.2.2). any relevant parameters in a group of at least 10 chickens
2016 Veterinary Vaccines Vet-265

receiving the material used for the 1st passage and another Any diluent supplied for reconstitution of the vaccine
similar group receiving the virus at the final passage level. complies with the test for sterility prescribed in the
The vaccine virus complies with the test if no indication of monograph Vaccines for veterinary use (0062).
increase in virulence of the virus at the final passage level 3-3 Mycoplasmas
compared with the material used for the 1st passage is The vaccine complies with the test for mycoplasmas (2.6.7).
observed. If virus is not recovered after an initial passage in 3-4 Extraneous agents
5 chickens and a subsequent repeat passage in 10 chickens, The vaccine complies with the tests for extraneous agents in
the vaccine virus also complies with the test. batches of finished product (2.6.25).
2-4-3 Immunogenicity 3-5 Virus titre
A test is carried out for each route and method of Titrate the vaccine virus by inoculation into embryonated
administration to be recommended using in each case hens' eggs from an SPF fiock (5.2.2) or into suitable cell
chickens not older than the minimum age to be cultures (5.2.4). The vaccine complies with the test if 1 dose
recommended for vaccination. The quantity of the vaccine contains not less than the minimum virus titre stated on the
virus administered to each chicken is not greater than the label.
minimum virus titre to be stated on the label and the virus is
at the most attenuated passage level that will be present in a 3-6 Potency
batch of the vaccine. Use for the test not fewer than The vaccine complies with the requirements of the test
30 chickens of the same origin and from an SPF fiock prescribed under Irnmunogenicity (section 2-4-3) when
(5.2.2). Vaccinate by a route to be recommended not fewer administered according to the recommended schedule by a
than 20 chickens. Maintain not fewer than 10 chickens as recommended route and method. It is not necessary to carry
controls. Challenge each chicken after 21 days by the feather- out the potency test for each batch of the vaccine if it has
follicle route with a sufficient quantity of virulent fowl-pox been carried out on a representative batch using a vaccinating
virus. Observe the chickens at least daily for 21 days after dos e containing not more than the minimum virus titre
challenge. Record the deaths and the number of surviving stated on the label.
_____________________________________________
chickens that show clinical signs of disease. Examine each PhE~

surviving chicken for macroscopic lesions: cutaneous lesions


of the comb, wattle and other unfeathered areas of the skin
and diphthericallesions of the mucous membranes of the
oro-pharyngeal area.
Furunculosis Vaccine for ***
The test is not valid if: *** ***
during the observation period after challenge fewer than Salmonids, Inactivated ***
90 per cent of the control chickens die or show severe (Furunculosis Vaccine (Illactivated) Oil-Adjuvanted)
clinical signs of fowl pox, including notable macroscopical Injectable) fo}' Salmollids) Ph Bu/' mOllograph 1521)
lesions of the skin or mucous membranes of the ~E~ ____________________________________________
oro-pharyngeal are a,
- and/or during the period between vaccination and 1 DEFINITION
challenge, more than 10 per cent of the control or Furunculosis vaccine (inactivated, oil-adjuvanted, injectable)
vaccinated chickens show abnormal clinical signs or die for salmonids is prepared from cultures of one or more
from causes not attributable to the vaccine. suitable strains of Aeromonas sabnOllicida subsp. salmollicida,
The vaccine virus complies with the test if during the inactivated while maintaining adequate immunogenic
observation period after challenge not less than 90 per cent properties. This monograph applies to vaccines intended for
of the vaccinated chickens survive and show no notable the active immunisation of salmonids against furunculosis.
clinical signs of disease, including macroscopicallesions of 2 PRODUCTION
the skin and mucous membranes of the oro-pharyngeal area. 2-1 PREPARATION OF THE VACCINE
3 BATCH TESTS The strains of A. salmonicida are cultured and harvested
3-1 Identification separately. The harvests are inactivated by a suitable method.
Carry out an irnmunostaining test in cell cultures to They may be purified and concentrated. Whole or disrupted
demonstrate the presence of the vaccine virus. For egg cells may be used and the vaccine may contain extracellular
adapted strains, inoculate the vaccine into eggs and notice products of the bacterium released into the growth medium.
the characteristic lesions. The vaccine contains an oily adjuvant.
3-2 Bacteria and fungi 2-2 CHOICE OF VACCINE STRAIN
Vaccines intended for administration by injection, The strains included in the vaccine are shown to be suitable
scarification or wing web piercing comply with the test for with respect to the production of antigens of assumed
sterility prescribed in the monograph Vaccines for veterina1Y protective importance. The vaccine is shown to be
use (0062). satisfactory with respect to safety (5.2.6) and efficacy (5.2.7)
Frozen or freeze-dried vaccines produced in embryonated in the species of fish for which it is intended.
eggs and not intended for administration by injection, The following tests for safety (section 2-2-1) and
scarification or wing web piercing either comply with the test immunogenicity (section 2-2-2) may be used during the
for sterility prescribed in the monograph Vaccines for demonstration of safety and efficacy.
veterina1Y use (0062) or with the following test: carry out a 2-2-1 Safety
quantitative test for bacterial and fungal contamination; carry 2-2-1-1 Laboratmy test. Carry-out the test in each species of
out identification tests for microorganisms detected in the fish for which the vaccine is intended, using fish of the
vaccine; the vaccine do es not contain pathogenic minimum body mas s to be recommended for vaccination.
microorganisms and contains not more than U se a batch of vaccine containing not less than the
1 non-pathogenic microorganism per dose.
Vet-266 Veterinary Vaccines 2016

maximum potency that may be expected in a batch of e = percentage of mortality in controls.


vaccine. The vaccine complies with the test if the RPS is not less than
Use not fewer than 50 fish fram a population that does not 70 per cent.
have specific antibodies against A. salmonicida subsp.
2-3 MANUFACTURER'S TESTS
salJ1zonicida and has not been vaccinated against or exposed
to furunculosis. The test is carried out in the conditions to be 2-3-1 Batch potency test
recommended for the use of the vaccine with a water The potency test (section 3-3) may be carried out for each
temperature not less than 10 oC. Administer to each fish by batch of vaccine, using fish of one of the species for which
the intraperitoneal raute 1 dose of the vaccine. Observe the the vaccine is intended. Where the test is not carried out, an
fish at least daily for 21 days. altemative validated method based on antibody response may
be used, the criteria for acceptance being set with reference
The test is not valid if more than 6 per cent of the fish die to a batch of vaccine that has given satisfactory results in the
from causes not attributable to the vaccine. The vaccine test described under Potency. The following test may be
complies with the test if no fish shows abnormal local or
used.
systemic reactions 01' dies from causes attributable to the
vaccine. Use not fewer than 35 fish from a population that does not
have specific antibodies against A sabnonicida subsp.
2-2-1-2 Field studies. Safety is also demonstrated in field trials salmonicida and that are within specified limits for body mass.
by administering the intended dose to a sufficient mimber of Carry out the test at a defined temperature not les s than
fish in not fewer than 2 sets of premises. Samples of 30 fish 12 oc. Inject intraperitoneally into each of not fewer than
are taken on 3 occasions (after vaccination, at the middle of 25 fish 1 dose of vaccine, according to the instructions for
the rearing period and at slaughter) and examined for local use. Perform mock vaccination on a control group of not
reactions in the body cavity. Moderate lesions involving fewer than 10 fish. Collect blood samples from vaccinates
localised adhesions between viscera or between viscera and and controls at a defined time not less than 500 degree days
the abdominal wall and slight opaqueness and/or sparse after vaccination. Determine for each sample the level of
pigmentation of the peritoneum are acceptable. Extensive specific antibodies against A sal1110nicida subsp. salmonicida by
lesions including adhesions between greater parts of the
a suitable irnmunochemical method (2.7.1).
abdominal organs, massive pigmentation andlor obvious
thickening and opaqueness of greater areas of the peritoneum The test is not valid if the control group shows antibodies
are unacceptable if they occur in more than 10 per cent of against A. sabnonicida subsp. salmonicida.
the fish in any sample. Such lesions include adhesions that The vaccine complies with the test if the mean level of
give the viscera a 'one-unit' appearance andlorlead to antibodies in the vaccinates is not significantly lower than
manifest laceration of the peritoneum following evisceration. that found for a batch that gave satisfactory results in the test
described under Potency.
2-2-2 Irnmunogenicity
Carry out the test according to a protocol defining limits of 3 BATCH TESTS
body mass for the fish, water source, water flow and 3-1 Identification
temperature limits, and preparation of a standardised When injected into fish that do not have specific antibodies
challenge. A test is carried out for the route and method of against A. salmonicida, the vaccine stimulates the production
administration to be recommended. The vaccine of such antibodies.
administered to each fish is of minimum potency. 3-2 Bacteria and fungi
Use for the test not fewer than 60 fish from a population that The vaccine, including where applicable the diluent supplied
does not have specific antibodies against A sabnonicida subsp. for reconstitution, complies with the test for sterility
salmonicida and has not been vaccinated against 01' exposed prescribed in the monograph Vaccines for veterinary use
to furunculosis. Vaccinate not fewer than 30 fish according to (0062).
the instructions for use. Perform mock vaccination on a 3-3 Potency
control group of not fewer than 30 fish; mark vaccinated and The vaccine complies with the requirements of the test
control fish for identification. Keep all the fish in the same mentioned under Irnmunogenicity (section 2-2-2) when
tank 01' mix equal numbers of conu'ols and vaccinates in each administered by the recommended route and method.
tank if more than one tank is used. Where justified and when
fish cannot be marked, non-marked fish may be used. 4 LABELLING
Vaccinates and controls may then be kept in the same tank The label states information on the time needed for
but physically separated (for example by fishing nets). development of irnmunity after vaccination under the range
Challenge each fish, by injection, at a fixed interval after of conditions corresponding to the recommended use.
vaccination corresponding to the onset of irnmunity claimed, _____________________________________________ ~E~

with a sufficient quantity of a culture of A salmonicida subsp.


salmonicida whose virulence has been verified. Observe the
fish at least daily until the end of mortality is reached in the
control group (no fish have died over a period of 2 days).
The test is not valid if the specific mortality is less than
60 per cent in the control group 21 days after the 1st death
in the fish. Calculate the relative percentage survival (RPS)
using the following expression:

V percentage of mortality in vaccinates;


2016 Veterinary Vaccines Vet-267

Infectious Avian Encephalomyelitis ***** Administer to each chicken of the 1st group by a route and
** ** method to be recommended a quantity of the vaccine virus
Vaccine, Living *** that will allow recovery of virus for the passages described
Epidemic Tremor Vaccine, Living below. 5-7 days later, prepare a suspension from the brain of
(Avian Infectious Encephalomyelitis Vaccine (Live) ,
each chicken and pool these samples. Administer a suitable
Ph Eu/" monograph 0588)
volume of the pooled samples by the oral route to each
PhE~ _____________________________________________
chicken of the next group. Carry out this passage operation
not fewer than 4 times; verify the presence of the virus at
1 DEFINITION each passage. If the virus is not found at a passage leve!,
Avian infectious encephalomyelitis vaccine (live) is a repeat the passage by administration to a group of 10
preparation of a suitable strain of avian encephalomye!itis chickens.
virus. This monograph applies to vaccines intended for If the 5th group of chickens shows no evidence of an increase
administration to non-laying breeder chickens to protect in virulence indicative of reversion during the observation
passively their future progeny andlor to prevent vertical period, further testing is not required. Othelwise, cany out
transmission of virus via the egg. an additional safety test and compare the c1inical signs and
2 PRODUCTION any relevant parameters in a group of at least 10 chickens
receiving the material used for the 1st passage and another
2-1 PREPARATION OF THE VACCINE
similar group receiving the virus at the final passage leve!.
The vaccine virus isgrown in embryonated hens' eggs or in
cell cultures. The vaccine virus complies with the test if no indication of
an increase in virulence of the virus recovered for the final
2-2 SUBSTRATE FOR VIRUS PROPAGATION passage compared with the material used for the 1st passage
2-2-1 Embryonated hens' eggs is observed. If virus is not recovered after an initial passage in
If the vaccine virus is grown in embryonated hens' eggs, they 5 chickens and a subsequent repeat passage in 10 chickens,
are obtained from fiocks free from specified pathogens (SPF) the vaccine virus also complies with the test.
(5.2.2).
2-4-3 Irnmunogenicity
2-2-2 Cell cultures If the vaccine is recornmended for passive protection of
If the vaccine virus is grown in cell cultures, they comply future progeny cany out test 2-4-3-1. If the vaccine is
with the requirements for cell cultures for production of recommended for prevention of vertical transmission of virus
veterinary vaccines (5.2.4). via the egg, cany out test 2-4-3-2. A test is carried out for
2-3 SEED LOTS each route and method of administration to be
2-3-1 Extraneous agents recommended, using in each case chickens from an SPF
The master seed lot complies with the tests for extraneous fiock (5.2.2) not older dlan the minimum age to be
agents in seed lots (2.6.24). In these tests on the master seed recommended for vaccination. The quantity of the vaccine
lot, the organisms used are not more dlan 5 passages from virus administered to each chicken is not greater than the
the master seed lot at the start of the tests. minimum titre to be stated on dle label and the virus is at
the most attenuated passage level that will be present in a
2-4 CHOICE OF VACCINE VIRUS batch of the vaccine.
The vaccine virus shall be shown to be satisfactory with
2-4-3-1 Passive i11lJ1lunity in chickens. Vaccinate not fewer than
respect to safety (5.2.6) and efficacy (5.2.7) for the chickens
20 breeder chickens from an SPF fiock (5.2.2). Maintain
for which it is intended.
separately not fewer than 10 breeder chickens of the same
The following tests for safety (section 2-4-1), increase in age and origin as controls. At the peak of lay, hatch not
virulence (section 2-4-2) and immunogenicity (section 2-4-3) fewer than 25 chickens from eggs from vaccinated breeder
may be used during the demonstration of safety and efficacy. chickens and 10 chickens from non-vaccinated breeder
2-4-1 Safety chickens. At 2 weeks of age, challenge each chicken by dle
Carry out the test for each route and method of intracerebral route with a sufficient quantity of virulent avian
administration to be recommended for vaccination using in encephalo'myelitis virus. Observe the chickens at least daily
each case non-laying breeder chickens not older than the for 21 days after challenge. Record the deaths and the
minimum age to be recommended for vaccination. number of surviving chickens that show c1inical signs of
Use vaccine virus at the least attenuated passage level that disease.
will be present in a batch of the vaccine. The test is not valid if:
For each test, use not fewer than 8 chickens from an SPF -- during the observation period after challenge fewer than
fiock (5.2.2). Administer to each chicken a quantity of the 80 per cent of the control chickens die or show severe
vaccine virus equivalent to not les s than 10 times the c1inical signs of avian infectious encephalomyelitis,
maximum virus titre likely to be contained in 1 dose of the and/or during the period between the vaccination and
vaccine. Observe the chickens at least daily for 21 days. challenge more than 15 per cent of control or vaccinated
The test is not valid if non-specific mortality occurs. chickens show abnormal c1inical signs or die from causes
not attributable to the vaccine.
The vaccine virus complies with the test if no chicken shows
abnormal signs of disease or dies from causes attributable to The vaccine virus complies widl the test if during the
the vaccine virus. observation period after challenge not fewer than 80 per cent
of the, progeny of vaccinated chickens survive and show no
2-4-2 Increase in virulence
notable c1inical signs of disease.
Carry out the test according to general chapter 5.2.6
using 1-day-old chickens from an SPF fiod: (5.2.2). If the 2-4-3-2 Passive immunity in émbryos. Vaccinate not fewer than
properties of the vaccine virus allow sequential passage 20 breeder chickens from an SPF fiod: (5.2.2). Maintain
through 5 groups via natural spreading, this method may be separately not fewer than 10 breeder chickens of the same
used, otherwise passage as described below is carried out. age and origin as controls. At dle peak of lay, incubate not
Vet-268 Veterinary Vaccines 2016

fewer than 36 eggs from the 2 groups, vaccinated and


Infectious Bovine Rhinotracheitis ***
controls, and carry out an embryo sensitivity test. On the *** ***
sixth day of incubation inoculate 100 E1Dso of the Van Vaccine, Living ***
Roekel strain of avian encephalomyelitis virus into the yolk (Infectious Bovine Rhinotracheitis Vaccine (Live) J
sacs of the eggs. 12 days after inoculation examine the Ph Eu/" 1110nograph 0696)
embryos for specific lesions of avian encephalomyelitis PhE~ _____________________________________________
(muscular atrophy). Deaths during the first 24 h are
considered to be non-specific. 1 DEFINITION
The test is not valid if fewer than 80 per cent of the control Infectious bovine rhinotracheitis vaccine (live) is a
embryos show lesions of avian encephalomyelitis. The test is preparation of one 01' more suitable strains of infectious
not valid if fewer than 80 per cent of the embryos can be bovine rhinotracheitis virus (bovine herpesvirus 1). This
given an assessment. monograph applies to vaccines intended for the active
irnmunisation of cattle against bovine rhinotracheitis caused
The vaccine virus complies with the test if not fewer than
80 per cent of the embryos in the vaccinated group show no by bovine herpesvirus 1.
lesions of avian encephalomyelitis. 2 PRODUCTION
3 BATCH TESTS 2-1 PREPARATION OF THE VACCINE
3-1 Identification The vaccine virus is grown in cell cultures.
The vaccine, diluted if necessary and mixed with a 2-2 SUBSTRATE FOR VIRUS PROPAGATION
monospecific avian encephalomyelitis virus antiserum, no 2-2-1 Cell cultures
longer infects embryonated hens' eggs from an SPF flock The cell cultures comply with the requirements for cell
(5.2.2) 01' susceptible cell cultures (5.2.4) into which it is cultures for production of veterinary vaccines (5.2.4).
inoculated.
2-3 CHOICE OF VACCINE VIRUS
3-2 Bacteria and fungi The vaccine virus is shown to be satisfactory with respect to
Vaccines intended for administration by injection comply safety (5.2.6) and efficacy (5. 2.7) for the catde for which it is
with the test for sterility presctibed in the monograph intended.
Vaccines for veterinaJY use (0062).
The following tests for safety (section 2-3-1), abortigenicity
Frozen 01' freeze-dried vaccines produced in embryonated and passage through the placenta (section 2-3-2), increase in
eggs and not intended for administration by injection either virulence (section 2-3-3) and immunogenicity (section 2-3-4)
comply with the test for stetility prescribed in the monograph may be used during the demonstration of safety and efficacy.
Vaccines for vetel'inaJy use (0062) 01' with the following test:
carry out a quantitative test for bactetial and fungal 2-3-1 Safety
contamination; carry out identification tests for Carry out the test fol' each route and method of
microorganisms detected in the vaccine; dle vaccine does not administration to be recommended for vaccination.
contain pathogenic microorganisms and contains not more U se vaccine virus at the least attenuated passage level that
than 1 non-pathogenic microorganism per dose. will be present in a batch of the vaccine.
Any diluent supplied for reconstitution of the vaccine For each test, use not fewer dlan S calves, 3 mondls old 01'
complies with the test for sterility presctibed in the of the minimum age to be recommended fol' vaccination if
monograph Vaccines for veterinaJY use (0062). this is less than 3 months, and that do not have antibodies
against infectious bovine l'hinotracheitis virus. Administer to
3-3 Mycoplasmas each calf a quantity of the vaccine virus equivalent to not less
The vaccine complies with the test for mycoplasmas (2.6.7). than 10 times the maximum virus titre likely to be contained
3-4 Extraneous agents in 1 dose of the vaccine. Observe the calves at least daily fol'
The vaccine complies with the tests for extraneous agents in at least 14 days.
batches of finished product (2.6.25). The vaccine virus complies with the test if no calf shows
3-5 Virus titre abnOlmal local or systemic reactions 01' dies from causes
Titrate the vaccine virus by inoculation into embryonated attributable to the vaccine virus.
hens' eggs from an SPF flock (5.2.2) or into suitable cell 2-3-2 Abortigenicity and passage through the placenta
cultures (5.2.4). The vaccine complies with the test if 1 dose Use not fewer than 24 pregnant cows that do not have
contains not less than the minimum virus titre stated on the antibodies against infectious bovine rhinotracheitis virus: 80f
label. the cows are in the 4th month of pregnancy, 8 in dle Sdl
3-6 Potency and 8 in the 6d1 or 7ili month. Administer to each cow by a
Depending on the indications, the vaccine complies with the route to be recommended a quantity of the vaccine virus
requirements of 1 01' both of the tests prescribed under equivalent to not less than 10 times the maxirnum virus titre
Irnmunogenicity (section 2-4-3-1, 2-4-3-2), when likely to be contained in 1 dose of the vaccine. Observe the
administered by a recommended route and method. It is not cows at least daily until the end of pregnancy.
necessary to carry out the potency test for each batch of the The vaccine virus complies with the test if:
vaccine if it has been carried out on a representative batch -- whel'e abortion occurs, tests show that neither virus nor
using a vaccinating dose containing not more than the viral antigens are pl'esent in the foetus or placenta;
minimum virus titre stated on the label. -- on calves born at term before ingestion of colostrum, a
--___________________________________________ PhE~
test for antibodies against infectious bovine rhinotracheitis
virus indicates no such antibodies are found.
2-3-3 Increase in virulence
Can-y out the test according to chapter 5.2.6. Evaluation of
safety of veterinaJY vaccines and i71l111UnOSera using calves
2016 Veterinary Vaccines Vet-269

3 months old or of the minimum age to be recommended for 3 BATCH TESTS


vaccination if this is less than 3 months, and that do not have 3-1 Identification
antibodies against infectious bovine rhinotracheitis virus. 3-1-1
If the properties of the vaccine virus allow sequential passage When mixed with a suitable quantity of a monospecific
through 5 groups via natural spreading, this method may be antiserum, the vaccine is no longer able to infect susceptible
used, otherwise passage as described below is carried out. cell cultures into which it is inoculated.
Take suitable samples from the calves used for the test for
3-1-2
safety at a time when the vaccinal virus can be easily
Any markers of the strain are verified.
detected, verify the presence and titre of the virus in the
samples and mix them. Administer to each calf of the 3-2 Bacteria and fungi
1st group by the intranasal route a quantity of the vaccine The vaccine, inc1uding where applicable the diluent supplied
virus that will allow recovery of virus for the passages for reconstitution, complies with the test for sterility
described below. Administer the virus by the intranasal route prescribed in the monograph Vaccines for veterinary
to each calf of the next group. Carry out this passage use (0062).
operation not fewer than 4 times; verify the presence of the 3-3 Mycoplasmas (2.6.7)
virus at each passage. If the virus is not found at a passage The vaccine complies with the test for mycoplasmas.
leve1, repeat the passage by administration to a group of 3-4 Extraneous agents
10 animals. Neutralise the vaccine virus with a suitable monospecific
If the 5th group of calves shows no evidence of an increase in antiserum against bovine rhinotracheitis virus and inoculate
virulence indicative of reversion during the observation into cell cultures known for their susceptibility to vu'uses
period, further testing is not required. Othelwise, carry out pathogenic for cattle. Carry out 1 passage at 7 days and
an additional safety test and compare the clinical signs and maintain the cultures for 14 days.
any relevant parameters in a group of at least 8 animals The vaccine complies with the test if no cytopathic effect
receiving the material used for the 1st passage and another develops and there is no sign of the presence of
similar group receiving the virus at the final passage level. haemadsorbing agents.
The vaccine virus complies with the test if no indication of 3-5 Virus titre
increased virulence of the virus recovered for the final Titrate the vaccine virus in susceptible cell cultures at a
passage compared with the material used for the 1st passage temperature favourable to replication of the virus.
is observed. If virus is not recovered after an initial passage in The vaccine complies with the test if 1 dose contains not less
2 animal s and a subsequent repeated passage in 10 animal s, than the minimum virus titre stated on the label.
the vaccine virus also complies with the test.
3-6 Potency
2-3-4 Irnmunogenicity The vaccine complies with the requirements of the test
A test is carried out for each route and method of prescribed under Immunogenicity (section 2-3-4) when
administration to be recommended for vaccination using in admu1istered by a recommended route and method. It is not
each case calves 2-3 months old. The quantity of vaccine to necessary to carry out the potency test for each batch of the
be administered to each calf is not greater than the minimum vaccine if it has been carried out on a representative batch
virus titre to be stated on the label and the virus is at the using a vaccinating dose containing not more than the
most attenuated passage level that will be present in a batch minimum virus titre stated on the label.
of vaccine. Use for the test not fewer than 7 calves that do
_____________________________________________ PhE~
not have antibodies against infectious bovine rhinotracheitis
virus. Vaccinate not fewer than 5 calves, according to the
schedule to be recommended. Maintain not fewer than
2 calves as controls. Challenge each calf after 20-22 days by
the intranasal route with a sufficient quantity of a virulent
Infectious Bursal Disease Vaccine, *****
infectious bovine rhinotracheitis virus. Observe the calves at ** **
least daily for 21 days after challenge, in particular for Inactivated ***
respiratory signs and virus shedding (by nasal swabs 01' Gumboro Disease Vaccine, Inactivated
tracheobronchial washing). (Avian bifectious Bursal Disease Vaccine (Inactivated))
The test is not valid if the controls do not show typical signs Ph Eur J11onogmph 0960)
of disease such as fever, ocular and nasal discharge and caution: Accidental Irljeciion of oily vaccine can cause serious local
ulceration of the nasal mucosa. reactions in mano Expert medical advice should be sought
The vaccine virus complies with the test if, during the imnzediately and the doctor should be informed that the vaccine is
observation period after challenge: an oil emulsiono
-- the vaccinated calves show no more than mild signs; PhE~ _____________________________________________
-- in not fewer than 4 of the 5 vaccinated calves, the
maximum virus titre found in the nasal mucus is at least 1 DEFINITION
100 times lower than the average of the ma.,'{imum titres Avian infectious bursal disease vaccine (inactivated) is a
found in the control calves; and preparation of a suitable strain of avian infectious bursal
-- the average number of days on which virus is excreted is disease virus type 1, inactivated while maintaining adequate
at least 3 days less in vaccinated calves than in the control immunogenic properties. This monograph applies to vaccines
calves. intended for use in breeding chickens to protect their
progeny from avian infectioús bursal disease.
Vet-270 Veterinary Vaccines 2016

2 PRODUCTION hen at the time of egg coIlection for hatching. The antibody
2-1 PREPARATION OF THE VACCINE response is measured in a serum-neutralisation test.
The vaccine virus is grown in embryonated hens' eggs or in Eggs are collected for hatching not less than 5 weeks after
cell cultures. vaccination and the test described below is carried out with
The vaccine may be adjuvanted. chickens at least 3 weeks old from that egg collection.
2-2 SUBSTRATE POR VIRUS PROPAGATION 25 chickens fram vaccinated (group B) hens and 10 control
chickens of the same breed and age from unvaccinated
2-2-1 Embryonated hens' eggs
(group A) hens are challenged with an eye-drop application
If the vaccine virus is grown in embryonated hens' eggs, they
of a quantity of a virulent strain of avian infectious bursal
are obtained from healthy fiocks.
disease virus sufficient to produce severe signs of disease,
2-2-2 Cell cultures including lesions of the bursa of Fabricius, in all
If the vaccine is grown in cell cultures, they comply with the unvaccinated chickens. 3-4 days after challenge, the bursa of
requirements for cell cultures for production of veterinary Fabricius is removed from each chicken. The bursae are
vaccines (5.2.4). examined for evidence of infection by histological
2-3 SEED LOTS examination and by testing for the presence of avian
2-3-1 Extraneous agents infectious bursal disease antigen by a suitable method.
The master seed lot complíes with the tests for exttaneous The vaccine complies with the test if 3 or fewer of the
agents in seed lots (2.6.24). In these tests on the master seed chickens from group B hens show evidence of avian
lot, the organisms used are not more than 5 passages from infectious bursal disease. The test is invalid unless all the
the master seed lot at the start of the test. chickens from group A hens show evidence of avian
infectious bursal disease.
2-4 CHOICE OF VACCINE COMPOSITION
Where there is more than one recommended route of
The vaccine is shown to be satisfactory with respect to safety
administration, the test described under Potency is carried
(5.2.6) and efficacy (5.2.7) for the birds for which it is
out in parallel with the aboye irnmunogenicity test, using
intended.
different groups of birds for each recommended raute.
The following tests for safety (section 2-4-1) and The serological response of the birds inoculated by rautes
immunogenicity (section 2-4-2) may be used during the other than thatused in the irnmunogenicity testis not
demonstration of safety and efficacy. significandy less than that of the group vaccinated by that
2-4-1 Safety route.
The test is carried out for each raute of administration to be 2-5 MANUFACTURER'S TESTS
recommended for vaccination. Use a batch of vaccine 2-5-1 Residuallive virus
containing not less than the maximum potency that may be An amplífication test for residuallíve avian infectious bursal
expected in a batch of vaccine. disease virus is carried out on each batch of antigen
For each test, use not fewer than 8 chickens not older than immediately after inactivation to confirm inactivation; the test
the minimum age to be recommended for vaccination and is canied out in embryonated hens' eggs or in suitable cell
fram a fiod: free fram specified pathogens (SPF) (5.2.2). cultures (5.2.4), whichever is the most sensitive for the
Administer by a raute and medlod to be recommended to vaccine strain; the quantity of inactivated virus harvest used
each chicken 1 dose of the vaccine. Observe the chickens at in the test is equivalent to not les s than 10 doses of the
least daily for at least 14 days after the administration of the vaccine. The vaccine complies with the test if no líve virus is
vaccine. detected.
The test is not valíd if non-specific mortality occurs. 2-5-2 Batch potency test
The vaccine complíes with the test if no chicken shows It is not necessary to carry out the potency test (section 3-5)
abnormal signs of disease ar dies from causes attributable to for each batch of the vaccine if it has been carried out using
the vaccine. a batch of vaccine with a minimum potency. Where the test
2-4-2 Irnmunogenicity is not carried out, an alternative validated method is used,
A test is carried out for each route and method of the criteria for acceptance being set with reference to a batch
administration to be recommended using in each case of vaccine that has given satisfactory results in the test
chickens from an SPF fiod: (5.2.2) and not older than the described under Potency. The foIlowing test may be used.
mínimum age to be recommended for vaccination (close to Vaccinate each of not fewer than 10 chickens, 14-28 days old
the point of lay). The dos e of vaccine administered to each and from an SPF fiod: (5.2.2), with 1 dose of vaccine by a
chicken contains not more than the minimum potency to be recommended route. 4-6 weeks later, collect serum samples
stated on the label. from each bird and 10 unvaccinated control birds of the
Where a challenge test is to be carried out, the foIlowing test same age and from the same source. Measure the antibody
may be used. Use 2 groups of not less than 20 hens treated response in a serum-neutralisation test.
as foIlows: The test is not valid if there are specific antibodies in the sera
- group A: unvaccinated controls; of the unvaccinated birds. The vaccine complies with the test
- group B: vaccinated with inactivated avian infectious if the mean antiboQY titre in fue sera from the vaccinated
bursal disease vaccine. birds is equal to or greater than the titres obtained with a
Serum samples are coIlected fram each unvaccinated control batch that has given satisfactory results in the test described
(group A) hen just befare administration of dle vaccine, under Potency.
4-6 weeks later, and at the time of egg coIlection for 3 BATCH TESTS
hatching. If a serological test is to be carried out for 3-1 Identification
demonstration of immunogenicity by other rautes, serum When injected into chickens that do not have antibodies
samples are also coIlected from each vaccinated (group B) against avian infectious bursal disease virus type 1, the
vaccine stimulates the production ofsuch antibodies.
2016 Veterinary Vaccines Vet-271

3-2 Bacteria and fungi


Infectious Bursal Disease Vaccine, *****
The vaccine, including where applicable the diluent supplied ** **
for reconstitution, complies with the test for sterility Living ***
prescribed in the monograph Vaccines for veterinalY Gumboro Disease Vaccine, Living
use (0062).
(Avian Infectious Bursal Disease Vaccine (Live)~
3-3 Residuallive virus Ph Bur J11onograph 0587)
A test for residual live virus is carried out to confirm PhE~ _____________________________________________
inactivation of avian infectious bursal disease type 1.
A. For vaccine prepared with embryo-adapted strains of 1 DEFINITION
virus, inject 2/5 of adose into the allantoic cavity or onto the Avian infectious bursal disease vaccine (live) [Gumboro
chorio-allantoic membrane of ten 9- to 11-day-old disease vaccine (live)] is a preparation of a suitable strain of
embryonated hen eggs from an SPF flod: (5.2.2). Incubate infectious bursal disease virus type 1. This monograph
the eggs and observe at least daily for 6 days. Pool separately applies to vaccines intended for administration to chickens
the allantoic liquid or membranes from eggs containing live for active immunisation; it applies to vaccines containing
embryos, and that from eggs containing dead embryos, strains of low virulence but not to those containing strains of
excluding those that die from non-specific causes within 24 h higher virulence that may be needed for disease control in
of the injection. certain epiderniological situations.
Inject into the aIlantoic cavity 01' onto the chorio-allantoic 2 PRODUCTION
membrane of each of ten 9- to 11-day-old SPF eggs 0.2 mL 2-1 PREPARATION OF THE VACCINE
of the po oled allantoic liquid or crushed chorio-allantoic The vaccine virus is grown in embryonated hens' eggs or in
membranes from the live embryos and, into each of cell cultures.
10 similar eggs, 0.2 mL of the pooled liquid or membranes
2-2 SUBSTRATE FOR VIRUS PROPAGATION
from the dead embryos and incubate for 6 days. Examine
2-2-1 Embryonated hens' eggs
each embryo for lesions of avian infectious bursal disease.
If the vaccine virus is grown in embryonated hens' eggs, they
If more than 20 per cent of the embryos die at either stage
are obtained from flocks free from specified pathogens (SPF)
repeat that stage.
(5.2.2).
The vaccine complies with the test if there is no evidence of
2-2-2 Cell cultures
lesions of avian infectious bursal disease and if, in any repeat
If the vaccine virus is grown in cell cultures, they comply
test, not more than 20 per cent of the embryos die from non-
with tlle requirements for cell cultures for production of
specific causes.
veterinary vaccines (5.2.4).
Antibiotics may be used in the test to control extraneous
bacterial infection. 2-3 SEED LOTS
E. For vaccine prepared with cell-culture-adapted strains of 2-3-1 Extraneous agents
virus, inoculate 10 doses of the vaccine into suitable cell The master seed lot complies with the tests for extraneous
cultures. If the vaccine contains an oil adjuvant, eliminate it agents in seed lots (2.6.24). In these tests on the master seed
by suitable means. Incubate at 38 ± 1 oC for 7 days. Make lot, the organisms used are not more than 5 passages from
a passage on another set of cell cultures and incubate at the master seed lot at the start of the tests.
38 ± 1 oC for 7 days. 2-4 CHOICE OF VACCINE vmus
The vaccine complies with the test if the cultures show no The vaccine virus shall be shown to be satisfactory with
signs of infection. respect to safety (5.2.6) and efficacy (5.2.7) for the chickens
3-4 Specified extraneous agents for which it is intended.
Use 10 chickens, 14-28 days old, from an SPF flock (5.2.2). The following tests for safety (section 2-4-1), damage to the
Vaccinate each chicken by a recommended route with a bursa of Fabricius (section 2-4-2), immunosuppression
double dose of the vaccine. After 3 weeks, administer 1 dos e (section 2-4-3), increase in virulence (section 2-4-4) and
by the same route. Collect serum samples from each chicken immunogenicity (section 2-4-5) may be used during the
2 weeks later and carry out tests for antibodies against the demonstration of safety and efficacy.
following agents by the methods prescribed in general 2-4-1 Safety
chapter 5.2.2. Chicken fiocks free fro112 specified pathogens for the Carry out the test for each route and method of
production and quality control of vaccines: avian administration to be recommended for vaccination using in
encephalomyelitis virus, avian leucosis virus es, egg-drop each case chickens not older than the minimum age to be
syndrome virus, avian infectious bronchitis virus, avian recommended for vaccination and from an SPF flod: (5.2.2).
infectious laryngotracheitis virus, influenza A virus, Marek's U se vaccine virus at the least attenuated passage level that
disease virus, Newcastle disease virus. will be present in a batch of the vaccine.
The vaccine complies with the test if it do es not stimulate the For each test performed in chickens younger than 3 weeks of
formation of antibodies against these agents. age, use not fewer than 10 chickens. For each test performed
3-5 Potency in chickens older than 3 weeks of age, use not fewer than
The vaccine complies with the requirements of the test 8 chickens. Administer to each chicken a quantity of the
mentioned under Immunogenicity (section 2-4-2) when vaccine virus' equivalent to not less than 10 times the
administered by a recommended route and method. maxim·um virus titre likely to be contained in 1 dose of the
vaccine. Observe the chickens at least daily for at least
4 LABELLING
14 days.
The label states whether the strain in the vaccine is embryo-
The test is not valid if more than 10 per cent of the chickens
adapted 01' cell-culture-adapted.
younger than 3 weeks of age show abnormal signs of disease
_____________________________________________ PhE~

01' die from causes not attributable to the vaccine.


Vet-272 Veterinary Vaccines 2016

For chickens older than 3 weeks of age, the test in not valid results obtained in the test for damage to the bursa of
if non-specific mortality occurs. Fabricius (section 2-4-2), administer to each vaccinated
The vaccine virus complies with the test if no chicken shows chicken and to each chicken of another group 1 dose of
abnormal signs of disease or dies from causes attributable to Hitchner B1 strain Newcastle disease vaccine (live).
the vaccine virus. Determine the seroresponse of each chicken of the 2 groups
to the Newcastle disease virus 14 days after administration.
2-4-2 Damage to the bursa of Fabricius
Challenge each chicleen of the 3 groups by the intramuscular
Cany out the test for the route to be recommended for
route with not 1ess than lOs EIDso of virulent Newcastle
vaccination likely to be the least safe using chickens not older
disease virus and note the degree of protection in the
than the minimum age to be recommended for vaccination.
2 groups vaccinated with Hitchner B1 strain Newcastle
Use virus at the least attenuated passage level that will be
vaccine compared with the non-vaccinated group.
present between the master seed lot and a batch of the
vaccine. Use not fewer than 20 chickens from an SPF flock The test is not va1id if 1 or more of the non-vaccinated
(5.2.2). Administer to each chicken a quantity of the vaccine chickens does not die within 7 days of challenge. The degree
virus equivalent to 10 times the maximum titre likely to be of irnmunosuppression is estimated from the comparative
contained in adose of the vaccine. On each of days 7, 14, 21 seroresponses and protection rates of the 2 Hitchner B 1
and 28 after administration of the vaccine virus, euthanise vaccinated groups.
not fewer than 5 chickens and prepare a section from the site The vaccine comp1ies with the test if there is no significant
with the greatest diameters of the bursa of Fabricius of each difference between the 2 groups.
chicken. Carry out histological examination of the section 2-4-4 Increase in virulence
and score the degree of bursal damage using the following Carry out the test according to general chapter 5.2.6
scale. using chickens from an SPF flock (5.2.2) and not older than
O No lesion, normal bursa. the minimum age to be recornmended for vaccination. If the
1 1 per cent to 25 per cent of the follicles show lymphoid properties of the vaccine virus allow sequential passage
depletion (i.e., less than 50 per cent depletion in 1 through 5 groups via natural spreading, this method may be
affected follicle) influx of heterophils in lesions. used, otherwise passage as described be10w is carried out.
2 26 per cent to 50 per cent of the follicles show nearly Administer to each chicleen of the 1st group by eye-drop a
complete lymphoid depletion (i.e., more than 75 per cent
quantity of the vaccine virus that will allow recovery of virus
dep1etion in 1 affected follicle), affected follicles show
for the passages described below. Prepare 3 to 4 days after
necrosis and severe influx of heterophils may be detected. administration a suspension from the bursa of Fabricius of
3 51 per cent to 75 per cent of the follicles show 1ymphoid
each chicleen and pool these samp1es. Administer 0.05 mL of
dep1etion; affected follicles show necrosis and severe the pooled samp1es by eye-drop to each chicleen of the next
influx of heterophils is detected.
group. Carry out this passage operation not fewer than
4 76 per cent to 100 per cent of the follieles show near1y
4 times; verify the presence of the virus at each passage.
complete 1ymphoid depletion, hyperp1asia and cyst
If the virus is not found at a passage 1eve1, repeat the passage
structures are detected; affected follicles show necrosis
by administration to a group of 10 chickens.
and severe influx of heterophi1s is detected.
5 100 per cent of the follicles show near1y complete Carry out the test for damage to the bursa of Fabricius
lymphoid dep1etion; complete 10ss of follicular structure (section 2-4-2) using the material used for the 1st passage
thickened and fo1ded epithelium, fibrosis of bursal tissu~. and the virus at the final passage. Administer the virus by the
route to be recommended for vaccination 1ike1y to be the
least safe.
Calculate the average score for each group of chickens.
The vaccine virus complies with the test if no indication of
The vaccine virus complies with the test if:
increasing virulence of the virus recovered for the final
- no chicken shows notable clinica1 signs of disease or dies
~assage compared with the material used for the 1st passage
from causes attributable to the vaccine virus,
1S observed. If virus is not recovered after an initia1 passage in
- the average score for bursal damage 21 days after
5 chickens and a subsequent repeat passage in 10 chickens,
administration of the vaccine virus is les s than or equa1
the vaccine virus a1so complies with the test.
to 2.0 and 28 days after administration is les s than or
equa1 to 0.6, 2-4-5 Immunogenicity
- during the 21 days after administration a notable A test is carried out for each route and method of
repopulation of the bursae by 1ymphocytes has taken administration to be recommended using in each case
place. chickens not older than the minimum age to be
recommended for vaccination. The quantity of vaccine virus
2-4-3 Immunosuppression
administered to each chicleen is not greater than the
Carry out the tests for the route to be recommended for
minimum virus titre to be stated on the 1abel and the virus is
vaccination likely to be the 1east safe using chickens not older
at the most attenuated passage leve1 that will be present in a
than the minimum age to be recommended for vaccination.
batch of the váccine. Use not fewer than 30 chickens of the
Use vaccine virus at the 1east attenuated passage leve1 that
same origin and from an SPF flock (5.2.2). Vaccinate by a
will be present between the master seed lot and a batch of
route to be recomrriended not fewer than 20 chickens.
the vaccine. Use not fewer than 30 chickens from an SPF
Maintain not fewer than 10 chickens as contro1s. Challenge
flock (5.2.2). Divide them random1y into 3 groups each of
each chicleen after 14 days by eye-drop with a sufficient
not fewer than 10 and maintain the groups separately.
quantity of viruient avian infectious bursal disease virus.
Administer by eye-drop to each chicken of 1 group a
Observe the chickens at 1east dai1y f01: 10 days after
quantity of the vaccine virus equivalent to not 1ess than the
challenge. Record the deaths due to infectious bursal disease
maximum titre likely to be contained in 1 dos e of the
and the surviving chickens that show clinical signs of disease.
vaccine. At the time after administration when maxima1
At the end of the observation period, euthanise all the
bursa1 damage is 1ike1y to be present, as judged from the
2016 Veterinary Vaccines Vet- 2 7 3

surviving chickens and carry out histological examination for


lesions of the bursa of Fabricius.
Infectious Chicken Anaemia
The test is not valid if one or more of the following applies: Vaccine (Live)
-- during the observation period following challenge, fewer (Ph. Eur. 7110nogmph 2038)
than 50 per cent of the control chickens show PhE~ ____________________________ ~ _______________
characteristic signs of avian infectious bursal disease;
-- 1 or more of the surviving control chickens does not show 1 DEFINITION
degree 3 lesions of the bursa of Fabricius; Infectious chicken anaemia vaccine (live) is a preparation of a
-- during the period between the vaccination and challenge suitable strain of chicken anaemia virus. This monograph
more than 10 per cent of the vaccinated or control applies to vaccines intended for administration to breeder
chickens show abnormal clinical signs or die from causes chickens for active immunisation, to prevent excretion of the
not attributable to the vaccine. virus, to prevent or reduce egg transmission and to protect
The vaccine virus complies with the test if during the passively their future prageny.
observation period after challenge not fewer than 90 per cent 2 PRODUCTION
of the vaccinated chickens survive and show no notable 2-1 PREPARATION OF THE VACCINE
clinical signs of disease nor degree 3 lesions of the bursa of The vaccine virus is grown in embryonated hens' eggs or in
Fabricius. cel1 cultures.
3 BATCH TESTS 2-2 SUBSTRATE FOR VIRUS PROPAGATION
3-1 Identification 2-2-1 Embryonated hens' eggs
The vaccine, diluted if necessary and mixed with a If the vaccine virus is grown in embryonated hens' eggs, they
monospecific infectious bursal disease virus type 1 antiserum, are obtained from flocks free from specified pathogens (SPF)
no longer infects embryonated hens' eggs from an SPF flock (5.2.2).
(5.2.2) or susceptible cell cultures (5.2.4) into which it is
2-2-2 CelI cultures
inoculated.
If the vaccine virus is grown in cell cultures, they comply
3-2 Bacteria and fungi with the requirements for cell cultures for production of
Vaccines intended for administration by injection comply veterinary vaccines (5.2.4).
with the test for sterility prescribed in the monograph
2-3 SEED LOTS
Vaccines for veterinalY use (0062).
2-3-1 Extraneous agents
Frozen or freeze-dried vaccines produced in embryonated
The master seed lot complies with the test for extraneous
eggs and not intended for administration by injection either
agents in seed lots (2.6.24). In these tests on the master seed
comply with the test for sterility prescribed in the monograph
lot, the organisms used are not more than 5 passages fram
Vaccines for veterinalY use (0062) or with the following test:
the master seed lot at the start of the tests.
carry out a quantitative test for bacterial and fungal
contamination; carry out identification tests for 2-4 CHOICE OF VACCINE VIRUS
microorganisms detected in the vaccine; the vaccine does not The vaccine virus is shown to be satisfactory with respect to
contain pathogenic microorganisms and contains not more safety (5.2.6) and efficacy (5.2.7) for the chickens for which
than 1 non-pathogenic microorganism per dose. it is intended.
Any diluent supplied for reconstitution of the vaccine The following tests for safety (section 2-4-1), increase in
complies with the test for sterility prescribed in the virulence (section 2-4-2) and immunogenicity (section 2-4-3)
monograph Vaccines for vetennalY use (0062). may be used during the demonstration of safety and efficacy.
3-3 Mycoplasmas 2-4-1 Safety
The vaccine complies with the test for mycoplasmas (2.6.7). Carry out the test for each route and method of
administration to be recommended for vaccination in
3-4 Extraneous agents
chickens not older than the minimum age to be
The vaccine complies with the tests for extraneous agents in
recommended for vaccination and fram an SPF flock (5.2.2).
batches of finished product (2.6.25).
U se vaccine virus at the least attenuated passage level that
3-5 Virus titre will be present in a batch of the vaccine.
Titrate the vaccine virus by inoculation into embryonated
2-4-1-1 General safet:y. For each test, use not fewer than
hens' eggs from an SPF flock (5.2.2) or into suitable cell
8 chickens. Administer to each chicken a quantity of the
cultures (5.2.4). The vaccine complies with the test if 1 dose
vaccine virus equivalent to not les s than 10 times the
contains not less than the minimum virus titre stated on the
maximum virus titre likely to be contained in 1 dose of the
label.
vaccine. 14 days after vaccination, collect blood samples fram
3-6 Potency half of the chickens and determine the haematocrit value.
The vaccine complies with the requirements of the test Euthanise these chickens and carry out post-mortem
prescribed under Immunogenicity (section 2-4-5) when examination. Note any pathological changes attributable to
administered by a recommended raute and method. It is not chicken anaemia virus, such as thymic atrophy and specific
necessary to carry out the potency test for each batch of the bone-marrow lesions. Observe the remaining chickens at least
vaccine if it has been carried out on a representative batch daily for at least 21 days.
using a vaccinating dose containing not more than the The test is not valid if non-specific mortality occurs.
minimum virus titre stated on the label.
_____________________________________________ ~E~
The.vaccine virus complies with the test if during the
observation period no chicken shows abnormal signs of
disease or dies from causes attributable to the vaccine virus.
2-4-1-2 Safety for young chickens. Use not fewer than twenty
1-day-old chickens from an SPF flock (5.2.2). Administer to
Vet-274 Veterinary Vaccines 2016

each chicken by the oculonasal route a quantity of the recommended for vaccination and from an SPF fiock (5.2.2);
vaccine virus equivalent to not less than the maximum titre keep not fewer than 10 unvaccinated breeder chickens of the
likely to be contained in 1 dos e of the vaccine. Observe the same origin and from an SPF fiock (5.2.2) as controls. At a
chickens at least daily. Record the incidence of any signs suitable time after excretion of vaccine virus has cea sed,
attributable to the vaccine virus, such as depression, and any collect fertilised eggs from each vaccinated and control
deaths. 14 days after vaccination, collect blood samples from breeder chicleen and incubate them. Challenge at least 3
half of the chickens and determine the haematocrit value. randomly chosen 1-day-old chickens from each vaccinated
Euthanise these chickens and carry out post-mortem and control breeder chicleen by intramuscular administration
examination. Note any pathological changes attributable to of a sufficient quantity of virulent chicleen anaemia virus.
chicleen anaemia virus, such as thymic atrophy and specific Observe the chickens at least daily for 14 days after
bone marrow lesions. Observe the remaining chickens at least challenge. Record the deaths and the surviving chickens that
daily for at least 21 days. Assess the extent to which the show signs of disease. At the end of the observation period
vaccine strain is pathogenic for 1-day-old susceptible chickens determine the haematocrit value of each surviving chicken.
from the results of the clinical observations and mortality Euthanise these chickens and carry out post-mortem
rates and the proportion of chickens examined at 14 days examination. Note any pathological signs attributable to
that show anaemia (haematocrit value less than 27 per cent) chicleen anaemia virus, such as thymic atrophy and specific
and signs of infectious chicleen anaemia on post-mortem bone-marrow lesions.
examination. The results are used to formulate the label The test is not valid if:
statement on safety for young chickens. - during the observation period after challenge fewer than
2-4-2 Increase in virulence 90 per cent of the chickens of the control breeder
Carry out the test according to general chapter 5.2.6 using chickens die 01' show severe signs of infectious chicleen
1-day-old chickens from an SPF fiock (5.2.2). If the anaemia, including haematocrit value under 27 per cent,
properties of the vaccine virus allow sequential passage and/or notable macroscopic lesions of the bone marrow
through 5 groups via natural spreading, this method may be and thymus;
used, otherwise passage as described below is carried out. and/or during the period between vaccination and egg
Administer to each chicleen of the 1st group by the collection more than 10 per cent of vaccinated or control
intramuscular route a quantity of the vaccine virus that will breeder chickens show notable signs of disease 01' die
allow recovery of virus for the passages described below. from causes not attributable to the vaccine.
Prepare 7-9 days after administration a suspension from the The vaccine complies with the test if during the observation
liver of each chicleen and pool these samples. Depending on period after challenge not fewer than 90 per cent of the
the tropism of the virus, other tissues such as spleen or bone chickens of the vaccinatedbreeder chickens survive and show
marrow may be used. Administer 0.1 mL of the pooled no notable signs of disease and/or macroscopic lesions of the
samples by the intramuscular route to each chicleen of the bone marrow and thymus.
next group. Carry out this passage operation not fewer than 2-4-3-2 Prevention 01 virus excretion. Vaccinate according to
4 times; verify the presence of the virus at each passage. the schedule to be recommended not fewer than 10 chickens
If the virus is not found at a passage level, repeat the passage not older than the minimum age to be recommended for
by administration to a group of 10 chickens. vaccination and from an SPF fiock (5.2.2). Maintain
If the 5th group of chickens shows no evidence of an increase separately not fewer than 10 chickens of the same age and
in virulence indicative of reversion during the observation origin as controls. At a suitable time after excretion of
period, further testing is not required. Otherwise, carry out vaccine virus has ceased, challenge all the chickens by
. an additional safety test and compare the clinical signs and intramuscular administration of a sufficient quantity of
any relevant parameters in a group of at least 10 chickens virulent chicleen anaemia virus. Collect blood samples from
receiving the material used for the 1st passage and another the chickens on days 3, 5 and 7 after challenge and faecal
similar group receiving the virus at the final passage level. samples from the chickens on days 7, 14 and 21 after
The vaccine virus complies with the test if no indication of challenge and carry out a test for presence of virus to
increased virulence of the virus at the final passage level determine whether or not the chickens are viraemic and are
compared with the material used for the 1st passage is excreting the virus.
observed. If virus is not recovered after an initial passage in The test is not valid if:
5 chickens and a subsequent repeat passage in 10 chickens, - fewer than 70 per cent of the control chickens are
the vaccine virus also complies with the test. viraemic and excrete the virus at one or more times of
2-4-3 Immunogenicity sampling;
A test is carried out for each route and method of - and/or during the period between vaccination and
administration to be recommended for vaccination using challenge more than 10 per cent of control or vaccinated
chickens not older than the minimum age to be chickens show abnormal clinical signs or die from causes
recommended for vaccination and from an SPF fiock (5.2.2). not attributable to the vaccine.
The test for prevention of virus excretion is intended to The vaccine complies with the test if not fewer than
demonstrate reduction of egg transmission through viraemia 90 per cent of the vaccinated chickens do not develop
and virus excretion in the faeces. The quantity of the vaccine viraemia or excrete the virus.
virus to be administered to each chicken is not greater than 3 BATCH TESTING
the minimum virus titre to be stated on the label and the 3-1 Identificatiún
virus is at the most attenuated passage level that will be The vaccine, diluted if necessaty and mixed with a
present in a batch of vaccine. monospecific chicleen anaemia virus antiserum, no longer
2-4-3-1 Passive im71nmisation 01 chickens. Vaccinate according infects susceptible cell cultures or eggs from an SPF
to the schedule to be recommended not fewer than 10 fiock (5.2.2) into which it is inoculated.
breeder chickens not older than the minimum age to be
2016 Veterinary Vaccines V et-2 75

3-2 Bacteria and fungi 2-2-2 Cell cultures


Vaccines intended for administration by injection comply If the vaccine virus is grown in cell cultures, they comply
with the test for sterility prescribed in the monograph with the requirements for cell cultures for production of
Vaccil1es for veteril1aJY use (0062). veterinary vaccines (5.2.4).
Frozen or freeze-dried vaccines produced in embryonated 2-3 SEED LOTS
eggs and not intended for administration by injection either 2-3-1 Extraneous agents
comply with the test for sterility prescribed in the monograph The master seed lot complies with the tests for extraneous
Vaccil1es for veteril1aJY use (0062) or with the following test: agents in seed lots (2.6.24). In these tests on the master seed
carry out a quantitative test for bacterial and fungal lot, the organisms used are not more than 5 passages from
contamination; cany out identification tests for the master seed lot at the start of the tests.
microorganisms detected in the vaccine; the vaccine does not
contain pathogenic microorganisms and contains not more 2-4 CHOICE OF VACCINE VIRUS
than 1 non-pathogenic microorganism per dose. The vaccine virus shall be shown to be satisfactory with
respect to safety (5.2.6) and efficacy (5.2.7) for the chickens
Any diluent supplied for reconstitution of the vaccine
for which it is intended.
complies with the test for sterility prescribed in the
monograph Vaccines for veterinaJY use (0062). The following tests for index of respiratory virulence
(section 2-4-1), safety (section 2-4-2), increase in virulence
3-3 Mycoplasmas
(section 2-4-3) and immunogenicity (section 2-4-4) may be
The vaccine complies with the test for mycoplasmas (2.6.7).
used during the demonstration of safety and efficacy.
3-4 Extraneous agents
2-4-1 Index of respiratory virulence
The vaccine complies with the tests for extraneous agents in
U se for the test not fewer than sixty 10-day-old chickens
batches of finished product (2.6.25).
from an SPF fiod: (5.2.2). Divide them randomly into
3-5 Virus titre 3 groups, maintained separately. Prepare 2 tenfold serial
Titrate the vaccine virus by inoculation into suitable cell dilutions starting from a suspension of the vaccine virus
cultures (5.2.4) or eggs from an SPF fiock (5.2.2). having a titre of.10 s EIDso 01' lOS CCID so per 0.2 mL or, if
The vaccine complies with the test if 1 dose contains no! less not possible, having the maximum attainable titre.
than the minimum virus titre stated on the label. U se vaccine virus at the least attenuated passage level that
3-6 Potency will be present between the master seed lot and a batch of
The vaccine complies with the requirements of the tests the vaccine. Allocate the undiluted virus suspension and the
prescribed under Immunogenicity (sections 2-4-3-1 and 2 virus dilutions each to a different group of chickens.
2-4-3-2) when administered by a recommended route and Administer by the intratracheal route to each chicken 0.2 mL
method. It is not necessaly to carry out the potency test for of the virus suspension attributed to its group. Observe the
each batch of the vaccine if it has been carried out on a chickens for 10 days after administration and record the
representative batch using a vaccinating dose containing not number of deaths. The index of respiratory virulence is the
more than the minimum virus titre stated on the label. total number of deaths in the 3 groups divided by the total
number of chickens. The vaccine virus complies with dle test
4 LABELLING
if its index of respiratory virulence is not greater than 0.33.
The label states to which extent the vaccine virus causes
disease if it spreads to susceptible young chickens. 2-4-2 Safety
_____________________________________________ PhE~
Carry out the test for each route and method of
administration to be recommended for vaccination, using in
each case chickens not older than the minimum age to be
recommended for vaccination and from an SPF fiod: (5.2.2).
U se vaccine virus at the least attenuated passage level that
Laryngotracheitis Vaccine, Living will be present in a batch of the vaccine. For each test
performed in chickens younger than 3 weeks of age, use not
(Avían Infectíous LaJyngotracheitis Vaccine (Live)J fewer than 10 chickens. For each test performed in chickens
Ph Eur 7110nograph 1068) older than 3 weeks of age, use not fewer than 8 chickens.
~E~ _____________________________________________ Administer to each chicleen a quantity of the vaccine virus
equivalent to not less than 10 times the maximum virus titre
1 DEFINITION lileely to be contained in 1 dose of the vaccine. Observe the
Avian infectious laryngotracheitis vaccine (live) is a chickens at least daily for at least 14 days.
preparation of a suitable strain of avian infectious
The test is not valid if more than 10 per cent of the chickens
laryngotracheitis virus (gallid herpesvirus 1). This monograph
younger than 3 weeks of age show abnormal signs of disease
applies to vaccines intended for administration to chickens
01' die from causes not attributable to the vaccine.
for active immunisation.
For chickens older than 3 weeks of age, the test is not valid if
2 PRODUCTION non-specific mortality occurs.
2-1 PREPARATION OF THE VACCINE The vaccine virus complies with the test if no chicleen shows
The vaccine virus is grown in embryonated hens' eggs or in abnormal signs of disease 01' dies from causes attributable to
cell cultures. the vaccine virus.
2-2 SUBSTRATE FOR VIRUS PROPAGATION 2-4-3 Increase in virulence
2-2-1 Embryonated hens' eggs Carry out the test according to general chapter 5.2.6 using
If the vaccine virus is grown in embryonated hens' eggs, they chickens not more than 2 wéeks old, from an SPF fiock
are obtained from fiocks free fram specified pathogens (SPF) (5.2.2). If the properties of the vaccine virus allow sequential
(5.2.2). passage ilirough 5 groups via natural spreading, this method
Vet-276 Veterinary Vaccines 2016

may be used, othelwise passage as described below is carried longer infects embryonated hens' eggs from an SPF
out. flock (5.2.2) or susceptible cell cultures (5.2.4) into which it
Administer to each chicken of the 1st group by eye-drop a is inoculated.
quantity of the vaccine virus that will allow recovery of virus 3-2 Bacteria and fungi
for the passages described below. After the period shown to Vaccines intended for administration by injection comply
correspond to maximum replication of the virus, prepare a with the test for sterility prescribed in the monograph
suspension from the mucosae of suitable parts of the Vaccines for veterinmy use (0062).
respiratory tract of each chicken and pool these samples. Frozen or freeze-dried vaccines produced in embryonated
Administer 0.05 mL of the pooled samples by eye-drop to eggs and not intended for administration by injection either
each 2 week-old SPF chicken (5.2.2) ofthe next group. comply with the test for sterility prescribed in the monograph
Carry out this passage operation not fewer than 4 times; Vaccines for veterinmy use (0062) or with the following test:
verify the presence of the virus at each passage. If the virus is carry out a quantitative test for bacterial and fungal
not found at a passage level, repeat the passage by contamination; carry out identification tests for micro-
administration to a group of 10 chickens. Determine the organisms detected in the vaccine; the vaccine does not
index of respiratory viru1ence (section 2-4-1) using the contain pathogenic micro-organisms and contains not more
material used for the 1st passage and the virus at the final than 1 non-pathogenic micro-organism per dose.
passage leve!; if the titre of the final passaged virus is less
Any diluent supplied for reconstitution of the vaccine
than lOS EID so or lOS CCID so , prepare the tenfold, serial
complies with the test for sterility prescribed in the
dilutions using the highest titre available.
monograph Vaccines for veterinmy use (0062).
The vaccine virus complies with the test if no indication of
3-3 Mycoplasmas
an increase in virulence of the virus recovered for the final
The vaccine complies with the test for mycoplasmas (2.6.7).
passage compared with the material used for the 1st passage
is observed. If virus is not recovered after an initial passage in 3-4 Extraneous agents
5 chickens and a subsequent repeat passage in 10 chickens, The vaccine complies with the tests for extraneous agents in
the vaccine virus also complies with the test. batches of finished product (2.6.25).
2-4-4 Irnmunogenicity 3-5 Virus titre
A test is carried out for each route and method of Titrate the vaccine virus by inoculation into embryonated
administration to be recommended for vaccination using in hens ' eggs from an SPF flock (5.2.2) or into suitable cell
each case chickens not older than the minimum age to be cultures (5.2.4). The vaccine complies with the test if 1 dose
recommended for vaccination. The quantity of the vaccine contains not les s than the minimum titre stated on the label.
virus administered to each chicken is not greater than the 3-6 Potency
minimum virus titre to be stated on the label and the virus is The vaccine complies with the requirements of the test
at the most attenuated passage level that will be present in a prescribed under Irnmunogenicity (section 2-4-4) when
batch of the vaccine. Use for the test not fewer than administered according 10 the recommended schedule by a
30 chickens of the same origin and from an SPF recommended route and method. It is not necessary to carry
flock (5.2.2). Vaccinate by a route to be recommended not out the potency test for each batch of the vaccine if it has
fewer than 20 chickens. Maintain not fewer than 10 chickens been carried out on a representative batch using a vaccinating
as controls. Challenge each chicken after 21 days by the dose containing not more than the minimum virus titre
intratracheal route with a sufficient quantity of virulent stated on the label.
infectious laryngotracheitis virus. Observe the chickens at _____________________________________________ ~Elf

least daily for 7 days after challenge. Record the deaths and
the number of surviving chickens that show clinical signs of
disease. At the end of the observation period euthanise all the
surviving chickens and carry out examination for macroscopic
lesions: mucoid, haemorrhagic and pseudomembraneous Bovine Leptospirosis Vaccine
inflammation of the trachea and orbital sinuses.
The test is not valid if:
(Inactivated)
-- during the observation period after challenge fewer than (Ph. Eur. monograph 1939)
_____________________________________________
90 per cent of the control chickens die or show severe ~Elf

clinical signs of avian infectious laryngotracheitis or


1 DEFINITION
notable macroscopic lesions of the u"achea and orbital
Eovine leptospirosis vaccine (inactivated) is a preparation of
sinuses;
inactivated whole organisms and/or antigenic extractes) of
-- or if during the period between the vaccination and
one or more suitable strains of one 01" more of Leptospira .
challenge more than 10 per cent of the vaccinated or
borgpetersenii serovar hardjo, Leptospira interrogans serovar
control chickens show notable clinical signs of disease or
hardjo or other L. interrogans serovars, inactivated while
die from causes not attributable to the vaccine.
maintaining adequate irnmunogenic properties. This
The vaccine virus complies with the test if during the monograph applies to vaccines intended for the active
observation period after challenge not fewer than 90 per cent immunisation of cattle against lep1Ospirosis.
of the vaccinated chickens survive and show no notable
clinical signs of disease and/or macroscopicallesions of the 2 PRODUCTION
trachea and orbital sinuses. 2-1 PREPARAIION OF THE VACCINE
The seed material is cultured in a suitable medium; each
3 BATCH TESTS strain is cultivated separately. During production, various
3-1 Identification parameters such as growth rate are monitored by suitable
The vaccine, diluted if necessary and mixed with a methods; the values are within the lirnits approved for the
monospecific infectious laryngotracheitis virus antiserum, no particular producto Purity and identity are verified on the
2016 Veterinary Vaccines Vet-277

harvest using suitable methods. After cultivation, the bacterial relevant serovar. Observe the cattle at least daily for a further
harvest is inactivated by a suitable method. The antigen may 35 days. Collect urine samples from each animal on days 0,
be concentrated. The vaccine may be adjuvanted. 14, 21, 28 and 35 post-challenge. Euthanise surviving cattle
2-2 CHOICE OF VACCINE COMPOSITION at the end of the observation periodo Carry out post-mortem
examination on any animal that dies and on those euthanised
The vaccine is shown to be satisfactory with respect to safety
at the end of the observation periodo In particular, examine
(5.2.6) and efficacy (5.2.7) for the cattle for which it is
the kidneys for macroscopic and microscopic signs of
intended.
leptospira infection. A sample of each kidney is collected and
The following tests for safety (section 2-2-1) and each urine and kidney sample is tested for the presence of
irnmunogenicity (section 2-2-2) may be used during the the challenge organisms by re-isolation 01' by another suitable
demonstration of safety and efficacy. method.
2-2-1 Safety For the test conducted with L. borgpete1"senii serovar hardjo,
2-2-1-1 Laborat01Y tests. Carry out the test for each route and control cattle are regarded as infected if the challenge
method of adminisu'ation to be recommended for vaccination organisms are re-isolated from at least 2 samples. The test is
and in cattle of each category for which the vaccine is not valid if infection has been established in fewer than
intended (for example, young calves, pregnant cattle). Use a 80 per cent of the control cattle.
batch of vaccine containing not les s than the maximum
The vaccine complies with the test if the challenge organisms
potency that may be expected in a batch of vaccine.
are re-isolated from any urine or kidney sample from not
For each test, use not fewer than 8 cattle that do not have more than 20 per cent ofthe vaccinated cattle.
antibodies against L. borgpetersenii serovar hardjo and the
2-2-2-2 Immunogenicity against other leptospira species.
principal serovars of L. imerrogans (icterohaemorrhagiae,
For leptospiral species other than L. b01"gpetel'senii serovar
canicola, grippotyphosa, sejroe, hardjo, hebdomonadis,
hardjo, the test is conducted as described in section 2-2-2-1
pomona, australis and autumnalis). Administer to each
but urine samples are collected on appropriate days,
animal 1 dose of the vaccine. If the schedule to be
determined by the characteristics of the challenge model.
recommended requires a 2nd dose, administer another dose
In the case of serovars for which there is published evidence
after an interval of at least 14 days. Observe the cattle at least
that the serovar has a lower tropism for tlle urinal)' tract, a
daily for at least 14 days after the last administration. Record
lower rate of infection may be justified. Depending on their
body temperatures the day before each vaccination, at
tissue tropism, for sorne leptospira serovars, samples from
vaccination, 4 h later and daily for 4 days.
other tissues/body fluids can be used to establish whether the
The vaccine complies with the test if no animal shows cattle are infected or not by the challenge organismo
abnormal local or systemic reactions, signs of disease, or dies
from causes attributable to the vaccine. 2-3 MANUFACTURER'S TESTS
2-3-1 Batch potency test
2-2-1-2 Field studies. The cattle used for the field trials are
It is not necessary to carry out the potency test (section 3-4)
also used to evaluate safety. Use not fewer than 3 groups of
for each batch of vaccine if it has been carried out using a
20 cattle with corresponding groups of not fewer than
batch of vaccine with a minimum potency. Where the test is
10 controls in 3 different locations. Examine tlle injection
not canied out, an alternative validated method is used, the
sites for local reactions after vaccination. Record body
criteria for acceptance being set with reference to a batch of
temperatures the day before vaccination, at vaccination and
vaccine that has given satisfactory results in the test described
on the 2 days following vaccination.
under Potency. The following test may be used.
The vaccine complies with the test if no animal shows
For each of the serovars for which protection is claimed, the
notable signs of disease or dies from causes attributable to
antibody response from vaccinated animals is measured.
the vaccine. In addition, if the vaccine is for use in pregnant
Use not fewer than 12 guinea-pigs weighing 250-350 g that
cattle, no adverse effects on the pregnancy and offspring are
do not have antibodies against L. bOl'gpete1"senii serovar hardjo
noted.
and the plincipal serovars of L. intel'rogans
2-2-2 Immunogenicity (icterohaemorrhagiae, canicola, grippotyphosa, sejroe, hardjo,
Carry out a separate test for each of the serovars for which a hebdomonadis, pomona, ausu'alis and autumnalis) and that
claim is made for a beneficial effect on the rates of infection have been obtained from a regularly tested and certified
and urinary excretion. If claims are to be made for protection leptospira-free source. The dose to be administered to the
against reproductive 01' production los ses, further specific guinea-pigs is that fraction of a cattle dose which has been
studies will be required. shown in the validation studies to provide a suitably sensitive
Each test is carried out for each route and method of test. Vaccinate each of 10 guinea-pigs with the suitable dose.
administration to be recommended, using in each case cattle Maintain not fewer than 2 guinea-pigs as controls. At a given
of the minimUll1 age to be recommended for vaccination. interval within the range of 19-23 days after the injection,
The vaccine administered to each animal is of minimum collect blood from each guinea-pig and prepare serum
potency. samples. Use a suitable validated method such as a micro-
2-2-2-1 Immunogenicity against L. borgpetersenii serovar hardjo. agglutination test to measure the antibodies in each sample.
Use not fewer than 15 cattle that do not have antibodies The vaccine complies with the test if antibody levels are
against L. bOl'gpetersenii serovar hardjo and the principal equal to or greater than those obtained with a batch that has
serovars of L. intel'rogans (icterohaemorrhagiae, canicola, given satisfactory results in the test described under Potency
grippotyphosa, sejroe, hardjo, hebdomonadis, pomona, and there is no significant increase in antibody titre in the
australis and autUll1nalis). Vaccinate not fewer than 10 cattle controls.
according to the schedule to be recommended. Maintain not
fewer than 5 cattle as controls. 20-22 days after the last
vaccination, challenge all the cattle by a suitable mucosal
route with a sufficient quantity of a virulent strain of the
Vet-278 Veterinary Vaccines 2016

3 BATCH TESTS 2-2-1 Safety


3-1 Identification Carry out the test for each route and method of
When injected into healthy animals that do not have specific administration to be recommended for vaccination and in
antibodies against the leptospira serovar(s) present in the dogs of each category for which the vaccine is to be intended,
vaccine, the vaccine stimulates the production of such using in each case dogs not older than the minimum age to
antibodies. be recommended for vaccination. Use a batch of vaccine
3-2 Bacteria and fungi containing not less than the maximum potency that may be
The vaccine, including where applicable the diluent supplied expected in a batch of vaccine.
for reconstitution, complies with the test for sterility For each test, use not fewer than 8 dogs that do not have
prescribed in the monograph Vaccines for veterina1Y antibodies against the principal L. interrogans serovars
use (0062). (icterohaemorrhagiae, canicola, grippotyphosa, sejroe, hardjo,
hebdomonadis, pomona, australis and autumnalis).
3-3 Residuallive bacteria
Administer to each dog 1 dose of the vaccine. If the schedule
Carry out a test for live leptospirae by inoculation of a
to be recommended requires a 2nd dose, administer 1 dose
specific medium. Inoculate 1 mL of the vaccine into 100 mL
after the recommended interval. Observe the dogs at least
of the medium. Incubate at 30 oC for 14 days, sub culture
daily for at least 14 days after the last administration. Record
into a further quantity of the medium and incubate both
body temperatures the day before each vaccination, at
media at 30 oC for 14 days: the vaccine complies with the
vaccination, 4 h later and daily for 4 days.
test if no growth occurs in either medium. At the same time,
carry out a control test by inoculating a further quantity of The vaccine complies with the test if no dog shows abnormal
the medium with the vaccine together with a quantity of a local or systemic reactions, signs of disease or dies from
culture containing approximately 100 leptospirae and causes attributable to the vaccine.
incubating at 30 oC: the test is not valid if growth of 2-2-2 Irnmunogenicity
leptospirae do es not occur within 14 days. For each type of the serovars against which protective
3-4 Potency immunity is claimed on the label, carry out a separate test
The vaccine complies with the requirements of the test with a challenge strain representative of that serovar.
mentioned under Immunogenicity (section 2-2-2) when Each test is carried out for each route and method of
administered by a recommended route and method. administration to be recommended for vaccination, using in
_____________________________________________ PhE~ each case dogs of the minimum age to be recommended.
The vaccine administered to each dog is of minimum
potency.
U se for the test not fewer than 12 dogs that do not have
antibodies against the principal serovars of L. interrogan s
Canine Leptospirosis Vaccine ******
*
(icterohaemorrhagiae, canicola, grippotyphosa, sejroe, hardjo,
(lnactivated) ***** hebdomonadis, pomona, australis and autumnalis). Vaccinate
not fewer than 6 dogs, according to the schedule to be
(Ph. Eur. 71Z01l0graph 0447) recommended. Maintain not fewer than 6 dogs as controls.
~E~ _____________________________________________
Challenge each dog after 25-28 days by the conjunctival
1 DEFINITION and/or intraperitoneal route with a sufficient quantity of a
Canine leptospirosis vaccine (inactivated) is a preparation of suspension of the relevant pathogenic L. interrogans serovar.
inactivated whole organisms and/or antigenic extractes) of Observe the dogs at least daily for 28 days after challenge.
one or more suitable strains of one or more of Leptospira Examine the dogs daily and record and score clinical signs
imerrogam serovar canicola, serovar icterohaemorrhagiae or observed post-challenge and any deaths that occur. If a dog
any other epidemiologically appropriate serovar, inactivated shows marked signs of disease, it is euthanised. Monitor
while maintaining adequate immunogenic properties. This body temperatures each day for the first week after challenge.
monograph applies to vaccines intended for the active Collect blood samples from each dog on days O, 2, 3, 4, 5, 8
immunisation of dogs against leptospirosis. and 11 post challenge. Collect urine samples from each dog
on days O, 3, 5, 8, 11, 14, 21 and 28 post challenge.
2 PRODUCTION
Euthanise surviving dogs at the end of the observation
2-1 PREPARATION OF THE VACCINE periodo Carry out post-mortem examination on any dog that
The seed material is cultured in a suitable medium; each dies during the observation period and on the remainder
strain is cultivated separately. During production, various when euthariised at the end of the observation periodo
parameters such as growth rate are monitored by suitable In particular, examine the liver and kidneys for macroscopic
methods; the values are within the limits approved for the and microscopic signs of leptospira infection. Take a sample
particular producto Purity and identity are verified on the of each kidney and test each blood, urine and kidney sample
harvest using suitable methods. After cultivation, the bacterial for the presence of challenge organisms by re-isolation or by
harvests are collected separately and inactivated by a suitable another suitable method. Analyse blood samples to detect
method. The antigen may be concentrated. The vaccine may biochemical and haematological changes indicative of
be adjuvanted. infection and score these.
2-2 CHOICE OF VACCINE COMPOSITION The test is not valid if: samples give positive results on day O;
The vaccine is shown to be satisfactory with respect to safety L. interrogans serovar challenge strain is re-isolated from or
(5.2.6) and efficacy (5. 2.7) for the dogs for which it is demonstrated by another suitable methodto be preserit in
intended. fewer than 2 samples on fewer than 2 different days, to show
The following tests for safety (section 2-2-1) and infection has been established in fewer than 80 per cent of
immunogenicity (section 2-2-2) may be used during the the control dogs.
demonstration of safety and efficacy.
2016 Veterinary Vaccines Vet-279

The vaccine complies with the test if: at least 80 per cent of more antigenic components which are indicators of
the vaccinates show no more than mild signs of disease (for protection and which are specific for that serovar.
example, transient hyperthermia) and, depending on the The criteria for acceptance are set with reference to a batch
L. interrogans serovar used for the challenge, one or more of of vaccine that has given satisfactory results in the test
the following is also shown: described under Potency.
-- where the vaccine is intended to have a beneficial effect
3 BATCH TESTS
against signs of disease, the clinical scores and
3-1 Identification
haematological and biochemical scores are statistically
When injected into healthy animals that do not have specific
lower for the vaccinates than for the controls,
antibodies against leptospira serovar(s) present in the vaccine,
-- where the vaccine is intended to have a beneficial effect
the vaccine stimulates the production of such antibodies.
against infection, the number of days that the organisms
If test 2-3-1-3 is used for the batch potency test, it also
are detected in the blood is statistically lower for the
serves to identify the vaccine.
vaccinates than for the controls,
-- where the vaccine is intended to have a beneficial effect 3-2 Bacteria and fungi
against urinary tract infection and excretion, the number The vaccine, including where applicable the diluent supplied
of days that the organisms are detected in the urine and for reconstitution, complies with the test for sterility
the number of kidney samples in which the organisms are prescribed in the monograph Vaccines for veterinary use
detected is statistically lower for the vaccinates than for (0062).
the controls. 3-3 Residuallive bacteria
2-3 MANUFACTURER'S TESTS Carry out a test for live leptospirae by inoculation of a
2-3-1 Batch potency test specific medium. Inoculate 1 mL of the vaccine into 100 mL
of the medium. Incubate at 30 oC for 14 days, sub culture
It is not necessary to carry out the Potency test (section 3-4)
for each batch of the vaccine if it has been canied out using into a further quantity of the medium and incubate both
a batch of vaccine with a minimum potency. Where the test media at 30 oC for 14 days: the vaccine complies with the
is not carried out, an altemative validated method is used, test if no growth occurs in either medium. At the same time,
the criteria for acceptance being set with reference to a batch carry out a control test by inoculating a further quantity of
of vaccine that has given satisfactory results in the test the medium with the vaccine together with a quantity of a
described under Potency. The following tests may be used. culture containing approximately 100 leptospirae and
incubating at 30 oC: the test is not valid if growth of
2-3-1-1 For vaccines zuith al' zuithout adJuvants. If leptospira leptospirae does not occur within 14 days.
from more than one serovar (for example L. interrogans
serovar canicola and serovar icterohaemorrhagiae) has been 3-4 Potency
used to prepare the vaccine, carry out a batch potency test The vaccine complies with the requirements of the test
for each serovar against which protective irnmunity is claimed mentioned under Immunogenicity (section 2-2-2) when
on the label. U se for the test 10 healthy hamsters not more administered by a recommended route and method.
than 3 months old, that do not have antibodies against the _____________________________________________ PhEw

principal serovars of L. imerrogans (icterohaemonhagiae,


canicola, grippotyphosa, sejroe, hardjo, hebdomonadis,
pomona, australis and autumnalis) and which have been
obtained from a regularly tested and certified leptospira-free
source. Administer 1/40 of the dose for dogs by the
Louping .. iII Vaccine
subcutaneous route to 5 hamsters. Maintain 5 hamsters as DEFINITION
controls. Challenge each hamster after 15-20 days by the Louping-ill Vaccine is a preparation of a suitable strain of
intraperitoneal route with a sufficient quantity of a virulent louping-ill virus which has been inactivated in such a manner
culture of leptospirae of the serovar against which protective that immunogenic activity is retained.
irnmunity is claimed on the label. The vaccine complies with PRODUCTION
the test if not fewer than 4 of the 5 control hamsters die
The virus strain is grown in suitable cell cultures,
showing typical signs of leptospira infection within 14 days of
Appendix XV J evet) 1. The viral suspension is harvested and
receiving the challenge suspension and if not fewer than 4 of
inactivated. A test for residual infectious louping-ill virus is
the 5 vaccinated hamsters remain in good health for 14 days
can'ied out on each batch of antigen immediately after
after the death of 4 control hamsters.
inactivation. A mouse inoculation test may provide a suitably
2-3-1-2 For vaccines 'lvith 01' 'lvithout adJuvants. A suitable sensitive test if there is no suitably sensitive in vitro test for
validated sero-response test may be canied out. Vaccinate the strain.
each animal in a group of experimental animals with a
The vaccine contains an adjuvant.
suitable dose. Collect blood samples after a suitable, fixed
time after vaccination. For each of the serovars present in the CHOICE OF VACCINE COMPOSITION
vaccine, an in vitro test is carried out on individual blood This vaccine is shown to be satisfactory with respect to safety
samples to determine the antibody response to one or more and irnmunogenicity for the animals for which the vaccine is
antigenic components which are indicators of protection and intended. The followihg tests may be used during the
which are specific for that serovar. The criteria for demonstration of safety, Appendix XV Jevet) 1 and
acceptance are set with reference to a batch of vaccine that irnmunogenicity, Appendix XV Jevet) 2.
has given satisfactory results in the test described under Safety
Potency. Can'] out a test in each category of each species of animal
2-3-1-3 For vaccines zuithout adJuva71ts. For each of the for which the vaccine is to be recommended and by each
serovars present in the vaccine, a suitable validated in vitro recommended route of administration. Vaccinate at least five
test may be carried out to determine the content of one or animals that do not have antibodies to louping-ill virus.
Use for the test a batch of vaccine with the maximum
Vet-280 Veterinary Vaccines 2016

potency likely to be included in adose of the vaccine. source that is monitored for freedom from certain diseases, as
Administer a double dose of vaccine to each animal and agreed with the competent authority. The calves are healthy,
observe them for two weeks. No abnormallocal 01' systemic have not been exposed previously to Dictyocaulus viviparus
reactions occur. If the vaccine is for use 01' may be used in and have been shown to be free from a range of infectious
pregnant animals, for the test in this category, administer the diseases, as agreed with the competent authority.
vaccine at the relevant stage 01' stages of pregnancy, pro long The third stage larvae are harvested from the faeces, purified,
the observation period up to the time of parturition and note partially inactivated by ionising radiation, then diluted as
any effects on gestation 01' on the offspring. necessary.
lnununogenicity CHOICE OF VACCINE STRAIN
The tests to demonstrate immunogenicity are carried out in The suspension of irradiated vaccinal organisms is shown to
each category of each species of animal for which the vaccine be satisfactory with respect to safety and immunogenicity for
is to be recommended and by each recommended route of the animals for which the vaccine is intended. The following
administration and using a batch 01' batches with the tests may be used during the demonstration of safety,
minimum potency likely to be included in adose of the Appendix XV K(Vet) 1 and immunogenicity,
vaccine. The efficacy claims made on the label Appendix XV K(Vet) 2.
(e.g. protection from clinical signs of the disease) reflect the
Safety
type of data generated.
Carry out a test in at least five calves of the minimum age to
BATCH TESTING be recommended for vaccination that do not have antibodies
Inactivation to Dictyocaulus viviparus. Administer orally, to each calf, a
Carry out a suitable validated test for residual louping-ill quantity of irradiated vaccinal organisms corresponding to
virus on the bulk antigen blend immediately before the twice the maximum number of organisms likely to be
addition of the adjuvant. No live virus is detected. included in adose of the vaccine. Observe the calves for
The vaccine complies with the requirements stated under 6 weeks. They remain in good health and show no more
Veterz'na¡y Vaccines with the following modifications. than transient respiratory signs approximately one week after
vaccination. Collect faecal samples from each calf 4, 5 and
IDENTIFICATION 6 weeks after vaccination and examine separately for the
When injected into healthy seronegative animals, the vaccine presence of D. viviparus larvae. No larvae are detected.
stimulates the production df specific haemagglutinating
lnununogenicity
antibodies against louping-ill virus.
The test described under Potency is suitable to demonstrate
TESTS immunogenicity when carried out using the minimum
Extraneous bacteria and fungi number of organisms likely to be included in adose of the
The vaccine complies with the test for sterility described vaccine.
under Veterinary Vaccines. BATCH TESTING
Safety If Identification test B and the test for Potency have been
Use two lambs of the minimum age recommended for carried out with satisfactory results on a representative batch
vaccination and that do not have antibodies to louping-ill of vaccine, these tests may be omitted as a routine control on
virus. Administer a double dose of vaccine to each of the other batches of vaccine subject to the agreement of the
lambs by a route recommended on the label and observe the competent authority.
animal s for two weeks. No abnormallocal 01' systemic Subject to the agreement of the competent authority, Test A
reactions occur. described below under Safety, may be conducted on
POTENCY representative batches, selected at defined periods during the
Inject subcutaneously each of no fewer than six healthy production season, rather than as a routine test on every
sheep, free from louping-ill haemagglutination-inhibiting (HI) batch.
antibodies, with the dose stated on the label. Between 14 and Extraneous bacteria
28 days after injection the serum of at least five of the sheep For each batch, the number of non-pathogenic organisms per
contains HI antibodies at dilutions of 1 in 20 01' greater dose is shown to be within the limits set for the product and
against 4 to 8 haemagglutinating units. shown to be safe.
STORAGE The vaccine complíes with the requirements stated undel'
When stored under the prescribed conditions the vaccine Veterina¡y Vaccines with the following modifications.
may be expected to retain its potency for at least 1 year. IDENTIFICATION
A. Produces petechial haemorrhages in the lungs of guinea-
pigs within 48 hours of oral administration. Adult worms do
not develop.
Lungworm (Dictyocaulus Viviparus) Oral B. Protects calves against D. vivipaJ"LlS infection and does not
cause parasitic bronchitis.
Vaccine, Living TESTS
DEFINITION Extraneous bacteria and fungi
Lungworm (Dictyocaulus Viviparus) Oral Vaccine, Living is The vaccine is shown by appropriate methods to be free from
an aqueous preparation containing approximately pathogenic organisms including Brucella, Mycobacteria and
1000 modified Dictyocaulus viviparus larva e per dose. Salmonella species.
PRODUCTION
The vaccinal organisms are produced in calves. The calves
used for production are obtained from a known, defined
2016 Veterinary Vaccines Vet-281

Extraneous virus es while maintaining adequate immunogenic properties. This


Inoculate the vaccine onto suitable celI cultures susceptible to monograph applies to vaccines intended for active
bovine virus es, make at least one passage and maintain the irnmunisation of cattle of different ages against respiratory
cultures for at least 14 days. No cytopathic effect develops. diseases caused by M. haemolytica.
Carry out a specific test for freedom from bovine viral 2 PRODUCTION
diarrhoea. No evidence of bovine viral diarrhoea is found. 2-1 PREPARATION OF THE VACClNE
Carry out a test for freedom from haemadsorbing agents. Production of the vaccine is based on a seed-Iot system.
The celI cultures show no signs of viral contamination. The seed material is cultured in a suitable medium; each
Safety strain is cultivated separately and identity is verified using a
A. Administer orally to each of two healthy susceptible calves suitable method. During production, various parameters such
of the minimum age for vaccination and free from antibodies as growth rate are monitored by suitable methods; the values
to D. viviparus, twice the dose stated on the label and repeat are within the limits approved for the particular producto
four weeks latero The animal s remain free from clinical signs Purity and identity of the harvest are verified using suitable
during 60 days after the second dose and show no post- methods. After cultivation, the bacterial suspensions are
mortem signs of parasitic bronchitis when subsequently colIected separately and inactivated by a suitable method.
killed. The vaccine may be adjuvanted.
B. Administer orally to each of four healthy susceptible 2-2 CHOICE OF VACCINE COMPOSITION
guinea-pigs the larval contents of 5 calf dos es of vaccine. KilI The choice of composition and the strains to be included in
two guinea-pigs on day 2 and the other two on day 10 after the vaccine is based on epidemiological data on the
dosing. Examine the lungs. The vaccine complies with the prevalence of the different serovars of M. hae11101ytica and on
test if at day 2 there are petechial haemorrhages in the lungs the claims being made.
and at day 10 there are no more than a very small number of The vaccine is shown to be satisfactory with respect to safety
fifth-stage larvae. (5.2.6) and efficacy (5.2.7) for the cattle for which it is
Viable larvae intended.
The nuniber of viable larvae is not les s than 1000 per dose The folIowing tests for safety (section 2-2-1) and
determined by microscopic examination. immunogenicity (section 2-2-2) may be used during the
POTENCY demonstration of safety and efficacy.
U se calves of the minimum age for vaccination 2-2-1 Safety
recommended on the label and that do not have antibodies 2-2-1-1 LaboratOlY tests. Carry out the test for each route and
to D. viviparus. Vaccinate at least ten calves as recommended method of administration to be recommended for vaccination
on the label. Maintain at least five calves as unvaccinated and in cattle of each category for which the vaccine is
controls. Observe the calves for 40 days and monitor and intended (for example, young calves, pregnant cattle). Use a
score the calves for signs of respiratory disease (e.g. increased batch of vaccine containing not less than the maximum
respiratory rate, coughing). Carry out post-mortem potency that may be expected in a batch of vaccine.
examination of the lungs of any animal that dies during the For each test, use not fewer than 8 cattle that preferably do
observation periodo At the end of the observation period, kilI not have antibodies against tl1e serovars of M. haemolytica or
the surviving calves and examine the lungs. The test is not against the leucotoxin present in the vaccine. Where justified,
valid unless the control calves show typical signs of cattle with a known history of no previous mannheimia
respiratory disease due to lungworm infection and, at post- vaccination and with low antibody titres (measured in a
mortem examination, there are noticeable, typical lesions in sensitive test system such as EUSA) may be used.
the lungs (e.g. areas of consolidation) and in calves submitted Administer to each animal 1 dose of the vaccine. If the
to post-mortem examination in the later stages, adult worms schedule to be recommended requires a 2nd dose, administer
are presento another dose after an interval of at least 14 days. Observe the
The vaccine complies with the test if the vaccinated calves cattle at least daily for at least 14 days after the last
show no more than very mild respiratory signs after chalIenge administration. Record body temperature the day before
and, at post-mortem examination, there are no or only very vaccination, at vaccination, 2 h, 4 h and 6 h later and then
limited lesions in the lungs (e.g. areas of consolidation) and daily for 4 days; note the maximum temperature increase for
no or very few adult worms are presento each animal.
STORAGE The vaccine complies with the test if no animal shows
When stored under the prescribed conditions the parasites abnormal local or systemic reactions or signs of disease, or
may be expected to survive for 45 days. dies from causes attributable to the vaccine, if the average
body temperature increase for all cattle does not exceed
1.5 oC, and if no animal shows a rise greater than 2.0 oC.
2-2-1-2 Field studies. The cattle used for the field trials are
also used to evaluate safety. Carry out a test in each category
Mannheimia Vaccine (Inactivated) ****
*** * of cattle for which the vaccine is intended. Use not fewer

****
for Cattte than 3 groups of 20 cattle with corresponding groups of not
fewer than 10 controls in 3 different locations. Examine the
(Ph. Eur. monograph 1944)
injection sites for local reactions after vaccination. Record
PhE~ _____________________________________________
body temperatures the day before vaccination, at vaccination
1 DEFINITION and on the 2 days folIowing vaccination.
Mannheimia vaccine (inactivated) for cattle is a preparation The vaccine complies with the test if no animal shows
from cultures of one or more suitable strains of Mannheimia abnormal local or systemic reactions 01' signs of disease, or
haemolytica (formerly Pasteurella haemolytica), inactivated dies from causes attributable to the vaccine. The average
Vet-282 Veterinary Vaccines 2016

body temperature increase for all cattle does not exceed acceptable level of endotoxin is used subsequently for testing
1.5 oC and no animal shows a rise greater than 2.0 oc. of each batch.
In addition, if the vaccine is intended for use in pregnant
3 BATCH TESTS
cows, no significant effects on gestation 01' the offspring are
3-1 Identification
demonstrated.
When injected into healthy animals that do not have specific
2-2-2 Irnmunogenicity antibodies against the serovars of M. haemolytica and/or
Carry out a test for each serovar for which protection is against the leucotoxin present in the vaccine, the vaccine
claimed on the label. stimulates the production of such antibodies.
Each test is carried out for each route and method of 3-2 Bacteria and fungi
administration to be recommended, using in each case cattle The vaccine, including where applicable the diluent supplied
of the minimum age to be recommended for vaccination. for reconstitution, complies with the test for sterility
The vaccine administered to each animal is of minimum prescribed in the monograph Vaccines for veterinaJY use
potency. (0062).
Use not fewer than 16 cattle that do not have antibodies 3-3 Potency
against M. haemolytica and against the leucotoxin of The vaccine complies with the requirements of the test
NI. haemolytica. Vaccinate not fewer than 8 of the cattle
mentioned under Immunogenicity (section 2-2-2) when
according to the schedule to be recommended. Maintain not administered by a recommended route and method.
fewer than 8 cattle as controls. Challenge each animal _____________________________________________ ~E~

20-22 days after the last vaccination by the intratracheal


route or by another appropriate route, with a sufficient
quantity of a low-passage, virulent strain of a serovar of
M. haemolytica. Observe the cattle at least dai1y for a further
7 days; to avoid unnecessary suffering, severely i11 cattle are Mannheimia Vaccine (Inactivated)
euthanised and are then considered to have died from the
disease. During ~e observation period, examine the cattle for tor Sheep
signs of disease (for example, increased body temperature, (Ph. Eur. nzonogmph 1946)
dullness, abnormal breathing) and record the mortality. PhE~ _____________________________________________
Euthanise surviving cattle at the end of the observation
1 DEFINITION
periodo Carry out post-mortem examination on any animal
Mannheimia vaccine (inactivated) for sheep is a preparation
that dies and those euthanised at the end of the observation
of one or more suitable strains of Mannheimia haemolytica
periodo Examine the lungs and evaluate the extent of lung
(former1y Pasteurella haemolytica), inactivated while
lesions due to mannheimiosis. Collect samples of lung tissue
maintaining adequate immunogenic properties. This
for re-isolation of the challenge organisms. Score the clinical
monograph applies to vaccines intended for the active
observations and lung lesions and compare the results
immunisation of sheep and/or for the passive protection of
obtained for these parameters and the bacterial re-isolation
their progeny against disease caused by M. haemolytica.
results for the 2 groups.
The test is not valid if signs of M. haemolytica infection occur 2 PRODUCTION
in less than 70 per cent of the control cattle. The vaccine 2-1 PREPARATION OF THE VACCINE
complies with the test if tl1ere is a significant difference Production of the vaccine is based on a seed-lot system.
between the scores obtained for the c1inical and post-mortem The seed material is cultured in a suitable medium; each
observations in the vaccinates compared to the controls. strain is cultivated separately and identity is verified using a
For vaccines with a claim for a beneficial effect on the extent suitable method. During production, various parameters such
of infection against the serovar, the results for the infection as growth rate are monitored by suitable methods; the values
rates are also significantly better for the vaccinates compared are within the limits approved for the particular product.
to the controls. Purity and identity of the harvest are verified using suitable
methods. After cultivation, the bacterial suspensions are
2-3 MANUFACTURER'S TESTS
collected separately and inactivated by a suitable method.
2-3-1 Batch potency test
The vaccine may be adjuvanted.
It is not neceSSaly to carry out the potency test (section 3-3)
for each batch of vaccine if it has been carried out using a 2-2 CHOICE OF VACCINE COMPOSITION
batch of vaccine with a minimum potency. Where the test is The choice of composition and the strains to be included in
not carried out, an alternative validated method is used, the the vaccine are based on epidemiological data on the
criteria for acceptance being set with reference to a batch of prevalence of the different serovars of M. haemolytica and on
vaccine that has given satisfactory results in the test described the claims being made for the product, for example active
under Potency. and/or passive protection.
2-3-2 Bacteria! endotoxins The vaccine is shown to be satisfactory with respect to safety
A test for bacterial endotoxins (2.6.14) is carried out on the (5.2.6) and efficacy (5.2.7) for the sheep for which it is
final lot or, where the nature of the adjuvant prevents intended.
performance of a satisfactory test, on the bulk antigen or the The following tests for safety (section 2-2-1) and
mixture of bulk antigens immediately before addition of the immunogenicity (section 2-2-2) may be used during the
adjuvant. The maximum acceptable amount of bacterial demonstration 6f safety and efficacy.
endotoxins is that found for a batch of vaccine that has been 2-2-1 Safety
shown satisfactory in safety test 2-2-1-1 given under Choice 2-2-1-1 Laboratory tests. Carry out the tests for each route
of vaccine composition. The method chosen for determining and method of administration to be recommended for
the amount of bacterial endotoxin present in the vaccine vaccination and in sheep of each category for which the
batch used in the safety test for determining the maximum vaccine is intended (for example, young sheep, pregnant
2016 Veterinary Vaccines Vet-283

ewes). U se a batch of vaccine containing not less than the disease. During the observation period, examine the lambs
maximum potency that may be expected in a batch of for signs of disease (for example, increased body temperature,
vaccine. dullness, abnormal respiration) and record the mortality.
For each test, use not fewer than 8 sheep that preferably do Euthanise surviving lambs at the end of the observation
not have antibodies against the serovars of M. haemolytica or periodo Carry out post-mortem examination on any lamb that
against the leucotoxin present in the vaccine. Where justified, dies and those euthanised at the end of the observation
sheep with a known history of no previous mannheimia periodo Examine the lungs and evaluate the extent of lung
vaccination and with low antibody titres (measured in a lesions due to mannheimiosis. Collect samples of lung tissue
sensitive test system such as ELISA) may be used. for re-isolation of the challenge organisms. Score the c1inical
Administer to each sheep 1 dose of the vaccine. If the observations and lung lesions and compare the results
schedule to be recommended requires a 2nd dose, administer obtained for these parameters and the bacterial re-isolation
another dose after an interval of at least 14 days. Observe the results for the 2 groups.
sheep at least daily for at least 14 days after the last The test is not valid if signs of M. haemolytica infection occur
administration. If the test is carried out in pregnant ewes, in less than 70 per cent of the controllambs. The vaccine
observe the ewes until 1 day after lambing. Record body complies with the test if there is a significant difference
temperature the day before vaccination, at vaccination, 2 h, between the scores obtained for the c1inical and post-mortem
4 h and 6 h later and then daily for 4 days; note the observations in the vaccinates compared to the controls.
maximum temperature increase for each sheep. For vaccines with a c1aim for a beneficial effect on the extent
The vaccine complies with the test if: of infection against the serovar, the results for the infection
- no sheep shows abnormal local reactions or notable signs rates are also significantly better for the vaccinates compared
of disease, or dies from causes attributable to the vaccine, to the controls.
- the average body temperatul'e increase for all sheep does 2-2-2-2 Passive protection. For vaccines with c1aims for passive
not exceed 1.5 oC and no sheep shows a rise greater than protection against mannheimiosis carry out a test for each
2.0 oC, and if serovar of M. haemolytica for which protection is to be
- no adverse effects on gestation or the offspring are noted c1aimed on the labe!'
if the test is carried out in pregnant ewes. A test is carried out for each route and method of
2-2-1-2 Field studies. The sheep used for the field trials are administration to be recommended for vaccination.
also used to evaluate safety. Carry out a test in each category The vaccine administered to each ewe is of minimum
of sheep for which the vaccine is intended. Use not fewer potency.
than 3 groups of 20 sheep with corresponding groups of not U se not fewer than 6 ewes that preferably do not have
fewer than 10 controls in 3 different locations. Examine the antibodies against the serovars of NI. hae7110lytica 01' against
injection sites for local reactions after vaccination. Record the leucotoxin present in the vaccine. Where justified, ewes
body temperatures the day before vaccination, at vaccination with a known history of no previous mamilieimia vaccination,
and on the 2 days following vaccination. from a source with a low incidence of respiratory disease and
The vaccine complies with the test if no sheep shows with low antibody titres (measured in a sensitive test system
abnormal local or systemic reactions or notable signs of such as EUSA) may be used. Vaccinate the ewes at the
disease, or dies from causes attributable to the vaccine. stages of pregnancy and according to the schedule to be
The average body temperature increase for all sheep does not recommended. A challenge study is conducted with
exceed 1.5 oC and no sheep shows a rise greater than 2.0 oc. 20 newbom, colostrum-deprived lambs. 10 of these lambs
In addition, if the vaccine is intended for use in pregnant are given colostrum from the vaccinated ewes and 10 control
ewes, no adverse effects on the gestation or offspring are lambs are given colostrum or colostrum substitute without
demonstrated. detectable antibodies to M. haemolytica. When the lambs are
2-2-2 Immunogenicity at the age to be c1aimed for the duration of the passive
2-2-2-1 Active immunisation. For vaccines with c1aims for protection, challenge each by the intratracheal route with a
active irnmunisation against mannheimiosis, carry out a test sufficient quantity of a low-passage, virulent strain of a
for each serovar of .NI. haemolytica for which protection is to serovar of M. haemolytica. Observe the lambs for a further
be c1aimed on the labe!' 7 days; to avoid unnecessary suffering, severely illlambs are
euthanised and are then considered to have died from the
A test is carried out for each route and method of
disease. Observe the lambs and assess the effect of the
administration to be recommended, using in each case lambs
challenge on the offspring of the vaccinates and the controls
of the minimum age to be recommended for vaccination.
as described in the test for active irnmunisation.
The vaccine administered to each lamb is of minimum
potency. The test is not val id if signs or lesions of M. haemolytica
infection occur in less than 70 per cent of the controllambs.
Use not fewer than 20 lambs that do not have antibodies
The vaccine complies with the test if there is a significant
against M. haemolytica and against the leucotoxin of
difference between the scores obtained for the c1inical and
M. haemolytica. Vaccinate not fewer than 10 lambs according
post-mortem observations in the lambs from the vaccinates
to the schedule to be recommended. Maintain not fewer than
comparedto those from the controls. For vaccines with a
10 lambs as controls. 20-22 days after the last vaccination,
claim for a beneficial effect on the extent of infection against
challenge each lamb by the intratracheal route or by another
the serovar, the results for the infection rates are also
appropriate route, with a sufficient quantity of a low-passage,
significantly better for the lambs from the vaccinates
virulent strain of a serovar of M. haemolytica. Where
compared to those from the controls.
necessary for a given serovar, prechallenge with parainfluenza
type 3 (PI3) virus or another appropriate respiratory 2-3 MANUFACTURER'S· TESTS
pathogen may be used. Observe the lambs for a further 2-3-1 Batch potency test
7 days; to avoid unnecessary suffering, severely ill lambs are It is not necessary to carry out the relevant potency test 01'
euthanised and are then considered to have died from the tests (section 3-3) for each batch of vaccine if they have been
Vet-284 Veterinary Vaccines 2016

carried out using a batch of vaccine with a minimum 2-3 SEED LOTS
potency. Where the relevant test or tests are not carried out, 2-3-1 Extraneous agents
an alternative validated method is used, the criteria for The master seed lot complies with the tests for extraneous
acceptance being set with reference to a batch of vaccine that agents in seed lots (2.6.24). In these tests on the master seed
has given satisfactory results in the testes) described under lot, the organisms used are not more than 5 passages from
Potency. the master seed lot at the start of the tests.
2-3-2 Bacterial endotoxins 2-4 CHOICE OF VACCINE VIRUS
A test for bacterial endotoxins (2.6.14) is canied out on the The vaccine virus shall be shown to be satisfactory with
finallot or, where the nature of the adjuvant prevents respect to safety (5.2.6) and efficacy (5.2.7) for the chickens
performance of a satisfactory test, on the bulk antigen or the and/or chicken emblyos for which it is intended.
mixture of bulk antigens immediately before addition of the
The tests shown below for residual pathogenicity of the strain
adjuvant. The maximum acceptable amount of bacterial
(section 2-4-1-1), increase in virulence (section 2-4-2) and
endotoxins is that found for a batch of vaccine that has been
immunogenicity (section 2-4-3) may be used during the
shown satisfactory in safety tests 2-2-1-1 given under Choice
demonstration of safety and efficacy. Additional testing may
of vaccine composition. The method chosen for determining
be needed to demonstrate safety in breeds of chickens known
the amount of bacterial endotoxin present in the vaccine
to be particularly susceptible to Marek' s disease virus, unless
batch used in the safety test for determining the maximum
the vaccine is to be contra-indicated.
acceptable level of endotoxin is used subsequently for testing
of each batch. 2-4-1 Safety
2-4-1-1 Residual pathogenicity of the strain. Carry out the test
3 BATCH TESTS for the route to be recommended for vaccination that is likely
3-1 Identification to be the least safe and in the category of chickens for which
When injected into healthy animal s that do not have specific the vaccine is intended that is likely to be the most
antibodies against the serovars M. haemolytica and/or against susceptible for Marek's disease.
the leucotoxin present in the vaccine, the vaccine stimulates
Carry out the test in chickens if the vaccine is intended for
the production of such antibodies.
chickens; cany out the test in chicken emblyos if the vaccine
3-2 Bacteria and fungi is intended for chicken embryos; carry out the test in
The vaccine, inc1uding where applicable the diluent supplied chickens andin chicken embryos if the vaccine is intended
for reconstitution, complies with the test for sterility for both.
prescribed in the monograph Vaccines fol' veterz'nalY U se vaccine virus at the least attenuated passage level that
use (0062).
will be present between the master seed lot and a batch of
3-3 Potency the vaccine.
The vaccine complies with the requirements of the test or Vaccines intended for use in chickens Use not fewer than 80
testes) mentioned under Immunogenicity (section 2-2-2) l-day-old chickens from a fiotk free from specified pathogens
when administered by a recommended route and method. (SPF) (5.2.2). Divide them randomly into 2 groups of not
_____________________________________________ PhEw fewer than 40 chickens and maintain the groups separately.
Administer by a suitable route to each chicken of one
group (1) a quantity of the vaccine virus equivalent to not
less than 10 times the maximum virus titre likely to be
*** contained in 1 dose of the vaccine. Administer by a suitable
Marek's Disease Vaccine, Living
*** *** route to each chicken of the other group (TI) a quantity of
virulent Marek's disease virus that will cause mortality andlor
Marek's Disease Vaccine (Turkey Herpes Virus) *** severe macroscopic lesions of Marek's disease in not fewer
Marek's Disease Vaccine, Living (HVT)
than 70 per cent of the effective number of chickens within
(Marek's Disease Vaccine (Live), Ph Eur ,1110nograph 0589) 70 days (initial number reduced by the number that die
~Ew _____________________________________________ within the first 7 days of the test).
1 DEFINITION Vaccines intendedfor use in chicken emblyos Use not fewer than
Marek's disease vaccine (live) is a preparation of a suitable 150 embryonated eggs from an SPF fiock (5.2.2). Divide
strain or strains of Marek's disease virus (gallid herpesvirus them randomly into 3 groups of not fewer than
2 or 3) andlor turkey herpesvirus (meleagrid herpesvirus 1). 50 embryonated eggs and maintain the groups separately but
This monograph applies to vaccines intended for under identical incubation conditions. Not later than the
administration to chickens and/or chicken embryos for active recommended day of vaccination, administer by the method
immunisation. to be recommended to each embryonated egg of one
group (1) a quantity of the vaccine virus equivalent to not
2 PRODUCTION less than 10 times the maximum virus titre likely to be
2-1 PREPARATION OF THE VACCINE contained in 1 dose of the vaccine. Administer by a suitable
The vaccine virus is grown in cell cultures. If the vaccine route to each embryonated egg of another group (TI) a
contains more than one type of virus, the different types are quantity of virulent Marek's disease virus that will cause
grown separately. The vaccine may be freeze-dried or stored mortality and/or severe macroscopic lesions of Marek's
in liquid nitro gen. disease in not fewer than 70 per cent of the effective number
2-2 SUBSTRATE FOR VIRUS PROPAGATION of hatched chickens within 70 days (initial number reduced
2-2-1 Cell cultures by the number that die within the first 7 days after hatching).
The cell cultures comply with the requirements for cell Keep the last group (TII) non-inoculated. The test is not
cultures for production of veterinary vaccines (5.2.4). valid if there is a significant difference in hatchability between
groups 1 and III and the hatchability in any of the 3 groups is
less than 80 per cent.
2016 Veterinary Vaccines Vet-285

Provided that the chickens and chicken embryos are derived virus administered to each chicleen or chicleen embryo is not
from the same flock, a common control group for in ovo and greater than the minimum virus titre to be stated on the label
parenteral administ:ration can be used. and the virus is at the most attenuated passage level that will
IlTespective of whether the vaccine was administered to be present in a batch of the vaccine.
chickens or chicken embryos, observe the chickens of Vaccines intended for use in chickens U se not fewer than
group II at least daily for 70 days and those of group 1 at 60 chickens of the same origin and from an SPF flock
least daily for 120 days. (5.2.2). Vaccinate by a route to be recommended not fewer
The test is not valid if one or more of the following apply: than 30 chickens. Maintain not fewer than 30 chickens as
- more than 10 per cent of the chickens in any of the controls.
3 groups die within the first 7 days; Vaccines intended f01" use in chicken embl)1os U se embryonated
- fewer than 70 per cent of the effective number of chickens of the same origin and from an SPF flock (5.2.2).
chickens in group II show macroscopic lesions of Marek's Vaccinate by the in ovo route using the method to be
disease; recommended, 50 per cent of the embryonated eggs.
The vaccine virus complies with the test if: Maintain 50 per cent of the embryonated eggs as controls.
- no chicleen of group 1 shows notable clinical signs or The test is not valid if any group consists of fewer than
macroscopic lesions of Marek's disease or dies from 30 hatched chicles.
causes attributable to the vaccine virus; IlTespective of whether the vaccine was administered to
- at 120 days the number of surviving chickens of group 1 chickens or chicleen embryos, challenge each chicken not
is not fewer than 80 per cent of the effective number. later than 9 days after vaccination by a suitable route with a
2-4-2 Increase in virulence sufficient quantity of virulent Marek' s disease virus. Observe
The test for increase in virulence is required for Marek' s the chickens at least daily for 70 days after challenge. Record
disease virus vaccine strains but not for turkey herpesvirus the deaths and the number of surviving chickens that show
vaccine strains, which are naturally apathogenic. clinical signs of disease. At the end of the observation period,
euthanise all the surviving chickens and carry out an
Carry out the test according to general chapter 5.2.6.
examination for macroscopic lesions of Marek's disease.
Vaccines intended for use in chickens Administer to each
The test is not valid if:
1-day-old SPF chicleen (5.2.2) by the intramuscular route a
during the observation period after challenge, fewer than
quantity of the vaccine virus that will allow recovery of virus
70 per cent of the control chickens die or show severe
for the passages described below.
clinical signs or macroscopic lesions of Marek's disease;
Vaccines intended for use onl)' in chicken e11lblJ1oS 01" intended for and/or, during the period between the vaccination and
use in chickens and in chicken embl)10s Administer to each challenge, more than 10 per cent of the control or
embryonated egg not later than the recommended day for vaccinated chickens show abnormal clinical signs or die
vaccination by the in ovo route, using the recommended from causes not attributable to the vaccine.
method, a quantity of the vaccine virus that will allow
The vaccine virus complies widl dle test if the relative
recovery of virus for the passages described below.
pl'otection percentage, calculated using the following
If the properties of the vaccine virus allow sequential passage expression, is not less than 80 per cent:
through 5 groups via natural spreading, this method may be
used, otherwise passage as described below is carried out.
5-7 days after administering the vaccine to chickens or
v-e x 100
5-7 days after hatching when the vaccine has been
e
100 -
administered in ovo, prepare a suspension of white blood cells
V percentage of challenged vaccinated chickens that
from each chicleen and pool these samples. Administer a
survive to the end of the observation period without
suitable volume of the pooled samples by the intraperitoneal
notable clinical signs or macroscopic lesions of
route to each 1-day-old SPF chicleen (5.2.2) of the next
Marek's disease;
group. Carry out this passage operation not fewer than
4 times; verify the presence of the virus at each passage.
e pel'centage of challenged control chickens that
survive to the end of the observation period without
If the virus is not found at a passage level, repeat the passage
notable clinical signs or macl'oscopic lesions of
by administration to a group of 10 animals. Carry out the
Mal'ek's disease.
test for residual pathogenicity (section 2-4-1-1) using the
material used for the 1st passage and the virus at the final 3 BATCH TESTS
passage level. Administer the virus by the route to be 3-1 Identification
recommended for vaccination that is likely to be the least safe Can-y out an immunostaining test in cell cultures using
for use in these chickens or chicleen embryos. monoclonal antibodies to demonstl'ate the presence of each
The vaccine virus complies with the test if no indication of type of virus stated on the label.
increase in virulence of the virus recovered for the final 3-2 Bacteria and fungi
passage compared with the material used for the 1st passage The vaccine, including where applicable the diluent supplied
is observed. If virus is not recovered after an initial passage in for reconstitution, camplies with the test fol' sterility
5 chickens or chicleen embryos and a subsequent repeat prescribed in the monograph Vaccines for veterinaJ)I
passage in 10 chickens or chicleen embryos, the vaccine virus use (0062).
also complies with the test.
3~3 Mycop1asmas
2-4-3 Irnmunogenicity The vaccine complies with the test for mycoplasmas (2.6.7).
A test is carried out for each route and method of
3-4 Extraneous agents
administration to be recommended, using in each case
The vaccine complies with the tests for extraneous agents in
chickens of the minimum age to be recommended for
batches of finished product (2.6.25).
vaccination or chicleen embryos. The quantity of the vaccine
Vet-286 Veterinary Vaccines 2016

3-5 Virus titre 2-2-1 Safety


3-5-1 Vaccines containing one t)'pe 01 virus. Titrate the vaccine The test is carried out for each route of administration to be
virus by inoculation into suitable cell cultures (5.2.4). If the recommended for vaccination and for each avian species for
virus titre is detennined in plaque-fonning units (PFU), only which the vaccine is intended. U se a batch of vaccine
primary plaques are taken into consideration. The vaccine containing not less than the maximum potency that may be
complies with the test if one dose contains not les s than the expected in a batch of vaccine.
minimum virus titre stated on the label. For each test performed in birds younger than 3 weeks of
3-5-2 Vaccines containing more than one t)'pe 01 virus. age, use not fewer than 10 birds not older than the minimum
For vaccines containing more than one type of virus, titrate age to be recommended for vaccination. For each test
each virus by inoculation into suitable cell cultures (5.2.4), perfonned in birds older than 3 weeks of age, use not fewer
reading the results by irnmunostaining using antibodies. than 8 birds not older than the minimum age to be
The vaccine complies with the test if one dose contains for recommended for vaccination. In the case of chickens, use
each vaccine virus not less than the minimum virus titre chickens from a fiod: free from specified pathogens (SPF)
stated on the label. (5.2.2) and in the case of turkeys, use birds that have not
3-6 Potency been vaccinated and that do not have antibodies against M.
The vaccine complies with the test for irnmunogenicity gallisepticum. Administer by a route and method to be
(section 2-4-3) when administered according to the recommended to each bird 1 dose of vaccine. If the schedule
recommended schedule by a recommended route and to be recommended requires a 2nd dose, administer 1 dose to
method. It is not necessary to carry out the potency test for each bird after an interval of at least 14 days. Observe the
each batch of the vaccine if it has been carried out on a birds at least daily for at least 14 days after the last
representative batch using a vaccinating dose containing not administration of the vaccine.
more than the minimum virus titre stated on the label. The test is not valid if more than 10 per cent of the birds
_____________________________________________ PhEw younger than 3 weeks of age show abnormal signs of disease
01' die from causes not attributable to the vaccine. For birds
older than 3 weeks of age, the test is not valid if non-specific
mortality occurs.
The vaccine complies with the test if no bird shows abnonnal
Mycoplasma Gallisepticum ***
*** ***
signs of disease 01' dies from causes attributable to the
vaccme.
Vaccine (Inactivated) ***
(Ph Bur monograph 1942) 2-2-2 Immunogenicity
The test is carried out for each recommended route of
PhEw _____________________________________________
administration and for each avian species for which the
1 DEFINITION vaccine is intended. U se for each test not fewer than 40 birds
Mycoplasma gallisepticum vaccine (inactivated) is a not older than the minimum age to be recommended for
preparation of one or more suitable strains of Mycoplasma vaccination. Use chickens from an SPF fiock (5.2.2) 01~
gallisepticuJ1l that have been inactivated while maintaining turkeys that have not been vaccinated and are free from
adequate immunogenic properties. This monograph applies antibodies against M. gallisepticum. For each test, administer
to vaccines intended for the active irnmunisation of chickens to each of not fewer than 20 birds a quantity of the vaccine
and/or turkeys. not greater than a single dose. If re-vaccination is
recommended, repeat this operation after the recommended
2 PRODUCTION
interval. Maintain not fewer than 20 birds as controls.
2-1 PREPARATION OF THE VACCINE
Challenge each bird from both groups not more than 28 days
Production of the vaccine is based on a seed-lot system. after the last administration by a suitable route with a
The seed material is cultured in a suitable solid and/or liquid sufficient quantity of virulent M. gallisepticum (R-strain).
medium to ensure optimal growth under the chosen Observe the birds at least daily for 14 days after challenge.
incubation conditions. Each strain is cultivated separately and Evaluation is carried out 14 days after challenge, at which
identity is verified using a suitable method. During point the birds are euthanised. Record the deaths and the
production, various parameters such as growth rate are number of surviving birds that show clinical signs of disease
monitored by suitable methods; the values are within the (e.g. respiratory distress, nasal discharge), and record air sac
limits approved for the particular vaccine. Purity and identity lesions.
of the harvest are verified using suitable methods. After
The test is not valid if:
cultivation, the mycoplasma suspensions are collected
-- during the observation period after challenge, fewer than
separately and inactivated by a suitable method.
70 per cent of the controls die 01' show lesions 01' clinical
The mycoplasma suspensions may be treated to fragment the
signs of disease; and/or
mycoplasmas and the fragments may be purified and
during the period between vaccination and challenge,
concentrated. The vaccine may contain an adjuvant.
more than 10 per cent of the birds from the control
2-2 CHOICE OF VACCINE COMPOSITION group 01' from the vaccinated group show abnonnal
The vaccine is shown to be satisfactory with respect to safety clinical signs of disease 01' die from causes not attributable
(5.2.6) and efficacy (5.2.7) in the target animals. to the vaccine.
The following tests for safety (section 2-2-1) and Thoracic and abdominal air sacs are evaluated individually
irnmunogenicity (section 2-2-2) may be used during the on each side of the animal. The scoring system presented
demonstration of safety and efficacy. If the indications for the below may be used. The vaccine complies with the test if the
vaccine include protection against a drop in laying score for the vaccinated birds is significantly lower than that
perfonnance 01' protection against infectious sinusitis in for the controls and if the reduction is not less than
turkeys, further suitable irnmunogenicity testing is necessary. 30 per cent.
2016 Veterinary Vaccines Vet-287

o no air sac lesions ***


Myxomatosis Vaccine (Live) for
in a limited area of 1 or 2 air sacs: cloudiness with slight *** ***
thickening of the air sac membrane or fiecks of yellowish Rabbits ***
exudate (Ph Eur monograph 1943)
2 in 1 air sac or portions of 2 air sacs: greyish or yellow, PhE~ _____________________________________________
sometimes foamy exudate, with thickening of the air sac
membrane 1 DEFINITION
3 in 3 air sacs: extensive exuda te, with clear thickening of Myxomatosis vaccine (live) for rabbits is a preparation of a
most air sacs suitable strain of either myxoma virus that is attenuated for
4 severe air-sacculitis with considerable exudate and rabbits or Shope fibroma virus. This monograph applies to
thickening of most air sacs. vaccines intended for the active immunisation of rabbits
against myxomatosis.
2-3 MANUFACTURER'S TESTS
2-3-1 Batch potency test 2 PRODUCTION
It is not necessary to carry out the potency test (section 3-4) 2-1 PREPARATION OF THE VACCINE
for each batch of the vaccine if it has been carried out using The vaccine virus is grown in cell cultures. The viral
a batch of vaccine with a minimum potency. Where the test suspension is harvested and titrated and may be mixed with a
is not carried out on a batch, an alternative validated method suitable stabilising solution.
is used, the criteria for acceptance being set with reference to 2-2 SUBSTRATE FOR VIRUS PROPAGATION
a batch of vaccine that has given satisfactory results in the 2-2-1 Cell cultures
potency test (section 3-4). The following test may be used. The cell cultures comply with the requirements for cell
U se not fewer than 15 chickens, 3-4 weeks old, from an SPF cultures for production of veterinary vaccines (5.2.4).
fiod::: (5.2.2) or not fewer than 15 turkeys, 3-4 weeks old,
that have not been vaccinated against M. gallisepticum, do not 2-3 CHOICE OF VACCINE VIRUS
have antibodies against M. gallisepticum, and are obtained The vaccine virus is shown to be satisfactory with respect to
from a healthy fiod::. Collect serum samples from each safety (5.2.6) and efficacy (5.2.7) for the rabbits for which it
vaccinate and control bird just before vaccination and check is intended.
for the absence of antibodies against M. gallisepticum. The following tests for safety (section 2-3-1), increase in
Administer to each of not fewer than 10 birds 1 dose of the virulence (section 2-3-2) and immunogenicity (2-3-3) may be
vaccine by a recommended route. Maintain not fewer than 5 used during the demonstration of safety and efficacy.
birds as controls. Collect serum samples 5 weeks after 2-3-1 Safety
vaccination from each vaccinated and control bird. Measure Carry out the test for each route and method of
the titres of serum antibodies against M. gallisepticUl1Z using a administration to be recommended for vaccination, using in
suitable method. Calculate the mean titres for the group of each case rabbits of the minimum age to be recommended
vaccinates. The test is not valid if specific M. gallisepticum for vaccination. Use vaccine virus at the least attenuated
antibodies are found in any serum samples from the control passage leve! that will be present in a batch of the vaccine.
birds 5 weeks after the time of administration of the vaccine. For each test, use not fewer than 8 rabbits that do not have
The vaccine complies with the test if the mean antibody antibodies against myxoma virus. Administer to each rabbit a
titres of the group of vaccinates are equal to or greater than quantity of the vaccine virus equivalent to not less than
the titres obtained with a batch that has given satisfactory 10 times the maximum virus titre likely to be contained in
results in the potency test (section 3-4). 1 dose of the vaccine. Observe the rabbits at least daily for
3 BATCH TESTS 28 days. Record the body temperature the day before
3-1 Identification vaccination, at vaccination, 4 h after vaccination and then
When injected into chickens from an SPF fiod:: (5.2.2) or daily for 4 days; note the maximum temperature increase for
turkeys from healthy fiocks, the vaccine stimulates the each rabbit.
production of antibodies against one or more strains of The vaccine virus complies with the test if no l'abbit shows
M. gallisepticw1Z. notable signs of disease or dies from causes attributable to
3-2 Bacteria and fungi the vaccine virus; the average temperature incl'ease do es not
The vaccine, including where applicable the diluent supplied exceed 1.0 oC and no l'abbit shows a rise greatel' than 2.0 oC.
for reconstitution, complies with the test for sterility A local reaction lasting less than 28 days may occur.
prescribed in the monograph Vaccines for veterinary use 2-3-2 Increase in virulence
(0062). (This test is pe¡jonned only for vaccines based on attenuated
3-3 Residual1ive mycoplasmas strains of myxoma virus). Carry out the test according to
A test for residual live mycoplasmas is carried out to confirm general chapter 5.2.6. Evaluation of safety of veterina¡J' vaccines
inactivation of M. gallisepticum. The vaccine complies with a and immunosera, using rabbits 5-7 weeks old that do not have
validated test for residual live M. gallisepticu111 carried out by antibodies against myxoma virus. If the properties of the
a culture method (see for example 2.6.7, using media shown vaccine virus allow sequential passage through 5 groups via
to be suitable for M. gallisepticum). natural spieading, tlÍis method may be used, otherwise
passage as described below is cal'ried out.
3-4 Potency
The vaccine complies with the test for immunogenicity Administer to each l'abbit by a route to be recommended a
(section 2-2-2). quantity of the vaccine virus that will allow l'ecovery of virus
_____________________________________________ PhE~
for the passages described below. Administer the virus by the
route to be recommended fol' vaccination most likely to lead
to reversion ofvirulence. Euthanise the l'abbits 5-10 days
after inoculation and l'emove from each rabbit organs or
tissues with sufficient virus. to allow passage; homogenise the
Vet-288 Veterinary Vaccines 2016

organs and tissues in a suitable buffer solution, centrifuge the have antibodies against myxoma virus and rabbit
suspension and use the supernatant for further passages. haemorrhagic disease virus and that have been reared in
Inoculate the supernatant into suitable cell culture to verify suitable isolation conditions to avoid contact with myxoma
the presence of virus. Adrninister by an appropriate route, at virus. Adrninister to each rabbit by a recommended route
a suitable rate, a suitable volume of the supernatant to each 10 doses of the vaccine. Observe the rabbits at least daily for
rabbit of the next group. Carry out this passage operation not 14 days. At the end of the observation period adrninister by a
fewer than 4 times; verify the presence of the virus at each recommended route to each rabbit a further 10 dos es of
passage. If the virus is not found at a passage level, repeat the vaccine. After 14 days take a blood sample from each rabbit
passage by adrninistration to a group of 10 animals. and carry out a test for antibodies against rabbit
If the 5th group of animal s shows no evidence of an increase haemorrhagic disease virus. The vaccine complies with the
in virulence indicative of reversion during the observation test if no antibodies are found.
period, further testing is not required. Otherwise, carry out 3-5 Virus titre
an additional safety test and compare the c1inical signs and Titrate the vaccine virus in suitable cell cultures. The vaccine
any relevant parameters in a group of at least 8 animals complies with the test if 1 dose contains not les s than the
receiving the material used for the 1sr passage and another minimum virus titre stated on the label.
similar group receiving the virus at the final passage level. 3-6 Potency
The vaccine virus complies with the test if no indication of The vaccine complies with the requirements of the test
increased virulence of the virus recovered for the final prescribed under Irnrnunogenicity (section 2-3-3) when
passage compared with the material used for the 1sr passage adrninistered by a recommended route and method. It is not
is observed. If virus is not recovered after an initial passage in necessary to carry out the potency test for each batch of the
2 animals and a. subsequent repeat passage in 10 animals , vaccine if it has been carried out on a representative batch
the vaccine virus also complies with the test. using a vaccinating dose containing not more than the
2-3-3 Immunogenicity minimum virus titre stated on the label.
A test is carried out for each route and method of _____________________________________________ ~E~

adrninistration to be recommended for vaccination using in


each case rabbits of the minimum age to be recornrnended.
The quantity of vaccine virus to be adrninistered to each
rabbit is not greater than the minimum virus titre to be
stated on the label and the virus is at the most attenuated Newcastle Disease and Avian Infectious
passage level that will be present in a batch of vaccine.
Bronchitis Vaccine, Living
U se for the test not fewer than 15 rabbits that do not have
The use oj Newcastle Disease and Avian Injectious Bronchitis
antibodies against myxoma virus and are reared in suitable
Vaccine Living is restricted in the United Kingd0111 by the
isolation conditions to ensure absence of contact with
Department jor Envimnment, Food and Rural Affairs.
myxoma virus. Adrninister 1 dose of vaccine to each of not
fewer than 10 of the rabbits according to the schedule to be DEFINITION
recommended. Maintain not fewer than 5 rabbits as controls. Newcastle Disease and Avian Infectious Bronchitis Vaccine,
Challenge each rabbit not less than 21 days after the last Living is a mixed preparation derived from separate groups
vaccination by a suitable route with a quantity of a virulent of eggs infected with suitable strains of Newcastle disease
strain of myxoma virus sufficient to cause typical signs of virus and of avian infectious bronchitis virus. The Newcastle
myxomatosis in a rabbit. Observe the rabbits at least daily for disease virus seed is either a modified strain, such as
a further 21 days after challenge and monitor each of them. Hitchner B 1 or La Sota, or a naturally occurring strain of
The test is not valid if fewer than 90 per cent of the control low pathogenicity. The avian infectious bronchitis virus seed
rabbits display typical signs of myxomatosis. A vaccine is a strain of the Massachusetts type 1 and it may be used at
containing myxoma virus complies with the test if, during the various levels of attenuation. The vaccine is prepared
observation period after challenge, not fewer than 90 per cent irnrnediately before use by reconstitution from the dried
of vaccinated rabbits show no signs of myxomatosis. vaccine with a suitable liquido
A vaccine containing Shope fibroma virus complies with the PRODUCTION
test if, during the observation period after challenge, not For vaccine production the virus is propagated in
fewer than 75 per cent of vaccinated rabbits show no signs of embryonated eggs derived from chicken fiocks free from
myxomatosis. specified pathogens. The final product is freeze dried.
3 BATCH TESTS Provided that the test for potency described below has been
3-1 Identification perforrned with satisfactory results on a representative batch
Carry out an immunofiuorescence test in suitable cell of vaccine it may be omitted by the manufacturer as a
cultures, using a monospecific antiserum. routine control on other batches of vaccine prepared from
3-2 Bacteria and fungi the same seed lot, subject to the agreement of the competent
The vaccine, inc1uding where applicable the diluent supplied authority.
for reconstitution, complies with the test for sterility CAUTION The vaccine is not dangerous to man, but it is
prescribed in the monograph Vaccines jor veterinalY use advisable to avoid undue exposure to the Newcastle disease virus
(0062). and to injected birds, as the Newcastle disease virus may cause a
type oj conjuctivitls or, more rarely, 111ay cause sympt0111s similar
3-3 Mycoplasmas (2.6.7)
to those charactenstic oj influenza.
The vaccine complies with the test for mycoplasmas.
The vaccine, reconstituted with a suitable liquid to provide a
3-4 Specified extraneous agents
concentration appropriate to the particular test, complies with the
Use not fewer than 2 rabbits that are not older than the
require11'lents stated under VeterinalY Vaccines with the jollowing
minimum age recommended for vaccination, that do not
modijications.
2016 Veterinary Vaccines Vet-289

IDENTIFICATION Between the 4th and 7th day after challenge, take a tracheal
When mixed with a mixture of monospecific Newcastle swab from each bird. Place each swab in a test tube
disease virus antiserum and avian infectious bronchitis virus containing 3 mL of broth to which suitable antibiotics have
antiserum, the vaccine no longer infects susceptible been added to inhibit the growth of bacterial contaminants
10- to 11-day-old embryonated eggs. and test for the presence of infectious bronchitis virus by the
inoculation of 0.2 mL of inoculum into the allantoic cavity of
TESTS
9- to 11-day-old emblyonated eggs using at least five eggs for
Mycop1asmas
each swab. A tracheal swab is positive if 20% or more of the
Complies with the test for absence of mycoplasmas,
embryos inoculated from it show lesions typical of infectious
Appendix XVI B(Vet) 3.
bronchitis virus. If more than one embryo but fewer than
Sterility 20% of those inoculated from any one swab show lesions
Carry out the test described under Veterinary Vaccines using similar to those of infectious bronchitis, inoculate at least five
solid media in place of liquid media. The vaccine contains no additional embryonated eggs with allantoic fluid from each of
pathogenic organisms and not more than one organism of a the suspect embryos. The swab is positive if 20% 01' more of
non-pathogenic species per bird dose. these additional embryos show lesions typical of infectious
Absence of extraneous pathogens bronchitis virus.
The neutralised vaccine complies with the test for A vian Live Not les s than 80% of the control birds give positive tracheal
Virus Vaccines: Tests for Bxtraneous Agents in Batches of swabs, and no more than 20% of the vaccinated chickens
Finished ProductJ Appendix XVI B(Vet) 5. give positive tracheal swabs.
Safety STORAGE
Inoculate each of 10 chicks of the minimum age for When stored under the prescribed conditions the dried
vaccination from a flock free from specified pathogens with vaccine may be expected to retain its potency for not less
10 dos es of the vaccine by intranasal instillation and observe than 12 months. The reconstituted vaccine should be used
for 14 days. No abnormal reaction develops. immediately.
Virus titre LABELLING
For the Newcastle disease component Neutralise the vaccine
The label states the names of the strains of virus used in the
with monospecific avian infectious bronchitis virus antiserum.
vaccine.
Inoculate serial dilutions of neutralised vaccine into the
allantoic cavity of 10- to 11-day-old embryonated eggs 1 Vaccine prepared using avian infectious bronchitis virus seed of the
derived fram chicken flocks free from specified pathogens. Connecticut type rnay be used in sorne countries but is not perrnitted
Incubate the eggs for 3 days at 37° and then examine the in the United Kingdorn.
embryos for evidence of virus infection, which is shown by
the presence of chick red cell haemagglutinins. The vaccine
contains not less than 10 6 . 0 EIDso of virus per bird dose.
For the avian infectious brol1chitis component Neutralise the ***
Newcastle Disease Vaccine,
vaccine with monospecific Newcastle disease virus antiserum.
*** ***
Inoculate serial dilutions of neutralised vaccine into the Inactivated ***
allantoic cavity of 10- to 11-day-old emblyonated eggs
(Newcastle Disease Vaccine (Inactivated),
derived from chicken flocks free from specified pathogens.
Ph Bur monograph 0870)
Incubate the eggs at 37° for 7 days and then examine tlle
PhEw _____________________________________________
embryos for lesions typical of avian infectious bronchitis.
The vaccine contains not les s than 10 3 .5 EIDso of virus per 1 DEFINITION
bird dose. Newcastle disease vaccine (inactivated) (also known as avian
POTENCY paramyxovirus 1 vaccine (inactivated) for vaccines intended
For the Newcasde disease component for sorne species) is a preparation of a suitable strain of
Vaccinate each of twenty-five 5- to 10-day-old chicks from Newcastle disease virus (avian paramyxovirus 1), inactivated
flocks free from specified pathogens, by the nasal instillation while maintaining adequate immunogenic properties. This
of one dose of vaccine. Twenty-one days later challenge the monograph applies to vaccines intended for active
vaccinated chicks as well as 10 control birds by the immunisation of birds against Newcastle disease.
intramuscular inoculation of at least 10 6 . 0 EIDso of Herts 2 PRODUCTION
(Weybridge 33/56) strain of Newcastle disease virus and 2-1 PREPARATION OF THE VACCINE
observe for 10 days. All the control birds die within 6 days The vaccine virus is grown in embryonated hens' eggs or in
and no fewer than 23 of the vaccinated birds survive the cell cultures. The virus harvest is inactivated. The vaccine
observation period without showing signs of Newcastle may be adjuvanted.
disease.
2-2 SUBSTRATE FOR VIRUS PROPAGATION
For the avian infectious bronchitis component
2-2-1 Embryonated hens' eggs
Vaccinate each of 10 healthy 3- to 4-week-old chickens from
If the vaccine virus is grown in embryonated hens' eggs, they
flocks free from specified pathogens, by nasal 01' ocular
are obtained from healthy flocks.
instillation such that each chicken receives one dose of
vaccine. Twenty-one to twenty-eight days later challenge the 2-2-2 Cell cultures
vaccinated chickens, as well as 10 control birds that are kept If the vaccine virus is grown in cell cultures, they comply
separate from the vaccinated birds, by nasal 01' ocular with the requirements for éell cultures for production of
instillation of 103 .0 to 10 3 .5. EIDso of the Massachusetts veterinary vaccines (5.2.4).
41 strain of virulent infectious bronchitis virus.
Vet-290 Veterinary Vaccines 2016

2-3 SEED LOTS vaccinated group without showing any signs of Newcastle
2-3-1 Extraneous agents disease during the 21 days.
The master seed lot complies with the test for extraneous The test is invalid unless all the control birds die within
agents in seed lots (2.6.24). In these tests on the master seed 6 days of challenge.
lot, the organisms used are not more than 5 passages from The vaccine complies with the test if the smallest dose stated
the master seed lot at the start of the test. on the label corresponds to not less than 50 PD so and the
2-4 CHOICE OF VACCINE COMPOSITION lower confidence limit is not les s than 35 PD so per dose.
The vaccine is shown to be satisfactory with respect to safety If the lower confidence limit is less than 35 PD so per dose,
(5.2.6) and efficacy (5.2.7) for each species and category of repeat the test; the vaccine must be shown to contain not les s
birds for which it is intended. than 50 PD so in the repeat test.
The following tests for safety (section 2-4-1) and 2-4-2-2 Vaccines for use in species other than the chicken.
irnmunogenicity (section 2-4-2) may be used during the U se not fewer than 30 birds of the target species, of the same
demonstration of safety and efficacy. origin and of the same age, that do not have antibodies
2-4-1 Safety against avian paramyxovirus 1. Vaccinate in accordance with
The test is carried out for each route of administration to be the recommendations for use not fewer than 20 birds.
recommended for vaccination and for each avian species for Maintain not fewer than 10 birds as controls. Challenge each
which the vaccine is intended. U se a batch of vaccine bird after 4 weeks by the intramuscular route with a
containing not less than the maximum potency that may be sufficient quantity of virulent avian paramyxovirus 1.
expected in a batch of vaccine. The test is not valid if serum samples obtained at the time of
For each test performed in birds younger than 3 weeks of the first vaccination show the presence of antibodies against
age, use not fewer than 10 birds not older than the minimum avian paramyxovirus 1 in either vaccinates or control s, or if
age to be recommended for vaccination. For each test tests carried out at the time of challenge show such
performed in birds older than 3 weeks ofage, use not fewer antibodies in controls.
than 8 birds not older than the minimum age to be The test is not valid if fewer than 70 per cent of the control
recommended for vaccination. In the case of chickens, use birds die or show serious signs of Newcastle disease.
chickens from a flock free from specified pathogens (SPF) The vaccine complies with the test if not fewer than
(5.2.2) and if the vaccine is used for species other than 90 per cent of the vaccinated birds survive and show no
chickens, they have not been vaccinated and do not have serious signs of avian paramyxovirus 1 infection.
antibodies against N ewcastle disease virus. Administer by a 2-5 MANUFACTURER'S TESTS
route and method to be recommended to each bird 1 dose of 2-5-1 Residuallive virus
vaccine. Observe the birds at least daily for at least 14 days The test is carried out in embryonated eggs or suitab1e cell
after the last administration of the vaccine. cultures (5.2.4), whichever is the most sensitive for the
The test is not val id if more than 10 per cent of the birds vaccine strain. The quantity of inactivated virus harvest used
younger than 3 weeks of age show abnormal signs of disease in the test is equivalent to not less than 10 dos es of vaccine.
or die from causes not attributable to the vaccine. For birds The vaccine complies with the test if no live virus is detected.
older than 3 weeks of age, the test is not valid if non-specific 2-5-2 Batch potency test
mortality occurS. It is not necessmy to carry out the potency test (section 3-5)
The vaccine complies Witl1 the test if no bird shows abnormal for each batch of vaccine if it has been carried out using a
signs of disease or dies from causes attributable to the vaccine. batch of vaccine with a minimum potency. Where the test is
2-4-2 Irnrnunogenicity not carried out, an alternative validated method is used, the
A test is carried out for each route and method of criteria for acceptance being set with reference to a batch of
administration to be recommended; the vaccine administered vaccine that has given satisfact01Y results in the test described
to each bird is of minimum potency. under Potency. The following tests may be used. Wherever
For chickens, the test for vaccines for use in chickens possible, carry out the test for antigen content (section 2-5-
(section 2-4-2-1) is suitable for demonstrating 2-1) together with the test for adjuvant (section 2-5-2-2).
immunogenicity. For other species of birds (for example, Vaccines for use in chickens The test for antigen content
pigeons or turkeys), the test for vaccines for use in species (section 2-5-2-1) together with the test for adjuvant
other than the chicken (section 2-4-2-2) is suitable for (section 2-5-2-2) may be carried out; if the nature of the
demonstrating immunogenicity. product does not allow valid results to be obtained with these
2-4-2-1 Vaccines for use in chickens. Use not fewer than tests, or if the vaccine does not comply, the test for
70 chickens, 21-28 days old, ofthe same origin and from an serological assay (section 2-5-2-3) may be carried out. If the
SPF flock (5.2.2). For vaccination, use not fewer than vaccine does not comply with the latter test, the test for
3 groups, each of not fewer than 20 chickens. Choose a vaccines for use in chickens (section 2-4-2-1) may be carried
number of different volumes of the vaccine corresponding to out. A test using fewer than 20 birds per group and a shorter
the number of groups: for example, volumes equivalent to observation period after challenge may be used if this has
1/25, 1/50 and 1/100 of adose. A1locate a different volume been shown to give a valid potency test.
to each vaccination group. Vaccinate each chicken by the Vaccines for use in species other than the chicken Cany out a
intramuscular route with the volume of vaccine allocated to suitable test for which a satisfactory correlation has been
its group. Maintain not fewer than 10 chickens as controls. established with the test for vaccines for use in species other
Challenge each chicken after 17-21 days by the intramuscular than the chicken (section 2-4-2-2), the criteria for acceptance
route with 6 10gIO embryo LDso of the Herts (Weybridge being set with reference to a batch that has given satisfact01Y
33/56) strain of avian paramyxovirus 1. Observe the chickens results in the latter test. A test in chickens from an SPF flock
at least daily for 21 days after challenge. At the end ofthe (5.2.2) consisting of a measure of the serological response to
observation period, ca1culate the PD so by standard statistical graded amounts ofvaccine (for example, 1/25, 1/50 and
methods from the number of chickens that survive in each 1/100 of adose with serum sampling 17-21 days later) may
2016 Veterinary Vaccines Vet-291

be used. Altematively, the test for antigen content 3-3 Specified extraneous agents
(section 2-5-2-1) together with the test for adjuvant Use 10 chickens, 14-28 days old, from an SPF flock (5.2.2).
(section 2-5-2-2) may be conducted if shown to provide a Vaccinate each chicken by a recommended route with a
valid potency test. double dose of the vaccine. After 3 weeks, administer 1 dose
2-5-2-1 Antigen contento The relative antigen content is by the same route. Collect serum samples from each chicken
determined by comparing the content ofhaemagglutinin- 2 weeks later and carry out tests for antibodies to the
neuraminidase antigen per dose of vaccine with a following agents by the methods prescribed in general
haemagglutinin-neuraminidase antigen reference preparation, chapter 5.2.2. Chicken fiocks free fr0111 specified pathogens for the
by enzyme-linked irnmunosorbent assay (2.7.1). For this production and quality control of vaccines: avian
comparison, Nezucastle disease virus reference antigen BRP, encephalomyelitis virus, avian infectious bronchitis virus,
Nezucastle disease virus control antigen BRP, Nezucastle disease avian leucosis viruses, egg-drop syndrome virus, avian
virus coating antibody BRP and Nezucastle disease virus conjugated infectious bursal disease virus, avian infectious
detection antibody BRP are suitable. Before estimation, the laryngotracheitis virus, influenza A virus, Marek's disease
antigen may be extracted from the emulsion using isopropyl virus.
myristate R or another suitable method. The vaccine complies The vaccine complies with the test if it does not stimulate the
with the test if the estimated antigen content is not formation of antibodies against these agents.
significantly lower than that of a batch that has been found to 3-4 Residuallive virus
be satisfactory with respect to irnmunogenicity (section 2-4-2). A test for residuallive virus is camed out to confirm
2-5-2-2 Adfuvant. If the irnmunochemical assay (section 2-5- inactivation of Newcastle disease virus.
2-1) is performed and if the vaccine is adjuvanted, the Inject 2/5 of adose into the allantoic cavity of each of 10
adjuvant is tested by suitable physical and chemical methods. embryonated hen eggs that are 9-11 days old and from SPF
For oil-adjuvanted vaccines, the adjuvant is tested in flocks (5.2.2) (SPF eggs), and incubate. Observe for 6 days
accordance with the monograph Vaccines for veterinary and pool separately the allantoic fluid from eggs containing
use (0062). If the adjuvant cannot be adequately live embryos and that from eggs containing dead embryos,
characterised, the antigen content determination cannot be exduding those dying within 24 h of the injection. Examine
used as the batch potency test. embryos that die within 24 h of injection for the presence of
2-5-2-3 Serological assay. Use not fewer than 15 chickens, Newcastle disease virus: the vaccine does not comply with
21-28 days old, ofthe same origin and from an SPF flock the test if Newcastle disease virus is found.
(5.2.2). Vaccinate by the intramuscular route not fewer than Inject into the allantoic cavity of each of 10 SPF eggs,
10 chickens with a volume of the vaccine equivalent to 1/50 9-11 days old, 0.2 mL of the pooled allantoic fluid from the
of adose. Maintain not fewer than 5 chickens as controls. live embryos and, into each of 10 similar eggs, 0.2 mL ofthe
Collect serum samples from each chicken after 17-21 days. pooled fluid from the dead embryos and incubate for
Measure the antibody levels in the sera by the 5-6 days. Test the allantoic fluid from each egg for the
haemagglutination-inhibition (HI) test using the technique presence of haemagglutinins using chicken erythrocytes.
described below or an equivalent technique with the same The vaccine complies witll tlle test if there is no evidence of
numbers of haemagglutinating units and red blood cells. haemagglutinating activity and if not more than 20 per cent
The test system used must indude negative and positive of the embryos die at either stage. If more than 20 per cent
control sera, the latter having an HI titre of 5.0 log2 to of the embryos die at one of the stages, repeat that stage;
6.0 log2' The vaccine complies with the test if the mean HI the vaccine complies with the test if there is no evidence of
titre of the vaccinated group is equal to or greater than haemagglutinating activity and not more than 20 per cent of
4.0 log2 and that of the unvaccinated group is 2.0 log2 or the embryos die at that stage.
less. If the HI titres are not satisfactory, carry out the test for
Antibiotics may be used in the test to control extraneous
vaccines for use in chickens (section 2-4-2-1).
bacterial infection.
Haemagglutination inhibition
Inactivate the test sera by heating at 56 oC for 30 min. 3-5 Potency
The vaccine complies with the requirements of the test
Add 25 ~lL of inactivated serum to the first row of wells in a
mentioned under Irnmunogenicity (section 2-4-2) when
microtitre plateo Add 25 ~lL of a buffered 9 gIL solution of
administer~d by a recommended route and method.
sodiu111 chlonde R at pH 7.2-7.4 to the rest ofthe wells. _____________________________________________ PhEm
Prepare twofold dilutions of the sera across the plateo
To each well add 25 ~L of a suspension containing
4 haemagglutinating units of inactivated Newcastle disease
virus. Incubate the plate at 4 oC for 1 h. Add 25 ~lL of a
1 per cent V/V suspension of red blood cells collected from Newcastle Disease Vaccine, Living *****
chickens that are 3-4 weeks old and free from antibodies ** **
against Newcastle disease virus. Incubate the plate at 4 oC
(Nezucastle Disease Vaccine (Live)) ***
Ph Eur 1110nograph 0450)
for 1 h. The HI titre is equal to the highest dilution that
produces complete inhibition. The use of Nezucastle Disease Vaccine) Living is restricted in the
United Kingd0111 by the Depart111ent for Environ111ent) Food and
3 BATCH TESTS
Rural Affairs.
3-1 Identification
~Em _____________________________________________
When injected into animal s that do not have antibodies
against Newcastle disease virus, the vaccine stimulates the 1 DEFINITION
production of such antibodies. Newcastle disease vaccine (live) is a preparation of a suitable
3-2 Bacteria and fungi strain of N ewcastle disease virus (avian paramyxovirus 1).
The vaccine, induding where applicable the diluent supplied This monograph applies to vaccines intended for
for reconstitution, complies with the test for sterility prescribed administration to chickens andlor other avian species for
in the monograph Vaccines for veterinmy use (0062). active immunisation.
Vet-292 Veterinary Vaccines 2016

2 PRODUCTION or equivalent with leucine at 117 and no basic amino acids at


2-1 PREPARATION OF THE VACCINE sites 111, 112, 114 and 115.
The vaccine virus is grown in embryonated hens' eggs or in 2-4-3 Safety
cell cultures. Carry out the test for each route and method of
2-2 SUBSTRATE FOR VIRUS PROPAGATION administration to be recommended for vaccination and in
2-2-1 Embryonated hens' eggs each avian species for which the vaccine is intended, using in
If the vaccine virus is grown in embryonated hens' eggs, they each case birds not older than the minimum age to be
are obtained from fiocks free fram specified pathogens (SPF) recommended for vaccination. If the test is performed in
(5.2.2). chickens, use chickens fram an SPF fiock (5.2.2). If the test
2-2-2 Cell cultures is pelformed in birds other than chickens, use birds that do
If the vaccine virus is grown in cell cultures, they comply not have antibodies against Newcastle disease virus.
with the requirements for cell cultures for production of Use vaccine virus at the least attenuated passage level that
veterinary vaccines (5.2.4). will be present in a batch of the vaccine.
2-3 SEED LOTS For each test performed in birds younger than 3 weeks of
2-3-1 Extraneous agents age, use not fewer than 10 birds. For each test performed in
The master seed lot complies with the tests for extraneous birds older than 3 weeks of age, use not fewer than 8 birds.
agents in seed lots (2.6.24). In these tests on the master seed Administer to each bird a quantity of the vaccine virus
lot, the organisms used are not more than 5 passages from equivalent to not less than 10 times the maximum virus titre
the master seed lot at the start of the tests. lil,ely to be contained in 1 dose of the vaccine. Observe the
birds at least daily for at least 14 days.
2-4 CHOICE OF VACCINE VIRUS
The test is not valid if more than 10 per cent of the birds
The vaccine virus shall be shown to be satisfactory wir.h
younger than 3 weeks of age show abnormal signs of disease
respect to safety (5.2.6) and efficacy (5.2.7) for the birds for
or die from causes not attributable to the vaccine virus.
which it is intended.
For bil"ds older than 3 weeks of age, the test is not valid if
The following tests for intracerebral pathogenicity index
non-specific mortality occurS.
(section 2-4-1), amino-acid sequence (section 2-4-2), safety
(section 2-4-3), increase in virulence (section 2-4-4) and The vaccine virus complies with the test if no bil"d shows
irnmunogenicity (section 2-4-5) may be used during the abnOlmal signs of disease or dies from causes attributable to
demonstration of safety and efficacy. the vaccine.
2-4-1 Intracerebral pathogenicity index 2-4-4 Increase in virulence
Use vaccine virus at the least attenuated passage level dlat Carry out the test according to general chapter 5.2.6 using
will be present between the master seed lot and a batch of birds not more than 2 weeks old. If the properties of the
the vaccine. Inoculate the vaccine virus into the allantoic vaccine virus allow sequential passage through 5 groups via
cavity of embryonated hens' eggs, 9- to 11- days-old, from natural spreading, this method may be used, otherwise
an SPF fiod: (5.2.2). Incubate dle inoculated eggs for a passage as described below is carried out. Carry out the test
suitable period and harvest and pool dle allantoic fiuids. in a target species, using the chicken if it is one of the target
Use not fewer than ten 1-day-old chickens (i.e. more dlan species. For the test in chickens, use chickens from an SPF
24 h but less than 40 h after hatching), from an SPF fiock fiock (5.2.2). For other species, cany out the test in birds
(5.2.2). Administer by the intracerebral raute to each chick that do not have antibodies against Newcastle disease virus.
0.05 mL of the pooled allantoic fiuids containing not less Administer to each bil"d of the 1st group by eye-drop a
than 10 8 .0 EIDso or, if this virus quantity cannot be achieved quantity of the vaccine vil"us that will allow recovery of virus
not less than 10 7 . 0 EIDso. Observe the chickens at least daily' for the passages described below. Observe the birds for the
for 8 days after administration and score them once every period shown to correspond to maximum replication of the
24 h. A score of O is attributed to a chicken if it is clinically vaccine virus, euthanise them and prepare a suspension from
normal, 1 if it shows clinical signs of disease and 2 if it is the brain of each bird and from a suitable organ depending
dead. The intracerebral pathogenicity index is the mean of on the tropism of the strain (for example, mucosa of the
the scores per chicken per observation over the 8-day periodo entire trachea, intestine, pancreas); pool the samples.
If an inoculum of not less than 10 8 . 0 EIDso is used, dle Administer 0.05 mL of the pooled samples by eye-drop to
vaccine virus complies widl the test if its intracerebral each bird of the next group. Carry out this passage operation
pathogenicity index is not greater than 0.5; if an inoculum of not fewer than 4 times; veri:fy the presence of the virus at
not less than 10 7 .0 EIDso but 1ess than 10 8 .0 EIDso is used, each passage. If the virus is not found at a passage level,
the vaccine vilus complies with dle test if its intracerebral repeat the passage by administration to a group of 10 birds.
pathogenicity index is not greater than 0.4. A. Carry out the test for intracerebral pathogenicity index
2-4-2 Amino-acid sequence (section 2-4-1) using the material used for the 1st passage
Determine the sequence of a fragment of RNA from the and the vil"us at the final passage level.
vaccine virus containing the region encoding for the FO B. Carry out the test for amino-acid sequence (section 2-4-2)
cleavage site by a suitable method. The encoded amino-acid using unpassaged vaccine virus the material used for the
sequence is shown to be one of the following: 1st passage and the virus at the final passage level.
C. Carry out the test for safety (section 2-4-3) using the
F2 Cleavage F1 material used for the 1st passage and the virus at the final
site
111 112 113 114 115 116 v 118 119
passage level. .
Site 117
Administer the virus by the raute to be recommended for
Gly Gly Lys Gln Gly Arg Leu He Gly
vaccination likely to be the least safe and to the avian species
or Gly Gly Arg Gln Gly Arg Leu He Gly for which the vaccine is intended that is likely to be the most
or Gly Glu Arg Gln Glu Arg Leu Val Gly susceptible to Newcastle disease.
2016 Veterinary Vaccines Vet-293

The vaccine virus complies with the test if, in the tests 3 BATCH TESTS
2-4-4A, 2-4-4B and 2-4-4C, no indication of increase in 3-1 Identification
virulence of the virus recovered for the final passage 3-1-1 Identification of the vaccine virus. The vaccine, diluted if
compared with the material used for the 1sr passage is neceSSa1y and mixed with a monospecific Newcastle disease
observed. If virus is not recovered after an initial passage in virus antiserum, no longer provokes haemagglutination of
5 birds and a subsequent repeat passage in 10 birds, the chicken red blood cells 01" infects embryonated hens I eggs
vaccine virus also complies with the test. from an SPF flock (5.2.2) 01" susceptible cell cultures (5.2.4)
2-4-5 Immunogenicity into which it is inoculated.
For each avian species for which the vaccine is intended, a 3-1-2 Identificarion of the virus strain. The strain of vaccine
test is carried out for each route and method of virus is identified by a suitable method, for example using
administration to be recommended using in each case birds monoc1onal antibodies.
not older than the minimum age to be recommended for 3-2 Bacteria and fungi
vaccination. The quantity of the vaccine virus administered Vaccines intended for administration by injection comply
to each bird is not greater than the minimum titre to be with the test for sterility prescribed in the monograph
stated on the label and the virus is at the most attenuated Vaccines for veterinary use (0062).
passage level that will be present in a batch of the vaccine.
Frozen 01" freeze-dried vaccines produced in embryonated
2-4-5-1 Vaccines for use in chickens. Use not fewer than eggs and not intended for administration by injection either
30 chickens of the same origin and from an SPF flock comply with the test for sterility prescribed in the monograph
(5.2.2). Vaccinate by a route to be recommended not fewer Vaccines for veterina1Y use (0062) 01" with the following test:
than 20 chickens. Maintain not fewer than 10 chickens as carry out a quantitative test for bacterial and fungal
controls. Challenge each chicken after 21 days by the contamination; carry out identification tests for
intramuscular route with not les s than 10 5 .0 emblyo LD 50 of microorganisms detected in the vaccine; the vaccine does not
the Herts (Weybridge 33/56) strain of Newcastle disease contain pathogenic microorganisms and contains not more
virus. Observe the chickens at least daily for 14 days after than 1 non-pathogenic microorganism per dose.
challenge. Record the deaths and the number of surviving
Any diluent supplied for reconstitution of the vaccine
chickens that show c1inical signs of disease.
complies with the test for sterility prescribed in the
The test is not valid if 6 days after challenge fewer than monograph Vaccines for veterina1Y use (0062).
100 per cent of the control chickens have died 01" if during
3-3 Mycoplasmas
the period between vaccination and challenge more than
The vaccine complies with the test for mycoplasmas (2.6.7).
10 per cent of the vaccinated 01" control chickens show
abnormal c1inical signs 01" die from causes not attributable to 3-4 Extraneous agents
the vaccine. The vaccine complies with tl1e tests for extraneous agents in
The vaccine virus complies Witl1 the test if during the batches of finished product (2.6.25).
observation period after challenge not fewer tl1an 90 per cent 3-5 Virus titre
of tl1e vaccinated chickens survive and show no notable Titrate the vaccine virus by inoculation into embryonated
c1inical signs of Newcastle disease. hens' eggs from an SPF flock (5.2.2) 01" into suitable cell
2-4-5-2 Vaccines for use in avian species other than the chicken. cultures (5.2.4). The vaccine complies with the test if 1 dose
Use not fewer than 30 birds of the species for which tl1e contains not less than the minimum virus titre stated on the
vaccine is intended for Newcastle disease, of the same origin label.
and that do not have antibodies against avian 3-6 Potency
paramyxovirus 1. Vaccinate by a route to be recommended Depending on the indications, tl1e vaccine complies with 1 01'
not fewer than 20 birds. Maintain not fewer than 10 birds as both of the tests prescribed under Immunogenicity (section
controls. Challenge each bird after 21 days by the 2-4-5) when administered according to the recommended
intramuscular route with a sufficient quantity of virulent schedule by a recommended route and method. If the test in
avian paramyxovirus 1. Observe the birds at least daily for section 2-4-5-2 Vaccine for use in avian species other than the
21 days after challenge. Record the deaths and tl1e surviving chicken is conducted and the vaccine is recommended for use
birds that show c1inical signs of disease. in more than 1 avian species, the test is carried out with
The test is not valid if: birds of that species for which the vaccine is recommended
-- during the observation period after challenge fewer than which is likely to be the most susceptible to avian
90 per cent of the control birds die 01" show severe c1inical paramyxovirus 1. It is not necessary to carry out the potency
signs of Newcastle disease; test for each batch of the vaccine if it has been carried out on
- - 01" if during the period between the vaccination and a representative batch using a vaccinating dose containing
challenge more than 10 per cent of the vaccinated 01" not more than the minimum virus titre stated on the label.
control birds show abnormal c1inical signs 01" die from _____________________________________________ PhE~

causes not attributable to the vaccine.


The vaccine virus complies with the test if during the
observation period after challenge not fewer than 90 per cent
of the vaccinated birds survive and show no notable c1inical Ovine Enzootic Abortion Vaccine,
signs of Newcastle dise¡:¡se. For species where there is
published evidence that it is not possible to achieve this level Inactivated
of protection, the vaccine complies with the test if there is a DEFINITION
significant reduction in morbidity and mortality of the Ovine Enzootic Abortion Vaccine, Inactivated is a suspension
vaccinated birds compared with the control birds. of one 01' more strains of the ch1amydia organisms of ovine
enzootic abortion which have been inactivated in such a
manner that the immunogenic activity is retained.
Vet-294 Veterinary Vaccines 2016

PRODUCTION animal s for two weeks. No abnormallocal or systemic


The Chlamydia psittaci organisms are grown in either suitable reactions occur.
cell cultures, Appendix XV Jevet) 1, or in the yolk sacs of POTENCY
embryonated eggs derived from healthy chicken fiocks. Inject each of five healthy susceptible sheep according to the
The organisms are harvested and inactivated. A validated, recommendations stated on the label. Maintain two
suitably sensitive test for residual chlamydia is carried out in
unvaccinated sheep from the same source as the
tissue cultures, on each batch of antigen immediately after
unvaccinated controls. Bleed all of the animals before
inactivation.
vaccination and again not less than 28 days latero Using an
The vaccine contains an adjuvant. appropriate serological test (complement-fixation or
CHOICE OF VACCINE COMPOSITION immunofiuorescence is suitable) the serum of each sheep
The vaccine is shown to be satisfactory with respect to safety before vaccination is negative at a 2-fold dilution and, not
and immunogenicity for the animals for which the vaccine is less than 28 days after vaccination, the serum of no fewer
intended. The following tests may be used during the than four of the vaccinated sheep gives a positive reaction at
demonstration of safety, Appendix XV Kevet) 1, and an 8-fold or greater dilution. The test is not valid if there is
immunogenicity, Appendix XV Kevet) 2. an increase in antibody levels in the controls.
Safety STORAGE
Carry out a test in each category of animal for which the When stored under the prescribed conditions the vaccine
vaccine is to be recommended and by each recommended may be expected to retain its potency for at least 1 year.
route of administration. Vaccinate at least five animals that
do not have antibodies to Chlamydia psittaci. U se for the test,
a batch of vaccine with the maximum potency likely to be
inc1uded in adose of the vaccine. Administer a double dose
of vaccine to each animal and observe them for two weeks., Pasteurella Vaccine (Inactivated)
No abnormallocal or systemic reactions occur. If the vaccine for Sheep
is for use or may be used in pregnant animals, for the test in
this category, administer the vaccine at the relevant stage 01' (Ph. Eur. monograph 2072)
_____________________________________________
stages of pregnancy, prolong the observation period up to the PhE~

time of parturition and note any effects on gestation or on 1 DEFINITION


the offspring. Pasteurella vaccine (inactivated) for sheep is a preparation of
Immunogenicity one or more suitable strains of PasteUJ'ella trehalosi, inactivated
The tests to demonstrate Immunogenicity are carried out in while maintaining adequate immunogenic properties. This
each category of animal for which the vaccine is to be monograph applies to vaccines intended for the active
recommended and by each recommended route of immunisation of sheep against disease caused by P. trehalosi.
administration and using a batch or batches with the
2 PRODUCTION
minimum potency likely to be inc1uded in adose of the
2-1 PREPARATION OF THE VACCINE
vaccine. The efficacy c1aims made on the label
(e.g. protection from abortion) refiect the type of data Production of the vaccine is based on a seed-lot system.
generated. The seed material is cultured in a suitable medium; each
strain is cultivated separately and identity is verified using a
BATCH TESTING suitable method. During production, various parameters such
Inactivation as growth rate are monitored by suitable methods; the values
Carry out a suitable validated test in tissue cultures for are within the limits approved for the particular producto
residual Chlamydia psittaci on the bulk antigen blend Purity and identity of the harvest are verified using suitable
immediately before the addition of the adjuvant. No live methods. After cultivation, the bacterial suspensions are
organisms are detected. collected separately and inactivated by a suitable method.
CAUTION Accidental i71y"ection of oil emulsion vaccines can The vaccine may be adjuvanted.
cause se1"ious local reactiolls in mano Expert medical advice should 2-2 CHOICE OF VACCINE COMPOSITION
be sought immediately and the doctor should be informed that the The choice of composition and the strains to be inc1uded in
vaccine is an oil emulsiono the vaccine are based on epidemiological data on the
The vaccine complies with the requirements stated undel' prevalence of the different serovars of P. trehalosi.
Veterinary Vaccines with the following modifications. The vaccine is shown to be satisfactory with respect to safety
IDENTIFICATION (S. 2. 6) and efficacy (5.2.7) for the sheep for which it is
When injected into healthy seronegative animals, the vaccine intended.
stimulates the production of specific antibodies against The following tests for safety (section 2-2-1) and
Chlamydia psittaci. immunogenicity (section 2-2-2) may be used during the
TESTS demonstration of safety and efficacy.
Extraneous bacteria and fungi 2-2-1 Safety
The vaccine complies with the test for sterility described 2-2-1-1 Laboratory tests. Carry out the tests for each route
under Veterinary Vaccines. and method of administration to be recommended for
vaccination and in sheep of each category for which the
Safety
vaccine is intended (for example, young sheep, pregnant
Use two lambs of the minimum age recommended for
ewes). U se a batch of vaccine containing not less than the
vaccination and that do not have antibodies to Chlamydia
maximum potency that may be expected in a batch of
psittaci. Administer by a route recommended on the label, a
vaccine.
double dose of vaccine to each of the lambs and observe the
2016 Veterinary Vaccines Vet-295

For each test, use not fewer than 8 sheep that preferably do obtained for these pa1'ameters and rile bacterial re-isolation
not have antibodies against the serovars of P. trehalosi 01' results for the 2 groups.
against the leucotoxin present in the vaccine. Where justified, The test is not valid if signs 01' lesions of P. trehalosi infection
sheep with a known history of no previous pasteurella occur in les s than 70 per cent of the controllambs.
vaccination and with low antibody titres (measured in a The vaccine complies with the test if there is a significant
sensitive test system such as EUSA) may be used. difference between the scores obtained for the c1inical and
Administer to each sheep 1 dose of the vaccine. If the post-mortem observations in the vaccinates compared to the
schedule to be recommended requires a 2nd dose, administer controls. For vaccines with a claim for a beneficial effect on
another dose after an interval of at least 14 days. Observe the the extent of infection against the serovar, the results for the
sheep at least daily for at least 14 days after the last infection rates are also significantly better for the vaccinates
administration. Record body temperature the day before compared to the controls.
vaccination, at vaccination, 2 h, 4 h and 6 h later and then
daily for 4 days; note the maximum temperature increase for 2-3 MANUFACTURER'S TESTS
each sheep. 2-3-1 Batch potency test
It is not necessary to carry out the potency test (section 3-3)
The vaccine complies with the test if no sheep shows
for each batch of vaccine if it has been ca1'ried out using a
abnormallocal reactions, notable signs of disease 01' dies
batch of vaccine with a minimum potency. Where the test is
from causes attributable to the vaccine, if the average body
not carried out, an alternative validated method is used, the
temperature increase for all sheep does not exceed 1.5 oC
criteria for acceptance being set with refe1'ence to a batch of
and no sheep shows a rise greater than 2.0 oc.
vaccine that has given satisfactory results in the test described
2-2-1-2 Field studies. The sheep used for the field trials are under Potency.
also used to evaluate safety. Carry out a test in each category
2-3-2 Bacterial endotoxins
of sheep for which the vaccine is intended. Use not fewer
A test for bacterial endotoxins (2.6.14) is ca1'ried out on the
than 3 groups of 20 sheep with corresponding groups of not
final lot 01', where the nature of the adjuvant prevents
fewer than 10 controls in 3 different locations. Examine the
perfOlmance of a satisfactory test, on the bulk antigen or the
injection sites for local reactions after vaccination. Record
mixture of bulk antigens immediately before addition of the
body temperatures the day before vaccination, at vaccination
adjuvant. The maximum acceptable amount of bacterial
and on the 2 days following vaccination.
endotoxins is that found for a batch of vaccine that has been
The vaccine complies with the test if no sheep shows shown satisfactory in safety tests 2-2-1-1 given under Choice
abnormal local or systemic reactions, notable signs of disease of vaccine composition. The method chosen for determining
01' dies from causes attributable to the vaccine. The average
the amount of bacterial endotoxin present in the vaccine
body temperature increase for all sheep does not exceed batch used in the safety test for determining the maximum
1.5 oC and no sheep shows a rise greater than 2.0 oc. acceptable level of endotoxin is used subsequently fo1' testil1g
In addition, if the vaccine is intended for use in pregnant of each batch.
ewes, no adverse effects on the pregnancy and offspring are
demonstrated. 3 BATCH TESTS
3-1 Identification
2-2-2 Immunogenicity
When injected into healthy animals that do not have specific
Carry out a test for each serovar of P. trehalosi for which
antibodies against the serovars of P. trelzalosi and/or against
protection is to be claimed on the label.
the leucotoxin present in the vaccine, the vaccine stimulates
A test is carried out for each route and method of the production of such antibodies.
administration to be recommended, using in each case lambs
of the minimum age to be recommended for vaccination. 3-2 Bacteria and fungi
The vaccine administered to each lamb is of minimum The vaccine, including where applicable tlle diluent supplied
potency. for reconstitution, complies with the test for sterility
prescribed in the monograph Vaccines for veterina1Y
Use not fewer than 20 lambs that do not have antibodies use (0062).
against P. trehalosi and agail1st the leucotoxin of P. trehalosi.
Vaccinate not fewer than 10 lambs according to the schedule 3-3 Potency
to be recommended. Maintain not fewer than 10 lambs as The vaccine complies with the requirements of the test
controls. 20-22 days after the last vaccination, challenge each mentioned under Immunogenicity (section 2-2-2) when
lamb by the subcutaneous or another suitable route, with a administered by a recommended route and method.
sufficient quantity of a low-passage, virulent strain of a __________________ ~ _________________________ PhE~

serovar of P. trehalosi. Observe the lambs for a further 7 days;


to avoid unnecessary suffering, severely ill lambs are
euthanised and are then considered to have died from the
disease. During the observation pe1'iod, examine the lambs ***
Porcine Actinobacillosis Vaccine,
for any signs of disease (for example, severe dullness, excess
*** ***
salivation) and record the mortality. Euthanise surviving Inactivated ***
lambs at the end of the observation periodo Carry out post-
(Porcine Aáinobacillosis Vaccine (In activa ted) )
mortem examination on any lamb that dies and those
Ph Bur monograph 1360)
euthanised at the end of the observation periodo Examine the
PhE~~ ___________________________________________
lungs, pleura, liver and spleen for haemorrhages and evaluate
the extent of lung consolidation due to pasteurellosis. Collect 1 DEFINITION
samples of lung, liver and spleen tissue for re-isolation of the Porcine actinobacillosis vacclne (inactivated) is a preparation
challenge organisms. Score the mortality, clinical observations which has one 01' more of the following components:
and the post-mOltem lesions and compare the results inactivated Actinobacillus pleuropneumoniae of a suitable strain
01' strains; toxins, proteins 01' polysaccharides derived from
Vet-296 Veterinary Vaccines 2016

suitable srrains of A. pleuropneumoniae, and treated to render pigs does not exceed 1.5 oC and no pig shows a rise greater
them harrnless while maintaining adequate irnmunogenic than 2.0 oC.
properties; fractions of toxins derived from suitable strains of 2-2-2 Immunogenicity
A. pleuropneumoniae and treated if necessary to render them The chalIenge strain for the folIowing test is chosen to ensure
harrnless while maintaining adequate irnmunogenic chalIenge with each Ap toxin 1 produced by the serotypes to
properties. This monograph applies to vaccines intended for be stated on the label; it may be necessmy to carry out more
the active irnmunisation of pigs against actinobacillosis. than one test using a different challenge strain for each test.
2 PRODUCTION Each test is carried out for each raute and method of
2-1 PREPARATION OF THE VACCINE administration to be recommended. The vaccine
The seed material is cultured in a suitable medium; each administered to each pig is of minimum potency.
strain is cultivated separately. During production, various For each test, use not fewer than 14 pigs that do not have
parameters such as growth rate, protein content and quantity antibodies against A. pleuropneumoniae and Ap toxins.
of relevant antigens are monitored by suitable methods; Vaccinate not fewer than 7 pigs according to the schedule to
the values are within the limits approved for the particular be recommended. Maintain not fewer than 7 pigs as controls.
producto Purity and identity are verified on the harvest using 3 weeks after the last vaccination, chalIenge all the pigs by
suitable methods. After cultivation, the bacterial harvests are the intranasal or intratracheal raute or by aerosol with a
collected separately and inactivated by a suitable method. sufficient quantity of a virulent serotype of
They may be detoxified, purified and concentrated. A. pleuropneu111oniae. Observe the pigs at least daily for
The vaccine may be adjuvanted. 7 days; to avoid unnecessary suffering, severely ill control
2-2 CHOICE OF VACCINE COMPOSITION pigs are euthanised and are then considered to have died
The choice of strains is based on epidemiological data. fram the disease. Euthanise all surviving pigs at the end of
The vaccine is shown to be satisfactory with respect to safety the observation periodo Carry out a post-mortem examination
(5.2.6) and efficacy (5.2.7) for the pigs for which it is on all pigs. Examine the lungs, the tracheobronchiallymph
intended. nodes and the tonsils for the presence of A. pleuropneu111oniae.
Evaluate the extent of lung lesions at post-mortem
The folIowing tests for safety (section 2-2-1) and
examination. Each of the 7 lobes of the lungs is alIotted a
irnmunogenicity (section 2-2-2) may be used during the
maximum possib1e lesion score2 of 5. The area showing
demonstration of safety and efficacy.
pneumonia and/or pleuritis of each lobe is assessed and
2-2-1 Safety expressed on a scale of O to 5 to give the pneumonic score
2-2-1-1 Laborat01Y tests Carry out the test for each raute and per lobe (the maximum total score possible for each
method of administration to be recornmended for vaccination complete lung is 35). Calculate separately for the vaccinated
and where applicable, in pigs of each category for which the and the control pigs the total score (the maximum score per
vaccine is intended, using in each case pigs not older than group is 245, if 7 pigs are used per group).
the minimum age to be recommended for vaccination. U se a The vaccine complies with the test if the vaccinated pigs,
batch containing not less than the maximum potency that when compared with controls, show lower incidence of:
may be expected in a batch of vaccine. mortality; typical signs (dyspnoea, coughing and vomiting);
For each test, use not fewer than 8 pigs that do not have typical lung lesions; re-isolation of A. pleuropneul1lOniae from
antibodies against the serotypes of A. pleuropneumoniae or its the lungs, the tracheobronchial lymph nodes and the tonsils.
toxins present in the vaccine. Administer to each pig 1 dose Where possible, the incidence is analysed statistically and
of the vaccine. If the schedule to be recommended requires a shown to be significantly lower for vaccinates.
2nd dose, administer another dose after an interval of at least
14 days. Observe the pigs at least daily until 14 days after the 2-3 MANUFACTURER'S TESTS
last administration. Record body temperature the day before 2-3-1 Batch potency test
vaccination, at vaccination, 2 h, 4 h and 6 h later and then It is not necessary to cany out the potency test (section 3-4)
daily for 4 days; note the maximum temperature increase for for each batch of vaécine if it has been carried out using a
each pig. batch of vaccine with a minimum potency. Where the test is
not carried out, an alternative validated method is used, the
The vaccine complies with the test if no pig shows abnormal
criteria for acceptance being set with reference to a batch of
local or systemic reactions or dies fram causes attributable to
vaccine that has given satisfactory results in the test described
the vaccine, and if the average temperature increase for alI
under Potency. The following test may be used.
pigs does not exceed 1.5 oC and no pig shows a rise greater
than 2.0 oc. Use 5 mice weighing 18-20 g and that do not have
antibodies against the serotypes of A. pleuropneumoniae or its
2-2-1-2 Field studies The pigs used for field trials are also
toxins present in the vaccine. Vaccinate each mouse by the
used to evaluate safety. Carry out a test in each category of
subcutaneous route with a suitable dose. Where the
pigs for which the vaccine is intended. U se not fewer than
recommended schedule requires a booster injection to be
3 groups each of not fewer than 20 pigs with corresponding
given, a booster vaccination may also be given in this test
groups of not fewer than 10 controls. Examine the injection
provided it has been demonstrated that this will still provide
site for local reactions after vaccination. Record body
a suitably sensitive test system. Before the vaccination and at
temperature the day before vaccination, at vaccination, at the
a given interval within the range of 14-21 days after the last
time interval after which a rise in temperature, if any, was
injection, collect blood from each mouse and prepare serum
seen in test 2-2-1-1, and daily during the 2 days following
samples. Determine individualIy for each serum the titre of
vaccination; note the maximum temperature increase for each
specific antibodies against each antigenic~ component stated
pig.
on the label, using a suitable validated test such as enzyme-
The vaccine complies with the test if no pig shows abnormal linked irnmunosorbent assay (2.7.1). The vaccine complies
local or systemic reactions or dies from causes attributable to with the test if the antibody levels are not significantly lower
the vaccine, and if the average temperature increase for alI
2016 Veterinary Vaccines Vet-297

than those obtained for a batch that has given satisfactory


Porcine Enzootic Pneumonia ***
results in the test described under Potency. *** ***
2-3-2 Bacterial endotoxins Vaccine (Inactivated) ***
A test for bacterial endotoxins (2.6.14) is carried out on the (Porcine Enzootic Pneumonia Vaccine (Inactivated) J
final bulk or, where the nature of the adjuvant prevents Ph Bur 1110nograph 2448)
performance of a satisfactory test, on the bulk antigen or PhE~ _____________________________________________
mixture of bulk antigens irnmediately before addition of the
adjuvant. The maximum acceptable amount of bacterial 1 DEFINITION
endotoxins is that found for a batch of vaccine that has been Porcine enzootic pneumonia vaccine (inactivated) is a
shown satisfactory in safety test 2-2-1-1 described under preparation of a suitable strain of Mycoplasma hyopneul1zoniae
Choice of vaccine composition or in the residual toxicity test that has been inactivated while maintaining adequate
described under Batch tests, carried out using 10 pigs. Where irnmunogenic properties. This monograph applies to vaccines
the latter test is used, note the maximum temperature intended for the active irnmunisation of pigs against enzootic
increase for each animal; the vaccine complies with the test if pneumonia caused by M. hyopneumoniae.
the average temperature increase for all animals do es not 2 PRODUCTION
exceed 1.5 oc. The method chosen for determining the 2-1 PREPARATION OF THE VACCINE
amount of bacterial endotoxin present in the vaccine batch Production of the vaccine is based on a seed-Iot system.
used in the safety test for determining the maximum The seed material is cultured in a suitable solid anci/or liquid
acceptable level of endotoxin is used subsequently for batch medium to ensure optimal growth under the chosen
testing. incubation conditions. The identity of the strain is verified
3 BATCH TESTS using a suitable method.
3-1 Identification During production, various parameters such as growth rate
When injected into healthy animals that do not have specific are monitored by suitable methods; the values are within the
antibodies against the antigenic components of A. limits approved for the particular vaccine. Purity of the
pleuropneUinoniae stated on the label, the vaccine stimulates harvest is verified using a suitable method.
the production of such antibodies. After cultivation, the mycoplasma suspension is collected and
3-2 Bacteria and fungi inactivated by a suitable method. The vaccine may contain
The vaccine, including where applicable the diluent supplied an adjuvant.
for reconstitution, complies with the test for sterility 2-2 CHOICE OF VACCINE COMPOSITION
prescribed in the monograph Vaccines for veterina1Y use The vaccine is shown to be satisfactory with respect to safety
(0062). (5.2.6) and efficacy (5.2.7) for the pigs for which it is
3-3 Residual toxicity intended.
U se 2 pigs of the minimum age recommended for The following tests for safety (section 2-2-1) and
vaccination and that do not have antibodies against the immunogenicity (section 2-2-2) may be used during the
serotypes of A. pleuropneunzoniae or its toxins present in the demonstration of safety and efficacy.
vaccine. Administer to each pig by a recornmended route a
2-2-1 Safety
double dose of the vaccine. Observe the pigs at least daily for
2-2-1-1 LaboratOlY tests. Carry out the test for each route and
14 days. Record body temperature the day before
method of administration to be recornmended for vaccination
vaccination, at vaccination, 2 h, 4 h and 6 h later and then
daily for 2 days. and where applicable, in pigs of each category for which the
vaccine is intended, using in each case pigs not older than
It is recommended to use the mean temperature of the days the minimum age to be recommended for vaccination. U se a
before administration ofthe vaccine (e.g. day -3 to day O) as batch of vaccine containing not less than the maximum
the baseline temperature to have clear guidance for potency that may be expected in a batch of vaccine.
evaluation of the test.
For each test, use not fewer than 8 pigs that do not have
The vaccine complies with the test if no pig shows notable antibodies against M. hyopneu1110niae. Administer to each pig
signs of disease or dies from causes attributable to the 1 dose of the vaccine. If the schedule to be recornmended
vaccine; a transient temperature increase not exceeding requires a 2nd dose, administer another dose after an interval
2.0 oC may occur. of at least 14 days. Observe the pigs at least daily until at
3-4 Potency least 14 days after the last administration. Record body
The vaccine complies with the requirements of the test temperature the day before vaccination, at vaccination, 4 h
mentioned under Irnmunogenicity (section 2-2-2) when later and then daily for 4 days; note the maximum
administered by a recommended route and method. temperature increase for each pig.
_____________________________________________ PhE~
The vaccine complies with the test if no pig shows notable
signs of disease or dies from causes attributable to the
(1)The nomenclature of the toxins of A. pleuropneumoniae is vaccine, and, in particular, if the average body temperature
described by J. Frey et al., Joumal of General Microbiology, 1993, increase for all pigs does not exceed 1.5 oC and no pig shows
139, 1723-1728. a rise greater than 2.0 oc.
(2) The system of lung scores is described in detail by P.C.T.
Hannan, B.S. Bhogal, J.P. Fish, Research in Veterinary Science, 1982, 2-2-1-2 Field studies. The animal s used for field trials are also
33, 76-88. used to evaluate safety. Carry out a test in each category of
animals for which the vaccin~ is intended. U se not fewer
than 3 groups each of not fewer than 20 animals with
corresponding groups of not fewer than 10 controls. Examine
the injection site for local reactions after vaccination. Record
body temperature the day before vaccination, at vaccination,
Vet-298 Veterinary Vaccines 2016

at the time interval after which a rise in temperature, if any, by the subcutaneous route with a suitable dose. Maintain not
was seen in test 2-2-1-1, and daily during the 2 days fewer than 5 mice as controls. Where the recommended
following vaccinatiol1; note the maximum temperature schedule requires a booster injection to be given, a booster
increase for each animal. vaccination may also be given in this test provided it has
The vaccine complies with the test if no animal shows been demonstrated that this will still provide a suitably
notable signs of disease or dies from causes attributable to sensitive test system. Before the vaccination and at a given
the vaccine, the average body temperatm'e increase for all interval within the range of 14-21 days after the last injection,
animals does not exceed 1.5 oC, and no animal shows a rise collect blood from each mouse and prepare serum samples.
in body temperature greater than 2.0 oc. Determine individually for each serum the titre of specific
antibodies against each antigenic component stated on the
2-2-2 Immunogenicity
label, using a suitable validated test such as enzyme-linked
A test is carried out for each route and method of
immunosorbent assay (2.7.1).
administration to be recommended using in each case pigs
not older than the minimum age to be recommended for The vaccine complies with the test if the mean antibody
vaccination. The vaccine to be administered to each pig is of levels are not significantly lower than those obtained for a
minimum potency. batch that has given satisfactory results in the test described
under Potency.
U se not fewer than 20 pigs that do not have antibodies
against M. hyopneumoniae and that are from a herd 01' herds 3 BATCH TESTS
where there are no signs of enzootic pneumonia and that 3-1 Identification
have not been vaccinated against M. hyopneu11l0niae. When injected into healthy animal s that do not have
Vaccinate not fewer than 12 pigs according to the schedule antibodies against M. hyopneumoniae, the vaccine stimulates
to be recommended. Maintain not fewer than 8 non- the production of such antibodies. SuÍtable molecular
vaccinated pigs as controls. Challenge each pig at least methods such as nucleic acid amplification techniques
14 days after the last vaccination by the intranasal or (2.6.21) may also serve for identification.
intratracheal route or by aerosol with a sufficient quantity of 3-2 Bacteria and fungi
a virulent strain of NI. hyopneu1110niae. The challenge strain The vaccine, including where applicable the diluent supplied
used is different from the vaccine strain. 21-30 days after for reconstitution, complies with the test for sterility
challenge, euthanise the pigs. Conduct a post-mortem prescribed in the monograph Vaccines for vetetina1Y
examination on each pig in order to evaluate the extent of use (0062).
lung lesions using a validated lung les ion scoring system that
3-3 Residual1ive mycoplasmas
is adapted to the age of the animals. The following scoring
A test for residuallive mycoplasmas is carried out to confirm
system may be used.
inactivation of NI. hyopneu1110niae. The vaccine complies with
A weighted score is allocated to each of the 7 lobes of the a validated test for residuallive M. hyopneu11l0niae carried out
lungs according to the relative weight of the lung lobes. by a culture method (see for example 2.6.7, using media
shown to be suitable for M. hyopnell1170niae).
Lobes Left Right 3-4 Potency
Apical 5 11 The vaccine complies with the requirements of the test
mentioned under Immunogenicity (section 2-2-2) when
Cardiac 6 10
administered by a recommended route and method.
Diaphragmatic 29 34 _____________________________________________ ~E~

Intermediate 5

The vaccine complies with the test if the vaccinated pigs,


when compared with\controls, show a significant reduction in
Porcine E. Coli Vaccine, ***
the lung les ion score. *** ***
2-3 MANUFACTURER'S TESTS Inactivated ***
2-3-1 Batch potency test Porcine Escherichia Coli Vaccine, Inactivated
It is not necessary to carry out the potency test (section 3-4) (Neonatal Piglet ColibaczIlosis Vaccine (Inactivated) J
for each batch of the vaccine if it has been carried out using Ph Bur monograph 0962)
a batch of vaccine with a minimum potency. Where the test ~E~ _____________________________________________
is not carried out, an altemative validated method is used,
the criteria for acceptance being set with reference to a batch 1 DEFINITION
of vaccine that has given satisfactory results in the test Neonatal piglet colibacillosis vaccine (inactivated) is a
described under Potency. A quantification of the antigen preparation from cultures of one 01' more suitable strains of
(i.e. an in vitro test using a reference vaccine that has given Bschetichia coli, carrying one 01' more adhesins or
satisfactory results in the test described under Potency) enterotoxins. This monograph applies to vaccines intended
together with a test for adjuvant quantification may be used for the active immunisation of sows and gilts for passive
as an altemative method provided the antigen that is protection of their newbom progeny against enteric forms of
measured has been proven to be protective and/or colibacillosis, administered by injection.
immunorelevant. 2 PRODUCTION
Altematively, a test measuring induction of antibody response 2-1 PREPARATION OF THE VACCINE
in laboratory animals may be used. The following method is The E. coli strains used for production are cultured separately
given as an example. in a suitable medium. The cells or toxins are processed to
U se at least 5 mice weighing 18-20 g and that do not have render them safe while maintaining adequate immunogenic
antibodies against M. hyopneu1110niae. Vaccinate each mouse properties and are blended. The vaccine may be adjuvanted.
2016 Veterinary Vaccines Vet -299

2-2 CHOICE OF VACCINE COMPOSITION on the label. Take not fewer than 4 at random and vaccinate
The E. colí strains used in the production of the vaccine are these at the stage of pregnancy and according to the schedule
shown to be satisfactory with respect to expression of to be recommended. Maintain not fewer than 4 gilts as
antigens and the vaccine is shown to be satisfactory with controls. Within 12 h of their giving birth, take not fewer
respect to safety (5.2.6) and efficacy (5. 2.7) for the sows and than 15 healthy piglets from the vaccinated gilts and
gilts for which it is intended. 15 healthy piglets from the controls, taking at least 3 from
The following tests for expression of antigens (section 2-2-1)., each litter. Challenge each piglet by the oral route with a
safety (section 2-2-2) and immunogenicity (section 2-2-3) sufficient quantity of a virulent strain of E. colí before or after
may be used during the demonstration of safety and efficacy. colostrum feeding and using the same conditions for
vaccinated piglets and controls. The strain used must not be
2-2-1 Expression of antigens
one used in the manufacture of the vaccine. Retum the
The expression of antigens that stimulate a protective
piglets to their dam and observe at least daily for 8 days.
irnmune response is verified by a suitable irnmunochemical
method (2.7.1) carried out on the antigen obtained from On each day, note signs in each piglet and score using the
each of the vaccine strains under the conditions used for the following scale.
production of the vaccine. O no signs
slight diarrhoea
2-2-2 Safety 2 marked diarrhoea (watery faeces)
2-2-2-1 Safety ín pregnant sows. Carry out the test for each
3 dead
route and method of administration to be recommended for
vaccination and in pregnant sows. Use a batch of vaccine Calculate total scores for each piglet over 8 days.
containing not les s than the maximum potency that may be The test is not valid if fewer than 40 per cent of the piglets
expected in a batch of vaccine. from the control gilts die and more than 15 per cent of the
For each test, use not fewer than 8 pregnant sows per group piglets from the control gilts show no signs of illness.
that have not been vaccinated against colibacillosis, at the The vaccine complies with the test if there is a significant
relevant stages of pregnancy in accordance with the schedule reduction in score in the group of piglets from the vaccinated
to be recommended or at different stages of pregnancy. gilts compared with the group from the unvaccinated
Administer to each sow 1 dose of the vaccine. If the schedule controls.
to be recommended requires a 2nd dose, administer another For sorne adhesins (for example, F5 and F41), there is
dose after an interval of at least 14 days. Observe the sows at published evidence that high mortality cannot be achieved
least daily until farrowing. Record body temperature the day under experimental conditions. If challenge has to be carried
before vaccination, at vaccination, 2 h, 4 h and 6 h later and out with a strain having such adhesins: the test is not valid if
then daily for 4 days; note the maximum temperature fewer than 70 per cent of the control piglets show signs
increase for each sow. expected with the challenge strain; the vaccine complies with
The vaccine complies with the test if: the test if there is a significant reduction in score in the
- no sow shows abnormal local or systemic reactions or die s group of piglets from the vaccinated gilts compared with the
from causes attributable to the vaccine; group from the unvaccinated controls.
- the average temperature increase for all sows does not 2-3 MANUFACTURER'S TESTS
exceed 1.5 oC and no sow shows a rise greater than 2-3-1 Batch potency test
2.0 oC; and It is not necessary to carry out the potency test (section 3-3)
- no adverse effects on gestation or the offspring are noted. for each batch of vaccine if it has been carried out using a
2-2-2-2 Fíeld studíes. The pigs used for field trials are also batch of vaccine with a minimum potency. Where the test is
used to evaluate safety. Use not fewer than 3 groups each of not carried out, an alternative validated method is used, the
not fewer than 20 pigs with corresponding groups of not criteria for acceptance being set with reference to a batch of
fewer than 10 controls. Examine the injection site for local vaccine that has given satisfactory results in the test described
reactions after vaccination. Record body temperature the day under Potency. The following test may be used.
before vaccination, at vaccination, at the time interval after Use 7 pigs not less than 3 weeks old and that do not have
which a rise in temperature, if any, was seen in test 2-2-2-1, antibodies against the antigens stated on the label. Vaccinate
and daily during the 2 days following vaccination; note the each of 5 pigs by the recommended route and according to
maximum temperature increase for each pig. the recommended schedule. Maintain 2 pigs as controls.
The vaccine complies with the test if no pig shows abnormal Alternatively, if the nature of the antigens allows reproducible
local or systemic reactions or dies from causes attributable to results to be obtained, a test in laboratory animals (for
the vaccine, the average temperature increase for all pigs does example, guinea-pigs, mice, rabbits 01' rats) may be carried
not exceed 1.5 oC, and no pig shows a rise greater than out. To obtain a valid assay, it may be necessary to carry out
2.0 oC. a test using several groups of animals, each receiving a
2-2-3 Immunogenicity different dose. For each dose, carry out the test as follows.
Carry out the test with a challenge strain representing each Vaccinate not fewer than 5 animals with a single injection of
type of antigen against which the vaccine is intended to a suitable dose. Maintain not fewer than 2 animal s as
protect: if a single strain with all the necessary antigens is not controls. Where the recommended schedule requires a
available, repeat the test using different challenge strains. booster injection to be given, a booster vaccination may also
be given in this test provided it has been demonstrated that
Each test is carried out for each route and method of
this w111 still provide a suitably sensitive test system. At a
administration to be recommended for vaccination.
given interval within the range of 14-21 days after the last
The vaccine administered to each gilt is of minimum
injection, collect blood from each animal and prepare serum
potency.
samples. Use a suitable validated test such as an enzyme-
Use not fewer than 8 gilts susceptible to E. colí infections and linked irnmunosorbent assay (2.7.1) to measure the antibody
that do not have antibodies against the antigens to be stated response to each of the antigens stated on the label.
Vet-300 Veterinary Vaccines 2016

The vaccine complies with the test if the antibody levels in method and may be fragmented (inactivation may be by
the vaccinates are not significantly less than those obtained fragmentation); the virus 01' viral fragments may be purified
with a batch that has given satisfactory results in the test and concentrated at a suitable stage of the process.
described under Potency and there is no significant increase The vaccine may be adjuvanted.
in antibody titre in the controls.
2-2 SUBSTRATE FOR VIRUS PROPAGATION
Where animals that do not have antibodies against the 2-2-1 Cell cultures
antigens stated on the label are not available, seropositive The cell cultures comply with the requirements for cell
animals may be used in the above test. During the cultures for production of veterinary vaccines (5.2.4).
development of a test with seropositive animals, particular
care will be required during the validation of the test system 2-3 CHOICE OF VACCINE COMPOSITION
to establish that the test is suitably sensitive and to specify The vaccine is shown to be satisfactory with respect to safety
acceptable pass, fail and retest criteria. It will be necessary to (5.2.6) (including absence of adverse effects on fertility,
take into account the range of possible prevaccination titres gestation, farrowing 01' offspring) and efficacy (5.2.7) for the
and establish the acceptable minimum titre rise after pigs for which it is intended.
vaccination in relation to these. The following tests for safety (section 2-3-1) and
2-3-2 Bacteria! endotoxins irnmunogenicity (section 2-3-2) may be used during the
A test for bacterial endotoxins (2.6.14) is carried out on the demonstration of safety and efficacy.
final lot 01', where the nature of the adjuvant prevents 2-3-1 Safety
pelformance of a satisfactory test, on the bulk antigen or the 2-3-1-1 Laboratory tests. Carry out the tests for each route
mixture of bulk antigens irnmediately before addition of the and method of administration to be recommended for
adjuvant. The maximum acceptable amount of bacterial vaccination and where applicable, in pigs of each category for
endotoxins is that found for a batch of vaccine that has been which the vaccine is intended, using in each case pigs not
shown satisfactory in safety test 2-2-2-1. given under Choice older than the minimum age to be recommended for
of vaccine composition. The method chosen for determining vaccination. U se a batch of vaccine containing not less than
the amount of bacterial endotoxin present in the vaccine the maximum potency that may be expected in a batch of
batch used in the safety test for determining the maximum vaccine.
acceptable level of endotoxin is used subsequently for testing 2-3-1-1-1 General safety. For each test, use not fewer than
of each batch. . 8 pigs that do not have antibodies against porcine parvovirus
3 BATCH TESTS 01' against a fraction of the virus. Administer to each pig

3-1 Identification 1 dose of the vaccine. If the schedule to be recommended


In animals that do not have antibodies against the antigens requires a 2nd dose, administer another dos e after an interval
stated on the label, the vaccine stimulates the production of of at least 14 days. Observe the pigs at least daily until
such antibodies. 14 days after the last administration.
3-2 Bacteria and fungi The vaccine complies with the test if no pig shows notable
The vaccine, including where applicable the diluent supplied signs of disease 01' dies from causes attributable to the
for reconstitution, complies with the test for sterility vaccine during the test.
prescribed in the monograph Vaccines for veterina1Y use 2-3-1-1-2 Safety in pregnant sows. If the vaccine is intended
(0062). for use in pregnant sows, use for the test not fewer than
8 pregnant sows at the stage 01' at different stages of
3-3 Potency
pregnancy according to the recommended schedule.
The vaccine complies with the requirements of the test
Administer to each sow 1 dose of the vaccine. If the schedule
mentioned under Irnmunogenicity (section 2-2-3) when
to be recommended requires a 2nd dose, administer another
administered by a recommended route and method.
dos e after an interval of at least 14 days. Observe the sows at
_____________________________________________ ~E~

least daily until farrowing.


The vaccine complies with the test if no sow shows abnormal
local 01' systemic reactions or dies from causes attributable to
the vaccine and if no adverse effects on gestation 01' the
Porcine Parvovirus Vaccine, offspring are noted.
Inactivated 2-3-1-1-3 Safety in the pigs used in test 2-3-2 for
(Porcine Parvovirosis Vaccine (Inactivated)) irnmunogenicity. The pigs used in the test for
Ph Bur 1110nograph 0965) irnmunogenicity are also used to evaluate safety. Measure the
~E~ _____________________________________________ body temperature of each vaccinated pig at the time of
vaccination 24 h and 48 h later. Examine the injection site
1 DEFINITION after vaccination and at slaughter for local reactions.
Porcine parvovirosis vaccine (inactivated) is a preparation of The vaccine complies with the test if no pig shows:
a suitable strain of porcine parvovirus, inactivated while -- abnormal body temperature;
maintaining adequate irnmunogenic properties, 01' of a non -- other systemic reactions (for example, anorexia);
infectious fraction of the virus. This monograph applies to -- abnormal local reactions attributable to the vaccine.
vaccines intended for the active irnmunisation of sows and
2-3-1-2 Field stL(dies. The pigs used for field trials are also
gilts for protection of their progeny against transplacental
used to evaluate safety. Carry out a test in each category of
infection.
pigs for which the vaccine is intended (sows, gilts). Use not
2 PRODUCTION fewer than 3 groups each of not fewer than 20 pigs with
2-1 PREPARATION OF THE VACCINE corresponding groups of not fewer than 10 controls. Measure
The vaccine virus is grown in cell cultures. The viral the body temperature of each vaccinated pig at the time of
suspension is harvested; the virus is inactivated by a suitable
2016 Veterinary Vaccines Vet-301

vaccination, 24 h and 48 h later. Examine the injection site 3 BATCH TESTS


after vaccination and at slaughter for local reactions. 3-1 Identification
The vaccine complies with the test if no pig shows: When injected into animals that do not have specific
-- abnormal body temperature; antibodies against porcine parvovirus or the fraction of the
-- abnormal local reactions attributable to the vaccine. virus used in the production of the vaccine, on one or, if
2-3-2 Immunogenicity necessary, more than one occasion, the vaccine stimulates the
A test is carried out for each route and method of formation of such antibodies.
administration to be recommended, using in each case gilts 3-2 Bacteria and fungi
of 5-6 months old. The vaccine administered to each gilt is The vaccine, including where applicable the diluent supplied
of minimum potency. for reconstitution, complies with the test for sterility
Use for the test not fewer than 12 gilts that do not have prescribed in the monograph Vaccines for veterinalY use
antibodies against porcine parvovirus or against a fraction of (0062).
the virus. Vaccinate not fewer than 7 gilts according to the 3-3 Residuallive virus
schedule to be recommended. Maintain not les s than U se a quantity of vaccine equivalent to 10 doses. If the
5 unvaccinated gilts of the same age as controls. The interval vaccine contains an oily adjuvant, break the emulsion and
between vaccination and service is that to be recommended. separate the phases. If the vaccine contains a mineral
Mate all the gilts on 2 consecutive days immediately adjuvant, carry out an elution to liberate the virus.
following signs of oestrus. At about the 40th day of gestation, Concentrate the viral suspension 100 times by ultrafiltration
challenge each gilt with a suitable quantity of a virulent strain or ultracentrifugation. N one of the above procedures must be
of porcine parvovirus. Euthanise the gilts at about the such as to inactivate or otherwise intelfere with detection of
90 th day of gestation and examine their foetuses for infection live virus. Carry out a test for residual live virus in suitable
with porcine parvovirus as demonstrated by the presence of non-confiuent cells; after incubation for 7 days, make a
either virus or antibodies. sub culture using trypsinised cells. After incubation for a
The test is not valid if: further 7 days, examine the cultures for residual live
-- fewer than 7 vaccinated gilts and 5 control gilts are parvovirus by an immunofiuorescence test. The vaccine
challenged; complies with the test if no live virus is detected.
-- fewer than 90 per cent of piglets from the control gilts are 3-4 Specified extraneous agents
infected; Use 2 pigs that do not have antibodies against porcine
-- and the average number of piglets per litter for the parvovirus or against a fraction of the virus, against
vaccinated gilts is fewer than 6. Aujeszky's disease virus or against pestiviruses. Administer to
The vaccine complies with the test if not fewer than each pig by a recommended route a double dose of the
80 per cent of the total number of piglets from vaccinated vaccine, then another dose after 14 days. 14 days after the
gilts are protected from infection. last administration, carry out tests for antibodies. The vaccine
complies with the test if it does not stimulate the formation
2-4 MANUFACTURER'S TESTS
of antibodies against pestiviruses and against Aujeszky's
2-4-1 Residuallive virus disease virus.
A test for residual live virus is carried out on each batch of
antigen immediately after inactivation. The quantity of 3-5 Potency
inactivated viral harvest used in the test is equivalent to not The vaccine complies with the requirements of the test
less than 100 doses of the vaccine. The bulk harvest is mentioned under Immunogenicity (section 2-3-2) when
inoculated into suitable non-confiuent cells; after incubation administered by a recommended route and method.
for 7 days, a sub culture is made using trypsinised cells. After _____________________________________________ PhE~

incubation for a further 7 days, the cultures are examined for


residual live parvovirus by an immunofiuorescence test.
The inactivated viral harvest complies with the test if no live
virus is detected.
Porcine Progressive Atrophic ***
2-4-2 Batch potency test *** ***
It is not necessary to carry out the potency test (section 3-5) Rhinitis Vaccine, Inactivated ***
for each batch of the vaccine if it has been carried out using (Porcine Progressive Atrophic Rhinitis Vaccine
a batch of vaccine with a minimum potency. Where the test (In activa ted) , Ph Bur monograph 1361)
is not carried out, an alternative validated method is used, PhE~ _____________________________________________
the criteria for acceptance being set with reference to a batch
of vaccine that has given satisfactory results in the test 1 DEFINITION
described under Potency. The following test may be used. Porcine progressive atrophic rhinitis vaccine (inactivated) is a
Use not fewer than 5 guinea-pigs, 5-7 weeks old and that do preparation containing either the dermonecrotic exotoxin of
not have antibodies against porcine parvovirus or against a Pasteurella multocida, treated to render it harmless while
fraction of the virus. Vaccinate each guinea-pig by the maintaining adequate immunogenic properties, or a
subcutaneous route with a quarter of the prescribed genetically modified form of the exotoxin that has adequate
dose volume. Take blood samples after the period immunogenic properties and that is free from toxic
corresponding to maximum antibody production and carry properties; the vaccine may also contain cells andlor antigenic
out tests on the serum for specific antibodies by a compónents of one or more suitable strains of P. multocida
haemagglutination-inhibition test or other suitable test. and/or Bordetella bronchisepti~a. This monograph applies to
The vaccine complies with the test if the level of antibodies is vaccines intended for the active immunisation of sows and
not lower than that found for a batch of vaccine that has gilts for passive protection of their progeny against porcine
given satisfactory results in the test described under Potency. progressive atrophic rhinitis.
Vet-302 Veterinary Vaccines 2016

2 PRODUCTION and no pregnant sow or gilt shows a rise greater than 2.0 oC,
2-1 PREPARATION OF THE VACCINE and if no adverse effects on gestation and offspring are noted.
The bacterial strains used for production are cultured 2-4-2-2 Field studies The pigs used for field trials are also
separately in suitable media. The toxins andlor cells are used to evaluate safety. Use not fewer than 3 groups each of
treated to render them safe. The vaccine may be adjuvanted. not fewer than 20 pigs with corresponding groups of not
2-2 DETOXIFICATION fewer than 10 controls. Examine the injection site for local
A test for detoxification of the dermonecrotic exotoxin of reactions after vaccination. Record body temperature the day
P. multocida is carried out immediately after detoxification. before vaccination, at vaccination, at the time interval after
which a rise in temperature, if any, was seen in test 2-4-2-1,
The concentration of detoxified exotoxin used in the test is
not less than that in the vaccine. The suspension complies and daily during the 2 days following vaccination; note the
with the test if no toxic dermonecrotic exotoxin is detected. maximum temperature increase for each pig.
The test for detoxification is not required where the vaccine The vaccine complies with the test if no pig shows abnormal
is prepared using a toxin-like protein free from toxic local or systemic reactions or dies from causes attributable to
propel"ties, produced by expression of a modified fOlm of the the vaccine and if the average temperature increase for all
corl"esponding gene. pigs does not exceed 1.5 oC and no pig shows a rise greater
than 2.0 oC.
2-3 ANTIGEN CONTENT
The content of the dermonecrotic exotoxin of P. nzultocida in 2-4-3 Immunogenicity
the detoxified suspension 01" the toxin-like protein in the Each test is carried out for each route and method of
harvest is detelmined by a suitable immunochemical administration to be recommended, using in each case pigs
method (2.7.1), such as an enzyme-linked irnmunosorbent that do not have antibodies against the components of the
assay, and the value found is used in the fol"mulation of the vaccine, that are from a herd or herds where there are no
vaccine. The content of othel" antigens stated on the label is signs of atrophic rhinitis and that have not been vaccinated
also determined (2.7.1). against atrophic rhinitis. The vaccine administered to each
pig is of minimum potency.
2-4 CHOICE OF VACCINE COMPOSITION 2-4-3-1 Vaccines comaining dermonecrotic exotoxin of
The strains used for the preparation of the vaccine are shown P. multocida (with 01' without cells of P. nzultocida) Use not
to be satisfactory with respect to the production of the fewer than 12 breeder pigs. Vaccinate not fewer than
dermonecrotic exotoxin and the other antigens daimed to be 6 randomly chosen pigs at the stage of pregnancy or non-
protective. The vaccine is shown to be satisfactory with pregnancy and according to the schedule to be
respect to safety (5.2.6) and efficacy (5.2.7) for the sows and recommended. Maintain not fewer than 6 pigs as controls.
gilts for which it is intended. From birth allow aH the piglets from the vaccinated and
The following tests for production of antigens (section unvaccinated breeder pigs to feed from their own dam.
2-4-1), safety (section 2-4-2) and immunogenicity Constitute from the progeny 2 challenge groups each of not
(section 2-4-3) may be used during the demonstration of fewer rilan 30 piglets chosen randomly, taking not fewer than
safety and efficacy. 3 piglets from each litter. On the 2 consecutive days
2-4-1 Production of antigens preceding challenge, the mucosa of the nasal cavity of the
The production of antigens claimed to be protective is piglets may be treated by instillation of 0.5 mL of a solution
verified by a suitable bioassay or irnmunochemical method of acetic acid (10 gIL C 2 H 4 0 2 ) in isotonic buffered saline
(2.7.1), carried out on the antigens obtained from each of the pH 7.2.
vaccine strains under the conditions to be used for the Challenge each piglet at 10 days of age by the intranasal
production of the vaccine. route with a sufficient quantity of a toxigenic strain of
2-4-2 Safety P. 111ultocida. At the age of 42 days, euthanise the piglets of
2-4-2-1 Safety in p1'egnam SO'lUS Carry out the test for each the 2 groups and dissect the nose of each of them
route and method of administration to be recommended for transversally at premolar-l. Examine the ventral and dorsal
vaccination using in each case pregnant sows or gilts that do turbina tes and the nasal septum for evidence of atrophy or
not have antibodies against the components of the vaccine, distortion and grade the observations on the following scales.
from a herd or herds where there are no signs of atrophic Tu1'binates
rhinitis and that have not been vaccinated against atrophic O no atrophy
rhinitis. U se a batch containing not less than the maximum slight atrophy
potency that may be expected in a batch of vaccine. 2 moderate atrophy
Use not fewer than 8 pregnant sows or gilts per group, at the 3 severe atrophy
stage or at different stages of pregnancy according to the 4 very severe atrophy with almost complete disappearance
schedule to be recommended. Administer to each pregnant of the turbinate
sow or gilt 1 dose of the vaccine. If the schedule to be The maximum score is 4 for each turbinate and 16 for the
recommended require.s a 2nd dose, administer another dos e sum of the 2 dorsal and 2 ventral turbinates.
after an interval of at least 14 days. Observe the pregnant Nasal septwl1
sows or gilts at least daily until farrowing. Record body O no deviation
temperature the day before vaccination, at vaccination, 2 h, very slight deviation
4 h and 6 h later andthen daily for 4 days; note the 2 deviation of the septum
maximum temperature increase for each pregnant sow or gilt.
The maximum total score for the turbinates and the nasal
The vaccine complies with the test if no pregnant sow or gilt
septum is 18.
shows abnormallocal or systemic reactions or dies from
causes attributable to the vaccine, if the average temperature The test is not valid if fewer than 80 per cent of the progeny
increase for aH pregnant sows or gilts does not exceed 1.5 oC of each litter of the unvaccinated breeder pigs have a total
score of at least 10. The vaccine complies with the test if a
2016 Veterinary Vaccines Vet-303

significant reduction in the total score has been demonstrated schedule requires a booster injection to be given, a booster
in the group from the vaccinated breeder pigs compared to vaccination may also be given in this test provided it has
that from the unvaccinated breeder pigs. been demonstrated that this will still provide a suitably
2-4-3-2 Vaccines containing P. multocida de17110necrotic exotoxin sensitive test system. At a given interval within the range of
(zuith 01' 'luithout cells of P. multocida) and cells and/or antigenic 14-21 days after the last administration, collect blood from
components of B. brol1chiseptica U se not fewer d1an 24 breeder each animal and prepare serum samples. Use a validated test
pigs. Vaccinate not fewer than 12 randomly chosen pigs at such as an enzyme-linked immunosorbent assay to measure
the stage of pregnancy 01' non-pregnancy and according to the antibody response to each of the antigens stated on the
the schedule to be recommended. Maintain not fewer label.
than 12 pigs as controls. From birth allow all the piglets from The test is not valid if there is a significant antibody titre in
the vaccinated and unvaccinated breeder pigs to feed from the controls. The vaccine complies with the test if the
their own dam. U sing groups of not fewer than 6 pigs, antibody responses of the vaccinated animals are not
constitute from their progeny 2 challenge groups from significandy less than those obtained with a batch of vaccine
vaccinated pigs and 2 groups from control pigs each group that has given satisfactory results in the test or tests (as
consisting of not fewer than 30 piglets chosen randomly, applicable) described under Potency.
taking not fewer than 3 piglets from each litter. On the Where animals that do not have antibodies against the
2 consecutive days preceding challenge, the mucosa of the antigens stated on the label are not available, seropositive
nasal cavity of the piglets may be treated by instillation of animals may be used in the above test. During the
0.5 mL of a solution of acetic acid (lO gIL C ZH 4 0z) in development of a test with seropositive animals, particular
isotonic buffered saline pH 7.2. For a group ofpiglets from care will be required during the validation of the test system
not fewer than 6 vaccinated pigs and a group from not fewer to establish that the test is suitably sensitive and to specify
than 6 controls, challenge each piglet by the intranasal route acceptable pass, fail and retest criteria. It will be necessary to
at 10 days of age with a sufficient quantity of a toxigenic take into account the range of prevaccination antibody titres
strain of P. 111ultocida. For the other group of piglets from not and to establish the acceptable minimum antibody titre rise
fewer than 6 vaccinated pigs and the other group from not after vaccination in relation to these.
fewer than 6 controls, challenge each piglet at 7 days of age
2-5-2 Bacterial endotoxins
by the intranasal route with a sufficient quantity of
A test for bacterial endotoxins (2.6.14) is carried out on the
B. bronchiseptica. In addition, challenge each piglet at 10 days
batch 01', where the nature of the adjuvant prevents
of age by the intranasal route with a sufficient quantity of a
performance of a satisfactory test, on the bulk antigen or the
toxigenic strain of P. multocida. At the age of 42 days,
mixture of bulk antigens irnmediately before addition of the
euthanise the piglets of the 4 groups and dissect the nose of
adjuvant. The maximum acceptable amount of bacterial
each of them transversally at premolar-l. Examine the ventral
endotoxins is that found for a batch of vaccine shown
and dorsal turbina tes and the nasal septum for evidence of
satisfactory in safety test 2-4-2-1 given under Choice of
atrophy or distortion and grade the observations on the scale
vaccine composition or in the residual toxicity test described
described above.
under Batch tests, carried out using 10 pigs. Where the latter
The test is not valid if fewer than 80 per cent of the progeny test is used, note the maximum temperature increase for each
of each litter of the unvaccinated breeder pigs have a total pig; the vaccine complies with the test if the average
score of at least 10. The vaccine complies with the test if a temperature increase for all pigs do es not exceed 1.5 oc.
significant reduction in the total score has been demonstrated The method chosen for determining the amount of bacterial
in the groups from the vaccinated breeder pigs compared to endotoxin present in the vaccine batch used in the safety test
the corresponding group from the unvaccinated breeder pigs. for determining the maximum acceptable level of endotoxin
2-5 MANUFACTURER'S TESTS is used subsequendy for testing of each batch.
2-5-1 Batch potency test 3 BATCH TESTS
It is not necessary to carry out the potency test (section 3-4) 3-1 Identification
for each batch of vaccine if it has been carried out using a In animals that do not have specific antibodies against the
batch of vaccine with a minimum potency. Where the test is antigens stated on the label, the vaccine stimulates the
not carried out, an alternative validated method is used, the production of such antibodies.
criteria for acceptance being set with reference to a batch of
3-2 Bacteria and fungi
vaccine that has given satisfactory results in the test described
under Potency. The following test may be used. The vaccine, including where applicable the diluent supplied
for reconstitution, complies with the test for sterility
Use not fewer than 7 pigs not less than 3 weeks old and that prescribed in the monograph Vaccil1es for veterinary
do not have antibodies against the components of the use (0062).
vaccine. Vaccinate not fewer than 5 pigs by a recommended
route and according to the recommended schedule. Maintain 3-3 Residual toxicity
not fewer than 2 pigs of the same origin as controls under U se not fewer than 2 pigs that do not have antibodies against
the same conditions. Alternatively, if the nature of the P. multocida and that preferably do not have antibodies
antigens allows reproducible results to be obtained, a test in against B. bronchiseptica. Administer to each pig by a
laboratory animals that do not have antibodies against the recommerided routé a double dose of the vaccine. Observe
components of the vaccine may be carried out. To obtain a the pigs at least daily for 14 days. Record body temperature
valid assay, it may be necessary to carry out a test using the day before vaccination, at vaccination, 2 h, 4 h and 6 h
several groups of animals, each receiving a different quantity later and then daily for 2 days.
of vaccine. For each quantity of vaccine, carry out the test as It is recommended to use the mean temperature of the days
follows: vaccinate not fewer than 5 animals with a suitable before administration of the vaccine (e.g. day -3 to day O) as
quantity of vaccine. Maintain not fewer than 2 animals of the the baseline temperature to have clear guidance for
same species and origin as controls. Where the recommended evaluation of the test.
Vet-304 Veterinary Vaccines 2016

The vaccine complies with the test if no pig shows notable les s than 10 times the maximum virus titre likely to be
signs of disease or dies from causes attributable to the contained in 1 vaccine bait. Observe the animal s at least daily
vaccine; a transient temperature increase not exceeding for 180 days.
2.0 oC may occur. For each test performed in the non-target species (dogs, cats,
3-4 Potency and if appropriate, raccoon dogs), use not fewer than
The vaccine complies with the requirements of the tests 10 animals that do not have antibodies against rabies virus.
mentioned under Irnmunogenicity (section 2-4-3) when Administer orally to each animal a quantity of the vaccine
administered by a recommended route and method. virus equivalent to not les s than 10 times the maximum virus
_____________________________________________ PhE~
titre likely to be contained in 1 vaccine bait. Observe the
animals at least daily for 180 days.
The vaccine virus complies with the test if no animal shows
signs of disease and if the presence of the vaccine virus is not
demonstrated in the brain of any animal. The presence of
Rabies Vaccine (live, Oral) for rabies virus in the brain is tested using reference diagnostic
Foxes and Raccoon Oogs tests (immunofluorescence test and cell-culture test).
(Rabies Vaccille (Live, Oral) for Foxes alld Racoon 2-3-2 Stability of the genetic marker
Dogs, Ph Bur mOl1ograph 0746) Carry out the test using suckling mice that have not been
PhE~ _____________________________________________ vaccinated against rabies. Passage the vaccine virus
sequentially through 5 groups via the intracerebral route.
1 DEFINITION
Inoculate each of the 5 mice of the 1st group with a quantity
Rabies vaccine (live, oral) for foxes (Vulpes vulpes) and of the vaccine virus that will allow recovery of virus for the
raccoon dogs (Nyctereutes procyonoides) is a preparation of a passages described be!ow (e.g. not more than 0.02 mL).
suitable irnmunogenic strain of an attenuated rabies virus. When the mice show signs of rabies, but not later than
The virus strain has one or more stable gene tic markers that 14 days after inoculation, euthanise the mice and remove the
discriminates the vaccine strain from other rabies virus brain of each mouse. Prepare a suspension from the brain of
strains. The vaccine is incorporated in bait in such a manner each mouse and pool the samples. Administer not more than
as to enable the tests prescribed below to be performed 0.02 mL of the pooled samples to each mouse of the next
aseptically. The bait casing, attractive to the target species, group. Carry out this passage operation not fewer than
may contain a biomarker (e.g. tetracycline). This monograph 4 times; verify the presence of the virus at each passage.
applies to vaccines intended for the active irnmunisation of If the virus is not found at a passage level, repeat the passage
foxes, or foxes and raccoon dogs against rabies. by administration to a group of 10 animals.
2 PRODUCTION Verify the genetic marker in the vaccine virus recovered from
2-1 PREPARATION OF THE VACCINE the last passage.
The vaccine virus is grown in cell cultures. The virus The vaccine virus complies with the test if the genetic marker
suspension is harvested on one 01' more occasions within remains stable.
14 days of inoculation. Multiple harvests from a single cell
2-3-3 Irnmunogenicity
lot may be pooled and considered as a single harvest. It may
A test is carried out for the oral route of administration and
be mixed with a suitable stabiliser.
with the bait to be stated on the label using animal s of the
2-2 SUBSTRATE FOR VIRUS PROPAGATION target species (foxes, or foxes and raccoon dogs) at least
2-2-1 Cell cultures 3 months old. The quantity of vaccine virus to be
The cell cultures comply with the requirements for cell administered to each fox, or fox and raccoon dog, is not
cultures for production of veterinary vaccines (5.2.4; ij the cell greater than the minimum virus titre to be stated on the labe!
cultures are of ma711711aliall origil1, they are shozvll to be free fr0711 and the virus is at the most attenuated passage level that will
rabies virus. be present in a batch of vaccine.
2-3 CHOICE OF VACCINE VIRUS U se for the test not fewer than 35 animal s of each target
The vaccine virus is shown to be satisfactory with respect to species, that do not have antibodies against rabies virus.
safety (5.2.6) for the target species and the non-target In each target species, apply the following protocol, validity
species, and efficacy (5.2.7) for the species for which it is criteria and acceptance limits.
intended. The vaccine strain is genetically characterised by Vaccinate not fewer than 25 animals, according to the
gene sequencing. schedule to be recommended. Maintain not fewer than
The following tests for safety of the virus strain (section 10 animals as controls. Observe the animals for 180 days
2-3-1), stability ofthe genetic marker (section 2-3-2) and after vaccination. The test is not valid if fewer than
irnmunogenicity (2-3-3) may be used during the 25 vaccinated animals survive after this observation periodo
demonstration of safety and efficacy. Challenge all the animals at least 180 days after vaccination
In natural and experimental conditions, the virus strain does by intramuscular injection of a sufficient quantity of a
not spread from one animal to another in wild rodents. virulent rabies virus strain approved by the competent
authority. Observe the animals at least daily for 90 days after
2-3-1 Safety ofthe virus strain challenge. Animals that die from causes not attributable to
Administer the virus strain by the oral route. Use vaccine rabies are eliminated.
virus at the least attenuated passage level that will be present
The test is not valid if the number of suchdeaths reduces the
in a batch of the vaccine.
number of vaccinated animals in the test to fewer than 25
For each test performed in the target species (foxes, or foxes and the test is invalid unless at least 9 control animals (or a
and raccoon dogs), use not fewer than 20 animals that do statistically equivalent number if more than 10 control
not have antibodies against rabies virus. Administer orally to animals are challenged) show signs of rabies and the presence
each animal a quantity of the vaccine virus equivalent to not
2016 Veterinary Vaccines Vet- 305

of rabies virus in their brain is demonstrated by the


irnmunofluorescence test or sorne other reEable method.
Rabies Veterinary Vaccine,
The vaccine virus compEes with the test if not more than 2 Inactivated
of 25 vaccinated animals (or a statistically equivalent number (Rabies Vaccine (Inactivated) fo1' Veterinary Use~
if more than 25 vaccinated animals are challenged) show Ph Bur monograph 0451)
signs of rabies. ~Ew _____________________________________________
2-4 BAIT STABILITY
1 DEFINITION
Incubate the bait at 25 oC for 5 days. Titrate the vaccine.
Rabies vaccine (inactivated) for veterinary use is a
The virus titre must be at least the minimum vims titre
preparation of a suitable strain of fixed rabies virus,
stated on the label. Heat the bait at 40 oC for 1 h. The bait
inactivated while maintaining adequate irnmunogenic
casing complies with the test if it remains in its original shape
properties. This monograph applies to vaccines intended for
and adheres to the vaccine container.
the active irnmunisation of animals against rabies.
3 BATCH TESTS
2 PRODUCTION
3-1 Identification
3-1-1 The vaccine vims is identified by a suitable method, 2-1 PREPARATION OF THE VACCINE
e.g. when mixed with a monospecific rabies antisemm, the The vaccine is prepared from virus grown either in suitable
vaccine is no longer able to infect susceptible cell cultures celllines or in primary cell cultures from healthy animals
into which it is inoculated. (5.2.4). The virus suspension is harvested on one or more
occasions within 28 days of inoculation. Multiple harvests
3-1-2 A test is carried out to demonstrate the presence of the
from a single production cell culture may be pooled and
genetic marker. .
considered as a single harvest.
3-2 Bacteria and fungi
The vaccine compEes with the test for sterility prescribed in The vims harvest is inactivated. The vaccine may be
the monograph Vaccines for veterinary use (0062). adjuvanted.
3-3 Mycoplasmas (2.6.7) 2-2 SUBSTRATE FOR VIRUS PROPAGATION
The vaccine complies with the test for mycoplasmas. 2-2-1 Cell cultures
3-4 Extraneous agents The cell cultures comply with the requirements for cell
3-4-1 Neutralise the vaccine vims with a suitable cultures for production ofveterinary vaccines (5.2.4).
monospecific neutralising rabies virus antiserum and 2-3 CHOICE OF VACCINE COMPOSITION
inoculate into susceptible cell cultures. The vaccine complies The vaccine vims is shown to be satisfactory with respect to
with the test if it no longer provokes cytopathic effects in safety (5.2.6) and efficacy (5.2.7) for the species for which it
susceptible cell cultures, and it shows no evidence of is intended.
haemagglutinating or haemadsorbing agents.
The following tests for safety (section 2-3-1) and
3-4-2 Inoculate 1 in 10 and 1 in 1000 dilutions of the irnmunogenicity (section 2-3-2) may be used during the
vaccine into susceptible cell cultures. Incubate at 37 oc. demonstration of safety and efficacy in cats and dogs.
After 2, 4 and 6 days, stain the cells with a panel of
The suitability of the vaccine with respect to irnmunogenicity
monoclonal antibodies that do not react with the vaccine
(section 2-3-2) for carnivores (cats and dogs) is demonstrated
strain but that react with other strains of rabies vaccine (for
by direct challenge. For other species, if a challenge test has
example, street vims, Pasteur strain). The vaccine compEes
been carried out for the vaccine in cats or dogs, an indirect
with the test if it shows no evidence of contaminating rabies
test is carried out by determining the antibody level following
virus.
vaccination of not fewer than 20 animals according to the
3-5 Virus titre schedule to be recommended; the vaccine is satisfactory if,
Titrate the vaccine virus in suitable cell cultures. The vaccine after the period to be claimed for protection, the mean rabies
complies with the test if 1 dose contains not less than the virus antibody level in the semm of the animals is not less
minimum virus titre stated on the label. than 0.5 IVIrnL and if not more than 10 per cent of the
3-6 Potency animals have an antibody levelless than 0.1 IV/rnL.
The vaccine complies with the requirements of the test
2-3-1 Safety
prescribed under Irnmunogenicity (section 2-3-3) when
Carry out the test for each route and method of
administered by a recommended route and method. It is not
administration to be recommended for vaccination. V se a
necessary to carry out the potency test for each batch of the
batch of vaccine containing not less than the maximum
vaccine if it has been carried out on a representative batch
potency that may be expected in a batch of vaccine.
using a vaccinating dose containing not more than the
minimum virus titre stated on the label. For each test, use not fewer than 8 animals of the minimum
3-7 Biomarker age to be recommended and that do not have antibodies
If the bait contains a biomarker, the stabÚity of the against rabies virus. Administer to each animal 1 dose of the
?iomarker is verified by a suitable method. When tetracycline vaccine. If the schedule to be recommended requires a
lS used, the vaccine complies with the test if chemical analysis
2nd dose, administer 1 dose after an interval of at least
of the bait casing shows less than 30 per cent conversion of 14 days. Observe the animals at least daily for at least
the total amount of tetracycline into the epitetracycline 14 days after the last administration.
isomer. The vaccine complies with the test if no animal shows
4 LABELLING abnormallocal or systemic reactions or dies from causes
attributable to the vaccine.
The label states:
-- the nature of the genetic marker of the vims strain' 2-3-2 Immunogenicity
-- where applicable, the nature of the biomarker of the bait. Each test is carried out for each route and method of
_____________________________________________ PhEw administration to be recommended, using in each case
animals of the minimum age to be recommended for
Vet-306 Veterinary Vaccines 2016

vaccination. The vaccine administered to each animal is of rabies vaccine (inactivated) jor veterinalY use BRP using
minimum potency. phosphate-buffered saline (PBS) for dilution. Vaccines with a
Use for the test not fewer than 35 animals. Take a blood minimum potency requirement of 1 IU/mL are used without
samp1e from each animal and test individually for antibodies further dilution. Vaccines with a minimum potency
against rabies virus to determine susceptibility. Vaccinate not requirement of more than 1 IU/mL are diluted Witll PBS to
fewer than 25 animals, according to the schedule to be contain approximately, but not less than, 1 IU/mL.
recommended. Maintain not fewer than 10 animals as Administer by the intraperitoneal route to each mouse of one
controls. Observe all the animals for a period equal to the group 0.2 mL of the vaccine, diluted where necessary, and to
claimed duration of irnmunity. No animal shows signs of each mouse of another group 0.2 mL of the suspension of
rabies. On the last day of the claimed period for duration of rabies vaccine (inactivated) jor veterinalY use BRP. Take blood
irnmunity or later, challenge each animal by intramuscular samples 14 days after the injection and test the sera
injection with a sufficient quantity of virulent rabies virus of a individually for rabies antibody using a suitable virus
strain approved by the competent authority. Observe the neutralisation test, for example the rapid fiuorescent focus
animal s at least dai1y for 90 days after challenge. Animals inhibition test (RFFIT) described for Human rabies
that die from causes not attributable to rabies are eliminated. immunoglobulin (0723) or a suitable validated modification of
The test is not valid if the number of such deaths reduces the the RFFIT. (1)
number of vaccinated animals in the test to fewer than 25 The test is not valid if more than 2 mice injected with
and the test is invalid unless at least 8 control animals (or a the suspension of rabies vaccine (inactivated) jor veterinalY
statistically equivalent number if more than 10 control use BRP show no antibodies in their serum.
animal s are challenged) show signs of rabies and the presence Individual serum titres are determined with an appropriate
of rabies virus in their brain is demonstrated by the anti-rabies immunoglobulin reference.
fiuorescent-antibody test 01' sorne other suitable method.
The antibody titre of mice receiving the suspension of rabies
The vaccine complies with the test if not more than 2 of the
vaccine (inactivated) jor veterinalY use BRP is compared to the
25 vaccinated animal s (or a statistically equivalent number if
antibody titre of mice receiving the vaccine using a suitable
more than 25 vaccinated animals are challenged) show signs
statistical approach (5.3).
of rabies.
The vaccine complies with the test if the antibody titre of
2-4 MANUFACTURER'S TESTS
mice injected with the vaccine is significantly higher than that
2-4-1 Residuallive virus of mice injected with the suspension of rabies vaccine
The test for residuallive virus is carried out by inocuÚltion of (inactivated) jor veterinal'Y use BRP.
the inactivated virus into the same type of cell culture as that
used in the production of the vaccine 01' a cell culture shown 3 BATCH TESTS
to be at least as sensitive. The quantity of inactivated virus 3-1 Identification
harvest used is equivalent to not less than 25 doses of the Administered to animals that do not have antibodies against
vaccine. After incubation for 4 days, a sub culture is made rabies virus, the vaccine stimulates tlle production of such
using trypsinised cells; after incubation for a further 4 days, antibodies.
the cultures are examined for residual live rabies virus by an 3-2 Bacteria and fungi
immunofiuorescence test. The inactivated virus harvest The vaccine, including where applicable the diluent supplied
complies with the test if no live virus is detected. for reconstitution, complies with the test for sterility
2-4-2 Antigen content of the harvest prescribed in the monograph Vaccines jor veterinalY
The content of rabies virus glycoprotein is determined by a use (0062).
suitable irnmunochemical method (2.7.1). The content is 3-3 Residuallive virus
within the limits approved for the particular preparation. Carry out the test using a pool of the contents of
2-4-3 Antigen content of the pooled harvest 5 containers.
The quantity of rabies virus glycoprotein per dose, For vaccines which do not contain an adjuvant, carry out a
determined by a suitable irnmunochemical method (2.7.1) on suitable amplification test for residuallive virus using the
the pooled harvest irnmediately before blending, is not same type of cell culture as that used in the production of
significantly lower tllan that of a batch of vaccine that gave the vaccine 01' a cell culture shown to be at least as sensitive.
satisfactory results in the test described under Potency. The vaccine complies with the test if no live virus is detected.
2-4-4 Batch potency test
For vaccines that contain an adjuvant, inject intracerebrally
It is not necessary to carry out the potency test (section 3-4)
into each of not fewer than 10 mice, each weighing 11-15 g,
for each batch of vaccine if it has been carried out using a
0.03 mL of a pool of at least 5 times the smallest
batch of vaccine with a minimum potency. Where the test is
stated dose. To avoid interference from any antimicrobial
not carried out, an alternative validated method is used, the
preservative or the adjuvant, the vaccine may be diluted not
criteria for acceptance being set with reference to a batch of
more than 10 times before injection. In this case or if the
vaccine that has given satisfactory results in the test described
vaccine strain is pathogenic only for unweaned mice, carry
under Potency. In accordance with the provisions of the
out the test on mice 1-4 days old. Observe the animal s for
European Convention for the Protection of Vertebra te
21 days. If more than 2 animals die during the first 48 h,
Animals Used for Experimental and Other Scientific
repeat the test. The vaccine complies with the test if, from
Purposes, such an alternative validated method should
the 3 rd to the 21 st days following the injection, the animals
preferably be used for routine testing. The following
show no signs ofrabies and immunofiuorescence tests carried
serological assay has been shown to be suitable and may be
out on the brains of the animals show no indication of tlle
used provided the test for antigen content of the pooled
presence of rabies virus.
harvest (section 2-4-3) has been carried out with satisfactory
results. 1 B. IZramer et al. The rapid fluorescenct focus inhibition test is a
Use groups of not fewer than 8 female mice (strain NMRI), suitable method for batch potency testing of inactivated rabies vaccine.
each weighing 18-20 g. Prepare a 1 IU/mL suspension of Biologicals 2009; 37: 119-126.
2016 Veterinary Vaccines Vet-307

3-4 Potency intracerebrally into each mouse 0.03 mL of the suspension or


The potency of rabies vaccine is determined by comparing one of the dilutions allocated to its group. Observe the
the dose necessary to protect mice against the clinical effects animals in each group at least daily for 14 days. The test is
of the dose of rabies virus defined below, administered not valid if more than 2 mice of any group die within the
intracerebrally, with the quantity of a reference preparation, first 4 days after challenge. Record the numbers in each
calibrated in International Units, necessary to provide the group that show signs of rabies in the period 5 days to
same protection. 14 days after challenge.
The International Unit is the activity of a stated quantity of The test is invalid unless:
the International Standard. The equivalence in International - for both the vaccine to be examined and the reference
U nits of the International Standard is stated by the W orld preparation the 50 per cent protective dose lies between
Health Organization. the smallest and the largest dose given to the mice;
Rabies vaccine (inactivated) for veterinary use BRP is calibrated - the titration of the challenge suspension shows that
in International Units against the International Standard. 0.03 mL of the suspension contained at least 10 IDso;
- the confidence limits (P = 0.95) are not less than
The test described below uses a parallel-line model with at
25 per cent and not more than 400 per cent of the
least 3 points for the vaccine to be examined and the
estimated potency; when this validity criteria is not met,
reference preparation. Once the analyst has experience with
the lower limit of the estimated potency must be at least
the method for a given vaccine, it is possible to carry out a
1 IU in the smallest prescribed dose;
simplified test using 1 dilution of the vaccine to be exarnined.
- the statistical analysis shows a significant slope (P = 0.95)
Such a test enables the analyst to determine that the vaccine
and no significant deviations from linearity or parallelism
has a potency significantly higher than the required minimum
ofthe dose-response lines (P = 0.99).
but will not give full information on the validity of each
individual potency determination. It allows a considerable The vaccine complies with the test if the estimated potency is
reduction in the number of animals required for the test and not less than 1 IU in the smallest prescribed dose.
should be considered by each laboratory in accordance with Application of alternative end-points Once a laboratory has
the provisions of the European Convention for the Protection established the above assay for routine use, the lethal
of Vertebrate Animals U sed for Experimental and Other end-point is replaced by an observation of clinical signs and
Scientific Purposes. application of an end-point earlier than death to reduce
Selection and distribution of the test animals U se in the test animal suffering. The following is given as an example.
healthy female mice about 4 weeks old and from the same The progress of rabies infection in mice following
stock. Distribute the mice into at least 10 groups of not fewer intracerebral injection can be represented by 5 stages defined
than 10 mice. by typical clinical signs:
Preparation of the challenge suspension Inoculate a group of Stage 1: ruffled fur, hunched back;
mice intracerebrally with the CVS strain of rabies virus and Stage 2: slow movements, los s of alertness (circular
when the mice show signs of rabies, but before they die, movements may also occur);
euthanise the mice and remove the brains and prepare a Stage 3: shaky movements, trembling, convulsions;
homogenate of the brain tissue in a suitable diluent. Separate
Stage 4: signs of paresis or paralysis;
gross particulate matter by centrifugation and use the
supernatant as challenge suspension. Distribute the Stage 5: moribund state.
suspension in small volumes in ampoules, seal and store at a Mice are observed at least twice daily from day 4 after
temperature below -60 oc. Thaw 1 ampoule of the challenge. Clinical signs are recorded using a chart such as
suspension and make serial dilutions in a suitable diluent. that shown in Table 0451.-1. Experience has shown that
Allocate each dilution to a group of mice and inject using stage 3 as an end-point yields assay results equivalent
intracerebrally into each mouse 0.03 mL of the dilution to those found when a lethal end-point is used. This must be
allocated to its group. Observe the animal s at least daily for verified by each laboratory by scoring a suitable number of
14 days and record the number in each group that, between assays using both clinical signs and the lethal end-point.
the 5d1 and the 14m days, develop signs of rabies. Calculate
the IDso of the undiluted suspension. Table 0451.-1. - Example 01 a chart used to record clinical signs
Determination of potency of the vaccine to be examined Prepare in the rabies vaccine potency test
at least 3 serial dilutions of the vaccine to be examined and Days after challenge
3 similar dilutions of the reference preparation. Prepare the
Clinical signs 4 5 6 7 8 9 10 11
dilutions such that those containing the largest quantity of
vaccine may be expected to protect more than 50 per cent of Ruffled fUf
the animal s into which they are injected and those containing Hunched back
the smallest quantities of vaccine may be expected to protect
Slow movements
less than 50 per cent of the animals into which they are Loss of alertness
injected. Allocate each dilution to a different group of mice Circular movements
and inject by the intraperitoneal route into each mouse
0.5 mL of the dilution allocated to its group. 14 days after Shak-y movements
the injection prepare a suspension of the challenge virus such Trembling
that, on the basis of the preliminary titration, it contains Convulsions
about 50 IDso in each 0.03 mL. Inject intracerebrally into Paresis
each vaccinated mouse 0.03 mL of this suspension. Prepare Paralysis
3 suitable serial dilutions of the challenge suspension.
Allocate the challenge suspension and the 3 dilutions one to Moribund state
each of 4 groups of 10 unvaccinated mice and inject
Vet-308 Veterinary Vaccines 2016

4. LABELLING The vaccine complies with the test if no rabbit shows


The label sta tes: abnormallocal or systemic reactions or signs of disease, or
- the type of cell culture used to prepare the vaccine and dies from causes attributable to the vaccine, the average body
the species of origin; temperature increase for all animals does not exceed 1.5 oC,
- the minimum number of International Units per dose; and no animal shows a temperature rise greater than 2.0 oC.
- the minimum period for which the vaccine provides 2-3-2 Immunogenicity
protection. A test is carried out for each route and method of
_ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ PhEur administration to be recommended for vaccination.
The test is carried out using in each case rabbits not les s
than 10 weeks old. The vaccine administered to each rabbit
is of minimum potency.
Use not fewer than 15 healthy, susceptible rabbits, free from
Rabbit Haemorrhagic Disease antibodies against RHDV, from the same healthy stock, and
Vaccine (Inactivated) reared in suitable isolation conditions to ensure absence of
(Ph. Eur. 1110nograph 2325) contact with RHDV. Administer 1 dose of vaccine to each of
PhE~ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ __ not fewer than 10 of the rabbits according to the instructions
for use to be stated on the labe!' Maintain not fewer than 5
1 DEFINITION other rabbits as controls. Not less than 7 days after
Rabbit haemorrhagic disease vaccine (inactivated) is a vaccination, challenge each rabbit by a suitable route with a
preparation of a suitable strain of rabbit haemorrhagic disease quantity of a virulent strain of RHDV sufficient to cause
virus (RHDV), inactivated while maintaining adequate signs of rabbit haemorrhagic disease (RHD) in a susceptible
irnmunogenic properties. This monograph applies to vaccines rabbit. Observe the rabbits for a further 14 days.
intended for active irnmunisation of rabbits. The test is not valid if fewer than 80 per cent of control
2 PRODUCTION rabbits die with typical signs of RHD within 120 h of
2-1 PREPARATION OF THE VACCINE challenge.
The vaccine virus is grown in rabbits. The rabbits must be The vaccine complies with the test if not fewer than
healthy, not vaccinated against RHDV, free from antibodies 90 per cent of vaccinated rabbits show no signs of RHD.
against RHDV, not treated with antibiotics within at least 2-4 MANUFACTURER'S TESTS
15 days of their use and from a healthy and monitored 2-4-1 Residuallive virus
breeding unit. A suspension is prepared from a homogenate A test for residual live virus is carried out on the bulk harvest
of suitable internal organs of those rabbits that are euthanised of each batch to confirm inactivation of the RHDV. The test
or that succumb to the infection within 120 h of inoculation. for inactivation is carried out in healthy, susceptible rabbits,
The virus in the suspension may be purified and not less than 10 weeks old, free from antibodies against
concentrated, and is inactivated by a suitable method. RHDV and from the same healthy stock. 5 rabbits are
2-2 SEED LOTS inoculated by a suitable parenteral route (subcutaneous or
2-2-1 Extraneous agents intramuscular) with at least a 5 mL dose of the suspension.
Each master seed lot complies with the tests for extraneous The rabbits are observed for not less than 7 days. At the end
agents in seed lots prescribed in the monograph Vaccines 101' of the observation period, the animals are euthanised and
veterina7Ji use (0062). liver extracts are tested by a suitable method for freedom
from RHDV.
2-3 CHOICE OF VACCINE COMPOSITION
The vaccine complies with the test if no rabbit die s and no
The vaccine is shown to be satisfactory with respect to safety
RHDV antigen is detected in the livers.
(5.2.6) and efficacy (5.2.7) for the rabbits for which it is
intended. 2-4-2 Batch potency test
The following tests for safety (section 2-3-1) and It is not necessary to carry out the potency test (section 3-4)
irnmunogenicity (section 2-3-2) may be used during the for each batch of the vaccine if it has been carried out using
demonstration of safety and efficacy. a batch of vaccine with a minimum potency. Where the test
is not carried out, an alternative validated method is used,
2-3-1 Safety the criteria for acceptance being set with reference to a batch
Carry out the test for each route and method of of vaccine that has given satisfactory results in the test
administration to be recommended for vaccination and in described under Potency.
rabbits of each category for which the vaccine is intended.
The following method is given as an example. Administer
U se a batch of vaccine containing not les s than the
1 dose of vaccine intramuscularly to each of 5 healthy
maximum potency that may be expected in a batch of
rabbits, 10 weeks old, free from antibodies against RHDV
vaccine.
and from the same healthy stock. Maintain 2 rabbits as
For each test, use not fewer than 8 healthy rabbits from the unvaccinated controls. Collect serum samples from each
same stock, not older than the minimum age to be rabbit just before administration of the vaccine and after the
recommended for vaccination and free from antibodies period defined when testing the reference vaccine; determine
against RHDV. Administer to each rabbit 1 dose of the the antibody titre of each serum by a suitable irnmunological
vaccine. If the schedule to be recommended requires a 2nd method, for example, ELISA. The antibody levels are not
dose, administer 1 dose after an interval of at least 14 days. significantly lower than those obtained with a batch that has
Observe the animals for at least 14 days after the last given satisfactory results in the test described under Potency.
administration. Record the body temperature the day before
The test is not valid if the sera coIlected from the
vaccination, at vaccination, 4 h after vaccination and then
unvaccinated controls and from the rabbits just before the
daily for 4 days; note the maximum temperature increase for
each animal.
2016 Veterinary Vaccines Vet-309

administration of the vaccine show detectable specific method (2.7.1) carried out on the antigen obtained from
antibodies. each of the vaccine strains under the conditions used for the
3 BATCH TESTS production of the vaccine.
3-1 Identification 2-2-2 Safety
When injected into susceptible animals, the vaccine 2-2-2-1 Safety in pregnant animals. Carry out the test for each
stimulates the production of specific antibodies against route and method of administration to be recommended for
RHDV, detectable by a haemagglutination-inhibition test or vaccination and in pregnant animals of each species for
enzyme irnmunoassay. which the vaccine is intended. Use a batch of vaccine
containing not les s than the maximum potency that may be
3-2 Bacteria and fungi
expected in a batch of vaccine.
The vaccine, inc1uding where applicable the diluent supplied
for reconstitution,. complies with me test for sterility F 01' each test, use not fewer than 8 pregnant animal s per
prescribed in the monograph Vaccines for vetennaIY use group that have not been vaccinated against colibacillosis.
(0062). Administer to each animal 1 dose of the vaccine. If the
schedule to be recommended requires a 2nd dose, administer
3-3 Residuallive virus
another dose after an interval of at least 14 days. Observe the
U se not fewer than 2 healthy rabbits, not les s than 10 weeks
animals at least daily until parturition. Record body
old, free from antibodies against RHDV and from the same
temperature the day before vaccination, at vaccination, 2 h,
healthy stock. Administer by a recommended route to each
4 h and 6 h later and then daily for 4 days; note the
rabbit 2 dos es of vaccine. Observe the rabbits for 14 days.
maximum temperature increase for each animal.
The vaccine complies with the test if no rabbit shows notable
The vaccine complies with the test if:
signs of disease or dies from causes attributable to the
-- no animal shows abnormal local or systemic reactions or
vaccine.
dies from causes attributable to the vaccine;
3-4 Potency -- the average temperature increase for all animals does not
The vaccine complies with the requirements of the test exceed 1.5 oC and no animal shows a rise greater than
mentioned under Irnmunogenicity (section 2-3-2), when 2.0 oC; and
administered by a recommended route and method. -- no adverse effects on gestation or the offspring are noted.
_____________________________________________ PhE~
2-2-2-2 Field studies. Safety is demonstrated in field trials for
each species for which the vaccine is intended. Administer
the dose to be recommended to not fewer than 60 animals
from 3 different stocks by the route and according to the
*** schedule to be recommended. Assign not fewer than
Ruminant E. Coli Vaccine,
*** *** 30 animals from the same stocks to control groups. Observe
Inactivated *** the animals at least daily for 14 days after the last
Ruminant Escherichia Coli Vaccine, Inactivated administration.
(Neonatal Rwninant Colibacillosis Vaccine (In activa ted) )
The vaccine complies with the test if no animal shows
Ph Eur monograph 0961)
abnormal local or systemic reactions or dies from causes
~E~ _____________________________________________
attributable to the vaccine and if no rise in temperature of
more than 1.5 oC occurs within 2 days of administration of
1 DEFINITION each dose of the vaccine.
Neonatal ruminant colibacillosis vaccine (inactivated) is a 2-2-3 Irnmunogenicity
preparation from cultures of one or more suitable strains of Carry out the test with a challenge strain representing each
Escherichia coli, carrying one 01' more adhesin factors or type of antigen against which the vaccine is intended to
enterotoxins. This monograph applies to vaccines intended protect: if a single strain with all the necessary antigens is not
for the active irnmunisation of dams for passive protection of available, repeat the test using different challenge strains.
their newbom progeny against enteric forms of colibacillosis, Each test is carried out for each route and method of
administered by injection. administration to be recommended for vaccination, using in
2 PRODUCTION each case animals of each species for which the vaccine is
2-1 PREPARATION OF THE VACCINE intended. The vaccine administered to each animal is of
The E. coli strains used for production are cultured separately minimum potency.
in a suitable medium. The cells or toxins are processed to For each test, use not fewer than 15 animals that do not
render them safe while maintaining adequate irnmunogenic have antibodies against the antigens to be stated on the label.
properties and are blended. The vaccine may be adjuvanted. Take not fewer than 10 at random and vaccinate these at the
stage of pregnancy and according to the schedule to be
2-2 CHOICE OF VACCINE COMPOSITION recommended. Maintain not fewer than 5 animals as
The E. coli strains used in the production of the vaccine are controls. Collect colostrum from all animal s after parturition
shown to be satisfactory with respect to expression of and store the samples individually in conditions that maintain
antigens and the vaccine is shown to be satisfact01Y with antibody lévels. Take not fewer than 15 newbom unsuckled
respect to safety (5. 2.6) and efficacy (5.2.7) for the ruminants animals and house them in an environment ensuring absence
for which it is intended. of enteric pathogens. Allocate a colostrum sample from not
The following tests for expression of antigens (section 2-2-1), fewer tb.an 10 vaccinated dams and not fewer than 5 controls
safety (section 2-2-2) and immunogenicity (section 2-2-3) to the offspring. After birth, feed the animals with the
may be used during the demonstration of safety and efficacy. colostrum sample allocated to it. After feeding the colostrum
2-2-1 Expression of antigens and within 12 h of birth, challenge all the animals by the oral
The expression of antigens that stimulate a protective route with a sufficient quantity of a virulent strain of E. coli
irnmune response is verified by a suitable irnmunochemical
Vet-310 Veterinary Vaccines 2016

and observe at least daily for 10 days. The strain must not be the amount of bacterial endotoxin present in the vaccine
one used in the manufacture of the vaccine. batch used in the safety test for determining the maximum
On each day, note daily signs in each animal and score using acceptable level of endotoxins is used subsequently for testing
the following scale. of each batch.
O no signs 3 BATCH TESTS
slight diarrhoea 3-1 Identification
2 marked diarrhoea (watery faeces) In animals that do not have antibodies against the antigens
3 dead stated on the label, the vaccine stimulates the production of
Calculate total scores for each animal over 10 days. such antibodies.
The test is not valid if fewer than 80 per cent of the animals 3-2 Bacteria and fungi
given colostrum from the controls die or show severe signs of The vaccine, including where applicable the diluent supplied
disease. The vaccine complies with the test if there is a for reconstitution, complies with the test for sterility
significant reduction in score in the group of animal s given prescribed in the monograph Vaccines for veterinmy
colostrum from vaccinated dams compared with the group use (0062).
given colostrum from the unvaccinated controls. 3-3 Potency
2-3 MANUFACTURER'S TESTS The vaccine complies with the requirements of the test
2-3-1 Batch potency test mentioned under Immunogenicity (section 2-2-3) when
It is not necessary to carry out the potency test (section 3-3) administered by a recommended route and method.
for each batch of vaccine if it has been carried out using a _____________________________________________ PhE~

batch of vaccine with a minimum potency. Where the test is


not carried out, an alternative validated method is used, the
criteria for acceptance being set with reference to a batch of
vaccine that has given satisfactory results in the test described
under Potency. The following test may be used. Salmonella Dublin Vaccine, Living
To obtain a valid assay, it may be necessary to carry out a Calf Paratyphoid Vaccine, Living
test using several groups of animals, each receiving a DEFINITION
different dose. For each dose required, carry out the test as Salmonella Dublin Vaccine, Living is a suspension of a
follows. Use not fewer than 7 animals (for example rabbits, suitably modified rough strain of Salmonella dublin.
guinea-pigs, rats or mice) that do not have antibodies against The vaccine is prepared immediately before use by
the antigens stated on the label. Vaccinate not fewer than reconstitution from the dried vaccine with a suitable liquido
5 animals, using 1 injection of a suitable dose. Maintain
2 animals as controls. Where the recommended schedule PRODUCTION
requires a booster injection to be given, a booster vaccination The vaccine may be prepared using suitable cultures grown
may also be given in this test provided it has been in solid or liquid media. The final product is freeze dried.
demonstrated that this will still provide a suitably sensitive CAUTION There is no evidence that the vaccine is harmful to
test system. At a given interval within the range of man) but it is advisable to avoid undue exposure.
14-21 days after the last injection, collect blood from each The reconstituted vaccine complies zuith the requirements stated
animal and prepare serum samples. Use a suitable validated under VeterinalY Vaccines) zuith the follozuing modifications.
test such as an enzyme-linked irnmunosorbent assay (2.7.1)
to measure the antibody response to each of the protective IDENTIFICATION
antigens stated on the label. The vaccine complies with the Consists of a suspension of Gram-negative bacilli having the
test if the antibody levels in the vaccinates are not morphological, cultural and serological characteristics of
significantly less than those obtained with a batch that has rough strains of S. dublin.
given satisfactory results in the test described under Potency TESTS
and there is no significant increase in antibody titre in the Extraneous micro-organisms
controls. Does not contain extraneous micro-organisms. Verify the
Where animal s that do not have antibodies against the absence of micro-organisms other than Salmonella dublin as
antigens stated on the label are not available, seropositive described in the test for sterility under Veterinary Vaccines.
animal s may be used in the above test. During the Safety
development of a test with seropositive animal s, particular Complies with the test described under Veterinary Vaccines.
care will be required during the validation of the test system
Viable count
to establish that the test is suitably sensitive and to specify
No fewer than 2.5 x 10 9 S. dublin organisms in the dose
acceptable pass, fail and retest criteria. It will be necessary to
stated on the label, determined by plate counts.
take into account the range of possible prevaccination titres
and establish the acceptable minimum titre rise after STORAGE
vaccination in relation to these. When stored under the prescribed conditions the dried
2-3-2 Bacterial endotoxins vaccine may be expécted to retain its potency for not less
A test for bacterial endotoxins (2.6.14) is carried out on the than 2 years. The reconstituted vaccine should be used
final lot 01', where the nature of the adjuvant prevents irnmediately.
performance of a satisfactory test, on the bulk antigen 01' the
mixture of bulk antigens irnmediately before addition of the
adjuvant. The maximum acceptable amount of bacterial
endotoxins is that found for a batch of vaccine that has been
shown satisfactory in safety test 2-2-2-1 given under Choice
of vaccine composition. The method chosen for determining
2016 Veterinary Vaccines Vet-311

*****
vaccination. Vaccinate not fewer than 30 chickens with no
Salmenella Enteritidis Vaccine
** ** more than the minimum recommended number of doses of
(Inactivated) ter Chickens *** vaccine. Maintain not fewer than 30 chickens as controls for
(Ph. Eur. monograph 1947) each group of vaccinates. Challenge both groups, 4 weeks
~E~ _____________________________________________ after the last administration of vaccine, by oral administration
to each chicleen of a sufficient quantity of a strain of
1 DEFINITION S. enterica Enteritidis that is able to colonise chickens. Take
Salmonella Enteritidis vaccine (inactivated) for chickens is a blood samples from control chickens on the day before
preparation of a suitable strain 01' strains of challenge. Observe the chickens at least daily for 4 weeks.
Salmonella enterica Enteritidis, inactivated while maintaining Take individual fresh faeces samples on day 1 after challenge
adequate immunogenic properties. This monograph applies and at least twice weeldy (including day 7) until 14 days after
to vaccines intended for administration to chickens for challenge. Test the fresh faeces samples for the presence of
reducing S. enterica Enteritidis colonisation and faecal S. enterica Enteritidis by direct plating. Euthanise all surviving
excretion of S. enterica Enteritidis. chickens at the end of the observation period, take samples of
2 PRODUCTION liver and spleen and test for the presence of S. entel'ica
Enteritidis by an appropriate method.
2-1 PREPARATION OF THE VACCINE
The seed material is cultured in a suitable medium; each The test is not valid if antibodies against S. entel'ica
strain is cultivated separately. During production, various Enteritidis are found in any control chicleen before challenge.
parameters such as growth rate are monitored by suitable The vaccine complies with the test if:
methods; the values are within the limits approved for the - the number of S. enterica Enteritidis in fresh faeces
particular vaccine. Purity of the cultures and identity are samples from vaccinated chickens after challenge at the
verified on the harvest using suitable methods. After different days of sampling is significantly lower in
cultivation, the bacterial harvests are collected separately, vaccinates than in controls and remains lower until the
inactivated by a suitable method, and blended. The vaccine end of the test;
may contain adjuvants. - the number of positive samples of liver and spleen is
significant1y lower in vaccinates than in controls.
2-2 CHOICE OF VACCINE COMPOSITION
The vaccine is shown to be satisfactory with respect to safety 2-3 MANUFACTURER'S TEST
(5. 2.6) and efficacy (5.2.7) for the birds for which it is 2-3-1 Batch potency test
intended. It is not necessary to carry out the potency test (section 3-3)
The following tests for safety (section 2-2-1) and for each batch of the vaccine if it has been carried out using
immunogenicity (section 2-2-2) may be used during the a batch of vaccine with a minimum potency. Where the test
demonstration of safety and efficacy. is not carried out, an altemative validated method is used,
the criteria for acceptance being set with reference to a batch
2-2-1 Safety of vaccine that has given satisfactory results in the test
The test is carried out for each route of administration to be described under Potency. The following test may be used.
recommended for vaccination, using in each case chickens
Use not fewer than 15 SPF chickens (5.2.2). Maintain not
not older than the minimum age to be recommended for
fewer than 5 SPF chickens as cont:rols. Administer to each of
vaccination and from a flock free from specified
10 chickens 1 dose of vaccine by a recommended route.
pathogens (SPF) (5.2.2). U se a batch of vaccine containing
Where the schedule stated on the label requires a booster
not less than the maximum potency that may be expected in
injection to be given, a booster vaccination may also be given
a batch of vaccine.
in this test provided it has been demonstrated that t11is will
For each test perfOlmed in chickens younger than 3 weeks of still provide a suitably sensitive test system. At a given
age, use not fewer than 10 chickens. For each test performed interval after the last injection, collect blood from each
in chickens older than 3 weeks of age, use not fewer than 8 vaccinated and control chicleen and prepare serum samples.
chickens. Administer by a route and method to be Measure the titre of antibodies against S. enterica Enteritidis
recommended to each chicleen 1 dose of the vaccine. If the in each serum sample using a suitable validated serological
schedule to be recommended requires a 2nd dose, administer method. Calculate the titre for t11e group of vaccinates.
1 dose to each chicleen after an interval of at least 14 days.
The test is not valid if specific S. enterica Enteritidis
Observe the chickens at least daily for at least 14 days after
antibodies are found in 1 01' more sera from control chickens
the last administration of the vaccine.
at a given interval afterthe time of administration of the
The test is not valid if more than 10 per cent of the chickens vaccine in the vaccinated group.
younger than 3 weeks of age show abnormal signs of disease
The vaccine complies with the test if the antibody titres of
01' die from causes not attributable to the vaccine.
the group of vaccinates at a given interval after each
For chickens older than 3 weeks of age, the test is not valid if
vaccination, where applicable, are not significantly lower than
non-specific mortality occurs.
the value obtained with a batch that has given satisfactory
The vaccine complies with the test if no chicleen shows results in the test described under Potency (section 3-3).
abnormal signs of disease or die s from causes attributable to
the vaccine. 3 BATCH TESTS
3-1 Identification
2-2-2 Irnmunogenicity
In ani;mals that do not have antibodies against S. enterica
A test is carried out for each route and method of
Enteritidis, the vaccine stimulates the production of such
administration to be recommended for vaccination.
antibodies.
The vaccine administered to each animal is of minimum
potency. 3-2 Bacteria and fungi
Use for the test not fewer than 60 SPF chickens (5.2.2) not The vaccine, including where applicable the diluent supplied
older than the minimum age to be recommended for for reconstitution, complies with the test for sterility
Vet-312 Veterinary Vaccines 2016

prescribed in the monograph Vaccines for vetel'inaJJ' use 2-2-1 Safety


(0062). Unless otherwise indicated below, carry out each test by the
3-3 Potency oral route of administration, using chickens from a flock free
The vaccine complies with the requirements of the test from specified pathogens (SPF) (5.2.2) not older than the
mentioned under Irnmunogenicity (section 2-2-2) when minimum age to be recommended for vaccination and that
administered by a recommended route and method. are free from antibodies against Salmonella spp. Where the
_____________________________________________ PhEw vaccine is recommended for administration to 1-day-old
chickens, the vaccine is administered before food is provided.
Use vaccine bacteria at the least attenuated passage level that
will be present in a batch of vaccine.
Measures taken to ensure absence of contamination by
Salmonella Enteritidis Vaccine ***
*** ***
Salmonella spp. fram the environment before the start of the
test and on a regular ongoing basis are described and
(Live, Oral) tor Chickens *** justified.
(Ph. Eur. monograph 2520) Whenever possible, items taken into the facilities are
~Ew _____________________________________________
sterilised.
1 DEFINITION For re-isolation of the vaccine strain, suitably sensitive
Salmonella Enteritidis vaccine (live, oral) for chickens is a validated methods that are optimal for the vaccine strain
preparation of a suitable strain of live Salmonella enterica concemed are used. The presence of relevant markers is
Enteritidis. This monograph applies to vaccines intended for confirmed to demonstrate that the organisms isolated are
the active irnmunisation of chickens against colonisation by vaccine-derived and not wild-type contaminants.
and faecal excretion of S. enterica Enteritidis. 2-2-1-1 General safety. For each test performed in chickens
2 PRODUCTION younger than 3 weeks of age, use not fewer than 10 chickens
that are free from antibodies against Salmonella spp.
2-1 PREPARATION OF THE VACCINE
Administer orally to each chicken a quantity of the vaccine
The vaccine strain is cultured in a suitable medium. During
strain equivalent to not less than 10 times the maximum
production, various parameters such as growth rate are
titre(s) likely to be contained in 1 dose of the vaccine.
monitored by suitable methods; the values are within the
Observe the cliickens at least daily for at least 14 days.
limits approved for the particular vaccine. Purity and identity
of the cultures are verified on the harvest using a The test is not valid if more than 10 per cent of the chickens
combination of methods such as morphological, serological younger than 3 weeks of age show abnormal signs or die
and biochemical methods and culture on appropriate from causes not attributable to the vaccine.
selective media. Suitable testes) are conducted to confirm the The vaccine complies with th,e test if no chicken shows
presence of relevant marker(s). The harvests are shown to be notable signs of disease or dies from causes attributable to
pure and the results obtained from the tests for identity are the vaccine.
in accordance with the documented characteristics of the 2-2-1-2 ExcretionJ duration of excretion and survival in the
strain. enviro711uent. The same animals can be used for the test for
2-2 CHOICE OF VACCINE STRAIN spread of the vaccine strain (section 2-2-1-3) provided they
The vaccine strain is shown to be satisfactory with respect to are of the minimum age to be recommended for vaccination.
safety (5.2.6) and efficacy (5.2.7) for the chickens for which Use for the test not fewer than 10 chickens. Administer
it is intended. During development, the safety of the vaccine orally to each chicken a quantity of the vaccine equivalent to
for the persons handling the vaccine or vaccinated chickens not less than the maximum titre of the strain under study
has to be addressed as well as, in accordance with the likely to be contained in 1 dose of the vaccine. Samples are
requirements of general chapter 5.2.6, the safety of the collected for re-isolation of vaccine from cloacal swabs from
spread of the vaccine to other susceptible species. The strain each chicleen and floor faeces on days 3, 7, 10 and 14 after
has a stable marker or markers to distinguish it from wild- vaccination and then weekly uiltil 3 consecutive negative
type strains. weekly samples are obtained from all vaccinated chickens.
Samples are collected for re-isolation of the vaccine strain
The following tests described under General safety
from the caeca of vaccinates at the end of the test.
(section 2-2-1-1), Excretion, duration of excretion and
survival in the environment (section 2-2-1-2), Spread of the The test is not valid if more than 10 per cent of vaccinated
vaccine strain (section 2-2-1-3), Dissemination and survival chickens show abnormal clinical signs or die from causes not
of the vaccine strain in vaccinated chickens after each attributable to the vaccine. The results are noted and used to
vaccination (section 2-2-1-4), Increase in virulence formulate the label statement on the length of time of
(section 2-2-1-5), Field trials (section 2-2-1-6), and excretion of the vaccine strain. '
Irnmunogenicity (sections 2-2-2-1 and 2-2-2-2) may be used 2-2-1-3 Spread ofthe vaccine strain. The same animal s can be
during the demonstration of safety and efficacy. used for the test for excretion, duration of excretion and
For vaccines intended to prevent and/or reduce colonisation survival in the environment(section 2-2-1-2). Use for the test
by and faecal excretion of S. enterica Enteritidis, the test for not fewer than 10 chickens of the minimum age to be
irnmunogenicity (section 2-2-2-1) is suitable to demonstrate recommended for vaccination. Use 10 chickens as controls.
that the vaccine is suitably irnmunogenic. Administer orally to each chicleen a quantity of me vaccine
When the vaccine is recommended for use in laying chickens, strain equivalent to not less than the maximum titre of the
the continuing irnmunogenicity of the vaccine until the end strain lileely to be contained in 1 dose of the vaccine. 1 day
of the laying period has to be demonstrated and the test for after vaccination, mix the 10 vaccinates with at least 10 non-
irnmunogenicity at the end of the laying period (section vaccinated chickens of the same age and source. Samples are
2-2-2-2) is suitable. collected for the re-isolation of the vaccine strain from cloacal
2016 Veterinary Vaccines Vet-313

swabs from each chicken and fioor faeces on days 3, 7, 10 evidence of an increase in virulence indicative of reversion
and 14 after vaccination and then weekly until 3 consecutive during the observation period, carry out the test for general
negative weekly samples are obtained from all chickens. safety (section 2-2-1-1), using the material used for the 1st
Collect samples of the caeca and spleens for re-isolation of passage and the bacteria at the last passage level where it was
the vaccine strain from 10 in-contact control chickens at the recovered. Test the bacteria recovered for the final passage
end of the test. The results are noted and used to formulate for the presence and stability of the marker(s).
the label statement on the extent to which the vaccine The vaccine strain complies with the test if no indication of
spreads to in-contact non-vaccinated chickens. increased virulence of the bacteria recovered for the final
2-2-1-4 Dissemination and survival 01 the vaccine strain in passage compared with the material used for the 1st passage
vaccinated chickens after each vaccination. Conduct the test is observed and the presence of the marker(s) is confirmed in
after each vaccination as prescribed by the vaccination the bacteria recovered for the final passage and remains
schedule to be recornmended in chickens of each category for identical to the material used for the 1st passage. If the
which the vaccine is intended with oral administration of the bacteria are not recovered after an initial passage in 5 animal s
test preparation. U se a sufficient number of chickens to and a subsequent repeat passage in 10 animals, the vaccine
conduct the sampling described below, the number of also complies with the test.
chickens required being dependent on the number of 2-2-1-6 Field tnals. The chickens used for field trials are also
vaccinations to be recommended, the interval between used to evaluate safety. A trial is carried out in each category
vaccinations and the length of time chickens are maintained of chickens for which the vaccine strain is intended, in not
after the last vaccination. Administer to each chicken a fewer than 2 sets of premises. Samples are taken from a
quantity of the vaccine strain equivalent to not les s than the significant number of chickens for re-isolation of bacteria to
maximum titre of the strain under study likely to be pravide information on the persistence, dissemination and
contained in 1 dose of the vaccine. Collect cloacal swabs spread of the bacteria, which can be used, with the data from
from each chicken for re-isolation of the vaccine strain the laboratory studies, to formulate the statements on the
on days 7 and 14 after each vaccination and at later label. The samples include cloacal swabs, fioor faeces, spleen
appropriate stages and with sufficient frequency to determine and liver and, in laying chickens, samples of ovaries and
the duration of dissemination. oviducts. Environmental samples are also tested at regular
For example, for broilers, samples are collected from 5 intervals.
chickens for re-isolation of the vaccine strain on days 7 and 2-2-2 Immunogenicity
14 after each vaccination and weekly until 8 weeks of age. The tests described in section 2-2-2-1 and, if appropriate, in
At the 7- and 14-day sampling points, samples are taken section 2-2-2-2 are carried out using chickens not older than
from the liver, caecum and spleen of 5 chickens. the mínimum age to be recommended for vaccination and
In the case of chickens intended for laying, samples are that are free from antibodies against Sal11l0nella spp.
collected from 5 chickens on days 7 and 14 after each The quantity of the vaccine strain to be administered orally
vaccination and weekly until 3 consecutive negative weeldy to each chicken is not greater than the minimun1 titre to be
samples are obtained or the time of the next vaccination is stated on the label.
reached, whichever is the sooner. At the 7- and 14-day Measures taken to ensure absence of contamination by
sampling points, samples are taken from the liver, caecum Salmonella spp. from the environment before the start of the
and spleen of 5 chickens. In addition, samples are collected test and on a regular ongoing basis are described and
from ovaries and oviducts where dissemination in vaccinated justified.
future layers is being investigated.
Whenever possible, items taken into the facilities are
The test is not valid if more than 10 per cent of vaccinated sterilised.
chickens in any group show abnormal clinical signs or die
Suitably sensitive validated methods are used for re-isolation
from causes not attributable to the vaccine. The results are
of bacteria derived from the challenge and for distinguishing
noted and used to formulate the label statement on the
these from the vaccine strain.
length of time the vaccine strain survives in the body and to
define a suitable withdrawal periodo 2-2-2-1 Immunogeniciry. Use for the test not fewer than
40 chickens of the same origin and from an SPF fiock
2-2-1-5 Increase in virulence. Carry out the test according to
(5.2.2). Vaccinate according to the schedule to be
general chapter 5.2.6 using SPF chickens (5.2.2) not older
recommended not fewer than 20 chickens with a single dose
than the mínimum age to be recommended for vaccination.
of vaccine. Maintain not fewer than 20 chickens as controls.
Administer orally to each chicken of the 1st group a quantity Challenge each chicken after 14 days by a suitable route with
of the vaccine strain of the strain under study that will allow a sufficient quantity of a virulent strain of S. enterica
recovery of bacteria for the passages described below. 4 to Enteritidis to give a valid test. Collect cloacal swabs from
7 days after administration of the vaccine strain, prepare a vaccinates and controls on days 3, 5, 7, 10 and 14 post-
suspension from the liver, spleen and caecum of chickens and challenge. Samples of caecum, liver and spleen are collected
pool these samples. Administer pooled samples orally to each from 10 chickens of each group on days 7 and 14 post-
chicken of the next group. Carry out this passage operation challenge for re-isolation of challenge bacteria. Collect the
not fewer than 4 times; verify the presence of the bacteria at same samples of internal organs from any chicken that dies.
each passage. If the bacteria are not found at a passage level, Examine the samples for the presence of the challenge
repeat the passage by administration to a group of organisms using a suitable sensitive culture medium and
10 chickens. Any mortalities are investigated for the presence compare results for vaccinates and controls.
of the vaccine strain and the properties of any re-isolated
The test is not valid if, during the observation period, fewer
vaccine strain determined.
than 80 per cent of the control chickens die or challenge
Carry out the test for excretion, duration of excretion and organisms are re-isolated fram fewer than 80 per cent of the
survival in the environment (section 2-2-1-2) and, if the last control chickens.
group of birds from which the bacteria was recovered shows
V et- 314 Veterinary Vaccines 2016

The vaccine complies with the test if there is a significant 3-4 Potency
reduction in the total number of cloacal swabs from The vaccine complies with the requirements of the test
vaccinates containing challenge organisms compared with the prescribed under Immunogenicity (section 2-2-2-1) when
number from the controls and there is a significant reduction administered by a recommended route and method. It is not
in the number of samples from internal organs from necessary to carry out the potency test for each batch of the
vaccinates containing challenge bacteria compared with the vaccine if it has been carried out on a representative batch
number from the controls. using a vaccinating dose containing not more than the
2-2-2-2 Immunogenicity at the end ofthe laying periodo Use for minimum titre stated on the labe!'
the test not fewer than 40 chickens of the same origin and 4 LABELLING
from an SPF fiod: (5.2.2). Vaccinate not fewer than 20 The label sta tes:
chickens according to the schedule to be recommended. -- the nature of the markers allowing the vaccine to be
Maintain not fewer than 20 chickens as controls. At the end distinguished from wild-type strains;
of the laying period, take serum samples and cloacal swabs -- the extent to which the vaccine spreads and is transmitted
from the chickens and environmental samples from the to non-vaccinated chickens and the time over which this
housing area. Test each serum sample individually for the could occur;
presence of antibodies to S. enterica Enteritidis and each -- the time that the vaccine survives in the body;
cloacal swab and environmental sample for the presence of -- the length of time of excretion and the time that the
Salmonella spp. Challenge each chicken by a suitable route vaccine survives in the environment;
with a sufficient quantity of a virulent strain of S. enterica -- the potential for spread to other susceptible species
Enteritidis to give a valid test. Collect cloacal swabs from including humans;
vaccinates and control s on days 3, 5, 7, 10 and 14 post- -- the withdrawal periodo
challenge. Samples of caecum, liver, spleen, ovaries and _____________________________________________ ~Ew

oviducts are collected from 10 chickens of each group for


re-isolation of challenge bacteria on days 7 and 14 post-
challenge. Collect the same samples of interna! organs from
any chicken that dies during the observation periodo Examine
the samples far the presence of the challenge organisms with
Salmonella Typhimurium Vaccine ***
growth in a suitable medium and compare results far ** **
vaccinates and controls. (Inactivated) tor Chickens *****
The test is not valid if, befare the challenge, antibodies to (Ph. Eur. monograph 2361)
Sa171l0nella spp. are found in the serum of the controls or ~Ew ___________________________________________
Sabnonella spp. bacteria are isolated from any of the chickens.
The test is also not valid if the challenge organisms are 1 DEFINITION
re-isolated from fewer than 80 per cent of the control Salmonella Typhimurium vaccine (inactivated) for chickens is
chickens. a preparation of a suitable strain or strains of
The vaccine complies with the test if there is a significant Salmonella enterica Typhimurium, inactivated while
reduction in the number of cloacal swabs from vaccinates maintaining adequate immunogenic properties. This
containing challenge organisms compared with the number monograph applies to vaccines intended for administration to
from the controls and there is a significant decrease in the chickens for reducing S. enterica Typhimurium colonisation
number of samples of internal organs from vaccinates and faecal excretion of S. enterica Typhimurium.
containing challenge bacteria compared with the number 2 PRODUCTION
from the controls. 2-1 PREPARATION OF THE VACCINE
3 BATCH TESTS The seed material is cultured in a suitable medium; each
3-1 Identification strain is cultivated separately. During production, various
The strain present in the vaccine is identified by a parameters such as growth rate are monitored by suitable
combination of methods such as morphological, serological methods; the values are within -fue limits approved for the
and biochemical methods and culture on appropriate particular vaccine. Purity of the cultures and identity are
selective media. Suitable testes) are conducted to confirm the verified on the harvest using suitable methods. After
presence of the relevant marker(s). cultivation, the bacterial harvests are collected separately,
inactivated by a suitable method, and blended. The vaccine
3-2 Bacteria and fungi may contain adjuvants.
Can)T out the test by microscopic examination and by
inoculation of suitable media, or verify the absence of micro- 2-2 CHOICE OF VACCINE COMPOSITION
organisms other than the vaccine strain present in the vaccine The vaccine is shown to be satisfactory with respect to safety
as described in the test for sterility prescribed in the (5.2.6) and efficacy (5.2.7) for the birds for which it is
monograph Vaccines for veterina/Y use (0062). The vaccine intended.
complies with the test if it do es not contain extraneous The following tests for safety (section 2-2-1) and
micro-organisms. immunogenicity (section 2-2-2) may be used during the
Any diluent supplied for reconstitution of the vaccine demonstration of safety and efficacy.
complies with the test for sterility prescribed in the 2-2-1 Safety
monograph Vaccines for veteril1a/Y use (0062). The test is carried out for each route of administration to be
3-3 Live bacteria recommended for vaccination, using il1 eaéh case chickens
Titrate the vaccine strain using a suitable medium for the not older than the minimum age to be recommended for
culture of the strain. The vaccine complies with the test if it vaccination and from a fiod: free from specified
contains not less than the titre stated on the labe!' pathogens (SPF) (5.2.2). Use a batch ofvaccine containing
2016 Veterinary Vaccines Vet-315

not less than the maximum potency that may be expected in 10 chickens 1 dose of vaccine by a recommended route.
a batch of vaccine. Where the schedule stated on the label requires a booster
For each test performed in chickens younger than 3 weeks of injection to be given, a booster vaccination may also be given
age, use not fewer than 10 chickens. For each test performed in this test provided it has been demonstrated that this will
in chickens older than 3 weeks of age, use not fewer than still provide a suitably sensitive test system. At a given
8 chickens. Administer by a route and method to be interval after the last injection, collect blood from each
recommended to each chicken 1 dose of the vaccine. If the vaccinated and control chicken and prepare serum samples.
schedule to be recommended requires a 2nd dose, administer Measure the titre of antibodies against S. enterica
1 dose to each chicken after an interval of at least 14 days. Typhimurium in each serum sample using a suitable
Observe the chickens at least daily for at least 14 days after validated serological method. Calculate the titre for the group
the last administration of the vaccine. of vaccinates.
The test is not valid if more than 10 per cent of the chickens The test is not valid if specific S. enterica Typhimurium
younger than 3 weeks of age show abnormal signs of disease antibodies are found in 1 or more sera from control chickens
01' die from causes not attributable to the vaccine. at a given interval after the time of administration of the
For chickens older than 3 weeks of age, the test is not valid if vaccine in the vaccinated group.
non-specific mortality occurs. The vaccine complies with the test if the antibody titres of
The vaccine complies with the test if no chicken shows the group of vaccinates at a given interval after each
abnormal signs of disease 01' dies from causes attributable to vaccination, where applicable, are not significantly lower than
the vaccine. the value obtained with a batch that has given satisfactory
2-2-2 Immunogenicity results in the test described under Potency (section 3-3).
A test is. carried out for each route and method of 3 BATCH TESTS
administration to be recommended for vaccination. 3-1 Identification
The vaccine administered to each animal is of minimum In animals that do not have antibodies against S. enterica
potency. Typhimurium, the vaccine stimulates the production of such
Use for the test not fewer than 60 SPF chickens (5.2.2) not antibodies.
older than the minimum age to be recommended for 3-2 Bacteria and fungi
vaccination. Vaccinate not fewer than 30 chickens with no The vaccine, including where applicable the diluent supplied
more than the minimum number of dos es of vaccine to be for reconstitution, complies with the test for sterility
recommended. Maintain not fewer than 30 chickens as prescribed in the monograph Vaccines for veterinary use
controls for each group of vaccinates. Challenge both groups, (0062).
4 weeks after the last administration of vaccine, by oral
3-3 Potency
administration to each chicken of a sufficient quantity of a
The vaccine complies with the requirements of the test
strain of S. enterica Typhimurium that is able to colonise
mentioned under Immul10genicity (section 2-2-2) when
chickens. Take blood samples from control chickens on the
administered by a recommended route and method.
day before challenge. Observe the chickens at least daily for
_____________________________________________ PhE~
4 weeks. Take individual fresh faeces samples on day 1 after
challenge and at least twice weekly (including day 7) until
14 days after challenge. Test the fresh faeces samples for the
presence of S. enterica Typhimurium by direct plating.
Euthanise all surviving chickens at the end of the observation
period, take samples of liver and spleen and test for the
Salmonella Typhimurium Vaccine
presence of S. enterica Typhimurium by an appropriate (live, Oral) for Chickens
method. (Ph. Eur. 11l0nograph 2521)
The test is not valid if antibodies against S. enterica PhE~ _____________________________________________
Typhimurium are found in any control chicken befare
challenge. 1 DEFINITION
Salmonella Typhimurium vaccine (live, oral) for chickens is a
The vaccine complies with the test if:
preparation of a suitable strain of live Salmonella enterica
-- the number of S. enterica Typhimurium in fresh faeces
Typhimurium. This monograph applies to vaccines intended
samples from vaccinated chickens after challenge at the
for the active immunisation of chickens against colonisation
different days of sampling is significantly lower in
by and faecal excretion of S. enterica Typhimurium.
vaccinates than in controls and remains lower until the
end of the test; 2 PRODUCTION
-- the number of positive samples of liver and spleen is 2-1 PREPARATION OF THE VACCINE
significantly lower in vaccinates than in controls. The vaccine strain is cultured in a suitable medium. During
2-3 MANUFACTURER'S TEST production, various parameters such as growth rate are
2-3-1 Batch potency test monitored by suitable methods; the values are within the
It is not necessary to carry out the potency test (section 3-3) limits approved for the particular vaccine. Purity and identity
for each batch of the vaccine if it has been carried out using of the cultures are verified on the harvest using a
a batch of vaccine with a minimum potency. Where the test combination of methods such as morphological, serological
is not carried out, an altemative validated method is used, and biochemical methods and culture on appropriate
the criteria for acceptance being set with reference to a batch selective media. Suitable testes) are conducted to confirm the
of vaccine that has given satisfactory results in the test presence of relevant marker(s). The harvests are shown to be
described under Potency. The following test may be used. pure and the results obtained from the tests for identity are
Use not fewer than 15 SPF chickens (5.2.2). Maintain not in accordance with the documented characteristics of the
fewer than 5 SPF chickens as controls. Administer to each of strain.
Vet-316 Veterinary Vaccines 2016

2-2 CHOICE OF VACCINE STRAIN 2-2-1-2 ExcretionJ duration of excretion and survival in the
The vaccine strain is shown to be satisfactory with respect to environJ1Zent. The same animal s can be used for the test for
safety (5.2.6) and efficacy (5.2.7) for the chickens for which spread of the vaccine strain (section 2-2-1-3) provided they
it is intended. During development, the safety of the vaccine are of the minimum age to be recommended for vaccination.
for the persons handling the vaccine or vaccinated chickens U se for the test not fewer than 10 chickens. Administer
has to be addressed as well as, in accordance with the orally to each chicken a quantity of the vaccine equivalent to
requirements of general chapter 5.2.6, the safety of the not less than the maximum titre of the strain under study
spread of the vaccine to other susceptible species. The strain likely to be contained in 1 dose of the vaccine. Samples are
has a stable marker or markers to distinguish it from wild- collected for reisolation of vaccine from cloacal swabs from
type strains. each chicken and fioor faeces on days 3, 7, 10 and 14 after
The following tests described under General safety vaccination and then weekly until 3 consecutive negative
(section 2-2-1-1), Excretion, duration of excretion and weekly samples are obtained fram all vaccinated chickens.
survival in the environment (section 2-2-1-2), Spread of the Samples are collected for re-isolation of the vaccine strain
vaccine strain (section 2-2-1-3), Dissemination and survival fram the caeca of vaccinates at the end of the test.
of the vaccine strain in vaccinated chickens after each The test is not valid if more than 10 per cent of vaccinated
vaccination (section 2-2-1-4), Increase in virulence chickens show abnormal clinical signs or die fram causes not
(section 2-2-1-5), Field trials (section 2-2-1-6), attributable to the vaccine. The results are noted and used to
Irnmunogenicity (sections 2-2-2-1 and 2-2-2-2) may be used formulate the label statement on the length of time of
during the demonstration of safety and efficacy. excretion of the vaccine strain.
For vaccines intended to prevent and/or reduce colonisation 2-2-1-3 Spread ofthe vaccine strain. The same animal s can be
by and faecal excretion of S. enterica Typhimurium, the test used for the test for excretion, duration of excretion and
for irnmunogenicity (section 2-2-2-1) is suitable to survival in the environment (section 2-2-1-2). Use for the test
demonstrate that the vaccine is suitably irnmunogenic. not fewer than 10 chickens of the minimum age to be
When the vaccine is recommended for use in laying chickens, recommended for vaccination. Use 10 chickens as controls.
the continuing irnmunogenicity ofthe vaccil1'e until the end Administer orally to each chicleen a quantity of the vaccine
of the laying period has to be demonstrated and the test for strain equivalent to not less than the maximum titre of the
immunogenicity at the end of the laying period (section strain likely to be contained in 1 dos e of the vaccine. 1 day
2-2-2-2) is suÍtable. after vaccination, mix the 10 vaccinates with at least 10 non-
vaccinated chickens of the same age and source. Samples are
2-2-1 Safety
collected for the re-isolation of the vaccine strain from cloacal
Unless otherwise indicated below, carry out each test by the
swabs from each chicleen and fioor faeces on days 3, 7, 10
oral route of administration, using chickens fram a fiock free
and 14 after vaccination and then weeldy until 3 consecutive
from specified pathogens (SPF) (5.2.2) not older than the
negative weeldy samples are obtained from aH chickens.
minimum age to be recommended for vaccination and that
Collect samples of the caeca and spleens for re-isolation of
are free from antibodies against Salman ella spp. Where the
the vaccine strain from 10 in-contact control chickens at the
vaccine is recommended for administration to 1-day-old
end of the test. The results are noted and used to fonnulate
chickens, the vaccine is administered before food is provided.
the label statement on the extent to which the vaccine
Use vaccine bacteria at the least attenuated passage level that
spreads to in-contact non-vaccinated chickens.
will be present in a batch of vaccine.
2-2-1-4 Dissemination and sUl'vival of the vaccine strain in
Measures taken to ensure absence of contamination by
vaccinated chickens afier each vaccination. Conduct the test
Salmonella spp. fram the environment before the start of the
after each vaccination as prescribed by the vaccination
test and on a regular ongoing basis are described and
schedule to be recommended in chickens of each category for
justified.
which the vaccine is intended with oral administration of the
Whenever possible, items taken into the facilities are test preparation. Use a sufficient number of chickens to
sterilised. conduct the sampling described below, the number of
For re-isolation of the vaccine strain, suitably sensitive chickens required being dependent on the number of
validated methods that are optimal for the vaccine strain vaccinations recommended, the interval between vaccinations
concemed are used. The presence of relevant markers is and the length of time chickens are maintained after the last
confirmed to demonstrate that the organisms isolated are vaccination. Administer to each chicleen a quantity of the
vaccine-derived and not wild-type contaminants. vaccine strain equivalent to not less than the maximum titre
2-2-1-1 General safety. For each test performed in chickens of the strain under study likely to be contained in 1 dose of
younger than 3 weeks of age, use not fewer than 10 chickens the vaccine. Collect cloacal swabs fram each chicleen for
that are free from antibodies against re-isolation of vaccinal bacteria on days 7 and 14 after each
Salmonella spp. Administer oralIy to each chicken a quantity vaccination and at later appropriate stages and with sufficient
of the vaccine strain equivalent to not less than 10 times the frequency to determine the duration of disseminatjon.
maximum titre(s) likely to be contained in 1 dose of the For example, for broilers, samples are collected from
vaccine. Observe the chickens at least daily for at least 5 chickens for re-isolation of the vaccine strain on days 7 and
14 days. 14 after each vacciriation andweeldy until 8 weeks of age.
The test is not valid if more than 10 per cent of the chickens At the 7- and 14-day sampling points, samples are taken
younger than 3 weeks of age show abnormal signs 01' die from the liver, caecum and spleen of 5 chickens.
from causes not attributable to the vaccine. In the case of chickens intended for laying, samples are
The vaccine complies with the test if no chicken shows collected from 5 chickens on days 7 and 14 after each
abnormal signs of disease 01' dies from causes attributable to vaccination and weeldy until 3 consecutive negative weeldy
the vaccine. samples are obtained or the time of the next vaccination is
reached, whichever is the sooner. At the 7- and 14- day
2016 Veterinary Vaccines Vet-31 7

sampling points, samples are taken from the liver, caecum Measures taken to ensure absence of contamination by
and spleen of 5 chickens. In addition, samples are collected Salmonella spp. from the environment before the statt of the
from ovaries and oviducts where dissemination in vaccinated test and on a regular ongoing basis are described and
future layers is being investigated. justified.
The test is not valid if more than 10 per cent of vaccinated Suitably sensitive validated methods are used for re-isolation
chickens in any group show abnormal clinical signs 01' die of bacteria derived from the challenge and for distinguishing
from causes not attributable to the vaccine. The results are these from the vaccine strain.
noted and used to formulate the label statement on the 2-2-2-1 Immunogenicity. Use for the test not fewer than
length of time the vaccine strain survives in the body and to 40 chickens of the same origin and from an SPF flock
define a suitable withdrawal periodo (5.2.2). Vaccinate according to the schedule to be
2-2-1-5 Increase in virulence. Carry out the test according to recommended not fewer than 20 chickens with a single dose
general chapter 5.2.6 using SPF chickens (5.2.2) not older of vaccine. Maintain not fewer than 20 chickens as controls.
than the minimum age to be recommended for vaccination. Challenge each chicken after 14 days by a suitable route with
Administer orally to each chicken of the 1st group a quantity a sufficient quantity of a virulent strain of S. enterica
of the vaccine strain under study that will allow recovery of Typhimurium to give a valid test. Collect cloacal swabs from
bacteria for the passages described below. 4 to 7 days after vaccinates and controls on days 3, 5, 7, 10 and 14 post-
administration of the vaccine strain, prepare a suspension challenge. Samples of caecum, liver and spleen are collected
from the liver, spleen and caecum of chickens and pool these from 10 chickens of each group on days 7 and 14 post-
samples. Administer pooled samples orally to each chicken of challenge for re-isolation of challenge bacteria. Collect the
the next group. Carry out this passage operation not fewer same samples of internal organs from any chicken that dies.
than 4 times; verify the presence of the bacteria at each Examine the samples for the presence of the challenge
passage. If the bacteria are not found at a passage level, organisms using a suitable sensitive culture medium and
repeat the passage by administration to a group of compare results for vaccinates and controls.
10 chickens. Any mortalities are investigated for the presence The test is not valid if, during the observation period, fewer
of the vaccine strain and the properties of any re-isolated than 80 per cent of the control chickens die 01' challenge
vaccine strain determined. organisms are re-isolated from fewer than 80 per cent of the
Carry out the test for excretion, duration of excretion and control chickens.
survival in the environment (section 2-2-1-2) and, if the last The vaccine complies with the test if there is a significant
group of birds from which the bacteria was recovered shows reduction in the number of cloacal swabs from vaccinates
evidence of an increase in virulence indicative of reversion containing challenge organisms compared with the number
during the observation period, carry out the test for general from the controls and there is a significant reduction in the
safety (section 2-2-1-1), using the material used for the number of samples from internal organs from vaccinates
1st passage and the bacteria at the last passage level where it containing challenge bacteria compared with the number
was recovered. Test the bacteria recovered for the final from the controls.
passage for the presence and stability of the marker(s). 2-2-2-2 Im7llzl71ogenicity at the end 01 the laying periodo Use for
The vaccine strain complies with the test if no indication of the test not fewer than 40 chickens of the same origin and
increased virulence of the bacteria recovered for the final from an SPF flock (5.2.2). Vaccinate not fewer than
passage compared with the material used for the 1st passage 20 chickens according to the schedule to be recommended.
is observed and the presence of the marker(s) is confirmed in Maintain not fewer than 20 chickens as controls. At the end
the bacteria recovered for the final passage and remains of the laying period, take serum samples and cloacal swabs
identical to the material used for the 1st passage. If the from the chickens and environmental samples from the
bacteria are not recovered after an initial passage in 5 animals housing area. Test each serum sample individually for the
and a subsequent repeat passage in 10 animals, the vaccine presence of antibodies to S. enterica Typhimurium and each
also complies with the test. cloacal swab and fresh environmental sample for the presence
2-2-1-6 Field tlials. The chickens used for field trials are also of Salmonella spp. Challenge each chicken by a suitable route
used to evaluate safety. A trial is carried out in each category with a sufficient quantity of a virulent strain of S. enterica
of chickens for which the vaccine strain is intended, in not Typhimurium to give a valid test. Collect cloacal swabs from
fewer than 2 sets of premises. Samples are taken from a vaccinates and controls on days 3, 5, 7, 10 and 14 post-
significant number of chickens for re-isolation of bacteria to challenge. Samples of caecum, liver, spleen, ovaries and
provide information on the persistence, dissemination and oviducts are collected from 10 chickens of each group for
spread of the bacteria, which can be used, with the data from re-isolation of challenge bacteria on days 7 and 14 post-
the laboratory studies, to formulate the statements on the challenge. Collect the same samples of internal organs from
label. The samples include cloacal swabs, floor fa,eces, spleen any chicken that dies during the observation periodo Examine
and liver and, in laying chickens, samples of ovarles and the samples for the presence of the challenge organisms with
oviducts. Environmental samples are also tested at regular growth in a suitable medium and compare results for
intervals. vaccinates and controls.
2-2-2 Irnmunogenicity The test is not validif, before the challenge, antibodies to
The tests described in section 2-2-2-1 and, if appropriate, in Salmonella spp. are found in the serum of the controls 01'
section 2-2-2-2, are carried out using chickens not older than Salmonella spp. bacteria are isolated from any of the chickens.
the minimum age to be recommended for vaccination and The test is also not valid if the challenge organisms are
that are free from antibodies against Sabnonella spp. re-isolated from fewer than 80 per cent of the control
The quantity of the vaccine strain to be administered orally chickens.
to each chicken is not greater than the minimum titre to be The vaccine complies with the test if there is a significant
stated on the label. reduction in the number of cloacal swabs from vaccinates
containing challenge organisms compared with the number
Vet-318 Veterinary Vaccines 2016

from the controls and there is a significant decrease in the 2 PRODUCTION


number of samples of internal organs from vaccinates The vaccine may be adjuvanted.
containing challenge bacteria compared with the number
2-1 CHOICE OF VACCINE COMPOSITION
from the controls.
The vaccine is shown to be satisfactory with respect to safety
3 BATCH TESTS (5.2.6) and efficacy (5.2.7) for the pigs for which iris
3-1 Identification intended.
The strain present in the vaccine is identified by a The following test for safety (section 2-1-1) and
combination of methods such as morphological, serological irnmunogenicity (section 2-1-2) may be used during the
and biochemical methods and culture on appropriate demonstration of safety and efficacy.
selective media. Suitable testes) are conducted to confirm the
2-1-1 Safety
presence of the relevant marker( s) .
Carry out the test for each route and method of
3-2 Bacteria and fungi administration to be recommended for vaccination and where
Carry out the test by microscopic examination and by applicable, in pigs of each category for which the vaccine is
inoculation of suitable media, or veri:fy the absence of micro- intended, using in each case pigs not older than the
organisms other than the vaccine strain present in the vaccine minimum age to be recommended for vaccination. Use a
as described in the test for sterility prescribed in the batch of vaccine containing not les s than the maximum
monograph Vaccines jor veterz"nmy use (0062). The vaccine potency that may be expected in a batch of vaccine.
complies with the test if it does not contain extraneous
For each test, use not fewer than 8 pigs that do not have
micro-organisms.
antibodies against swine erysipelas. Administer to each pig
Any diluent supplied for reconstitution of the vaccine 1 dose of the vaccine. If the schedule to be recornmended
complies with the test for sterility prescribed in the requires a 2nd dose, administer another dose after an interval
monograph Vaccines jor veterz"nmy use (0062). of at leas! 14 days. Observe the pigs at least daily until at
3-3 Live bacteria least 14 days after the last administration. Record body
Titrate the vaccine strain using a suitable medium for the temperatures the day before each vaccination, at vaccination,
culture of the strain. The vaccine complies with the test if it 2 h, 4 h and 6 h later and daily for 4 days; note the
contains not less than the titre stated on the label. maximum temperature increase for each pig.
3-4 Potency The vaccine complies with the test if no pig shows abnormal
The vaccine complies with the requirements of the test local or systemic reactions or signs of disease, or dies from
prescribed under Irnmunogenicity (section 2-2-2-1) when causes attributable to the vaccine, if the average body
administered by a recommended route and method. Ir is not temperature increase for all pigs does not exceed 1.5 oC, and
necessary to carry out the potency test for each batch of the if no pig shows a rise greater than 2.0 oC.
vaccine if it has been carried out on a representative batch 2-1-2 Immunogenicity
using a vaccinating dose containing not more than the The test described below is suitable to demonstrate
minimum titre stated on the label. irnmunogenicity of the vaccine with respect to
4 LABELLING E. rhusiopathiae serotypes 1 and 2. If claims are made
The label sta tes: conceming another serotype, then a further test to
-- the nature of the markers allowing the vaccine to be demonstrate irnmunogenicity against this serotype is
distinguished from wild-type strains; necessary.
-- the extent to which the vaccine spreads and is transmitted If the vaccine contains more than 1 serotype, a test for
to non-vaccinated chickens and the time over which this 2 serotypes may be carried out on a single group by injecting
could OCCUf; each challenge serotype on different flanks of the pigs.
-- the time that the vaccine survives in the body; Validation and acceptance criteria are applied separately to
-- the length of time of excretion and the time that the the respective injection sites. If the vaccine contains more
vaccine survives in the environment; than 1 serotype, the immunogenicity test may also be carried
-- the potential for spread to other susceptible species out using a separate group for each serotype.
including humans; A test is carried out for each route and method of
-- the withdrawal periodo administration to be recommended, using in each case pigs
_____________________________ PhE~
not les s than 12 weeks old and weighing not les s than 20 kg.
The vaccine administered to eachpig is of minimum
potency.
For each test, use not fewer than 15 pigs that do not have
antibodies against swine erysipelas. Divide the pigs into
Swine Erysipelas Vaccine, ***
*** ***
2 groups. Vaccinate a group ofnot fewer than 10 pigs
according to the schedule to be recommended. Maintain a
Inactivated *** group of not fewer than 5 pigs as controls. Challenge each
(Swine Elysipelas Vaccine (Inactivated), pig 3 weeks after vaccination by the intradermal route by
Ph Eur monogmph 0064) separate injections of 0.1 mL of a virulent strain of each of
Ph Eur _______________________________ serotype 1 and serotype 2 of E. rhusiopathiae. Observe the
1 DEFINITION pigs at least daily for 7 days.
Swine erysipelas vaccine (inactivated) is a preparation of one The test is not valid if fewer than 80 per cent of control pigs
or more suitable strains of Elysipelothrix rhusiopathiae, show typical signs of disease, i.e. diamond skin lesions at the
inactivated while maintaining adequate immunogenic injection sites. The vaccine complies with the test if not fewer
properties. This monograph applies to vaccines intended for than 90 per cent of the vaccinated pigs remain free from
the active irnmunisation of pigs against swine erysipelas. diamond skin lesions at the injection site.
2016 Veterinary Vaccines Vet-319

Szuine elysipelas bacteria serotype 1 BRP and szuine elysipelas 2-3 CHOICE OF VACCINE VIRUS
bactel1a serotype 2 BRP are suitable for use as challenge The vaccine virus is shown to be satisfactory with respect to
strains. safety (5.2.6) and efficacy (5.2.7) for the swine for which it is
2-2 MANUFACTURER'S TESTS intended.
2-2-1 Batch potency test The fol1owing tests described under Safety test in piglets
It is not necessary to carry out the potency test (section 3-3) (section 2-3-1), Safety test in pregnant sows and test for
for each batch of vaccine if it has been carried out using a transplacental transmission (section 2-3-2), Non-
batch of vaccine with a minimum potency. Where the test is transmissibility (section 2-3-3), Increase in virulence
not carried out, an alternative validated method is used, the (section 2-3-4) and Irnmunogenicity (section 2-3-5) may be
criteria for acceptance being set with reference to a batch of used during the demonstration of safety and immunogenicity.
vaccine that has given satisfactory results in the test described 2-3-1 Safety test in piglets
under Potency. The following test may be used. Can-y out the test for each route to be recommended using
Use 10 mice of a suitable strain (for example, NMRI) in each case piglets not older than the minimum age to be
weighing 17-20 g, from a uniform stock and that do not have recommended for vaccination. Use vaccine virus at the least
antibodies against swine erysipelas. Vaccinate each mouse by attenuated passage level that will be present in a batch of the
the subcutaneous route with a suitable dose (usually 1/10 of vaccine.
the pig dose). At a given interval (for example, 21-28 days), U se not fewer than 8 healthy piglets that do not have
depending on the vaccine to be examined, bleed the mice antibodies against pestiviruses. Administer to not fewer dlan
under anaesthesia. Pool the sera, using an equal volume from 8 piglets a quantity of the vaccine virus equivalent to not less
each mouse. Determine the level of antibodies by a suitable than 10 times the maximum virus titre likely to be contained
irnmunochemical method (2.7.1), for example, enzyme-linked in 1 dose of the vaccine. Observe the piglets at least daily for
immunosorbent assay with elysipelas BLISA coating at least 14 days. The body temperature of each vaccinated
antigen BRP. The vaccine complies with the test if the piglet is measured on at least the 3 days preceding
antibody level is not significandy less than that obtained with administration of the vaccine, at the time of administration,
a batch that has given satisfactory results in the test described 4 h after and then dai1y for at least 14 days. The vaccine
under Potency. complies with the test if the average body temperature
3 BATCH TESTS increase for all piglets does not exceed 1.5 oC, no piglet
3-1 Identification shows a temperature rise greater than 1.5 oC for a period
Injected into animals that do not have antibodies against exceeding 3 days, and no piglet shows notable signs of
E. rhusiopathiae, the vaccine stimulates the production of disease 01' dies from causes attributable to the vaccine.
such antibodies. 2-3-2 Safety test in pregnant sows and test for
3-2 Bacteria and fungi transplacental transmission
The vaccine, including where applicable the diluent supplied Carry out the test by a route to be recommended using not
for reconstitution, complies widl the test for sterility fewer than 8 healthy sows 01' gilts of the same age and origin,
prescribed in the monograph Vaccines for veterina1Y use between dle 55 th and 80 th days of gestation, and that do not
(0062). have antibodies against pestiviruses. Use vaccine virus at the
least attenuated passage level that will be present in a batch
3-3 Potency
of the vaccine.
The vaccine complies with the requirements of the tests
mentioned under Immunogenicity (section 2-1-2) when Administer to not fewer than 8 sows or gilts a quantity of the
administered by a recommended route and method. vaccine virus equivalent to not less than the maximum virus
_____________________________________________ PhE~
titre likely to be contained in 1 dose of the vaccine. Record
the body temperature on at least the 3 days preceding
administration of the vaccine, at the time of administration,
4 h after and then daily for at least 15 days. Observe until
*** farrowing. .
Swine-Fever Vaccine (Live,
*** *** Can-y out tests for serum antibodies against classical swine-
Prepared in Cell Cultures), *** fever virus. No antibodies against classical swine-fever virus
are found in sera taken from the newborn piglets before
Classical ingestion of colostrum. The test is not valid if the vaccinated
(Ph Bur 1110nograph 0065) sows do not seroconvert. The vaccine virus complies with the
PhE~ _____________________________________________ test if no abnormalities in the gestation 01' in the piglets are
1 DEFINITION noted, no sow or gilt shows a temperature rise greater than
1.5 oC for a period exceeding 5 days, and no sow or gilt
Classical swine-fever vaccine (live, prepared in cell cultures)
shows notable signs of disease 01' die s from causes
is a preparation obtained from a strain of classical swine-fever
attributable to the vaccine.
virus that has lost its pathogenicity for the pig by in vivo
and/or in vitro passage and has been adapted to growth in 2-3-3 Non-transmissibility
cell cultures. Keep together for the test not fewer than 12 healthy piglets,
6-10 weeks old and of the same origin, and that do not have
2 PRODUCTION antibodies against pestiviruses. Use vaccine virus at the least
2-1 PREPARATION OF THE VACCINE attenuated passage level that will be present between the
The vaccine virus is grown in cel1 cultures. master seed lot and a batch of the vaccine. Administer by a
2-2 SUBSTRATE FOR VIRUS PROPAGATION route to be recommended to not fewer than 6 piglets a
Cell cultures quantity of the vaccine virus equivalent to not less than the
Cell cultures comply with the requirements for cell cultures maximum virus titre li1zely to be contained in 1 dose of the
for production of veterinary vaccines (5.2.4). vaccine. Maintain not fewer than 6 piglets as contact
Vet-320 Veterinary Vaccines 2016

controls. The mixing of vaccinated piglets and contact piglets fever virus, including cutaneous lesions, or die, and if fewer
is done 24 h after vaccination. than 100 per cent of the control piglets show clinical signs of
After 45 days, euthanise all piglets. Carry out appropriate disease within the 21 days following chaIlenge. The vaccine
tests on the piglets to detect antibodies to classical swine- complies with the test if the minimum dose stated on the
fever. Carry out appropriate tests on the control piglets to label corresponds to not less than 100 PD so .
detect classical swine-fever virus in the tonsils. The vaccine 2-3-5-2 Protection against transplacental infection Use at least 8
complies with the test if antibodies are found in aIl sows that do not have antibodies against pestiviruses,
vaccinated piglets and if no antibodies and no virus are found randomIy allocated to either the vaccine group (n = 6) or the
in the control piglets. control group (n = 2).
2-3-4 Increase in virulence Between the 40 th and 50th day of gestation, aH sows aHocated
Can-y out the test according to chapter 5.2.6. Evaluation of to the vaccine group are vaccinated once with 1 dose of
safety of veterina1Y vaccines and immunosera, using piglets vaccine containing not more than the minimum titre stated
6-10 weeks old that do not have antibodies against on the label. On day 60 of gestation, aH sows are challenged
pestiviruses. If the properties of the vaccine virus allow by a route to be recommended with a suitable strain of
sequential passage through 5 groups via natural spreading, virulent virus. Just before farrowing and about 5-6 weeks
this method may be used, otherwise passage as described after challenge, the sows are euthanised and their foetuses are
below is carried out. examined for classical swine-fever virus. Serum samples from
Administer to each piglet of the 1st group by a route to be sows and foetuses are tested for the presence of antibodies
recommended a quantity of the vaccine virus equivalent to against classical swine-fever virus. Isolation of classical swine-
not less than the maximum virus titre likely to be contained fever virus is carried out from blood of the sows (collected 7
in 1 dose of the vaccine. Collect an appropriate quantity of and 9 days after challenge and at euthanasia), and from
blood from each piglet daily between day 2 and day 7 after homogenised organ material (spleen, kidneys, lymph nodes)
administration of the vaccine virus, and pool the samples of the foetuses.
taken on the same day. Administer 2 mL of the pooled blood The test is not valid if one or more of the vaccinated sows do
with the highest virus titre by a route to be recommended to not seroconvert after the vaccination and the control sows do
each piglet of the next group. Carry out this passage not seroconvert after the challenge, or if no virus is found in
operation not fewer than 4 times, verliying the presence of more than 50 per cent of the foetuses from the control sows
the virus at each passage. If no virus is found, repeat the test (excluding mummified foetuses).
once. If virus is found, can-y out a 2nd series of passages by The vaccine complies with the test if no virus is found in the
administering 2 mL of positive blood by a route to be blood of vaccinated sows and in foetuses from the vaccinated
recommended to each piglet of a group of 10 animals. sows, and no antibodies against classical swine-fever virus are
If the 5th group of animals shows no evidence of an increase found in the serum of the foetuses from the vaccinated sows.
in virulence indicative of reversion during the observation 3 BATCH TESTS
period, further testing is not required. Otherwise, carry out 3-1 Identification
an additional safety test and compare the clinical signs and Specific classical swine-fever monoclonal antibodies are used
any relevant parameters in a group of at least 8 animals to identify the vaccinal strain.
receiving the material used for the 1st passage and another
similar group receiving the virus at the final passage level. 3-2 Bacteria and fungi
The vaccine, including where applicable, the diluent supplied
The vaccine virus complies with the test if no indication of
for reconstitution, complies with the test for sterility
increasing virulence of the virus recovered for the final
prescribed in the monograph Vaccines for veterina1Y use
passage compared with the material used for the 1st passage
(0062).
is observed. If virus is not recovered after an initial passage in
2 animal s and a subsequent repeat passage in 10 animals, the 3-3 Mycoplasmas (2.6.7)
vaccine virus also complies with the test. The vaccine complies with the test for mycoplasmas.
2-3-5 Immunogenicity 3-4 Extraneous agents
2-3-5-1 Protective dose The efficacy of the vaccine is expressed N eutralise the vaccine virus using monoclonal antibodies to
by the number of 50 per cent protective doses (PD so ) for the vaccine virus. Inoculate into ceH cultures known to be
pigs contained in the vaccinal dos e as indicated on the label. sensitive to viruses pathogenic for pigs and to pestiviruses.
The vaccine contains at least 100 PD so per dose. Maintain these cultures for not les s than 14 days and carry
out at least 3 passages during thisperiod.
U se 1 01' more groups of piglets aged 6-10 weeks and that do
not have antibodies against pestiviruses, and use an The vaccine complies with the test if no cytopathic effect is
additional group of piglets of the same age and origin as produced and if the cells show no evidence of the presence of
controls. Each group of piglets is vaccinated with 1 dilution haemadsorbing agents.
of the vaccine dose. 14 days after the single injection of U se monoclonal antibodies that can identify possible
vaccine, chaIlenge the piglets by a suitable route with a contamination with pestiviruses. No virus is detected by an
suitable strain of virulent virus and adose that kills not fewer appropriate method.
than 50 per cent of the non-vaccinated piglets in less than 3-5 Virus titre
21 days. Observe the piglets for 21 days and record the body Titrate the vaccine virus in suitable cell cultures (5.2.4).
temperature 3 days before challenge and daily after challenge The vaccine complies with the test if 1 dose contains not less
for 21 days. The PD so is calculated by the usual statistical than the minimum virus titre stated on the label.
methods (for example, 5.3), taking into account the surviving
3-6 Potency
piglets that have no clinical signs of swine fever, including
The vaccine complies with the requirements of the test
cutaneous lesions or an increase in body temperature.
prescribed under Irnmunogenicity (section 2-3-5) when
The test is not valid if fewer than 50 per cent of the control administered by a recommended route and method. It is not
piglets display typical signs of serious infection with swine-
2016 Veterinary Vaccines Vet-321

necessary to carry out the potency test for each batch of the 2-3-1-1-2 Safety in the pigs used in test 2-3-2 for
vaccine if it has been carried out on a representative batch irnmunogenicity. The pigs used in the test for
using a vaccinating dose containing not more than the irnmunogenicity are also used to evaluate safety. Measure the
minimum virus titre stated on the label. body temperature of each vaccinated pig at the time of
_____________________________________________ PhE~ vaccination and 24 h and 48 h later. Examine the injection
site at slaughter for local reactions.
The vaccine complies with the test if no pig shows:
abnormal body temperature;
-- other systemic reactions (for example, anorexia);
Swine Influenza Vaccine, -- abnormal local reactions attributable to the vaccine.
Inactivated 2-3-1-2 Field studies. The pigs used for field trials are also
(Porcine Influenza Vaccine (Inactivated) , used to evaluate safety. Carry out a test in each category of
Ph Bur monograph 0963) pigs for which the vaccine is intended (sows, fattening pigs).
~E~ _____________________________________________ U se not fewer than 3 groups each of not fewer than 20 pigs
in at least 2 locations with corresponding groups of not fewer
1 DEFINITION than 10 controls. Measure the body temperature of each
Porcine influenza vaccine (inactivated) is a preparation of one vaccinated pig at the time of vaccination and 24 h and 48 h
or more suitable strains of swine or human influenza virus latero Examine the injection site at slaughter for local
inactivated while maintaining adequate irnmunogenic reactions.
properties. Suitable strains contain both haemagglutinin and The vaccine complies with the test if no pig shows:
neuraminidase. This monograph applies to vaccines intended -- abnormal body temperature;
for the active irnmunisation of pigs against porcine influenza. -- abnormal local reactions attributable to the vaccine.
2 PRODUCTION 2-3-2 Immunogenicity
2-1 PREPARATION OF THE VACCINE The following test carried out using an epidemiologically
The vaccine virus is grown in embryonated hens' eggs or in relevant challenge strain or strains is suitable to demonstrate
cell cultures. Each virus strain is cultivated separately. After the irnmunogenicity of the vaccine. It is carried out for each
cultivation, the viral suspensions are collected separately and subtype used in the preparation of the vaccine.
inactivated by a suitable method. If necessary, they may be A test is carried out for each route and method of
purified. The vaccine may be adjuvanted. administration to be recommended, using in each case pigs
2-2 SUBSTRATE FOR VIRUS PROPAGATION of the minimum age to be recommended for vaccination.
The vaccine administered to each pig is of minimum
2-2-1 Embryonated hens' eggs
potency.
If the vaccine virus is grown in embryonated hens' eggs, they
are obtained from a healthy flock. Use for the test not fewer than 20 pigs that do not have
antibodies against swine influenza virus. Vaccinate not fewer
2-2-2 Cell cultures
than 10 pigs according to the schedule to be recommended.
If the vaccine virus is grown in cell cultures, they comply
Maintain not fewer than 10 pigs as controls. Take a blood
with the requirements for cell cultures for production of
sample from all control pigs irnmediately before challenge.
veterinary vaccines (5.2.4).
3 weeks after the last administration of vaccine, challenge all
2-3 CHOICE OF VACCINE COMPOSITION the pigs by the intratracheal route with a sufficient quantity
The choice of strains is based on the antigenic types and sub- of a virulent influenza field virus. Euthanise half of the
types observed in Europe. The vaccine is shown to be vaccinated and control pigs 24 h after challenge and the
satisfactory with respect to safety (5.2.6) and efficacy (5.2.7) other half 72 h after challenge. For each pig, measure the
for the pigs for which it is intended. quantity of influenza virus in 2 lung tissue homogenates, one
The following tests for safety (section 2-3-1) and from the left apical, cardiac and diaphragmatic lobes, and the
irnmunogenicity (section 2-3-2) may be used during the other from the corresponding right lung lobes. Take
demonstration of safety and efficacy. equivalent samples from each pig.
2-3-1 Safety The test is not valid if antibodies against influenza virus are
2-3-1-1 Laboratory tests. Carry out the tests for each route found in any control pig irnmediately before challenge.
and method of administration to be recornmended for The vaccine complies with the test if, at both times of
vaccination and where applicable, in pigs of each category for measurement, the mean virus titre in the pooled lung tissue
which the vaccine is intended (sows, fattening pigs), using in samples of vaccinated pigs is significantly lower than that for
each case pigs not older than the minimum age to be control pigs, when analysed by a suitable statistical method
recommended for vaccination. Use a batch of vaccine such as the Wilcoxon Mann-Whitney test.
containing not less than the maximum potency that may be 2-4 MANUFACTURER'S TESTS
expected in a batch of vaccine. 2-4-1 Residuallive virus
2-3-1-1-1 General safety. For each test, use not fewer than 8 An amplification test for residual live virus is carried out on
pigs that do not have antibodies against swine influenza virus. each batch of antigen irnmediately after inactivation by
Administer to each pig 1 dos e of the vaccine. If the schedule passage in the same type of substrate as that used for
to be recommended requires a 2nd dose, administer another production (eggs or cell cultures) or a substrate shown to be
dose after an interval of at least 14 days. Observe the pigs at at least as sensitive. The quantity of inactivated virus harvest
least daily until 14 days after the last administration. used in the test is equivalent to not less than 10 doses of the
The vaccine complies with the test if no pig shows abnormal vaccine. The inactivated viral harvest complies with the test if
local or systemic reactions or dies from causes attributable to no live virus is detected.
the vaccine during the test.
Vet-322 Veterinary Vaccines 2016

2-4-2 Batch potency test complies with the test if it does not stimulate the formation
It is not necessary to carry out the potency test (section 3-5) of antibodies against pestiviruses and Aujeszky's disease virus.
for each batch of vaccine if it has been carried out using a 3-5 Potency
batch of vaccine with a minimum potency. Where the test is The vaccine complies with the requirements of the test
not carried out, an alternative validated method is used, the mentioned under Irnmunogenicity (section 2-3-2) when
criteria for acceptance being set with reference to a batch of administered by a recommended route and method.
vaccine that has given satisfactory results in the test described _____________________________________________ PhE~

under Potency. The following test may be used.


Use 5 guinea-pigs, 5-7 weeks old and that do not have
antibodies against swine influenza virus. Vaccinate each
guinea-pig by the subcutaneous route with a quarter of the
Turkey Infectious Rhinotracheitis ***
** **
recommended dose. Collect blood samples before the
vaccination and 21 days after vaccination. Determine for
each sample the level of specific antibodies against each virus Vaccine (Live) *****
subtype in the vaccine by haemagglutination-inhibition or (Ph. Eur. 1110nograph 2461)
another suitable test. The vaccine complies with the test if ~E~ _____________________________________________
the level of antibodies is not lower than that found for a
1 DEFINITION
batch of vaccine that gave satisfactory results in the potency
test in pigs (see Potency). Turkey infectious rhinotracheitis vaccine (live) is a
preparation of a suitable strain of turkey rhinotracheitis virus.
2-4-3 Bacteria! endotoxins This monograph applies ro vaccines intended for
For vaccines produced in eggs, the content of bacterial administration to turkeys for active irnmunisation against
endoroxins is determined on the virus harvest to monitor turkey infectious rhinotracheitis.
production.
2 PRODUCTION
3 BATCH TESTS 2-1 PREPARATION OF THE VACCINE
3-1 Identification The vaccine virus is grown in ce11 cultures.
When injected into healthy animals that do not have specific
antibodies against the influenza virus subtypes included in 2-2 SUBSTRATE FOR VIRUS PROPAGATION
the vaccine, the vaccine stimulates the production of such 2-2-1 Cell cultures
antibodies. The antibodies may be detected by a suitable The vaccine virus is grown in cell cultures that comply with
irnmunochemical method (2.7.1). the requirements for cell cultures for production of veterinary
vaccines (5.2.4).
3-2 Bacteria and fungi
The vaccine, inc1uding where applicable the diluent supplied 2-3 SEED LOTS
for reconstitution, complies with the test for sterility 2-3-1 Extraneous agents
prescribed in the monograph Vaccines fol' veterinary use The master seed lot complies widl the tests for extraneous
(0062). agents in seed lots (2.6.24). In these tests on the master seed
3-3 Residuallive virus lot, the organisms used are not more than 5 passages from
3-3-1 Vaccines prepared in eggs. If the vaccine has been the master seed lot at the start of the test.
prepared in eggs, inoculate 0.2 mL into the allantoic cavity of 2-4 CHOICE OF VACCINE VIRUS
each of 10 fertilised hen eggs, 9-11 days old. Incubate at a The vaccine virus shall be shown ro be satisfactory with
suitable temperature for 3 days. The death of any embryo respect to safety (5.2.6) and efficacy (5.2.7) for the turkeys
within 24 h of inoculation is considered as non-specific for which it is intended.
mortality and the egg is discarded. The test is not valid if
The fo11owing tests for safety (section 2-4-1), increase in
fewer than 80 per cent of the eggs survive. Collect the
virulence (section 2-4-2) and irnmunogenicity (section 2-4-3)
allantoic fluid of each egg, pool equal quantities and carry
may be used during the demonstration of safety and efficacy.
out a 2nd passage on fertilised eggs in the same manner.
Incubate for 4 days; the vaccine complies with the test if the 2-4-1 Safety
allantoic fluid of these eggs shows no haemagglutinating Safety for the respiratOly tract Carry out the test using turkeys
activity. not older than the minimum age to be recommended for
vaccination and free from antibodies against turkey
3-3-2 Vaccines prepared in cell cultures. If the vaccine has been
rhinotracheitis virus. Use vaccine virus at the least attenuated
prepared in ce11 cultures, carry out a suitable test for residual
passage level that will be present in a batch of vaccine.
live virus using 2 passages in the same type of ce11 culture as
used in the production of vaccine. The vaccine complies with For each test performed in turkeys younger than 3 weeks of
the test if no live virus is detected. If the vaccine contains an age, use not fewer than 10 turkeys. For each test perfOlmed
oi1y adjuvant that intetferes with this test, where possible in turkeys older than 3 weeks of age, use not fewer than
separate the aqueous phase from the vaccine by means that 8 turkeys. Administer to each turkey, by the oculonasal
do not diminish the capacity to detect residual infectious route, a quantity of the vaccine virus equivalent to' not less
influenza virus. than 10 times the maximum virus titre likely to be contained
in 1 dose of the vaccine. Observe the turkeys at least daily for
3-4 Specified extraneous agents
at least 14 days and monitor clinical signs individually by a
Use not fewer than 2 pigs that do not have antibodies against
suitable scoring system. Mortality should be taken into
swine influenza virus, against Aujeszky's disease virus and
account when calculating clinical scores: Record the death of
against pestiviruses. Administer to each pig by a
any turkey and check for lesions of the respiratory tracto
recommended route a double dose of the vaccine, then
another dose after 14 days. 14 days after the last The test is not val id if more than 10 per cent of the turkeys
administration, carry out tests for antibodies. The vaccine younger than 3 weeks of age show abnormal signs of disease
or die from causes not attributable ro the vaccine virus.
2016 Veterinary Vaccines Vet-323

For turkeys older than 3 weeks of age, dle test is not val id if The test is not val id if one or more of the following applies:
non-specific mortality occurs. -- fewer than 80 per cent of the unvaccinated turkeys show
The vaccine virus complies with the test if no vaccinated typical signs of respiratory disease following challenge
turkey shows notable signs of disease or dies from causes with the virulent turkey rhinotracheitis virus;
anributable to the vaccine virus. -- during the period between vaccination and challenge,
more than 10 per cent of vaccinated 01' control turkeys
The clinical scores are used in the test described under
2-4-2. show abnormal clinical signs 01' die from causes not
anributable to the vaccine.
2-4-2 Increase in virulence
The vaccine virus complies with the test if during the
Carry out the test according to general chapter 5.2.6 using
observation period after challenge not fewer than 90 per cent
turkeys younger than 3 weeks of age and free from antibodies
of the vaccinated turkeys survive and show no typical clinical
against turkey rhinotracheitis virus. If the properties of the
signs or lesions of an infection with turkey rhinotracheitis
vaccine virus allow sequential passage through 5 groups via
virus.
natural spreading, this method may be used, otherwise,
passage as described below is carried out. 3 BATCH TESTS
Administer to each turkey of the 1st group, by the oculonasal 3-1 Identification
route, a quantity of the vaccine virus that will allow recovery The vaccine, diluted if necessary and mixed with turkey
of virus for the passages described below. 2-6 days after rhinotracheitis virus antiserum specific for the virus subgroup,
administration of the vaccine virus, prepare a suspension no longer infects susceptible cell cultures (5.2.4) into which it
from the mucosa of the turbinates or the upper trachea, or is inoculated. The vaccine may also be identified using
from an oro-pharyngal or tracheal swab from not less than appropriate molecular biology techniques (for example
5 inoculated turkeys and pool these samples. Administer RT-PCR).
0.1 mL of the pooled samples by the oculonasal route to 3-2 Bacteria and fungi
each turkey of the next group. Carry out this passage Vaccines intended for administration by injection comply
operation not fewer than 4 times; verify the presence of the with the test for sterility prescribed in the monograph
virus at each passage. If the virus is not found at a passage Vaccines for veterina1Y use (0062).
level, repeat the passage by administration to a group of Any diluent supplied for reconstitution of the vaccine
10 turkeys. complies with the test for sterility prescribed in the
If the 5th group of turkeys shows no evidence of an increase monograph Vaccines for veterina1Y use (0062).
in virulence during the observation period, further testing is 3-3 Mycoplasmas
not required. Otherwise, carry out an additional safety test The vaccine complies with the test for mycoplasmas (2.6.7).
for the respiratory tract and compare the clinical signs and
any relevant parameters in a group of at least 10 turkeys 3-4 Extraneous agents
receiving dle material used for the 1st passage and another The vaccine complies with the tests for extraneous agents in
similar group receiving the virus at the final passage level. batches of finished product (2.6.25).
The vaccine virus complies with the test if no indication of 3-5 Virus titre
an increased virulence of the virus at the final passage level Titrate the vaccine virus by inoculation into suitable cell
compared with the material used for the 1st passage is cultures (5. 2. 4). The vaccine complies with the test if 1 dose
observed or a slight increase in virulence of the virus at the contains not les s than the minimum titre of vaccine virus
final passage level may be observed for a vaccine which stated on the label.
complies with the safety test (section 2-4-1). If virus is not 3-6 Potency
recovered after an initial passage in 5 turkeys and a The vaccine complies with the requirements of the test
subsequent repeat passage in 10 turkeys, dle vaccine virus prescribed under Irnmunogenicity (section 2-4-3) when
also complies with the test. administered according to the recommended schedule by a
2-4-3 Immunogenicity recommended 'route and method. It is not necessary to carry
A test is carried out for each route and method of out the potency test f6'r each batch of the vaccine if it has
administration to be recommended using turkeys not older been carried out on a representative batch using a vaccinating
than the minimum age to be recommended for vaccination dose containing not more than the minimum virus titre
and that are free from antibodies against turkey stated on the label.
rhinotracheitis virus. The quantity of vaccine virus to be _____________________________________________ PhE~

administered to each turkey is not greater than the minimum


virus titre to be stated on the label and the virus is at the
most anenuated passage level that will be present in a batch
of the vaccine. '
Cold-water Vibriosis Vaccine for *****
Clinical protection against virulent challenge U se not fewer than * *
30 turkeys of the same origin and free from antibodies Salmonids, Inactivated *****
against turkey rhinotracheitis virus. Vaccinate by a route to (Vibriosis (Cold-wate1) Vaccine (Inactivated) for
be recommended not fewer than 20 turkeys according to the Sabnonids) Ph Bur monograph 1580)
schedule to be recommended. Maintain not fewer than PhE~ _____________________________________________
10 turkeys as controls. Challenge each turkey after 21 days
by the oculonasal route with a sufficient quantity of a suitable 1 DEFINITION
strain of virulent turkey rhinotracheitis virus. Observe the Cold-water vibriosis vaccine (inactivated) for salmonids is
turkeys at least daily for 10 days and monitor clinical signs prepared from cultures of one 01' more suitable strains of
individually. Record the death of any turkey and check for Vib'70 salmonicida, inactivated while maintaining adequate
lesions of the respiratory tract. immunogenic properties. This monograph applies to vaccines
Vet-324 Veterinary Vaccines 2016

intended for administration by injection or irnmersion for the route and method of administration to be recornmended.
active irnmunisation of salmonids against cold-water vibriosis. The vaccine administered to each fish is of minimum
2 PRODUCTION potency.
2-1 PREPARATION OF THE VACCINE U se for the test not fewer than 60 fish of the minimum body
The strains of V. salmonicida are cultured and harvested mass to be recommended for vaccination, from a population
separately. The harvests are inactivated by a suitable method. that do es not have specific antibodies against V. salmonicida
They may be purified and concentrated. Whole or disrupted and has not been vaccinated against or exposed to cold-water
cells may be used and the vaccine may contain extracellular vibriosis. Vaccinate not fewer than 30 fish according to the
products of the bacterium released into the growth medium. instructions for use. Perform mock vaccination on a control
group of not fewer than 30 fish; mark vaccinated and control
2-2 CHOICE OF VACCINE COMPOSITION fish for identification. Keep a11 the fish in the same tank or
The strain or strains of V. salmonicida used are shown to be mix equal numbers of controls and vaccinates in each tank if
suitable with respect to production of antigens of assumed more than 1 tank is used. Where justified and when fish
protective importance. The vaccine is shown to be cannot be marked, non-marked fish may be used. Vaccinates
satisfactory with respect to safety (5.2.6) and efficacy (5.2.7) and controls may then be kept in the same tank but
in the species of fish for which it is intended. physica11y separated (for example by fishing nets). Challenge
The following tests for safety (section 2-2-1) and each fish at a fixed interval after vaccination, corresponding
irnmunogenicity (section 2-2-2) may be used during the to the onset of irnmunity c1aimed, by a suitable route, with a
demonstration of safety and efficacy. sufficient quantity of a culture of V. salmonicida whose
2-2-1 Safety virulence has been verified. Observe the fish at least daily
2-2-1-1 Laborat01Y tests. Safety is tested using test 2-2-1-1-1, until at least 60 per cent specific mortality is reached in the
test 2-2-1-1-2, or both, depending on the recommendations control group. Plot for both vaccinates and controls a curve
for use. of specific mortality against time from challenge and
determine by interpolation the time corresponding to
Carry out the test in each species of fish for which the
60 per cent specific mortality in controls.
vaccine is intended, using fish of the minimum body mas s to
be recommended for vaccination. Use a batch of vaccine The test is not valid if the specific mortality is les s than
containing not less than the maximum potency that may be 60 per cent in the control group 21 days after the 1st death
expected in a batch of vaccine. The test is carried out in the in the fish. Read from the curve for vaccinates the
conditions to be recommended for use of the vaccine with a mortality (M) at the time corresponding to 60 per cent
water temperature not less than 10°C. mortality in controls. Calculate the relative percentage
survival (RPS) using the. following expression:
2-2-1-1-1 Vaccines intended for administration by injection.
Use not fewer than 50 fish from a population that does not
have specific antibodies against V. sabnonicida and has not
been vaccinated against or exposed to cold-water vibriosis.
Administer to each fish by the intraperitoneal route 1 dos e of
the vaccine. Observe the fish at least daily for 21 days. The vaccine complies with the test if the RPS is not less than
The test is not valid if more than 6 per cent of the fish die 60 per cent for vaccines administered by immersion and
from causes not attributable to the vaccine. The vaccine 90 per cent for vaccines administered by injection.
complies with the test if no fish shows abnormal local or
2-3 MANUFACTURER'S TESTS
systemic reactions or dies from causes attributable to the
vaccme. 2-3-1 Batch potency test
The potency test (section 3-3) may be carried out for each
2-2-1-1-2 Vaccines imended for administration by imnzersion. batch of vaccine using fish of one of the species for which the
Use not fewer than 50 fish from a population that does not vaccine is intended. Where the test is not carried out, an
have specific antibodies against V. salmonicida and has not altemative validated method based on antibody response may
been vaccinated against or exposed to cold-water vibriosis. be used, the criteria for acceptance being set with reference
Prepare an irnmersion bath at twice the concentration to be to a batch of vaccine that has given satisfactory results in the
recommended. Bathe the fish for twice the time to be test described under Potency. The following test may be
recommended. Observe the fish at least daily for 21 days. used.
The test is not valid if more than 6 per cent of the fish die Use not fewer than 35 fish from a population that does not
from causes not attributable to the vaccine. The vaccine have specific antibodies against V. sabnonicida and that are
complies with the test if no fish shows abnormal local or within specified limits for body mass. Carry out the test at a
systemic reactions or dies from causes attributable to the , defined temperature. Inject into each of not fewer than
vaccine. 25 fish 1 dose of vaccine, according to the instructions for
2-2-1-2 Field studies. Safety is demonstrated in addition in use. Perform mock vaccination on a control group of not
field trials by administering the dose to be recommended to a fewer than 10 fish. Collect blood samples at a defined time
sufficient number of fish distributed in not fewer than 2 sets after vaccination. Determine for each sample the level of
of premises. specific antibodies against V. salmonicida by a suitable
The vaccine complies with the test if no fish shows abnormal immunochemical method (2.7.1). The test is not valid ifthe
reactions or dies from causes attributable to the vaccine. control group shows antibodies against V. salmonicida.
2-2-2 Immunogenicity The vaccine complies with the test if the mean level of
Carry out a separate test for each fish species and each strain antibodies inthe vaccinates is not significantly lower than
inc1uded in the vaccine, according to a protocol defining that found for a batch that gave satisfactory results in the test
water source, water flow, temperature limits, and preparation described under Potency.
of a standardised cha11enge. Each test is carried out for each
2016 Veterinary Vaccines Vet-325

3 BATCH TESTS conditions to be recommended for use of the vaccine with a


3-1 Identification water temperature not less than 10 oC.
When injected into fish that do not have specific antibodies 2-2-1-1-1 Vaccines imended fo1' administration by injection.
against V salmonicida, the vaccine stimulates the production Use not fewer than 50 fish from a population that does not
of such antibodies. have specific antibodies against L. anguillanmz or where
3-2 Bacteria and fungi applicable V o1'dalii and has not been vaccinated against or
The vaccine, inc1uding where applicable the diluent supplied exposed to vibriosis. Administer to each fish by the
for reconstitution, complies with the test for sterility intraperitoneal route 1 dose of the vaccine. Observe the fish
prescribed in the monograph Vaccines for veterina1Y use at least daily for 21 days.
(0062). The test is not valid if more than 6 per cent of the fish die
3-3 Potency from causes not attributable to the vaccine. The vaccine
The vaccine complies with the requirements of the test complies with the test if no fish shows abnormallocal or
mentioned under Irnmunogenicity (section 2-2-2) when systemic reactions or die s from causes attributable to the
administered by a recommended route and method. vaccine.
2-2-1-1-2 Vaccines intendedfo1' administration by immersion.
4 LABELLING
Use not fewer than 50 fish from a population that does not
The label states information on the time needed for
have specific antibodies against L. anguillamm or where
development of irnmunity after vaccination under the range
applicable V ordalii and has not been vaccinated against or
of conditions corresponding to the recommended use.
exposed to vibriosis. Prepare an immersion bath at twice the
_____________________________________________ ~Ew

concentration to be recornmended. Bathe the fish for twice


the time to be recommended. Observe the fish at least daily
for 21 days.
The test is not valid if more than 6 per cent of the fish die
Vibriosis Vaccine tor Salmonids, ***** from causes not attributable to the vaccine. The vaccine
* * complies with the test if no fish shows abnormallocal or
Inactivated ***** systemic reactions or dies from causes attributable to the
(Vibriosis Vaccine (Inactivated) for Salmonids) vaccine.
Ph Bur 7110nograph 1581)
2-2-1-2 Field studies. Safety is demonstrated in addition in
~Ew _____________________________________________
field trials by administering the dose to be recommended to a
1 DEFINITION sufficient number of fish distributed in not fewer than 2 sets
Vibriosis vaccine (inactivated) for salmonids is prepared from of premises.
cultures of one or more suitable strains or serovars of The vaccine complies with the test if no fish shows abnormal
Listonella anguillanmz (Vibrio anguillarum), inactivated while reactions or dies from causes attributable to the vaccine.
maintaining adequate immunogenic properties; the vaccine 2-2-2 Irnmunogenicity
may also inc1ude Vibrio o1'dalii. This monograph applies to Carry out a separate test for each fish species and each
vaccines intended for administration by injection or serovar inc1uded in the vaccine, according to a protocol
immersion for the active immunisation of salmonids against defining water source, water fiow and temperature limits, and
vibriosis. preparation of a standardised challenge. Each test is carried
2 PRODUCTION out for each route and method of administration to be
2-1 PREPARATION OF THE VACCINE recommended. The vaccine administered to each fish is of
The strains of L. anguillarum and V o1'dalii are cultured and minimum potency.
harvested separately. The harvests are inactivated by a U se for the test not fewer than 60 fish of the minimum body
suitable method. They may be purified and concentrated. mass to be recommended for vaccination, from a population
Whole or disrupted cells may be used and the vaccine may that does not have specific antibodies against L. anguillantm
contain extracellular products of the bacterium released into or where applicable V o1'dalii and has not been vaccinated
the growth medium. against or exposed to vibriosis. Vaccinate not fewer than
30 fish according to the instructions for use. Perform mock
2-2 CHOICE OF VACCINE COMPOSITION
vaccination on a control group of not fewer than 30 fish;
The strains of L. anguillanmz and V o1'dalii used are shown
mark vaccinated and control fish for identification. Keep all
to be suitable with respect to production of antigens of
the fish in the same tank or mix equal numbers of controls
assumed protective importance. The vaccine is shown to be
and vaccinates in each tank if more than 1 tank is used.
satisfactory with respect to safety (5.2.6) and efficacy (5.2.7)
Where justified and when fish cannot be marked, non-
in the species of fish for which it is intended.
marked fish may be used. Vaccinates and controls may then
The following tests for safety (section 2-2-1) and be kept in the same tank but physically separated (for
irnmunogenicity (section 2-2-2) may be used during the example by fishing nets). Challenge each fish at a fixed
demonstration of safety and efficacy. interval after vaccination, corresponding to the onset of
2-2-1 Safety irnmunity c1aimed, by a suitable route with a sufficient
2-2-1-1 Laborat01Y tests. Safety is tested using test 2-2-1-1-1, quantity of cultures of L. anguillanmz or V o1'dalii whose
test 2-2-1-1-2, or both, depending on the recommendations virulence has been verified. Observe the fish at least daily
for use. until at least 60 per cent specific mortality is reached in the
Carry out the test in each species of fish for which the control group. Plot for both yaccinates and control s a curve
vaccine is intended, using fish of the minimum body mass to of specific mortality against time from challenge and
be recommended for vaccination. U se a batch of vaccine detennine by interpolation the time corresponding to
containing not less than the maximum potency that may be 60 per cent specific mortality in controls.
expected in a batch of vaccine. The test is carried out in the
Vet-326 Veterinary Vaccines 2016

The test is not valid if the specific mortality is less than


60 per cent in the control group 21 days after the 1st death
Enteric Redmouth Disease
in the fish. Read from the curve for vaccinates the Vaccine for Rainbow Trout
mortality (M) at the time corresponding to 60 per cent
mortality in controls. Calculate the relative percentage
(Inactivated)
survival (RPS) using the following expression: Yersiniosis Vaccine (Inactivated) jOl' Salmonids
(Ph. Eur. monograph 1950)
~E~ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ __

1 DEFINITION
Yersiniosis vaccine (inactivated) for salmonids is prepared
The vaccine complies with the test if the RPS is not less than from cultures of serovars 1 or 2 of Yersinia l"Llckeri, inactivated
60 per cent for vaccines administered by immersion and while maintaining adequate immunogenic properties. This
75 per cent for vaccines administered by injection. monograph applies to vaccines intended for administration by
injection or irnmersion for the active immunisation of
2-3 MANUFACTURER'S TESTS salmonids against yersiniosis.
2-3-1 Batch potency test
The potency test (section 3-3) may be carried out for each 2 PRODUCTION
batch of vaccine, using fish of one of the species for which 2-1 PREPARATION OF THE VACCINE
the vaccine is intended. Where the test is not carried out, an The strains of Y. rucke17 are harvested and inactivated by a
altemative validated method based on antibody response may suitable method. They may be purified and concentrated.
be used, the criteria for acceptance being set with reference Whole or disrupted cells may be used and the vaccine may
to a batch of vaccine that has given satisfactory results in the contain extracellular products of the bacterium relea sed into
test described under Potency. The following test may be the growth medium.
used. 2-2 CHOICE OF VACCINE COMPOSITION
Use not fewer than 35 fish from a population that does not The strains of Y. ruckeri used are shown to be suitable with
have specific antibodies against L. anguillal"um inc1uded in the respect to the production of antigens of assumed protective
vaccine and where ápplicable against V. ol"dalii, and that ~re importance. The vaccine is shown to be satisfactory with
within specified limits for body mass. Carry out the test at a respect to safety (5.2. 6) and efficacy (5.2. 7) in the species of
defined temperature. Inject into each of not fewer than fish for which it is intended.
25 fish 1 dose of vaccine, according to the instructions for The following tests for safety (section 2-2-1) and
use. Perform mock vaccination on a control group of not irnmunogenicity (section 2-2-2) may be used during the
fewer than 10 fish. Collect blood samples at a defined time demonstration of safety and efficacy.
after vaccination. Determine for each sample the level of
2-2-1 Safety
specific antibodies against L. anguillarum inc1uded in the
2-2-1-1 Laborato1J) tests. Safety is tested using test 2-2-1-1-1,
vaccine and where applicable against V. ol"dalii, by a suitable
test 2-2-1-1-2, or both, depending on the recommendations
immunochemical method (2.7.1). The test is not valid if the
for use.
control group shows antibodies against L. anguillarum or,
where applicable, against V. ordalii. The vaccine complies Carry out the test in each species of fish for which the
with the test if the mean level of antibodies in the vaccinates vaccine is intended, using in each case fish of the minimum
is not significantly lower than that found for a batch that body mas s to be recommended for vaccination. U se a batch
gave satisfactory results in the test described under Potency. of vaccine containing not less than the maximum potency
that may be expected in a batch of vaccine. The test is
3 BATCH TESTS carried out :in the conditions to be recommended for use of
3-1 Identification the vaccine with a water temperature not less than 10°C.
When injected into fish that do not have specific antibodies
2-2-1-1-1 Vaccines intended jor administration by injection.
against L. anguillarum and, where applicable, V. ordalii, the
Use not fewer than 50 fish from a population that does not
vaccine stimulates the production of such antibodies.
have specific antibodies against the relevant serovars of
3-2 Bacteria and fungi Y. rucke17 and has not been vaccinated against or exposed to
The vaccine, including where applicable the diluent supplied yersiniosis. Where the size of the fish for the test is such that
for reconstitution, complies with the test for sterility a blood sample cannot be removed for antibody testing, a
prescribed in the monograph Vaccines jor veterina1Y use number of larger fish may be kept with the group for this
(0062). purpose. Administer to each fish by the intraperitoneal route
3-3 Potency 1 dose of the vaccine. Observe the fish at least daily for
The vaccine complies with the requirements of the test 21 days.
mentioned under Immunogenicity (section 2-2-2) when The test is not valid if more than 6 per cent of the fish die
administered by a recommended route and method. from causes not attributable to the vaccine. The vaccine
4 LABELLING complies with the test if no fish shows abnormal local or
The label states information on the time needed for the systemic reactions or dies from causes attributable to the
development of immunity after vaccination under the range vaccine.
of conditions corresponding to the recommended use. 2-2-1-1-2 Vaccines intendedjol' administration by immel'sion.
_ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ PhEur Use not fewer th~m 50 fish from a population that does not
have specific antibodies against the relevant serovars of
Y. rucken and has not been vaccinated against or exposed to
yersiniosis. Where the size of the fish for the test is such that
a blood sample cannot be removed for antibody testing, a
number of larger fish may be kept with the group for this
2016 Veterinary Vaccines Vet-327

purpose. Prepare an imrnersion bath at twice the 2-3 MANUFACTURER'S TESTS


concentration to be recomrnended. Bathe the fish for twice 2-3-1 Batch potency test
the time to be recommended. Observe the fish at least daily The potency test (section 3-3) may be carried out for each
for 21 days. batch of the vaccine, using fish of one of the species for
The test is not valid if more than 6 per cent of the fish die which the vaccine is intended. Where the test is not carried
from causes not atu-ibutable to the vaccine. The vaccine out, an altemative validated method based on antibody
complies with the test if no fish shows abnormal local or response in fish or other vertebrate animals may be used, the
systemic reactions or dies from causes attributable to the criteria for acceptance being set with reference to a batch of
vaccine. vaccine that has given satisfactory results in the test described
2-2-2 Immunogenicity under Potency. The following test may be used.
Carry out a separate test for each fish species and each Use not fewer than 25 fish from a population that do es not
serovar included in the vaccine, according to a protocol have specific antibodies against the relevant serovars of
defining water source, water flow and temperature limits, and Y. ruckeri and that are within specified limits for body mass.
preparation of a standardised challenge. Each test is carried Carry out the test at a defined temperature. Inject into each
out for each route and method of administration to be of not fewer than 20 fish 1 dos e of vaccine, according to the
recommended. Where the size of the fish for the test is such instructions for use. Perform mock vaccination on a control
that a blood sample cannot be removed for antibody testing, group of not fewer than 5 fish. Collect blood samples at a
a number of larger fish of the same origin may be selected for defined time after vaccination. Determine for each sample
this purpose. The vaccine administered to each fish is of the level of specific antibodies against the relevant serovars of
minimum potency. Y. rucken: included in the vaccine by a suitable
Use for the test not fewer than 60 fish of the minimum body irnmunochemical method (2. 7.1).
mas s to be recommended for vaccination, from a population The test is not valid if the control group shows antibodies
that does not have specific antibodies against the relevant against the relevant serovars of Y. ruckeri. The vaccine
serovars of Y. ruckeri and has not been vaccinated against or complies with the test if the mean level of antibodies in the
exposed to yersiniosis. Vaccinate not fewer than 30 fish vaccinates is not significantly lower than that found for a
according to the instructions for use. Perform mock batch that gave satisfactory results in the test described under
vaccination on a control group of not fewer than 30 fish; Potency.
mark vaccinated and control fish for identification. Keep all 3 BATCH TESTS
the fish in the same tank or mix equal numbers of controls 3-1 Identification
and vaccinates in each tank if more than 1 tank is used. When injected into animals that do not have specific
Where justified and when fish cannot be marked, non- antibodies against the relevant serovars of Y. ruckeri, the
marked fish may be used. Vaccinates and controls may then vaccine stimulates the production of such antibodies or
be kept in the same tank but physically separated (for protects against virulent challenge with Y. rucker7.
example by fishing nets). Challenge each fish at a fixed
3-2 Bacteria and fungi
interval after vaccination, corresponding to the onset of
The vaccine, including where applicable the diluent supplied
immunity claimed, by injection or immersion, with a
for reconstitution, complies with the test for sterility
sufficient quantity of cultures of Y. ruckeri whose virulence
prescribed in the monograph Vaccines for veterinary use
has been verified or, where all fish are kept in the same tank,
(0062).
with a sufficient challenge by cohabitation. Observe the fish
at least daily until at least 60 per cent specific mortality is 3-3 Potency
reached in the control group. Plot for both vaccinates and The vaccine complies with the requirements of the test
controls a curve of specific mortality against time and mentioned under Immunogenicity (section 2-2-2) when
determine by interpolation the time corresponding to administered by a recommended route and method.
60 per cent specific mortality in controls. 4 LABELLING
The test is not valid if the specific mortality is les s than The label states information on the time needed for
60 per cent in the control group 21 days after the 1sr death development of imrnunity after vaccination under the range
in the fish. Read from the curve for vaccinates the of conditions corresponding to the recommended use.
mortality (M) at the time corresponding to 60 per cent _____________________________________________ PhE~

mortality in controls. Calculate the relative percentage


survival (RPS) using the following expression:

For serovar 1 vaccines, the vaccine complies with the test if


the RPS is not less than 75 per cent for vaccines
administered by immersion and 90 per cent for vaccines
administered by injection.
For serovar 2 vaccines, the vaccine complies with the test if
the RPS is not less than 60 per cent for vaccines
administered by imrnersion and 85 per cent for vaccines
administered by injection.
Vet-328 Diagnostic Preparations 2016

Sterility
DIAGNOSTIC PREPARATIONS It complíes with the test for sterility prescribed in the
monograph Vaccines f01" veterina1Y use (0062).
POTENCY
Avian Tuberculin Purified Protein *** The potency of avian tuberculin purified protein derivative is
*** *** detetmined by comparing the reactions produced in
Derivative *** sensitised guinea-pigs by the intradermal injection of a series
Avian Tuberculin P.P.D. of dilutions of the preparation to be examined with those
(Ph. Eur. monograph 0535) produced by known concentrations of a reference preparation
PhE~ _____________________________________________ calibrated in International Units.
The International Unit is the activity contained in a stated
DEFINITION amount of the International Standard. The equivalence in
Avian tuberculin purified protein derivative (avian tuberculin International Units of the International Standard is stated by
PPD) is a preparation obtained from the heat-treated the World Health Organization.
products of growth and lysis of Mycobacteriu111 aviu111 capable
Sensitise not fewer than 8 albino guinea-pigs, each weighing
of revealing a delayed hypersensitivity in an animal sensitised
400-600 g, by the deep intramuscular injection of a
to micro-organisms of the same species.
suitable dose of inactivated or live M. aviu111. Not les s than
PRODUCTION 4 weeks after the sensitisation of the guinea-pigs, shave their
It is obtained from the water-soluble fractions prepared by flanks to provide space for not more than 4 injection sites on
heating in free-flowing steam and subsequently filtering each side. Prepare dilutions of the preparation to be
cultures of M. aviu111 grown in a liquid synthetic medium. examined and of the reference preparation using isotonic
The active fraction of the filtrate, consisting mainly of phosphate-buffered saline (pH 6.5-7.5) containing 0.005 giL
protein, is isolated by precipitation, washed and re-dissolved. of polysorbate 80 R. Use not fewer than 3 doses of the
An antimicrobial preservative that does not give rise to false reference preparation and not fewer than 3 doses of the
positive reactions, such as phenol, may be added. The final preparation to be examined. Choose the doses such that the
sterile preparation, free from mycobacteria, is distributed lesions produced have a di ame ter of not les s than 8 mm and
aseptically into sterile tamper-proof glass containers, which not more than 25 mm. Allocate the dilutions randomly to the
are then closed so as to prevent contamination. sites, for example using a Latin square designo Inject
The preparation may be freeze-dried. each doseintradermally in a constant volume of 0.1 mL or
The identification, the tests and the determination of potency apply 0.2 mL. Measure the diameters of the lesions after 24-28 h
to the liquid fo1'm and to the freeze-dried form after reconstitution and calculate the results of the test using the usual statistical
as stated 011 the label. methods (for example, 5.3) and assuming that the diameters
of the lesions are directly proportional to the logarithm of the
IDENTIFICATION concentration of the tuberculins.
Inject a range of graded doses intradermally at different sites
The test is not valid unless the confidence limits (P = 0.95)
into suitably sensitised albino guinea-pigs, each weighing not
are not less than 50 per cent and not more than 200 per cent
les s than 250 g. After 24-28 h, reactions appear in the form
of the estimated potency. The estimated potency is not less
of oedematous swellings with erythema, with or without
than 75 per cent and not more than 133 per cent of the
necrosis, at the points of injection. The size and severity of
stated potency. The stated potency is not les s than
the reactions vary according to the dose. Unsensitised guinea-
20 000 IU/rnL.
pigs show no reactions to similar injections.
STORAGE
TESTS
pH (2.2.3)
Protected from light, at a temperature of 5 ± 3 oc.
6.5 to 7.5. LABELLING
Phenol (2.5.15) The label states:
Maximum 5 gIL, if the preparation to be examined contains - the potency in International Units per millilitre;
phenol. - the na me and quantity of any excipient;
- for freeze-dried preparations:
Sensitising effect
- the name and volume of the reconstituting líquid to
Use a group of 3 guinea-pigs that have not been treated with
be added;
any material that will interfere with the test. On 3 occasions
- that the product is to be used irnmediately after
at intervals of 5 days, inject intradermally into each guinea-
reconstitution.
pig adose of the preparation to be examined equivalent to
_____________________________________________
500 IU in 0.1 mL. 15-21 days after the 3rd injection, inject
PhE~

the same dos e (500 IV) intradermally into these animals and
into a control group of 3 guinea-pigs of the same mass,
which have not previously received injections of tuberculin.
24-28 h after the last injections, the reactions of the 2 groups
are not significantly different.
Toxicity
Use 2 guinea-pigs, each weighing not less than 250 g, that
have not previously been treated with any material that will
interfere with the test. Inject subcutaneously into each
guinea-pig 0.5 mL of the preparation to be examined.
Observe the animals for 7 days. No abnormal effects occur
during the observation periodo
2016 Diagnostic Preparations Vet-329

Bovine Tuberculin Purified Protein ***** POTENCY


** ** The potency of bovine tuberculin purified protein derivative
Derivative *** is determined by comparing the reactions produced in
Bovine Tuberculin P.P.D. sensitised guinea-pigs by the intradermal injection of a series
(Ph. Eur. monograph 0536) of dilutions of the preparation to be examined with those
PhE~ ___________________________________________
produced by known concentrations of a reference preparation
calibrated in International Units.
DEFINITION The International Unit is the activity contained in a stated
Bovine tuberculin purified protein derivative (bovine amount of the International Standard. The equivalence in
tuberculin PPD) is a preparation obtained from the heat- International Units of the International Standard is stated by
treated products of growth and lysis of Mycobacterium bovis the World Health Organization.
capable of revealing a delayed hypersensitivity in an animal Sensitise not fewer than 8 albino guinea-pigs, each weighing
sensitised to micro-organisms of the same species. 400-600 g, by the deep intramuscular injection of 0.0001 mg
PRODUCTION of wet mas s of living M. bovis of strain AN5 suspended in
It is obtained from the water-soluble fractions prepared by 0.5 mL of a 9 gIL solution of sodium chloride R. Not less than
heating in free-flowing steam and subsequently filtering 4 weeks after the sensitisation of the guinea-pigs, shave their
cultures of M. bovis grown in a liquid synthetic medium. flanks to provide space for not more than 4 injection sites on
The active fraction of the filtrate, consisting mainIy of each side. Prepare dilutions of the preparation to be
protein, is isolated by precipitation, washed and re-dissolved. examined and of the reference preparation using isotonic
An antimicrobial preservative that does not give rise to false phosphate-buffered saline (pH 6.5-7.5) containing 0.005 giL
positive reactions, such as phenol, may be added. The final of polysorbate 80 R. Use not fewer than 3 doses of the
sterile preparation, free from mycobacteria, is distributed reference preparation and not fewer than 3 doses of the
aseptically into sterile, tamper-proof glass containers, which preparation to be examined. Choose the doses such that the
are then c10sed so as to prevent contamination. lesions produced have a diameter of not less than 8 mm and
The preparation may be freeze-dried. not more than 25 mm. Allocate the dilutions randomly to the
The identificationJ the tests and the detennination of potency apply sites, for example using a Latin square designo Inject each
to the liquid fonn and to the freeze-dried for111 after reconstitution
dose intradermally in a constant volume of 0.1 mL or
as stated on the labe!' 0.2 mL. Measure the diameters of the lesions after 24-28 h
and calculate the results of the test using the usual statistical
IDENTIFICATION methods (for example, 5.3) and assuming that the diameters
Inject a range of graded doses intradermally at different sites of the lesions are directly proportional to the logarithm of the
into suitably sensitised albino guinea-pigs, each weighing not concentration of the tuberculins.
les s than 250 g. After 24-28 h, reactions appear in the form The test is not valid unless the confidence limits (P = 0.95)
of oedematous swellings with erythema, with or without are not less than 50 per cent and not more than 200 per cent
necrosis, at the points of injection. The size and severity of of the estimated potency. The estimated potency is not les s
the reactions vary according to the dose. Unsensitised guinea- than 66 per cent and not more than 150 per cent of the
pigs show no reactions to similar injections. stated potency. The stated potency is not less than
TESTS 20 000 IU/mL.
pH (2.2.3) STORAGE
6.5 to 7.5. Protected from light, at a temperature of 5 ± 3 oC.
Phenol (2.5.15)
LABELLING
Maximum 5 gIL, if the preparation to be examined contains
The label states:
phenol.
-- the potency in International Units per millilitre;
Sensitising efIect -- the na me and quan~ity of any excipient;
U se a group of 3 guinea-pigs that have not been treated with -- for freeze-dried preparations:
any material that will interfere with the test. On 3 occasions -- the name and volume of the reconstituting liquid to
at intervals of 5 days, inject intradermally into each guinea- be added;
pig adose of the preparation to be examined equivalent to -- that the product is to be used irnmediately after
500 IU in 0.1 mL. 15-21 days after the 3rd injection, inject reconstitution.
the same dose (500 ID) intradermally into these animals and ___________________________________________ PhE~

into a control group of 3 guinea-pigs of the same mass,


which have not previously received injections of tuberculin.
24-28 h after the last injections, the reactions of the 2 groups
are not significantly different.
Toxicity Mallein Purified Protein Derivative
Use 2 guinea-pigs, each weighing not less than 250 g, that Mallein P.P.D.
have not previously been treated with any material that will
interfere with the test. Inject subcutaneously into each DEFINITION
guinea-pig 0.5 mL of the preparation to be examined. Mallein Purified Protein Derivative is a preparation of the
Observe the animals for 7 days. No abnormal effects occur heat treated products of growth and lysis of Pseudomonas
during the observation periodo mallei. It contains not less than 0.95 mg per mL and not
more than 1.05 mg per mL óf purified protein derivative.
Sterility
It complies with the test for sterility prescribed in the PRODUCTION
monograph Vaccines fol' veterina1Y use (0062). It is prepared from the water-soluble fractions obtained by
heating in free-flowing steam and subsequently filtering
Vet-330 Diagnostic Preparations 2016

cultures of the glanders bacillus grown in a liquid synthetic Repeat the operation using 2.5 mL of water in place of the
medium. The active fraction of the filtrate, which is preparation being examined. The difference between the
predominantly protein, is isolated by precipitation, washed titrations represents the arnmonia liberated by the substance
and redissolved in phosphate buffered saline at neutral pH. being tested. Each mL of 0.00447M sulfiuic acid VS is
Ir is then distributed in sterile containers that are inert equivalent to 0.875 mg of purified protein derivative.
towards the contents and sealed so as to exclude micro-
STORAGE
organisms. A suitable preservative may be added.
Mallein Purified Protein Derivative should be protected from
CAUTION Mallein P.P.D. is not dangerous to mall, but the light and stored at a temperature between 2° and 8°. Under
organism fram which it is prepared is pathogenic to man and may these conditions it may be expected to retain its potency for
be fatal zf an infection is untreated. Jf an infection is suspected not less than 6 months.
treat111ent should begin without delay.
LABELLING
IDENTIFICATION The label states (1) the volume of the contents; (2) the date
Inject small doses intradermally into suitable guinea-pigs that after which the preparation is not intended to be used;
have been sensitised with killed P. mallei in oily adjuvant. (3) that the preparation is to be used for animals only;
Oedematous swellings occur at the point of injection after (4) the conditions under which it should be stored; (5) the
48 hours. name and percentage of any added preservative; (6) the dose.
TESTS ANNEX
Acidity or alkalinity Guidance to manufacturers performing the test for sterility.
pH, 6.5 to 7.5, Appendix V L. In determining the number of containers to be tested, the
Phenol manufacturer should have regard to the environmental
For preparations containing phenol as a preservative, not conditions of manufacture, the volume of preparation per
more than 0.5% w/v, determined by the method described container and any other special considerations applying to
under Veterinary Antisera. the preparation concerned. With respect to diagnostic
Sterility preparations for veterinary use, 1 % of the containers in a
Complies with the test for sterility, Appendix XVI A, using batch, with a minimum of three and a maximum of 10 is
Method 1: Membrane filtration, whenever possible and considered a suitable number assuming that the preparation
particularly when the volume in a container is greater than has been manufactured under appropriately validated
100 mL, with the following modifications. conditions designed to exclude contamination.
Incubate the media for not less than 14 days at 30° to 35° in 1 Guidance to manufacturers on the number of containers is provided
the test intended to detect bacteria and at 20° to 25° in the in the Annex to this monograph.
test intended to detect fungi.
U se the quantities stated under Application of the test to
injectable preparations except that when the quantity in each
container l is 20 mL or more of a liquid, the minimum
quantity to be used for each medium is 10% of the contents
or 5 mL, whichever is the less.
Abnormal toxicity
Inject 0.5 mL subcutaneously into each of two guinea-pigs.
Neither shows a significant local 01' systemic reaction within
7 days.
ASSAY
To 2.5 mL add 2.5 mL of water and 2.5 mL of a 40% w/v
solution of trichloroacetic acid, mix, allow to stand for
30 minutes and centrifuge for 15 minutes. Discard the
supernatant liquid and dissolve the residue in 0.5 mL of
5M sodiu111 hydroxide. Transfer the solution to a Kjeldahl flask
with the aid of 6 mL of wate/" and add about 0.1 g of a
mixture of 100 parts of potassium sulfate, 10 parts of copper(lI)
sulfate and 5 parts of seleniu111 dioxide and 1 mL of nitrogen-
free sulfuric acid. Evaporate the water and continue heating
until a brown deposit appears. Dissolve the deposit by the
addition of 0.5 mL of hydragen peraxide solution (100 vol),
continue heating until white fumes of sulfur trioxide appear
and boil rapidly for at least 10 minutes. (If while heating a
brown deposit again appears, add a further 0.5 mL of
hydrogen peraxide solution (100 vol). Transfer to an arnmonia
distillation apparatus with the aid of 5 mL of water and add
5 mL of a 50% w/v solution of sodium hydroxide to form a
lower layer. Distil for 3 minutes, collecting the distillate in a
mixture of 5 mL of a 2% w/v solution of boric acid and
0.05 mL of a solution containing 0.066% w/v of methyl red
and 0.033% w/v of bromocresol green in ethanol (96%) and
titrate with 0.00447M sulfuric acid VS (prepared by diluting
89.3 mL of 0.05M sulfuric acid VS to 1000 mL with water).
Monographs

Surgical Materials
2016 Surgical Materials Vet-333

hand ayer the other end held in the left hand, passing one
SUTURES end ayer the strand and through the loop so formed (see
Figure 0660.-1) and pulling the knot tight.

Sterile Catgut in Distributor ***


*** ***
(Catgut, Sterile, in Distributor for Veterina1Y Use, ***
Ph Bur monograph 0660)
PhEif _____________________________________________

DEFINITION
Sterile catgut in distributor for veterinary use consists of
strands prepared from collagen taken from the intestinal
membranes of mammals. After cleaning, the membranes are
split longitudinally into strips of varying width, which, when
assembled in small numbers, according to the di ame ter
required, are twisted under tension, dried, polished, se1ected
and sterilised. The strands may be treated with chemical
substances such as chromium salts to prolong absorption and
glycerol to make them supple, provided such substances do
not reduce tissue acceptability.
The strand is presented in a distributor that aHows the
withdrawal and use of aH or part of it in aseptic conditions. Figure 0660.-1. - Simple knot
The design of the distributor is such that with suitable Make not fewer than one measurement per 2 m of length.
handling the sterility of the content is maintained even when If the strand consists of several sections joined by knots,
part of the strand has been withdrawn. It may be stored dry make not fewer than three measurements per section and, in
or in a preserving liquid to which an antimicrobial any case, not fewer than one measurement per 2 m of length
preservative but not an antibiotic may be added. at points even1y spaced along the strand or along each
TESTS section. Determine the breaking load using a suitable
1f stored in a preserving liquid, remove the strand froln the tensilometer. The apparatus has two clamps for holding the
distributor and measure promptly and in succession the length, su"and, one of which is mobile and is driven at a constant
diameter and breaking load 1f stored in the dly state, im11lerse the rate of 30 cm per minute. The clamps are designed so that
strand in alcohol Rora 90 per cent V/V solution of the strand being tested can be attached without any
2-propanol R for 24 h and proceed with the measurements as possibility of slipping. At the beginning of the test the length
indicated aboye. of strand between the clamps is 12.5 cm to 20 cm and the
knot is midway between the clamps. Set the mobile clamp in
Length
motion and note the force required to break the strand. If the
Measure the length without applying to the strand more
strand breaks in a clamp 01' within 1 cm of it, the result is
tension than is necessary to keep it straight. The length is not
discarded and the test repeated on another part of the strand.
less than 95 per cent of the length stated on the label. If the
The average of aH the results, excluding those legitimately
strand consists of several sections joined by knots, the length
discarded, is equal to or greater than the value in column C
of each section is not les s than 2.5 m.
and no value is les s than that given in column D in
Diameter Table 0660.-1 for the gauge number concemed.
Carry out the test using a suitable instrument capable of
measuring with an accuracy of at least 0.002 mm and having
a circular pressor foot 10 mm to 15 mm in diameter. Table 0660.-1. :. . Diameters and breaking loads
The pressor foot and the moving parts attached to it are Diameter Breaking load
(millimetres) (newtons)
weighted so as to apply a total load of 100 ± 10 g to the Gauge
strand being tested. When making the measurements, lower number A B e D
the pressor foot slowly to avoid crushing the strand. Make not mino max. mino max.
fewer than one measurement per 2 m of length. If the strand 1 0.100 0.149 0.085 0.175 1.8 004
consists of several sections joined by knots, make not fewer
1.5 0.150 0.199 0.125 0.225 3.8 0.7
than three measurements per section. In any case make not
fewer than twe1ve measurements. Make the measurements at 2 0.200 0.249 0.175 0.275 7.5 1.8
points even1y spaced along the strand 01' along each section. 2.5 0.250 0.299 0.225 0.325 10 3.8
The strand is not subjected to more tension than is necessary
3 0.300 0.349 0.275 0.375 12.5 7.5
to keep it straight during measurement. The average of the
measurements carried out on the strand being tested and not 3.5 0.350 0.399 0.325 0.450 20 10
less than two-thirds of the individual measurements are within 4 00400 0.499 0.375 0.550 27.5 12.5
the limits given in the column under A in Table 0660.-1 for
the gauge number concemed. None of the measurements is 5 0.500 0.599 0.450 0.650 38.4 20.0

outside the limits given in the columns under B in 6 0.600 0.699 _0.550 0.750 45.0 27.5
Table 0660.-1 for the gauge number concemed. 7 0.700 0.799 0.650 0.850 60.0 38.0
Minimum breaking load
8 0.800 0.899 0.750 0.950 70.0 45.0
The minimum breaking load is determined ayer a simple
knot formed by placing one end of a strand held in the right
Vet-334 Surgical Materials 2016

Soluble chromium compounds Length


Place 0.25 g in a conical flask containing 1 mL of 'Water R Measure the length in the condition in which the strand is
per 10 mg of catgut. Stopper the flask, allow to stand at presented and without applying more tension than is
37 ± 0.5 oC for 24 h, cool and decant the liquido Transfer necessary to keep it straight. The length of the strand is not
5 mL to a small test tube and add 2 mL of a 10 giL solution less than 95 per cent of the length stated on the label.
of diphenylcarbazide R in alcohol R and 2 mL of dilute sulfuric Diameter
acid R. The solution is not more intensely coloured than a Unless otherwise presClibed, measure the diameter by the
standard prepared at the same time using 5 mL of a solution following method using the strand in the condition in which
containing 2.83 ~lg of potassium dichromate R per millilitre, it is presented. Use a suitable instrument capable of
2 mL of dilute sulfuric acid R and 2 mL of a 10 giL solution measuring with an accuracy of at least 0.002 mm and having
of diphenylcarbazide R in alcohol R (1 ppm of Cr). a circular pressor foot 10 mm to 15 mm in diameter.
Sterility (2.6.1) The pressor foot and the moving parts attached to it are
It complies with the test for sterility as applied to catgut and weighted so as to apply a total load of 100 ± 10 g to the
other surgical sutures. Carry out the test on three sections, strand being tested. When making the measurements, lower
each 30 cm long, cut off respectively from the beginning, the the pressor foot slowly to avoid crushing the strand. Make
centre and the end of the strand. not fewer than one measurement per 2 m of length and in
STORAGE any case not fewer than 12 measurements at points evenly
spaced along the strand. DUling the measurement submit
Store protected from light and heat.
monofilament strands to a tension not greater than that
LABELLING required to keep them straight. Submit multifilament strands
The label sta tes: to a tension not greater than one-fifth of the minimum
-- the gauge number, breaking load shown in column C of Table 0605.-1
-- the length in centimetres or in metres. appropriate to the gauge number and type of material
_____________________________________________ ~E~
concemed or ION whichever is less. For multifilament
strands of gauge number above 1.5 make two measurements
at each point, the second measurement being made after
rotating the strand through 90°. The di ame ter of that point is
the average of the two measurements. The average of the
Sterile Non-absorbable Strands in measurements carried out on the strand being tested and not
less than two-thirds of the individual measurements are
Distributor within the limits given in the columns under A in
(Strands) Sterile N on-absorbable) in Distributor for Table 0605.-1 for the gauge number concemed. None ofthe
Veteri71a7Y Use) Ph Bur 71l0nograph 0605) measurements is outside the limits given in the columns
Ph Eur _____________________________________________ under B in Table 0605.-1 for the gauge number concemed.
DEFINITION
rabIe 0605.-1. - Diameters and minimum breaking loads
The statements in this monograph are intended to be read in
conjunction 'With the individual monographs on sterile non- Diameter Minimum breaking load
(millimetres) (newtons)
absorbable strands in distributor for veterinalY use in the
Gauge AH other
Pharmacopoeia The require11lents do not necessarily apply to sterile A 8 Linen thread non-absorbable
number
non-absorbable strands 'Which are not the subject of such strands
monographs. mino max. mino max. 'e D e D
Sterile non-absorbable strands in distributor for veterinary 0.5 0.050 0.069 0.045 0.085 1.0 0.35
use are strands which, when introduced into a living 0.7 0.070 0.099 0.060 0.125 1.0 0.3 1.5 0.60
organism, are not metabolised by that organism.Sterile non-
1 0.100 0.149 0.085 0.175. 2.5 0.6 3.0 1.0
absorbable strands vary in origin, which may be animal,
vegetable or synthetic. They occur as cylindrical 1.5 0.150 0.199 0.125 0.225 5.0 1.0 5.0 1.5
monofilaments or as multifilament strands. Multifilament 2 0.200 0.249 0.175 0.275 8.0 2.5 9.0 3.0
strands consist of elementary fibres which are assembled by
2.5 0.250 0.299 0.225 0.325 9.0 5.0 13.0 5.0
twisting, cabling or braiding. Suchstrands may be sheathed.
Sterile non- absorbable strands may be treated to render 3 0.300 0.349 0.275 0.375 11.0 8.0 15.0 9.0
them non-capillary, and they may be coloured with colouring 3.5 0.350 0.399 0.325 0.450 15.0 9.0 22.0 l3.0
matter or pigments authorised by the competent authority.
The strands are sterilised. 4 0.400 0.499 0.375 0.550 18.0 n.o 27.0 15.0

They are presented in a suitable distributor that allows the 5 0.500 0.599 0.450 0.650 26.0 15.0 35.0 22.0
withdrawal and use of all or part of the strand in aseptic 6 0.600 0.699 0.550 0.750 37.0 18.0 50.0 27.0
conditions. The design of the distributor is such that with
7 0.700 0.799 0.650 0.850 50.0 26.0 62.0 35.0
suitable handling the sterility of the content is maintained
even when part of the strand has been removed. They may 8 0.800 0.899 0.750 0.950 65.0 37.0 73.0 50.0
be stored dry or in a preserving liquid to which an
antimicrobial preservative but not an antibiotic may be
Minimum breaking load
added.
Unless otherwise prescribed, determine- the minimum
TESTS breaking load by the following method using the strand in
Remove the strand fr0111 the distributor and measure promptly and the condition in which it is presented. The minimum
in succession the length) diameter and minimum breaking load. breaking load is detelmined over a simple knot formed by
placing one end of a strand held in the right hand over the
2016 Surgical Materials Vet-335

other end held in the left hand, passing one end over the The test solution is not more intensely coloured than the
strand and through the loop so formed (see Figure 0605.-1) appropriate reference solution.
and pulling the knot tight.
STORAGE
Make not fewer than one measurement per 2 m of length at Store protected from light and heat.
points evenly spaced along the strand. Determine the
breaking load using a suitable tensilometer. The apparatus LABELLING
has two clamps for holding the strand, one of which is The label states:
mobile and is driven at a constant rate of 30 cm per minute. - the gauge nurnber,
The clamps are designed so that the strand being tested can - the length in centimetres or in metres,
be artached with-out any possibility of slipping. At the - where appropriate, that the strand is coloured and
beginning of the test the length of strand between the clamps intended ro remain so during use.
is 12.5 cm to 20 cm and the knot is midway between the _____________________________________________ PhE~

clamps. Set the mobile clamp in motion and note the force
required to break the strand. If the strand breaks in a clamp
or within 1 cm of it, the result is discarded and the test
repeated on another part of the strand. The average of all the
results, excluding those legitimately dis-carded, is equal to or Sterile Linen Thread in Distributor
greater than the value in colurnn C and no value is less than
that given in colurnn D in Table 0605.-1 for the gauge (Linen ThreadJ SterileJ in Distributor for VeterinaJJ'
nurnber and type of material concemed. UseJ Ph Bur monograph 0608)
PhE~ _____________________________________________

DEFINITION
Sterile linen thread in distributor for veterinary use consists
of the pericyclic fibres of the stem of Linum usitatissimum L.
The elementary fibres, 2.5 cm to 5 cm long, are assembled in
bundle s 30 cm to 80 cm long and spun into continuous
lengths of suitable diameter. The thread may be creamy-
white or may be coloured with colouring marter authorised
by the competent authority. The thread is sterilised.
IDENTIFICATION
A. Dissect the end of a thread, using a needle or fine
tweezers, to iso late a few individual fibres. Examined under a
microscope, the fibres are seen to be 12 ~lm to 31 ~lm wide
and, along the greater part of their length, have thick walls,
sometimes marked with fine longitudinal striations, and a
Figure 0605.-1. - Simple knof
narrow lurnen. The fibres gradually narrow to a long, fine
point. Sometimes there are unilateral swellings with
Sterility (2.6.1) transverse lines.
They comply with the test for sterility as applied to catgut B. Impregnate isolated fibres with iodinated zinc chloride
and other surgical sutures. Carry out the test on three solution R. The fibres are coloured violet-blue.
sections each 30 cm long, cut off respectively from the TESTS
beginning, the centre and the end of the strand.
It complies with the tests prescribed in the monograph on
Extractable colour Strands sterile non-absorbable in distributor for veterinaJY
J J

Strands that are dyed and intended to remain so during use use (0605).
comply with the test for extractable colour. Place 0.25 g of Jf stored in a dry stateJ expose to an at1110sphere with a relative
the strand to be examined in a conical flask, add 25.0 mL of hU111idity of 65 ± 5 per cent at 20 ± 2 oC for 4 h i111mediately
water R and cover the mouth of the flask with a short- O

befare 111easun ng the diameter and for the determination of


sternmed funnel. Boil for 15 min, cool and adjust to the minimum breaking load im111erse in water R at room temperature
original volurne with water R. Depending on the colour of the for 30 min i111111ediately befare canying out the test.
strand, prepare the appropriate reference solution as
described in Table 0605.-2 using the primary colour STORAGE
solutions (2.2.2). See the monograph on Strands sterile non-absorbable in
J J

distributor for veterinaJY use (0605).


Table 0605.-2. - Colour reference solutions LABELLING
Colour oC strand Composition oC reCerence solution See the monograph on Strands sterile non-absorbable in
J J

(parts by volume) distributor for veterinary use (0605).


Red Yellow Blue Water ____________________________________________ PhE~

primary primary primary


solution solution solution
YeIlow - brown 0.2 1.2 8.6
Pink - red 1.0 9.0

Creen - blue 2.0 8.0


Violet 1.6 804
Vet-336 Surgical Materials 2016

100 giL solution of sodium hydroxide) but is attacked by


Sterile Poly(ethylene terephthalate) dilute mineral acids (for example 20 gIL sulfuric acid), by
Suture in Distributor hot glacial acetic acid and by 70 per cent mlm formic acid.
(Poly(ethylene terephthalate) Suture) Sterile) in IDENTIFICATION
Distributor for VeterinalY Use) A. Heat about 50 mg with 0.5 mL of hydrochloric acid R1 in a
Ph Bur monograph 0607) sealed glass tube at 110 oC for 18 h and allow to stand for
~E~ ___________________________________________ 6 h. No crystals appear.
DEFINITION B. To about 50 mg add 10 mL of hydrochlO1ic acid R1.
Sterile poly(ethylene terephthalate) suture in distributor for The material disintegrates in the cold and dissolves
veterinary use is obtained by drawing poly(ethylene completely within a few minutes.
terephthalate) through a suitable die. The suture is prepared C. It dissolves in a 70 per cent mlm solution of anhydrous
by braiding very fine filaments in suitable numbers, fonnic acid R.
depending on the gauge required. It may be whitish in TESTS
colour, or may be coloured with authorised colouring matter It complies with the tests prescribed in the monograph on
or pigments authorised by the competent authority. Strands) stenle non-absorbable) in distributor for vetennary
The suture is sterilised. use (0605) and with the following test:
CHARACTERS Monomer and oligomers
It is practically insoluble in most of the usual organic In a continuous-extraction apparatus, treat 1.00 g with
solvents, but is attacked by strong alkaline solutions. It is 30 mL of methanol R at arate of at least three extractions per
incompatible with phenols. hour for 7 h. Evaporate the extract to dryness, dry the
IDENTIFICATION residue at 110°C for 10 min, allow to cool in a desiccator
A. It dissolves with difficulty when heated in and weigh. The residue weighs not more than 20 mg
dimethylformamide R and in dichlorobenzene R. (2 per cent).
B. To about 50 mg add 10 mL of hydrochloric acid R1. STORAGE
The material remains intact even after irnmersion for 6 h. See the monograph on Stl"ands) stenle non-absorbable) in
distributor for vetennary use (0605).
TESTS
It complies with the tests prescribed in the monograph on LABELLING
Strands) stelile non-absorbable) in disuibutor for vetelinary See the monograph on Su"ands) stenle non-absorbable) in
use (0605). disuibutor for vetennalY use (0605).
STORAGE The label states whether the suture is braided, monofilament
See the monograph on Strands) sterile non-absorbable) in or sheathed.
distributor for veterinary use (0605). ___________________________________________ ~E~

LABELLING
See the monograph on Strands) stelile non-absorbable) in
disuibut01" for vetelinalY use (0605).
***
___________________________________________ Sterile Polyamide 6/6 Suture in
*** ***
PhE~

Distributor ***
(Polyamide 616 Suture) Stenle) in Disuibutor for
VetennalY Use) Ph Bur monograph 0610)
***
Sterile Polyamide 6 Suture in
** ** NOTE: The name Nylon 6/6 as a synonyrn for Polyamide 6/6

Distributor ***** may be used freely in many countries including Great Britain
and N orthern Ireland, but exclusive proprietary rights in this
(Polyamide 6 Suture) Sterile) in Dismbutor for
name are claimed in certain other countries.
VetelinalY Use) Ph Bur monograph 0609)
~E~ ___________________________________________
NOTE: The name Nylon 6 as a synonym for Polyamide 6 may
be used freely in many countries, including Great Britain and DEFINITION
Northern Ireland, but exclusive proprietary rights in this Sterile polyamide 6/6 suture in distributor for veterinary use
name are claimed in certain other countries. is obtained by drawing through a suitable die a synthetic
~E~ ___________________________________________ plastic material formed by the polycondensation of
hexamethylene-diamine and adipic acid. It consists of
DEFINITION smooth, cylindrical monofilaments or braided filaments, or
Sterile polyamide 6 suture in distributor for veterinary use is lightly twisted strands sheathed with the same material.
obtained by drawing through a suitable die a synthetic plastic It may be coloured with authorised colouring matter or
material formed by the polyrnerisation of ¡:;-caprolactam. pigments authorised by the cómpetent authority. The suture
It consists of smooth, cylindrical monofilaments or braided is sterilised.
filaments, or lightly twisted strands sheathed with the same
CHARACTE~S
material. It may be coloured with colouring matter
authorised by the competent authority. The suture· is It is practically insoluble in the usual organic solvents; it is
sterilised. not attacked by dilute alkaline solutions (for example a
100 giL solution of sodium hydroxide) but is attacked by
CHARACTERS dilute mineral acids (for example 20 gIL sulfuric acid), by
It is practically insoluble in the usual organic solvents; it is hot glacial acetic acid and by 80 per cent 1111m formic acid.
not attacked by dilute alkaline solutions (for example a
2016 Surgical Materials Vet-337

IDENTIFICATION LABELLING
A. In contact with a fiame it melts and burns, forming a hard See the monograph on Strands~ sterile non-absorbable, in
globule of residue and gives off a characteristic odour distlibutor for veterinary use (0605).
resembling that of celery. ___________________________________________ PhE~

B. Place about 50 mg in an ignition tube held vertically and


heat gently until thick fumes are evolved. When the fumes fill
the tube, withdraw it from the fiame and insert a strip of
nitrobenzaldehyde paper R. A violet-brown colour slowly
appears on the paper and fades slowly in air; it disappears
irnmediately on washing with dilute sulfuric acid R.
C. To about 50 mg add 10 rnL of hydrochloric acid Rl.
The material disintegrates in the cold and dissolves within a
few minutes.
D. It does not dissolve in a 70 per cent nzl1n solution of
anhydrous fornzic acid R but dissolves in an 80 per cent nzl1n
solution of anhydrous formic acid R.
TESTS
It complies with the tests prescribed in the monograph on
Strands~ sterile non-absorbable~ in distributor for veterinary
use (0605).
STORAGE
See the monograph on Strands, stel'üe non-absorbable~ in
distributor for veterinary use (0605).
LABELLING
See the monograph on Strands, sterile non-absorbable~ in
distributor for vetelina¡y use (0605).
The label states whether the suture is braided, monofilament
or sheathed.
___________________________________________ ~E~

Sterile Braided Silk Suture in


Distributor
(Silk Suture~ Sterile~ Braided, in Distributor for
Veterinary Use~ Ph Bur nzonograph 0606)
PhE~ ___________________________________________

DEFINITION
Sterile braided silk suture in distributor for veterinary use is
obtained by braiding a variable number of threads, according
to tlle diameter required, of degummed silk obtained from
the cocoons of the silkworm Bombyx mori L. It may be
coloured with colouring matter authorised by the competent
authority. The suture is sterilised.
IDENTIFICATION
A. Dissect the end of a strand, using a needle or fine
tweezers, to isolate a few individual fibres. The fibres are
sometimes marked with very fine longitudinal striations
parallel to the axis of the strand. Examined under a
microscope, a cross-section is more or les s triangular or semi-
circular, with rounded edges and without a lumen.
B. Impregnate isolated fibres with iodinated potassium iodide
solution R. The fibres are coloured pale yellow.
TESTS
It complies with the tests prescribed in the monograph on
Strands~sterile non-absorbable, in dist1ibutor for veterina¡y
use (0605).
STORAGE
See the monograph on Strands~ sterile non-absorbable~ in
distributor for vetelina¡y use (0605).
2016

Infrared Reference Spectra


Vet-S2 Infrared Reference Spectra 2016

AH spectra presented in this section were recorded using either a


Perkin-Elmer model 682 dispersive infrared spectrophotometer or
a Perkin Elmer model 16PC Fourier transform infrared
spectrophotometer.
Pressed discs, 13 mm in diameter, were prepared using
potassium bromide or potassium chloride. Liquid paraffin muHs
and thin films were prepared between potassium bromide plates,
and gas and solution spectra were prepared using ceHs with
potassium bromide windows. Solution spectra were prepared
against a solvent reference and aH other spectra were recorded
against airo
For solution spectra the regions of the spectrum within which
the solvent shows strong absorption should be disregarded. Solvent
'cut-offs' in the reference spectra may be recorded as horizontal
straight lines or may appear as blank regions on the spectrum.
2016 Infrared Reference Spectra Vet-S3

Polystyrene Instrument: Dispersive Phase: Thin Film Thickness: O.038mm


100.0

80 .~ ~"\
/V"JWv
j\ ~
'" (

60

40

cf2.
ID
() 20
c:
ro
:::::
"E
en
c:
ro
¡::: 0.0 J
4000.0 3600 3200 2800 2400 2000.0
Wavenumber (cm-1)
100.0

80
/ rv'
\(t \ AAV\
A

r rf\ An ~
f'/\
~r
ftP
j
JI) n I
60 ft
V V l ti V
\ I
\ I
AV V fI

40 .

V ~
~
c:
20 \
\
ro
:::::
"E
en
c:
ro
¡::: 0.0 ~
2000.0 1800 1600 1400 1200 1000 800 600 400.0
Wavenumber (cm-1)
Vet-S4 Infrared Reference Spectra 2016

Polystyrene Instrument: Fourier Transform Phase: Thin Film Thickness: O.038mm


100.0

80
~
~
--------- - ~ ~
60 I

~
40 ..... ....

~ ~
Q)
ü 20
c:
ro
~
E
CJ)
c:
~
1- 0.0 \J
4000.0 3600 3200 2800 2400 2000.0
Wavenumber (cm-1)
100.0

80 A
(1(\
60 ~(\A '0fV1 M f"
n ~~ ~ ~ h~ 0r 1\ (

\! ~ ~\ /
(\
V I f'\
\
40 y
t .'

cf2-
Q)
ü
c:
ro
:::::
20 \~
"E
CJ)
c:
ro
¡.=: 0.0 .. v \j
2000.0 1800 1600 1400 1200 1000 800 600 400.0
Wavenumber (cm-1)

Acepromazine RSVOl Instrument: Fourier Transform Phase: Thin Film


100.0 . . ,. ,.,"'' ' ' ' ' ' ' ' ' ' ' ' ' ' ' ' ' ' ' ' ' ' ' ' , . . ,."".,'' ' ' '""""'' ' ' ' ' ' ' ' ' ' ' ' ' '.' ' ' ' ' ' ' ' ' ' ' ' ' ' ' ' ' ' ' ' ' ' ' ' ' ' ' ' '.""'' ' ' ' ' ' ' ' ' ' ' ' ' ' ' ' ' ' ' ' ' ' ' ' ' ' .' ' ' ' ' ' ' ' ' ' ' ' ' ' ' ' ' ' """""""'' ' ' ' ' ' ' ' ' ' ' ' ' ' ' ' ' ' ' ' ' ' ' ' ' ' ' ' ' ' ' '.' ' ' ' ' ' ' ' ' ' """''''''''''''''''''''''''''''T''''''''''''''''''''''''''''''''''''''''''''''''''''',

80, . . . ,."."., . ,. "" ". . ,. . ,. , ! \ " . " , .. ",.", .. ,.,., .. ". '~""""""""""""""<""""""""""'",.".".",." .••.•• , • • • ",.""",.,'., .• , •• " •••• ,.".,.,."".'", • • • • • • • . " •• , •• ",."",.,."., •••.••. , •••••• , ••• +"."."".'''I\l\i0~w

60 .".,. ,.".""""""., . . ". ". ", ... +1 f 11,1 . j " " . , ..... <.""".",.,., ... , ..... , .. """,."", .. "." ++IHI"M·,·,'"""H,··········· "., ,,1 J+I,,·····,···, 11""".,·"·""""""···,···"···"··,,,·,,,,,,',," .. " .

40;., . "",."., . ,. . ". ,. . ", . . ,., . . ",' •... "·'·"··"""'··'II·'··'···"'·IIU·'·"""·'·I··"·"""f""~"""""'~'"IIH'''''''''''''''''''''''''''""·"l"·,·"··,·"""",···,········,··,,,,,···,,,,.': 1 1 " " " " " " " " " " " " " " " " " " " " " " " " " " , . , ' , •• ,", •. ,." •..• , •.• ,.".".'.

~ 20."""""""",..,,, ,,' ,,,1/,,,,,,.. ,,,, ..·,.,·,..,·. "+/"+'¡",,,,,,,,·,,,Y.'lll.,,; ... ,,,,,,,,,,, .. ,,,,;,,,,,,,,,,,,,,,,,;' ""'" '"" ... ' ' ' ''''''','
c:
ro
:::::
"E
CJ)
c:
ro
¡.=: 0,0""., . ". . """", . .". ". . ", .• '"",."."."., . . ,.""","""", . . ,."" .•'""""", . ,. ,. . ,.", . "",.",. . . . """.".".,., . . . ",.""."""""., . .,. . . ,., • """" . ,. ,. . ,.,.".""""",,, ."."",."",."."., . "., . . "",,,,,;"",., . .,. . . "".",""""""""""'" , ",.".""""""""""',.,",.,.,., . . ,""',.,".
2000,0 1800 1600 1400 1200 1000 800 600 400,0
Wavenumber (cm-1)
2016 Infrared Reference Spectra Vet-S5

Alfadolone Acetate RSV02 Instrument: Dispersive Phase: Potassium Bromide Disc


100.0 ..•.........................................................•..............................................•............................................... c................................................•......................................................•................................................... c ........................ u ............ c ..................................... u

60 , ............................................ ,¡ ........... , ...........•................................... 1 .......; ............ +uIH·············+u~

40 . ,.......................................................... \ /\./ . . . . . . ,............................................ 1.111...................... 1/

~ 20 •...................... uu. IlIJu ........... ¡ ................................. u •................................. 11' ............................................ ( ......................................................•..............................................•................................................. ,


c:
ro
:::::
'EU)
c:
ro
~ 0.0 •................................................ ,. . . . . . :...........................................,.................................................•.................................................... , ............................................ } ...........................................•............................................•............................................ ,
2000.0 1800 1600 1400 1200 1000 800 600 400.0
Wavenumber (cm-1)

Alfaxalone RSV03 Instrument: Dispersive Phase: Potassium Bromide Disc


100.0

80 ---~ ,./ ~ Mn 1\1\ n((VV(íf\(tJ r-,


\I \¡ ~ rf1 ~ ~ NV"V~\ j ~V iV~
60 ~
N
I~
1)
40

~
o

ID
ü 20
c:
ro
:::::
'E
U)
c:
ro ~
~ 0.0
2000.0 1800 1600 1400 1200 1000 800 600 400.0
Wavenumber (cm-1)

Amitraz RSV04 Instrument: Fourier Transform Phase: Potassium Bromide Disc


100.0

- ...... y---
~

~ ¡"')l"'j ~
r.tfot
(V~ í\( Nr
80 0dt'r\R~
~~I .~

\
H

trvV JVfI
60 ,
I
~
40
r

cf2.
ID
ü 20
c:
ro
:::::
'E
U)
c:
ro v
~ 0.0
2000.0 1800 1600 1400 1200 1000 800 600 400.0
Wavenumber (cm-1)
Vet-S6 Infrared Reference Spectra 2016

Amoxicillin Trihydrate RSV05 Instrument: Dispersive Phase: Potassium Bromide Disc


100.0 . "........................................................ ,......................................................... ,........................................................"........................................................,........................................................ ,.........................................................,........................................................,.........................................................,

60+ .......................... ········l ..······.. ····I~·········.. ···················, .................................................. , ...................................... ,;Hf~

?fl.
Q)
ü 20
e
ro
::::::
"E
en
e
ro
.= 0.0
2000.0 1800 1600 1400 1200 1000 800 600 400.0
Wavenumber (cm-1)

Ampicillin Trihydrate RSV06 Instrument: Dispersive Phase: Potassium Bromide Disc


100.0 .,.......................... ........................................ ....................................... ............ ......................... ............................................... . ............................................................................................................... ,.........................................................,

80+ ........... ~ ...., .... , .............................. , ................................................................................................................................................ ~ . . . . . . . ~, .................•................................................. , ............................................... ,

60 . .". . . . . . . . . . . . . . . . . . . . . . . . . . . \ ........... 1' .........................•................................... I

40 . .••.. . . . . . . . . . . . . . . . . . . . . . . '11 1+1 • 1 1 IIY··········H.NIFH 1/·······.············································ ....................................................................................................................................................................................... '

~
Q)
ü 20
e
ro
::::::
"E
en
e
ro
.= 0.0
2000.0 1800 1600 1400 1200 1000 800. 600 400.0
Wavenumber (cm- 1
)

Amprolium Hydrochloride RSV07 Instrument: Fourier Transform Phase: Potassium Chloride Disc
100.0, ..........................., ...............................,................................................ , . . . . . . . . . . . . . . . . . . . . . . . . . . . . ,......................................................... ,...................................................•................................................ , .................................................,

60 ~·I··········~·················\I ...... +··········· ..................................... j .................................................... fl {"t.········.. ·.·········" 11·········· 1·~··················/VIl·········· •....................................................•

~ 20+ ......................................................................... \/1


e
ro
::::::
"E
en
e
ro
.= 0.0+ .............................. , ....................... ..;. ....................................................................................................................,...........................................,...........................................................•.......................................... ,
2000.0 1800 1600 1400 1200 1000 800 600 400.0
Wavenumber (cm- ) 1
2016 Infrared Reference Spectra Vet-S7

Azaperone RSV08 Instrument: Fourier Transform Phase: Potassium Bromide Disc


100.0

80
~ - '\ rVV y
~f\ I"l

~~~~ v ~
ftt\A

Al ~(V1M
't'I

60
(\ r~ A nA A n,
11

~. I~ (V Ir
11 1V

N
40 WA ~
~~ W

~ 20
e
ro
:::::
"E(f)
e
ro
¡.:: 0.0
2000.0 1800 1600 1400 1200 1000 800 600 400.0
Wavenumber (cm-1)

Cefalonium RSV09 Instrument: Dispersive Phase: Potassium Bromide Disc


100.0 ...•...................................................................................................................•........................................................•................................................. , .................................................................................................................•...................................................................................................
H.

80 •.......................................... ~. . . ,........................................................•........................................................•.........................................................•................................ ~. . . " \ r \ •..................•................................................... + .................................................. ,

60 •......................................... \···········/11···················· • ·········· IV\I +I+A································~I···Y···IFII'\,LIIIv··········· y ..•............................................ !Il~n····················· f·····.··············································· .........,

40

~
Q)
u 20
e
ro
:::::
"E(f)
e
ro
¡.:: 0.0
2000.0 1800 1600 1400 1200 1000 800 600 400.0
Wavenumber (cm-1)

Chlo.-amphenicol RSVI0 Instrument: Fourier Transform Phase: Potassium Bromide Disc


100.0 ..•......................................................•......................................................•..................................................................................................................•................................................ H .. H • • H .. HHH .. HHH .. H .... H ...... H .... HHHH.HH ..... H .. H .... H .......... HH ... HH .... H .. H ...... H ...... H .. HHH.HH .. H .. H ...... H .... H .. HH .. HH ... .

60 . ,.........................................................•................. \ . . . . . . ,~

~ 20 ..•................................................. ·······.··························11·················· .•........... +1·····················. H·······································.··································\1············.·················· ...................................•.....................................................•.......................................... :


e
ro
:::::
"E
(f)
e
ro
¡.:: 0.0 ...........................................................•.......................................................•........................................................•........................................... + .........................................................................................................•............................................•.......................................................•
2000.0 1800 1600 1400 1200 1000 800 600 400.0
Wavenumber (cm-1)
Vet-S8 Infrared Reference Spectra 2016

C1opl'ostenol Sodium RSVll Instrument: Fourier Transform Phase: Liquid Paraffin Mull
100,0 . ;,................... "., . . . . . ,. . . . . . ,. . ,. . .,. . . . ,. . . ,......... ,......................,.,. . . ,; . ,. . . ,..................,. . . . ,. . ,."."., . . . ,." . . ,. . . ,. . . . . . . ,. . .,. . ,................. T" ... '.' .... ,...... ,...... """ ..."",, .. ,.. ,",, .. ,;." ........ " ..... " .. ",." ...... "'" .. ,...... ,.... "';''', .. "'',, ...... ,, ..... ,, .... "." .. " .... """" .. "" ..... " ....... "." .... ,,, .. ,,, ............ ,.... ,,""":'

60..;,., . "., . ,,,,,,,,,,,. . ,,.,,,.,,;,,.,.,,. " " A " .. ,.. ,... ,.,¡ .. ,." ......... ,....... ,.... ,... " .. ,.. ,.. ,.. )",,,,,,,,,,.,,.,,,,,,,,;.,,,, . . .,,., ... ,..•. ,..... , " . ¡ ,.... ,.. ,.. " ............... ,.... ' ¡ .. ,.. "... ,... ,.... ,.,."."., .... " .. ,.... ,... , " ...... " .. ,.... ",.", .. ,,(

~
ID
ü 20
e
ro
::::::
"E(j)
e
ro
¡.=: 0,0
2000,0 1800 1600 1400 1200 1000 800 600 400,0
Wavenumber (cm-1)

Cloxacillin Benzathine RSV12 Instrument: Fourier Transform Phase: Liquid Paraffin Mull

60+ . ,. "."." . ,. ,.",., . . ". ". ".""., . , f ,',.,",."." .. ,.. : ,........... ,.. ,........... ".. ,..."." .. ,.... + . ".,",.,',.".,"",.".,',... ".,'~ . . ,.~

?f?
ID
ü 20
e
ro
::::::
"E(j)
e
ro
¡.=: 0,0
2000,0 1800 1600 1400 1200 1000 800 600 400,0
Wavenumber (cm-1)

Cloxacillin Sodium RSV13 Instrument: Dispersive Phase: Potassium Bromide Disc


100,0"",.... "."."."., ..... ,. . ".,,,,. . ,,.,.,,,,,.",., . . ,. ".,',." . ,.,',., . ". ,. . ,. ,. . ,. . . ,., . . , """ "",.".".".,,, ".,.... ,. ".' . "."". ,",.,.,."., . . . . . . ,.....".......... , ;,. . ,. . . ,., . ,., . . . . . . ". . . ,. . ,. . , . . ,. . ,. . . . . . ". . . . . ,. . . . . ,. .

60}, . . . ". . . ,. . . .". ,.""""",. "", . :I +."t

8e 20 ¡ ,.. ,........... ,.. ,......... ,. . ,... ,... ,. ¡ H,·,··,,·,·····,"",·,"',·,',·· Ihl,,···,,······,··,,·····,··,,··,···,··,,·,' '¡.".,',.,',." . . ".,', .. " .......... ¡ ...... ,... " ".. ,.. ,... ,..... ,.. "" .. ,., .... ¡,."., .. ,... " .. ".",."", .. ,."".,., .. ,.. ,.. ,.•.. ,.,." ... ,, .. "." .... ",.", .. ""." .. ",." ... ;,."".",., .. ,.. ".", .. " .. "".", ... ,. ,

ro
::::::
"E
(j)
e
ro
¡.=: O,O< ............."......,.~, .......... ,.... "... "" . ".... ".;~.....,.... ,.............. " .."" ..;..."...." .............. ".~:''',., ..,........". ,.",., ... "." . . . , .......... ,.,... ,."""., . ".......,. ,. ", . ". . ,. . ,. ,., . ",."" . . ".,.".,.",.,.", . ,.", ...... """, . . ,. . "." . ,. ,....¡
2000,0 1800 1600 1400 1200 1000 800 600 400,0
Wavenumber (cm-1)
2016 Infrared Reference Spectra Vet-S9

Decoquinate RSV14 Instrument: Fourier Transform Phase: Potassium Bromide Disc


100,0

80
'"'"'
(f'y (W An /\ .-'\~

'\ ~ AA~ ~ AA!n~


60

40 ~ / ~ ~r f' r r
11 ~ V

~ V V
~ 20 1/
e V
ro
::=
-E
(J)
e
ro
.= 0,0
2000,0 1800 1600 1400 1200 1000 800 600 400.0
Wavenumber (cm-1)

Deltamethrin RSV 47 Instrument: Dispersive Phase: Potassium Bromide Disc


100,0

80 ~ \ /\
¡l{v vy /'\ ""

Irfv~
A

(~
\

n fI~ ~ n r ~ f I ¡~
60
f A~ d
IV~
~ r
(

M f .~ ~ •.

40 ........ ~ \
~ 20
e
ro
::=
-E
(J)
e
ro
.= 0,0 ••

2000,0 1800 1600 1400 1200 1000 800 600 400.0


Wavenumber (cm-1)

Dimetl'idazole RSV16 Instrument: Fourier Transform Phase: Potassium Bromide Disc


100.0

1\
~ v'

80 JC'\ r\(1 /lr1í VI


¡
.....

v1
~~
V I
60 \ l
40
\ A
M
I

Al •

~ N('
IV
~ 20
e
ro
::=
-E
(J)
e
V VVV
~
ro
.= 0,0

2000,0 1800 1600 1400 1200 1000 800 600 400.0


Wavenumber (cm-1)
Vet-S 1O Infrared Reference Spectra 2016

Dimpylate RSV49 Instrument: Fourier Transform Phase: Thin Film


100.0 .. ,........................................................ ,................................................... ".................................................................................................... ,....................................................... ,.................................................. ,.............................................................................................................,

80 ..••....................................................•.............................

60 .. ".................................................. ....•............................ ..... ........... ¡ ........... / ..... +,I

40 ......................... ww .•...•.•..•..•........•.•.... ' >.w·······················.·· .•······ + l·.·············

-¿¿
CD 20
ü
e
ro
~
E
en
e
ro
~ 0.0
2000.0 1800 1600 1400 1200 1000 800 600 400.0
Wavenumber (cm-1)

Dinitolmide RSV17 Instrument: Fourier Transform Phase: Potassium Bromide Disc


100.0

80

60

40

-¿¿
CD 20
ü
e
ro
:::::
"E
en
e
ro
~ 0.0
2000.0 1800 1600 1400 1200 1000 800 600 400.0
Wavenumber (cm- 1
)

Dip,'enorphine Hydrochloride RSV18 Instrument: Fourier Transform Phase: Potassium Chloride Disc
100.0 ..•................................................... ,.................................. .

80

60

40

~ 20
e
ro
~
E
en
e
ro
~ 0.0........................................................................................................................................................ ......•.................................................................................................................... '.......................................................................................................................................................................... '
2000.0 1800 1600 1400 1200 1000 800 600 400.0
Wavenumber (cm- 1
)
2016 Infrared Reference Spectra Vet-S 11

Etamiphylline RSV19 Instrument: Dispersive Phase: Liquid Paraffin Mull


100.0 ............................................................................................................................................................................................................................................................................................................................................................................................................,.........................................................

80

60

40 > ...................................... , .......... 11111 ............................................ h)

cf2.
Q)
ü 20
e
ca
:::::
"E
en
e
ca
r= 0.0
2000.0 1800 1600 1400 1200 ' 1000 800 600 400.0
Wavenumber (cm-I )

Etamiphyllinc Camsilate RSV51 Instrument: Dispersive Phase: Liquid Paraffin Mull


100.0 . ,........................................................ ,............................................................................................................................................................................ ,............................................................................................................................................................................................................................

80

60

40

cf2.
Q)
ü 20
e
ca
:::::
"E
en
e
ca
r= 0.0
2000.0 1800 1600 1400 1200 1000 800 600 400.0
Wavenumber (cm-I )

Ethopabate RSV20 Instrument: Fourier Transform Phase: Potassium Bromide Disc


100.0

80

60

40

25 20
e
ca
.E
E
en
e
ca
r= 0.0
2000.0 1800 1600 1400 1200 1000 800 600 400.0
Wavenumber (cm-I )
Vet-S 12 Infrared Reference Spectra 2016

Fenthion RSV21 Instrument: Dispersive Phase: Thin Film


100.0

80 ~~

'f"\
--... f\

n /V V\
...... 01\A
"""'" N 1, nr vv \

60 r
I \IA~
40
N
~
Q)
ü
e
20
\ ~ ~ ~j
ro
::::::
'E
en
e
ro
V
.= 0.0
2000.0 1800 1600 1400 1200 1000 800 .600 400.0
Wavenumber (cm-1)

Fluanisone RSV22 Instrument: Fourier Transform Phase: Potassium Bromide Disc


100.0

80 L~ .-'\ A/vo.~

f f' ~rv
~(\ (\ nNr~
~
,~f
ti

60
~ A 11, h h

~ I~
I~ ~ ~~ ~
V

N
40 ~ ~
/' ~
~
Q)
ü 20
e
ro
::::::
'E
en V
e
ro
.= 0.0
2000.0 1800 1600 1400 1200 1000 800 600 400.0
Wavenumber (cm-1)

Flunixin Meglumine RSV23 Instrument: Fourier Transform Phase: Liquid Paraffin Mull
100.0 . .,........................................................,..........................................................,........................................................"........................................................,.........................................................,.........................................................,........................................................ ,......................................................... ,

80+ ............................................... ; ............................................... i ............................................... + .........................................,.................................................. , ................................................·. . i ................................................. + .................................................... ;

60 . .,................................................... , . . . . . . . . . . . . . . . . . . l········;············································ ............ ,•.......................................................¡ ............................................ iI"~,JII

40 > .................................................. '......................................... \ , ++ . . . . . <..............··.·.····. ·. .f·.·······•.·¡IIHI.I·.·..···.··.. IIJI .. ,·.·····.····.····.······· ...··.····....If·HH.,·.····,·,". , .....,............. ,......... ,.... ,..... ,

~ 20 > ................................................ ' ................................................. L/Al) I


e
ro
::::::
'E
en
e
ro
.= 0.0 . .;.................................................... , ....................................................;.......................................................•;.........................................................•............................... ,.................... , ..................................................•........................................................•........................................................•
2000.0 1800 1600 1400 1200 1000 800 600 400.0
Wavenumber (cm- ) 1
2016 Infrared Reference Spectra Vet-S 13

Griseofulvin RSV24 Instrument: Fourier Transform Phase: 1.5% w/v Solution in Chloroform Thickness: 0.1 mm

r
100.0

~r() n/\ .IV


h
80 f\
I
(I~ . . ~ Nl(V
, I···~···v , ......

60 1
u

IJ
40

?f?
Q)
ü 20
e
ro
::=:
"E
rn
e
ro
¡.=: 0.0
2000.0 1800 1600 1400 1200 1000 800 600 400.0
Wavenumber (cm-1)

Levomepromazine RSV25 Instrument: Fourier Transform Phase: Liquid Paraffin Mull


100.0 ; . . . . . . u........................ , ................................... ¿u.uu ........................ ; u ............ u ...... u.... ,. uu . .uu u ............ uu'uuuu ............ u uu, uuuu . u ' u u ......................... ,

80 ¡ ........................................ +......................................... / \ + .......................................... ; ............................... u •••••••••••••••••••••• ; ••••••••••••••••••••••• u ••••••••.••••••••••••••••• + .......................................... j .................... Iw··········· 11."···········'11.·······.···.·,

60 . ;.........................................................;............................................. ········f··' l························;···························· ....... ~A····;····························iH\p·······~·······............ \

40 , ........................................... , . . . . . . . . . . . . . . . . . . . . . . '11. Jlu+;H.¡

?f?
Q)
ü 20
e
ro
::=:
"E
rn
e
ro
¡.=: 0.0
2000.0 1800 1600 1400 1200 1000 800 600 400.0
Wavenumber (cm-1)

Lincomycin Hydrochloride RSV52 Instrument: Dispersive Phase: Potassium Chloride Disc


100.0 . ,.........................................................;" ................................................ u ••••• , •••••••••••••••••••••••••••••••••••••••••••••••••••••••••• , ........................................................., ••••••••••••••••••••••••••••••••••••••••••••••.•••••••••• " •••••••••••••••••••••••••.••••••••••••••••••••••••.•••••• , ••••.••••••••••••••••••••••••••••••••••••••••••••••••••• , ••••••••••••••••••••••••••••••••••••••••••••••••••••••••,.

60 . ;................................................. ; .................... 1 ........... ++lu.u/ ........... ¡ ............ u..... ;.......... u.................... u~·l·+····················u ............. ,. + '~·I 1··IHI·········\u/l fl : u u·····················1~;

~ 20 . ;................................. u....... , .................... d .1J.+.II.......·................ u .: ............. u................. +............ u j


e
ro
::=:
"E
rn
e
ro
¡.=: 0.0 . .,................................................. ; ...................................................... > ...............................................;................................................. uj ....u.... uu.u.............................. L .............................................. , ......................................................, ........................................ u l
2000.0 1800 1600 1400 1200 1000 800 600 400.0
Wavenumber (cm-1)
Vet-S 14 Infrared Reference Spectra 2016

Meclofenamic Acid RSV26 Instrument: Fourier Transform Phase: Potassium Bromide Disc
100.0

80
~
~ ~ pD . "(\(\,rl
(1/
A.

60
\ ~
~ F. V~ A
A
f \
f 1\
)
40 A
I
;g
o

<D 20
W
ü
e
ro
:::::
~
'E
CJ)
e
.=ro 0.0
V V
2000.0 1800 1600 1400 1200 1000 800 600 400.0
Wavenumber (cm-1)

Methyltestosterone RSV28 Instrument: Dispersive Phase: Potassium Bromide Disc


100.0 . ,...................................................... ,........................................................ ,........................................................ ,........................................................ ,......................................................... ,........................................................ ,..................................................................................................................,

80 ............................................................................ , ...................... f······················ \ •••••••••.•••••.•••. '••.•.•......•.. J'l~······ h·································(V·'ll

60

40 ...•........................................................•.............................. ++ ...•........................................................•.................................................... , ..................................................•.........................................................•.........................................................•........................................................•

~ 20
e
ro
:::::
'E
CJ)
e
ro
.= 0.0 . ,........................................................'. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . ,. . . . . . . . . . . . . . . . . . . . . . . . . . >.......................................................,.........................................................•.........................................................,.................................................•............................................•
2000.0 1800 1600 1400 1200 1000 800· 600 400.0
Wavenumber (cm-1)

Metronidazole RSV29 Instrument: Dispersive Phase: Potassium Bromide Disc


100.0

80 ..... ....., .IV>. ~('


r
n r ·
( !~( I~.~((i
\r ~.~
I ~ \

60 \ \
A t1
~
(
40
~ \
.( JI
\ (1

AJ ~
?ft. N
<D
ü 20
e
~
ro W
E
CJ)
e
.=ro 0.0
2000.0 1800 1600 1400 1200 1000 800 600 400.0
Wavenumber (cm- 1
)
2016 Infrared Reference Spectra Vet-S15

Nandrolone Lau."ate RSV30 Instrument: Fourier Transform Phase: Potassium Bromide Disc
100.0, ..................................................................................................................................................................•................................................. , ............................................................................................................................................................................................................. .

80 ..•...................................................................................................................••..................................................... + ...................................................................................................... ;················································h···· ...................... ~;~ .··················.··························Á

60 ..••.......................................................•..... \ ................................. l······················· l··········· A··········· I!l~t···········.··········· fll\"j II············IHI································ 111·······················.···························· .............. ,

40; .................................... ;·············11+ Illi······················· liIV'<11 H'fI+IHIHI···································.········· ..................................................................................................... + ...................................................•

ü
Q)
20
e
ro
~
'E
en
e
ro
¡-:: 0.0
2000.0 1800 1600 1400 1200 1000 800 600 400.0
Wavenumber (cm- 1
)

Nitroxinil RSV31 Instrument: Fourier Transform Phase: Liquid Paraffin Mull


100.0 .................................................•.............................................................................................................................................................................•................................................................................................................................................................................................................................

80 ............................................................................................................................................................................................................................................................................................................................................................................................... :~l•.......... L.

60

40

:;R:
o
Q)
ü 20
e
ro
~
'E
en
e
ro
¡-:: 0.0
2000.0 1800 1600 1400 1200 1000 800 600 400.0
Wavenumber (cm- 1
)

Oxfendazole RSV32 Instrument: Fourier Transform Phase: Potassium Bromide Disc


100.0 i.................................................. ........................................................................................................•......................................................... ,........................................................................................................................................................................................................................ :

80 . ~:~~~......................•........................................................•................................................•................................................ j ........................................................•......................................................•............................

60 ..•................................................• ··········1···································.·························11··········· ¡.................... \ ....................• J··········I·····················,··········.········ ··············\11 I 1+ 1/····················+1+;11·

40

~ 20 .•............................................. ················/1 / I···!;/······· .......................................•............................ +···(·1,'· .. ······································· ..............•..............................................•.............•.......................................•...............................................•


e
ro
~
'E
en
e
ro
¡-:: 0.0
2000.0 1800 1600 1400 1200 1000 800 600 400.0
Wavenumber (cm- 1
)
Vet-S 16 Infrared Reference Spectra 2016

Oxyclozanidc RSV33 Instrument: Fourier Transform Phase: Potassium Bromide Disc


100,0 ...•...................................................................................................................................................................................................................................................................................................................................................................................................................................................................................•

80··.·················~.~~~~··'~r~\N/\~·········· •...................................................•.........................................................•................................................. ~~~ .......................... l~·······.+ .1I1++I,I~ H'lhll\f\/·····.

60 ........................................................................................... Illfl J 1II,···.·······llI y lll····················. 11.1····························· 1.··········\1··.1·.······· .. +1·········· 11····.····· 1,1·······························.

40 ..•................................................................................................................. il,·····················HI············································IJIlV··································..................................................................................................................................................... •

~
o
(])
u 20
e
ro
:::::
"E
U)
e
ro
.= 0,0
2000,0 1800 1600 1400 1200 1000 800 600 400,0
Wavenumber (cm-1)

Pcntobarbital RSV34 Instrument: Dispersive Phase: Potassium Bromide Disc


100,0 ....................................•...............

80

60

40

cf2.
(])
U 20
e
ro
:::::
"E
U)
e
ro
.= 0,0
2000,0 1800 1600 1400 1200 1000 800·· 600 400,0
Wavenumber (cm- 1
)

Phcnylbutazonc RSV35 Instrument: Dispersive Phase: Potassium Bromide Disc


100,0

80 .•..................................................... ~ ....................................... _

60

40 .............................. .

~ 20 .................................................. 1+ .............................................................•........................................
e
ro
:::::
"E
U)
e
ro
.= 0,0
2000,0 1800 1600 1400 1200 1000 800 600 400,0
Wavenumber (cm- 1
)
2016 Infrared Reference Spectra Vet-S 1 7

Ronidazole RSV36 Instrument: Fourier Transform Phase: Potassium Bromide Disc


100.0

80 -- .•
~

60
"\

1\ A
V\
~ \ / / ~ A rf (Wlr\ Al
/~

40
\ J V A
f \h f I '~~ ( ~ VP
~
I
V
~ 20 \n I
e ~
ro
:::::
y
V
"E
en
e
ro
.= 0.0
2000.0 1800 1600 1400 1200 1000 800 600 400.0
Wavenumber (cm-1)

Sulfadoxine RSV37 Instrument: Dispersive Phase: Potassium Bromide Disc


100.0 .......................................................... ,.........................................................................................................................................................................................................................................................................................................................................................................................................

80 .•. . . . . . . . . . . . . . . . . . . . . . . . , ......................... \···············.···············~.t············· .. A.························F\······,··················· ························r:'~········I:~. Akr\ I~·~,.··············································,

60 .................................................. ................................. 11· l'" l······· ·Y·III ·I··H·

40 > ..................................... +······································111············ 11111············· 11···················.·····11 11·············.···········

~ 20
e
ro
:::::
"E
en
e
ro
.= 0.0
2000.0 1800 1600 1400 1200 1000 800 600 400.0
Wavenumber (cm-1)

Sulfadimidine RSV50 Instrument: Dispersive Phase: Potassium Bromide Disc


100.0 ..

~
80
\
••

••

(I A
(~
Ir ~ r ~{\ ¡ r\(\ r
\/
60

~ ~ ~\ ~~ ~
VJ
I1
MI
40
"
'. ~ ~
~
Q)
ü 20
e
ro
~
E
en
e
ro
.= 0.0
600 400.0

2000.0 1800 1600 1400 1200 1000 800


Wavenumber (cm- 1
)
Vet-S 18 Infrared Reference Spectra 2016

Sulfamctoxypyridazinc RSV38 Instrument: Dispersive Phase: Potassium Bromide Disc


100.0

80

60

40

cf2.
Q)
ü 20
e
~
'E
U)
e
m
r= 0.0
2000.0 1800 1600 1400 1200 1000 800 600 400.0
Wavenumber (cm-1)

Sulfaquinoxalinc RSV41 Instrument: Fourier Transform Phase: Liquid Paraffin Mull


100.0

80

60

40

cf2.
Q)
ü 20
e
m
-~
E
U)
e
m
r= 0.0
2000.0 1800 1600 1400 1200 1000 800 600 400.0
Wavenumber (cm- 1)

Sulfathiazolc Sodium RSV42 Instrument: Fourier Transform Phase: Potassium Bromide Disc
100.0

80

60

40

~ 20
e
m
.~
E
U)
e
m
r= 0.0
2000.0 1800 1600 1400 1200 1000 800 600 400.0
Wavenumber (cm- 1)
2016 Infrared Reference Spectra Vet-S19

Testosterone Phenylpropionate RSV 43 Instrument: Dispersive Phase: Potassium Bromide Disc


100.0

80

60

40

cf2-
Q)
u 20
e
~
E
CfJ
e
ro
t-= 0.0
2000.0 1800 1600 1400 1200 1000 800 600 400.0
Wavenumber (cm-1)
Trimethoprim RSV 45 Instrument: Dispersive Phase: Potassium Bromide Disc
100.0

80

60

40

cf2-
Q)
u 20
e
~
E
CfJ
e
ro
t-= 0.0
2000.0 1800 1600 1400 1200 1000 800 600 400.0
Wavenumber (cm-1)
Tylosin RSV46 Instrument: Fourier Transform Phase: Potassium Bromide Disc
100.0

80

60

40

25 20
e
ro
E
E
CfJ
e
ro
t-= 0.0
2000.0 1800 1600 1400 1200 1000 800 600 400.0
Wavenumber (cm- 1)
Appendices
Contents of the Appendices
When a method, test or other matter described in an appendix is invoked in a
monograph reproduced from the European Pharmacopoeia, Part IJI of the General
Notices applies. When a method, test or other matter described in an appendix is
invoked in any other monograph, Part JI of General Notices applies.
Any reference to an appendix that is not contained within this edition of the British
Pharmacopoeia (Veterinary) is to be construed as a reference to the said appendix
contained within the British Pharmacopoeia 2011 modified as necessary by
amendments.

The following appendices are included in this section of the British Pha1"Jnacopoeia
(Veterinary) .

APPENDIX XV PRODUCTION AND TESTING OF VACCINES


A (Vet). Terminology Used in Monographs on Biological Products
H (Vet). Chicken Flocks Free From Specified Pathogens for the Production and
Quality Control of Vaccines
J (Vet) 1. Cell Cultures for the Production of Veterinary Vaccines
J (Vet) 2. Substances of Animal Origin for the Production of Immunological
Veterinary Medicinal Products
K (Vet) 1. Evaluation of Safety of Veterinary Vaccines and Immunosera
K (Vet) 2. Evaluation of Efficacy of Veterinary Vaccines and Immunosera
K (Vet) 3. Evaluation of Safety of Each Batch of Immunosera for Veterinary Use

APPENDIX XVI
B (Vet) 1. Vacant
B (Vet) 2. Vacant
B (Vet) 3. Mycoplasmas
B (Vet) 4. Avian Viral Vaccines: Tests for Extraneous' Agents in Seed Lots
B (Vet) 5. Avian Live Virus Vaccines: Tests for Extraneous Agents in Batches of
Finished Product
B (Vet) 6. Vacara
B (Vet) 7. Vacant
B (Vet) 8. Vacant

APPENDIX XXI
B (Vet). Approved Synonyms (Veterinary)
2016 European Pharmacopoeia Equivalent Texts Vet-A3

European Pharmacopoeia Equivalent Texts


In monographs reproduced from the European Pharmacopoeia the analytical
methods, tests and other supporting texts are invoked by means of the reference
number of the text in the General Chapters of the European Pharmacopoeia.
The table below lists the contents of the General Chapters of the European
Pharmacopoeia and gives the British Pharmacopoeia or British Pharmacopoeia
(Veterinary) equivalents. It is provided for information but it is emphasised that,
for texts of the European Pharmacopoeia, in cases of doubt or dispute the text
published by the Council of Europe is authoritative. Appendices of the British
Pharmacopoeia (Veterinary) are identified by inclusion of '(Vet)' after the
Appendix letter, for example, Appendix XV J (Vet) l.

Ph. Eur. Subject of text British Pharmacopoeia reference


1 General N otices
2.1.1 Droppers Appendix I A
2.1.2 Sintered-glass Filters Appendix XVII B2
2.1.3 UV lamps Appendix lIT A
2.1.4 Sieves Appendix XVII B 1
2.1.5 Tubes for Comparative Tests Appendix VII
2.1.6 Gas detector tubes Appendix IX K
2.2.1 Clarity and Degree of Opalescence of Liquids Appendix IV A
2.2.2 Degree of Coloration of Liquids Appendix IV B
2.2.3 Potentiometric Determination of pH Appendix V L
2.2.4 Reaction of Solution, pH and Indicator Colour Appendix V K
2.2.5 Relative Density Appendix V G
2.2.6 Refractive Index Appendix V E
2.2.7 Optical Rotation Appendix V F
2.2.8 Viscosity Appendix V H
2.2.9 Capillary Viscometer Method Appendix V H, Method IT
2.2.10 Rotating Viscometer Method Appendix V H, Method ITI
2.2.11 Distillation Range Appendix V C
2.2.12 Boiling Point Appendix V D
2.2.13 Water by Distillation Range Appendix IX C, Method II
2.2.14 Melting point: Capillary Method Appendix V A, Method I
2.2.15 Melting point: Open Capillary Method Appendix V A, Method IV
2.2.16 Melting point: Instantaneous Method Appendix V A, Method V
2.2.17 Drop Point Appendix V A, Method III
2.2.18 Freezing Point Appendix V B
2.2.19 Amperometric Titration Appendix VIII B
2.2.20 Potentiometric Titration Appendix VITI B
2.2.21 Fluorimetry Appendix II E
2.2.22 Atomic Emission Spectrometry Appendix IT D
2.2.23 Atomic Absorption Spectrometry Appendix II D
2.2.24 Infrared Spectrophotometry Appendix II A
2.2.25 Visible and Ultraviolet Spectrophotometry Appendix II B
2.2.26 Paper Chromatography Appendix lIT E
2.2.27 Thin-layer Chromatography Appendix lIT A
2.2.28 Gas Chromatography Appendix lIT B
2.2.29 Liquid Chromatography Appendix lIT D
2.2.30 Size-exclusion Chromatography Appendix lIT C
2.2.31 Electrophoresis Appendix III F
2.2.32 Loss on Drying Appendix IX D
2.2.33 Nuclear Magnetic Resonance Spectrometry Appendix II C
2.2.34 Thermogravimetry . Appendix V M
2.2.35 Osmolality AppendixVN
2.2.36 Ion-selective Potentiometry Appendix VIII E
2.2.37 X-ray Fluorescent Spectrophotometry Appendix II F
2.2.38 Conductivity Appendix V O
2.2.39 Molecular Mass Distribution in Dextrans Appendix III C
2.2.40 N ear-Infrared Spectroscopy Appendix II A

t Reproduced in fuU as Pan JII of the General N atices of the Bl'itish Phal'rnacopoeia and Bl'itish Phamzacopoeia (Vetel'ina¡~y).
Vet-A4 European Pharmacopoeia Equivalent Texts 2016

2.2.41 Circular Dichroism Appendix V J


2.2.42 Density of Solids Appendix V Q
2.2.43 Mass Spectrometry Appendix II G
2.2.44 Total Organic Cm'bon in Water Appendix V P
2.2.45 Supercritical Fluid Chromatography Appendix III H
2.2.46 Chromatographic Separation Techniques Appendix III
2.2.47 Capillary Electrophoresis Appendix III G
2.2.48 Raman Spectroscopy Appendix II H
2.2.49 Falling Ball Viscometer Method Appendix V H, Method IV
2.2.54 Isoelectric Focussing Appendix III J
2.2.55 Peptide Mapping Appendix III K
2.2.56 Amino Acid Analysis Appendix III L
2.2.57 Inductively Coupled Plasma-Atomic Emission Spectrometry Appendix II D
2.2.58 Inductively Coupled Plasma-Mass Spectrometry Appendix II G 1
2.2.59 Glycan Analysis of Glycoproteins Appendix III M
2.2.60 Melting Point: Instrumental Method Appendix V A, Method VI
2.2.61 Characterisation of Crystalline Solids by Microcalorimetry and Solution Appendix XVII V
Calorimetry
2.2.64 Peptide Identification by Nuclear Magnetic Resonance Spectrometry Appendix II K
2.2.65 Voltametric Titration . Appendix VIII B
2.2.66 Detection and Measurement of Radioactivity Appendix V R
2.3.1 Identification Reactions Appendix VI
2.3.2 Identification of Fatty Oils by TLC Appendix X N
2.3.3 Identification of Phenothiazines by TLC Appendix III A
2.3.4 Odour Appendix VI
Limit test for:
2.4.1 - Ammonium Appendix VII
2.4.2 - Arsenic Appendix VII
2.4.3 - Calcium Appendix VII
2.4.4 - Chlorides Appendix VII
2.4.5 - Fluorides Appendix VII
2.4.6 - Magnesium Appendix VII
2.4.7 - Magnesium and Alkaline-earth Metals Appendix VII
2.4.8 - Heavy Metals Appendix VII
2.4.9 - Iron Appendix VII
2.4.10 - Lead in Sugars Appendix VII
2.4.11 - Phosphates Appendix VII
2.4.12 - Potassium Appendix VII
2.4.13 - Sulfate Appendix VII
2.4.14 Sulfated Ash Appendix IX A, Method II
2.4.15 Limit test for Nickel in Polyols Appendix VII
2.4.16 Total Ash Appendix XI J, Method II
2.4.17 Limit test for Aluminium Appendix VII
2.4.18 Free Formaldehyde Appendix XV D
2.4.19 Alkaline Impurities in Fatty Oils AppendixXN
2.4.20 Determination of Metal Catalyst 01' Metal Reagent Residues Appendix VIII W
2.4.21 Foreign Oils in Fatty Oils by TLC Appendix X N
2.4.22 Composition of Fatty Acids by Gas Chromatography Appendix X N
2.4.23 Sterols in Fatty Oils AppendixX Q
2.4.24 Residual Solvents Appendix VIII L
2.4.25 Residual Ethylene Oxide and Dioxan Appendix VIII M
2.4.26 N,N- Dimethylaniline Appendix VIII N
2.4.27 Heavy Metals in Herbal Drugs and Fatty Oils Appendix VII
2.4.28 2-Ethylhexanoic acid Appendix VIII O
2.4.29 Composition of Fatty Acids in Oils Rich in Omega-3-acids Appendix X PI
2.4.30 Ethylene Glycol and Diethylene Glycol in Ethoxylated Substances Appendix VIII K
2.4.31 Nickel in Hydrogenated Vegetable Oils Appendix VIII R
2.4.32 Total Cholesterol in Oils Rich in Omega-3 Acids Appendix X P2
2.5.1 Acid Value Appendix X B
2.5.2 Ester Value Appendix XC
2.5.3 Hydroxyl Value AppendixX D
2.5.4 Iodine Value Appendix X E
2.5.5 Peroxide Value Appendix X F
2.5.6 Saponification Value Appendix X G, Method II
2.5.7 Unsaponifiable Matter Appendix X H, Method II
2.5.8 Assay of Primary Aromatic Amino Nitrogen Appendix VIII B
2016 European Pharmacopoeia Equivalent Texts Vet-A5

2.5.9 Semi-micro Determination of Nitrogen by Sulfuric Acid Digestion Appendix VIII H


2.5.10 Oxygen-flask Method Appendix VIII C
2.5.11 Complexometric Titrations Appendix VIII D
2.5.12 Semi-micro Determination of Water Appendix IX C, Method I
2.5.13 Aluminium in Adsorbed Vaccines Appendix XV B
2.5.14 Calcium in Adsorbed Vaccines Appendix XV C
2.5.15 Phenol in Immunosera and Vaccines Appendix XV E
Polysaccharide Vaccines:
2.5.16 - Protein Appendix XV G
2.5.17 - Nucleic Acids Appendix XV G
2.5.18 - Phosphorus Appendix XV G
2.5.19 - O-acetyl Appendix XV G
2.5.20 - Hexosamines Appendix XV G
2.5.21 - Methylpentoses Appendix XV G
2.5.22 - Uronic Acids Appendix XV G
2.5.23 - Sialic Acid Appendix XV G
2.5.24 Carbon Dioxide in Medicinal Gases Appendix IX F
2.5.25 Carbon Monoxide in Medicinal Gases Appendix IX E
2.5.26 Nitrogen Monoxide and Nitrogen Dioxide in Medicinal Gases Appendix IX G
2.5.27 Oxygen in Medicinal Gases Appendix IX H
2.5.28 Water in Medicinal Gases Appendix IX J
2.5.29 Sulfur Dioxide Appendix IX B, Method II
2.5.30 Oxidising Substances Appendix X L
2.5.31 Ribose in Polysaccharide Vaccines Appendix XV G
2.5.32 Micro Determination of Water Appendix IX C, Method III
2.5.33 Total Protein Assay Appendix VIII P
2.5.34 Acetic Acid in Synthetic Peptides Appendix VIII Q
2.5.35 Nitrous Oxide in Gases Appendix IX L
2.5.36 Anisidine value Appendix X O
2.5.37 Methyl, Ethyl and Isopropyl Methanesulfonate in Methanesulfonic Acid Appendix VIII S
2.5.38 Methyl, Ethyl and Isopropyl Methanesulfonate in Active Substances Appendix VIII T
2.5.39 Methanesulfonyl Chloride in Methanesulfonic Acid Appendix VIII V
2.5.40 Methyl, Ethyl and Isopropyl Toluenesulfonate in Active Substances Appendix VIII X
2.6.1 Sterility Appendix XVI A
2.6.2 Mycobacteria Appendix XVI B4
2.6.3 Vacam
2.6.4 Vacant
2.6.5 Vacant
2.6.6 Vacant
2.6.7 Mycoplasmas Appendix XVI B3 and
Appendix XVI B(Vet)3
2.6.8 Pyrogens Appendix XN D
2.6.9 Abnormal Toxicity Appendix XN E
2.6.10 Histamine Appendix XN G
2.6.11 Depressor Substances Appendix XN F
2.6.12 Microbiological Examination of Non-sterile Products: Microbial Appendix XVI B2
Enumeration Tests
2.6.13 Microbiological Examination of Non-sterile Products: Test for Appendix XVI B 1
Specified Micro-organisms
2.6.14 Bacterial Endotoxins Appendix XN C
2.6.15 Prekallikrein Activator Appendix XN J A9
2.6.16 Extraneous Agents in Viral Vaccines Appendix XVI B5
2.6.17 Anticomplementary Activity of Immunoglobulins Appendix XN J C2
2.6.18 Neurovirulence of Live Viral Vaccines Appendix XV F1
2.6.19 Neurovirulence of Poliomyelitis Vaccine (Oral) Appendix XV F2
2.6.20 Anti-A and Anti-B Haemagglutinins Appendix XN J C5
2.6.21 Nucleic Acid Amplification Appendix XN L
2.6.22 Activated Coagulation Factors Appendix XN J A7
2.6.24 Avian Viral Vaccines: Tests for Extraneous Agents in Seed Lots Appendix XVI B(Vet)4
2.6.25 Avian Live Virus Vaccines: Tests for Extraneous Agents in Batches of Appendix XVI B(Vet)5
Finished Product
2.6.26 Test for Anti-D Antibodies in Human Immunoglobulin Appendix XN J C4
2.6.27 Microbiological Control of Cellular Products Appendix XVI E
2.6.30 Monocyte-Activation Test Appendix XN H
2.6.31 Microbiological Examination of Herbal Medicinal Products for Oral Appendix XVI F
Use and Extracts used in their Preparations
Vet-A6 European Pharmacopoeia Equivalent Texts 2016

2.6.33 Residual Pertussis Toxin and Irreversibility of Pertussis Toxoid Appendix XIV K9
2.7.1 Immunochemical Methods Appendix XIV B
2.7.2 Microbiological Assay of Antibiotics Appendix XIV A
2.7.3 Vacant
2.7.4 Assay of Coagulation Factor VIII Appendix XIV J A3
2.7.5 Assay of Heparin Appendix XIV J B2
2.7.6 Assay of Diphtheria Vaccine (Adsorbed) Appendix XIV K1
2.7.7 Assay of Pertussis Vaccine (Whole Cell) Appendix XIV K2
2.7.8 Assay of Tetanus Vaccine (Adsorbed) Appendix XIV K3
2.7.9 Fc Function of Immunoglobulins Appendix XIV J C 1
2.7.10 Assay of Human Coagulation Factor VII Appendix XIV J A2
2.7.11 Assay of Coagulation Factor IX Appendix XIV J A4
2.7.12 Assay of Heparin in Coagulation Factors Appendix XIV J B 1
2.7.13 Assay of Anti-D Immunoglobulin Appendix XIV J C3
2.7.14 Assay of Hepatitis A Vaccine Appendix XIV K4
2.7.15 Assay of Hepatitis B (rDNA) Vaccine Appendix XIV K5
2.7.16 Assay of Pertussis Vaccine (Acellular) Appendix XIV K6
2.7.17 Assay of Human Antithrombin III Appendix XIV J B3
2.7.18 Assay of Human Coagulation Factor II Appendix XIV J Al
2.7.19 Assay of Human Coagulation Factor X Appendix XIV J A5
2.7.20 In vivo Assay of Poliomyelitis Vaccine (Inactivated) Appendix XIV K7
2.7.21 Assay of Human von Willebrand factor Appendix XIV J A8
2.7.22 Assay of Human Coagulation Factor XI Appendix XIV J A6
2.7.23 Numeration of CD34/CD45+ Cells in Haematopoietic Products Appendix XIV N
2.7.24 Flow Cytometry Appendix II J
2.7.25 Assay of Human Plasmin Inhibitor Appendix XIV JAlO
2.7.27 Flocculation Value (Lf) of Diphtheria and Tetanus Toxins and Toxoids Appendix XIV K8
(Ramon Assay)
2.7.28 Colony-forming Cell Assay for Human Haematopoietic Progenitor Appendix XIV N2
Cells
2.7.29 Nucleated Cell Count and Viability Appendix XIV N3
2.7.30 Assay of Human Protein C Appendix XIV J B4
2.7.31 Assay of Human Protein S Appendix XIV J B5
2.7.32 Assay of Human a.-1-Proteinase Inhibitor Appendix XIV J D 1
2.8.1 Ash Insoluble in Hydrochloric Acid Appendix XI K, Method II
2.8.2 F oreign Matter Appendix XI D
2.8.3 Stomata and Stomatal Index Appendix XI H
2.8.4 Swelling Index Appendix XI C
2.8.5 Water in Essential Oils Appendix X M
2.8.6 F oreign Esters Appendix X M
2.8.7 Fatty Oils and Resinified Volatile Oils AppendixX M
2.8.8 Odour and Taste of Volatile Oils AppendixX M
2.8.9 Residue on Evaporation AppendixXM
2.8.10 Solubility in Ethanol (Volatile Oils) Appendix X M
2.8.11 Determination of Cineole Appendix X J
2.8.12 Essential Oils in Herbal Drugs Appendix XI E
2.8.13 Pesticide Residues Appendix XI L
2.8.14 Tannins in Herbal Drugs Appendix XI M
2.8.15 Bittemess value Appendix XI N
2.8.16 Dry Residue of Extracts Appendix XI P
2.8.17 Loss on Drying of Extracts Appendix XI Q
2.8.18 Determination of Aflatoxin Bl in Herbal Drugs Appendix XI S 1
2.8.20 Herbal Drugs: Sampling and Sample Preparation Appendix XI T
2.8.21 Test for Aristolochic Acids in Herbal Drugs Appendix XI R1
2.8.22 Determination of Ochratoxin A in Herbal Drugs Appendix XI S2
2.8.23 Microscopic Examination of Herbal Drugs Appendix XI U
2.9.1 Disintegration of Tablets and Capsules Appendix XII Al
2.9.2 Disintegration of Suppositories and Pessaries Appendix XII A2
2.9.3 Dissolution Test for Solid Dosage Forms Appendix XII B 1
2.9.4 Dissolution Test for Transdermal Patches Appendix XII B2
2.9.5 Uniformity of Mass Appendix XII C 1
2.9.6 Uniformity of Content Appendix XII C3
2.9.7 Friability of Uncoated Tablets Appendix XVII G 1
2.9.8 Resistance to Crushing of Tablets Appendix XVII H
2.9.9 Measurement of Consistency by Penetrometry Appendix XVII F
2.9.10 Ethanol Content Appendix VIII F, Method III
2016 European Pharmacopoeia Equivalent Texts Vet-A 7

2.9.11 Methanol and 2-propanol Appendix VIII G


2.9.12 Particle Size Classification of Powders (Sieve Test) Appendix XVII Al
2.9.13 Vacant
2.9.14 Specific Surface Area by Gas Permeability Appendix XVII C
2.9.15 Vacant
2.9.16 Flowability Appendix XVII E
2.9.17 Extractable Volume of Parenteral Preparations Appendix XII C5
2.9.18 Aerodynamic Assessment of Fine Particles Appendix XII C7
2.9.19 Particulate Contamination: Sub-visible Particles Appendix XIII A
2.9.20 Particulate Contamination: Visible Particles Appendix XIII B
2.9.21 Vacant
2.9.22 Softening Time of Lipophilic Suppositories Appendix XVII J
2.9.23 Pycnometric Density of Solids Appendix XVII K
2.9.24 Vacant
2.9.25 Dissolution Test for Medicated Chewing Gums Appendix XII B4
2.9.26 Specific Surface Area By Gas Adsorption Appendix XVII M
2.9.27 Uniformity of Mass of Delivered Doses from Multidose Containers Appendix XII C2
2.9.28 Vacant
2.9.29 Intrinsic Dissolution Appendix XII B5
2.9.31 Particle Size Analysis by Laser Light Diffraction Appendix XVII P
2.9.32 Porosity and Pore-size Distribution of Solids by Mercury Porosimetry Appendix XVII R
2.9.33 Characterisation of Crystalline and Partially Crystalline Solids by X-ray Appendix XVII Q
Powder Diffractlon (XRPD)
2.9.34 Bulk Density and Tapped Density of Powders Appendix XVII S
2.9.35 Powder Fineness Appendix XVII A2
2.9.36 Powder Flow Appendix XVII N
2.9.37 Optical Microscopy Appendix XVII O
2.9.38 Particle-size Distribution Estimation by Analytical Sieving Appendix XVII B3
2.9.39 Water-Solid Interactions: Determination of Sorption-Desorption Appendix IX M
Isotherms and of Water Activity
2.9.40 UnifOlmity of Dosage Units Appendix XII C4
2.9.41 Friability of Granules and Spheroids Appendix XVII G2
2.9.42 Dissolution Test for Lipophilic Solid Dosage Forms Appendix XII B3
2.9.43 Apparent Dissolution Appendix XII B6
2.9.44 Preparations for Nebulisation: Characterisation Appendix XII C8
2.9.45 Wettability of Porous Solids Including Powders Appendix XVII T
2.9.46 Vaca11t
2.9.47 Demonstration of UnifOlmity of Dosage Units Using Larger Sample Appendix XII C9
Sizes
3.1.1 Material Used for Containers for Human Blood and Blood Appendix XX A
Components
3.1.1.1 Materials Based on PVC for Blood and Blood Components Appendix XX Al
3.1.1.2 Materials Based on PVC for Tubing U sed in Sets for the Transfusion Appendix XX A2
of Blood and Blood Components
3.1.3 Polyolefins Appendix XX B
3.1.4 Polyethylene - Without Additives Appendix XX C1
3.1.5 Polyethylene - With Additives Appendix XX C2
3.1.6 Polypropylene Appendix XX D
3.1.7 Poly(ethylene-vinyl acetate) Appendix XX E
3.1.8 Silicone Oil U sed as a Lubricant Appendix XX F1
3.1.9 Silicone Elastomer for Closures and Tubing Appendix XX F2
3.1.10 Materials Based on Non-plasticised PVC for Containers for Non- Appendix XX A3
injectable, Aqueous Solutions
3.1.11 Materials Based on Non-plasticised Poly(vinyl chloride) for Containers Appendix XX A4
for Solid Dosage forms for Oral Administration
3.1.12 Vacant
3.1.13 Plastic Additives Appendix XX G
3.1.14 Materials Based on Plasticised Poly(vinyl chloride) for Containers for Appendix XX A5
Aqueous Solutions for Intravenous Infusion
3.1.15 Polyethylene Terephthalate for Containers for Preparations Not for Appendix XX H
Parenteral U se
3.2 Containers Appendix XIX A
3.2.1 Glass Containers for Pharmaceutical U se Appendix XIX B
3.2.2 Plastic Containers and Closures Appendix XIX C
3.2.2.1 Plastic Containers for Aqueous Solutions for Parenteral Infusions Appendix XIX C1
3.2.3 Sterile Plastic Containers for Blood and Blood Components Appendix XIX D1
Vet-A8 European Pharmacopoeia Equivalent Texts 2016

3.2.4 Empty Sterile Plastic Containers for BIood and Blood Components Appendix XIX D2
3.2.5 Sterile Plastic Containers with Anticoagulant for BIood and BIood Appendix XIX D3
Components
3.2.6 Sets for the Transfusion of BIood and BIood Components Appendix XIX F
3.2.7 Vacant
3.2.8 Sterile Single-Use Plastic Syringes Appendix XIX G
3.2.9 Rubber Closures for Containers for Aqueous Preparations for Appendix XIX E
Parenteral Use
4.1.1 Reagents Appendix I A
4.1.2 Standard Solutions Appendix I C
4.1.3 Buffer Solutions Appendix I D
4.2.1 Reference Substances for Volumetric Solutions Appendix lB
4.2.2 Volumetric Solutions Appendix I B
5.1.1 Methods of Preparation of Sterile Products Appendix XVIII
5.1.2 Biological Indicators (sterilisation) Appendix XVIII
5.1.3 Efficacy of Antimicrobial Preservation Appendix XVI C
5.1.4 Microbiological Quality of Non-sterile Pharmaceutical Preparations and Appendix XVI D
Substances for Pharmaceutical U se
5.1.5 Po Concept (sterilisation) Appendix XVIII
5.1.6 Alternative Methods for Control of Microbiological Quality Supplementary Chapter IV L
5.1.7 Viral Safety Appendix XXII A
5.1.8 Microbiological Quality of Herbal Medicinal Product,s for Oral U se and Appendix XVI G
Extracts used in their Preparation
5.1.9 Guidelines for using the Test for Sterility Supplementary Chapter IV P
5.1.10 Guidelines for using the test for bacterial endotoxins Supplementary Chapter I C
5.2.1 Terminology: Vaccines Appendix XV A and
Appendix XV A(Vet)
5.2.2 SPF Chicken Flocks Appendix XV H and
Appendix XV H(Vet)
5.2.3 Cell Substrates for the Production ofVaccines for Human Use Appendix XV J
5.2.4 Cell Cultures (Veterinary Vaccine Production) Appendix XV J (Vet) 1
5.2.5 Substances of Animal Origin for the Production of Irnmunological Appendix XV J(Vet)2
Veterinary Medicinal Products
5.2.6 Safety: Veterinary Vaccines Appendix XV K (Vet) 1
5.2.7 Efficacy: Veterinary Vaccines Appendix XV K(Vet) 2
5.2.8 Minimising the Risk of Transmitting Animal Spongiform Appendix XXII B
Encephalopathy Agents via Medicinal Products
5.2.9 Evaluation of Safety of Each Batch of Immunosera for Veterinary Use Appendix XV K (Vet) 3
5.2.11 Carrier Proteins for the Production of Conjugated Polysaccharide Appendix XV K
Vaccines for Human Use
5.3 Statistical Analysis of Biological Assays and Tests Supplementary Chapter IV G
5.4 Residual Solvents Supplementary Chapter IV D
5.5 Alcoholimetric Tables Supplementary Chapter IV E
5.6 Assay of Intelferons Appendix XIV MI
5.7 Physical Characteristics of Radionuc1ides Radiopharmaceutical Preparations
5.8 Pharmacopoeial Harmonisation Supplementary Chapter IV F
5.9 Polymorphism Appendix I F
5.10 Control of Impurities in Substances for Pharmaceutical U se Supplementary Chapter IV J
5.11 Characters Section in Monographs Supplementary Chapter IV K
5.12 Reference Standards Supplementary Chapter IV M
5.14 Gene Transfer Medicinal Products for Human Use Supplementary Chapter IV N
5.16 Crystallinity Appendix XVII U
5.17.1 Recommendations on dissolution testing Supplementary Chapter I E
5.20 Metal Catalyst or Metal Reagent Residues Supplementary Chapter IV Q
5.22 Names of Herbal Drugs Used in Traditional Chinese Medicine Supplementary Chapter VII B
5.23 Monographs on Herbal Drug Extracts (Information Chapter) Supplementary Chapter VII C
2016 Vet-A9

same working seed lot 01" a suspension derived from the


A (Vet). Terminology used in working seed lot, incubated, and harvested in a single
Monographs on Biological Products production runo
(Ph. EUI". method 5.2.1) Monovalent pooled harvest Po oled material containing a
single strain or type of micro-organism or antigen and
For sorne items, alternative terms commonly used in
derived from a number of eggs, cell culture containers etc.
connection with veterinary vaccines are shown in parenthesis.
that are processed at the same time.
Seed-lot system A seed-Iot system is a system according to
Final buIk vaccine Material that has undergone all the steps
which successive batches of a product are derived from the
of production except for the final filling. Ir consists of one 01"
same master seed loto For routine production, a working seed
more monovalent pooled harvests, from cultures of one 01"
lot may be prepared from the master seed loto The origin and
more species or types of micro-organism, after clarification,
the passage history of the master seed lot and the working
dilution or addition of any adjuvant or other auxiliary
seed lot are recorded.
substance. Ir is treated to ensure its homogeneity and is used
Master seed 10t A culture of a micro-organism distributed for filling the containers of one 01" more finallots (batches).
from a single bulk into containers and processed together in
Finallot (Batch) A collection of closed, final containers or
a single operation in such a manner as to ensure uniformity
other final dosage units that are expected to be homogeneous
and stability and to prevent contamination. A master seed lot
and equivalent with respect to risk of contamination during
in liquid form is usually stored at 01" below - 70 oC.
filling or preparation of the final producto The dosage units
A freeze-dried master seed lot is stored at a temperature
are filled, or otherwise prepared, from the same final bulk
known to ensure stability.
vaccine, freeze-dried together (if applicable) and closed in
Working seed 10t A culture of a micro-organism derived one continuous working session. They bear a distinctive
from the master seed lot and intended for use in production. number or code identifying the final lot (batch). Where a
Working seed lots are distributed into containers and stored final bulk vaccine is filled andl or freeze-dried on several
as described above for master seed lots. separate sessions, there results a related set of finallots
Cell-banl, system (Cell-seed system) A system whereby (batches) that are usually identified by the use of a common
successive final lots (batches) of a product are manufactured part in the distinctive number or code; these related final lots
by culture in cells derived from the same master cell bank (batches) are sometimes referred to as sub-batches, sub-Iots
(master cell seed). A number of containers from the master or filling lots.
cell bank (master cell seed) are used to prepare a working Combined vaccine A multicomponent preparation
cell bank (working cell seed). The cell-banl, system (cell-seed formulated so that different antigens are administered
system) is validated for the highest passage level achieved simultaneously. The different antigenic components are
during routine production. intended to protect against different strains or types of the
Master cell bank (Master cell seed) A culture of cells same organism and/or different organisms. A combined
distributed into containers in a single operation, processed vaccine may be supplied by the manufacturer either as a
together and stored in such a manner as to ensure uniformity single liquid or freeze-dried preparation or as several
and stability and to prevent contamination. A master cell constituents with directions for admixture before use.
bank (master cell seed) is usually stored at - 70 oC or lower.
Working cell bank (Worldng cell seed) A culture of cells
derived from the master cell bank (master cell seed) and
intended for use in the preparation of production cell H (Vet). Chicken Flocks Free from
cultures. The working cell bank (working cell seed) is
distributed into containers, processed and stored as described
Specified Pathogens for the Production
for the master cell bank (master cell seed). and Quality Control of Vaccines
Primary cell cultures Cultures of cells obtained by (Ph. EUI". method 5.2.2)
trypsination of a suitable tissue or organ. The cells are
Where specified, chickens, embryos or cell cultures used for
essentially identical to those of the tissue of origin and are no
the production or quality control of vaccines are derived from
more than 5 in vitm passages from the initial preparation
eggs produced by chicken flocks free from specified
from the animal tissue.
pathogens (SPF). The SPF status of a flock is ensured by
Celllines Cultures of cells that have a high capacity for means of the system described below. The list of micro-
multiplication in vitJ"o. In diploid cell lines, the cells have organisms given is based on current knowledge and will be
essentially the same characteristics as those of the tissue of updated as necessary.
origino In continuous cell lines, the cells are able to multiply
A flock is defined as a group of birds sharing a common
indefinitely in culture and may be obtained from healthy or
environment and having their own caretakers who have no
tumoral tissue. Sorne continuous cell lines have oncogenic
contact with non-SPF flocks. Once a flock is defined, no
potential under certain conditions.
non-SPF birds are added to it.
Production cell culture A culture of cells intended for use
Each flock is housed so as to minimise the risk of
in production; it may be derived from one or more
contamination. The facility in which the flock is housed must
containers of the working cell bank (working cell seed) or it
not be sited near to any non-SPF flocks of birds with the
may be a primary cell culture.
exception of flocks that are in the process of being
Control cells A quantity of cells set aside, at the time of established as SPF flocks and that are housed in facilities and
virus inoculation, as uninfected cell cultures. The uninfected conditions appropriate to SP-F flocks. The SPF flock is
cells are incubated under similar conditions to those used for housed within an isolator or in a building with filtered air
the production cell cultures. under positive pressure. Appropriate mea sures are taken to
Single harvest Material derived on one or more occasions prevent entry of rodents, wild birds, insects and unauthorised
from a single production cell culture inoculated with the personnel.
Vet-A10 2016

Table 5.2.2.-1
Agent Test Vertical Rapid/slow
to be used** transmission spread
Avian adenoviruses, group 1 AGP, EIA yes slow
Avian encephalomyelitis virus AGP, EIA yes rapid
Avian infectious bronchitis virus HI, EIA no rapid
Avian infectious laryngotracheitis virus VN,EIA no slow
Avian leucosis viruses EIA for virus, yes slow
VN, EIA for antibody
Avian nephritis virus IS no slow
Avian orthoreoviruses IS, EIA yes slow
Avian reticuloendotheliosis virus AGP, IS, EIA yes slow
Chicken anaemia virus IS, EIA, VN yes slow
Egg drop syndrome virus HI, EIA yes slow
Infectious bursal disease virus Serotype 1: AGP, EIA, VN no rapid
Serotype 2: VN
Influenza A virus AGP, EIA, HI no rapid
Marek's disease virus AGP no rapid
Newcastle disease virus HI, EIA no rapid
Turkey rhinotracheitis virus EIA no slow
Mycoplasma gallisepticum Agg and HI to confirm a yes slow
positive test,
EIA, HI
Mycoplasma synoviae Agg and HI to confirm a yes rapid
positive test,
EIA, HI
Salmonella pullorum Agg yes slow
Agg: agglutination HI: haemagglutination inhibition
AGP: agar gel precipitation; the technique is suitable where testing is carried IS: ímmunostaining
out weekly VN: virus neutralisation
EIA: enzyme immunoassay
**Subject to agreement by the competent authority, other types of test may be used provided they are at least as sensitive as those indicated and of
appropriate specificity.

Personnel authorised to enter the facility must have no A positive result for chicken anaemia virus (CAV) does not
contact with other birds or with agents potentially capable of necessarily exc1ude use of material derived from the flock,
infecting the fiock. It is advisable for personnel to shower and but live vaccines for use in birds less than 7 days old shall be
change c10thing or to wear protective c10thing before entering produced using material from CAV-negative fiocks.
the controlled facility. Inactivated vaccines for use in birds less than 7 days old may
Wherever possible, items tak~n into the facility are sterilised. be praduced using material from fiocks that have not been
In particular it is recommended that the feed is suitably shown to be free from CAV, provided it has been
treated to avoid introduction of undesirable micro-organisms demonstrated that the inactivation process inactivates CAVo
and that water is at least of potable quality, for example from
a chlorinated supply. No medication is administered to birds Establishment of an SPF flock
within the fiock that might interfere with detection of any A designated SPF fiod: is derived fram chickens shown to be
disease. free fram vertically-transmissible agents listed in
Table 5.2.2-1. This is achieved by testing of 2 generations
A permanent record is kept of the general health of the fiod:
prior to the designated SPF fiod::. A general scheme for the
and any abnormality is investigated. Factors to be monitored
pracedure to be followed in establishing and maintaining an
inc1ude morbidity, mortality, general physical condition, feed
SPF fiock is shown diagrammatically in Table 5.2.2.-2.
consumption, daily egg production and egg quality, fertility
In order to establish a new SPF fiod:, a series of tests must
and hatchability. Records are maintained for a period of at
be conducted on 3 generations of birds. AH birds in the 1st
least 5 years. Details of any deviation from normal in these
generation must be tested at least once before the age of
performance parameters or detection of any infection are
20 weeks for freedom from avian leucosis group-antigen and
notified to the users of the eggs as soon as practicable.
tested by an enzyme irnmunoassay (EIA) or by virus
The tests or combination of tests described below must have neutralisation (VN) for freedom of antibodies to avian
suitable specificity and sensitivity with respect to relevant leucosis virus subtypes A, B and J. AH birds must also be
serotypes of the viruses. Samples for testing are taken at tested for freedom from antibodies to the vertically-
random. transmissible agents listed in Table 5.2.2-1. From the age of
8 weeks the fiock is tested for freedom from Salmonella.
2016 Vet-A11

Table 5.2.2-2. - Schematic description of the establishment and maintenance of SPF flocks
NEW STOCK Establish freedom from vertical!y-transmissible agents

Test al! birds for avian leucosis antigen and antibodies prior to 20 weeks of age

Test for Salmonella spp. and perform general clinical observation fram 8 weeks af age
Carry out routine testing for specified agents from 20 weeks af age
nd
2 GENERATION Test all birds for avian leucosis antigen and antibodies prior to 20 weeks of age

Test for Salmonella spp. and perform general clinical observation from 8 weeks of age
Carry out routine testing for specified agents from 20 weeks of age

3rd GENERATION Test all birds for avian leucosis antigen and antibadies prior to 20 weeks of age

Test for Salmonella spp. and perform general clinical abservation from 8 weeks af age
DESIGNATE FLOCK AS SPF IF ALL TESTS ARE SATISFACTORY

3rd GENERATION Carry out routine testing for specified agents from 20 weeks of age

Carry out post-lay testing for vertically-transmissible agents

SUBSEQUENT GENERATIONS Test twa 5 per cent samples for avian leucosis antigen and for antibodies against specified
agents between 12 and 20 weeks of age
Test far Salmonella spp. and perform general clinical observation from 8 weeks af age

Carry out routine testing for specified agents from 20 weeks af age

Carry out past-lay testing for vertical!y-transmissible agents

Clinical examination is carried out on the flod: from 8 weeks infection. In the event of mortality exceeding 0.2 per cent per
of age and the birds must not exhibit any signs of infectious week, necropsy is performed on aH available carcasses to
disease. The test methods to be used for these tests are given verify dlat dlere is no sign of infection. Where appropriate,
in the table and further guidance is also given in the section histopadlological and/or microbiologicallvirological studies are
below on routine testing of designated SPF flocks. From performed to confirm diagnosis. Specific examination for
20 weeks of age, the flock is tested as described under tuberculosis lesions is carried out and histological samples
Routine testing of designated SPF flocks. AH stages of this from any suspected lesions are specificaHy stained to verify
testing regime are also applied to the subsequent freedom from lVlycobacterúl771 avil/77l. Caecal contents of aH
2 generations, except the testing of every bird before lay for available carcasses are examined microbiologically for dle
vertically-transmissible agents. AH test results must indicate presence of Sal7710llella spp. using dle techniques described
freedom from pathogens in aH 3 generations for the flock below. Where appropriate, caecal samples from up to 5 birds
consisting of the 3rd generation to be designated as SPF. may be pooled.
SPF embryos derived from another designated SPF flock Cultural testing for Sabnonella spp
contained within a separate facility on the same site may be Cultural testing for SallllOnella spp. is perfOTI1led either by
introduced. From 8 weeks of age, these replacement birds are testing samples of droppings 01' cloacal swabs or by testing of
regarded as a flock and are tested in accordance with test drag swabs. Where droppings 01' cloacal swabs are tested, a
procedures described aboye. total of 60 samples within each 4-week period is tested
throughout the entire life of dle flock. Tests may be
Initial testing requirements for subsequent
performed on pools of up to 10 samples. Where drag swabs
generations derived from a designated SPF ftock
are tested, a minimum' of 2 drag swabs are tested during each
Where a replacement flock is derived exclusively from a fully
4-week period dlroughout dle entire life of the flock.
established SPF flock the new generation is tested prior to
Detection of Sa171lonella spp. in these samples is perfonned by
being designated as SPF. In addition to the tests for
pre-enrichment of the samples followed by culture using
Salmonella and monitoring of the general health and
SaI7l1071ella-selective media.
performance of the flock, further specific testing from the age
of 8 weeks is required. Tests are performed on two Tests f01" avian leucosis antigen
5 per cent samples of the flock (minimum 10, maximum 200 Prior to dle commencement of laying, cloacal swabs or blood
birds) taken with an interval of at least 4 weeks between the samples (using buffy coat cultivation) are tested for the
ages of 12-16 weeks and 16-20 weeks. presence of group-specific leucosis antigen. A total of
AH samples are coHected and tested individuaHy. BIood 5 per cent (minimum 10, maximurn 200) of dle flock is
samples for antibody tests and suitable samples for testing for sampled during each 4-week periodo During lay, albumen
leucosis antigen are collected. The test methods to be used samples from 5 per cent (minimum 10, maximum 200) of
are as described under Routine testing of designated SPF the eggs are tested ih each 4-week periodo Tests are
flocks. Only when aH tests have confirmed the absence of performed by EIA for group-specific antigen using medl0ds
infection may the new generation be designated as SPF. that are capable of detecting antigen from subgroups A, B
and J.'
Routine testing of designated SPF ftocks
Testfor antibodies to other agents
General examination and necropsy Tests for antibodies to aH agents listed in Table 5.2.2.-1 are
Clinical examination is carried out at least once per week performed d1roughout the laying period of the fiod:. In each
throughout the life of the flock in order to verify that the 4-week period, samples are taken from 5 per cent (minimum
birds are free from fowl-pox virus and signs of any other
Vet-A12 Appendix XV JeVet) 1 2016

10, maximum 200) of the fiock. It is recommended that


1.25 per cent of the fiod: is sampled each week since sorne
J (Vet) 1. Cell Cultures tor the
test methods for sorne agents must be conducted on a weekly Production of Veterinary Vaccines
basis. Table 5.2.2.-1 c1assifies the agents into those that
spread rapidly through the fiod: and those that spread slowly
01' may not infect the entire fiod:. For those agents listed as
slowly spreading, each sample is tested individually. Cell Cultures (Veterinary Vaccine
For those agents listed as rapidly spreading, at least
20 per cent of the samples collected in each 4-week period Production)
are tested individually 01', where serum neutralisation 01' (Ph Bur general texts 5.2.4)
EUSA tests are employed, aH of the samples may be tested Cell cultures for the production of vaccines for veterinary use
individually 01' by preparing pools of 5 samples, collected at comply with the requirements of this section. It may also be
the same time. neceSSa1y that cell cultures used for testing of vaccines for
Suitable methods to be used for detection of the agents are veterina1Y use also comply with so~e or aH of these
shown in Table 5.2.2.-1. Subject to agreement by the requirements.
competent authority, other test methods may be used For most mammalian viruses, propagation in celllines is
provided they are shown to be at least as sensitive as those possible and the use of primary cells is then not acceptable.
indicated and of appropriate specificity. Permanently infected cells used for production of veterinary
vaccines comply with the appropriate requirements described
Tests to be conducted at the end of the laying period be1ow. The cells shall be shown to be infected only with the
Following the last egg collection, final testing to confirm the agent stated.
absence of verticaHy-transmissible agents indicated in
Table 5.2.2.-1 is perfonned. After the last egg collection, a CELL LINES
minimum of 5 per cent of the fiod: (minimum 10, 'maximum Celllines are normally handled according to a cell-seed
200) is retained for at least 4 weeks. BIood samples are system. Each master cell seed is assigned a specific code for
coHected from every bird in the group during the 4-week identification purposes. The master cell seed is stored in
period with at least 1.25 per cent of the birds (25 per cent of aliquots at - 70 oC or lower. Production of vaccine is not
the sample) being bled not earlier than 4 weeks after the final normaHy undertaken on cells more tllan twenty passages
egg collection. Semm samples are tested for verticaHy- from the master cell seed. Where suspension cultures are
transmissible agents (as defined by Table 5.2.2.-1) using the used, an increase in cell numbers equivalent to approximately
methods indicated. Where sampling is performed on a weekly three population doublings is considered equivalent to one
basis, at least 1.25 per cent of the birds (25 per cent of the passage. If cells beyond twenty passage levels are to be used
sample) are tested each week during this periodo for production, it shaH be demonstrated, by validation or
Alternatively, within 4 weeks of the final egg coHection blood further testing, that the production cell cultures are
and/or other suitable sample material s are collected from at essentially similar to tlle master cell seed with regard to their
least 5 per cent of the fiod: and tested for the presence of biological characteristics and purity and that the use of such
vertically-transmissible agents using validated nuc1eic acid cells has no deleterious effect on vaccine production.
amplification techniques (2.6.21). The history of the cell1ine shall be known and recorded in
detail (for example, origin, number of passages and media
Action to be taken in the event of detection of a used for multiplication, storage conditions).
specified agent The method of storing and using the cells, inc1uding details
If evidence is found of contamination of the fiock by an of how it is ensured that the maximum number of passages
agent listed as slowly spreading in Table 5.2.2.-1, aH permitted is not exceeded during product manufacture, are
materials derived from the fiod: during the 4-week period recorded. A sufficient quantity of the master cell seed and
irnmediately preceding the date on which the positive sample each working cell seed are kept for analytical purposes.
was collected are considered unsatisfactory. Similarly, if The tests described below are cairied out (as prescribed in
evidence is found of contamination of the fiock by an agent Table 5.2.4.-1) on a culture ofthe master cell seed and the
listed as rapidly spreading in Table 5.2.2.-1, aH materials working cell seed 01' on cell cultures from the working cell
derived from the fiock during the 2-week period immediately
seed at the highest passage level used for production and
preceding the date on which the positive sample was derived from a homogeneous sample demonstrated to be
collected are considered unsatisfact01Y. Any product representative.
manufactured with such materials, and for which the use of
SPF materials is required, is considered unsatisfactory and TabIe 5.2.4.-1. - Cel! culture stage at which tests are carried
must be discarded; any quality control tests conducted using out
the materials are invalid.
Master Working Cell from working
Producers must notify users of aH eggs of the evidence of cell seed cell seed cell seed at highest
contamination as soon as possible following the outbreak. passage IeveI
General microscopy + + +
Any fiod: in which an outbreak of any specified agent is
confirmed may not be redesignated as an SPF fiod:. Bacteria and fungi + +
Any progeny derived from that fiod: during or after the + +
Mycoplasmas
4-week period prior to the last negative sample being
collected may not be designated as SPF. Viruses + +

Identification of species + +

Karyotype + +

Tumorigenicity +
2016 Appendix XV J(Vet) 1 Vet-A13

Characteristics of culture The appearance of cell buffer and a sufficient volume of a suspension of suitable red
monolayers, before and after histological staining, is blood cells added to cover the surface of the monolayer
described. Information, if possible numerical data, is evenly. After different incubation times cells are examined for
provided especially on the speed and rate of growth. the presence of haemadsorption.
Similady, the presence or absence of contact inhibition, Detection of specified virus es Tests are carried out for
polynucleated cells and any other cellular abnormalities are freedom from contaminants specific for the species of origin
specified. of the cell line and for the species for which the product is
Karyotype A chromosomal examination is made of not intended. Sufficient cells on suitable supports are prepared to
fewer than fifty cells undergoing mitosis in the master cell carry out tests for the agents specified. Suitable positive
seed and at a passage level at least as high as that to be used controls are included in each test. The cells are subjected to
in production. Any chromosomal marker present in the suitable tests, for example using fiuorescein-conjugated
master cell seed must also be found in the high passage cells antibodies or similar reagents.
and the modal number of chromosomes in these cells must Tests in other cell cultures Monolayers totalling at least
not be more than 15 per c'eJ:~t higher than of cells of the 140 cm2 are required. The cells are frozen and thawed at
master cell seed. The karyotypes must be identical. If the least three times and then centrifuged to remove cellular
modal number exceeds the level stated, if the chromosomal debris. Inoculate aliquots onto the following cells at any time
markers are not found in the working cell seed at the highest up to 70 per cent confiuency:
level used for production or if the karyotype differs, the cell - primary cells of the source species;
line shall not be used for manufacture. cells sensitive to vu'uses pathogenic for the species for
Identification of the species It shall be shown, by one which the vaccine is intended;
validated method, that the master cell seed and the cells from cells sensitive to pestiviruses.
the working cell seed at the highest passage level used for The inoculated cells are maintained in culture for at least
production come from the species of origin specified. When a 7 days, after which freeze-thawed extracts are prepared as
fiuorescence test is carried out and the corresponding serum above and inoculated onto sufficient fresh cultures of the
to the species of origin of cells is used and shows that all the same cell types to allow for the testing as described below.
tested cells are fiuorescent, it is not necessary to carry out The cells are incubated for at least a further 7 days.
other tests with reagents able to detect contamination by cells The cultures are examined regulady for the presence of any
of other species. cytopathic changes indicative of living organisms.
Bacterial and fungal contamination The cells comply At the end of this period of 14 days, the inoculated cells are
with the test for sterility (2.6.1). The sample of cells to be subjected to the following checks:
examined consists of not less than the number of cells in a - freedom from cytopathic and haemadsorbent organisms,
monolayer with an area of 70 cm2 or, for cells grown in using the methods specified in the relevant paragraphs
suspension, an approximately equivalent number of cells. above,
The cells are maintained in culture for at least 15 days - absence of pestiviruses and other specific contaminants by
without antibiotics before carrying out the test. immunofiuorescence or other validated methods as
Mycoplasmas (2.6.7) The cells comply with the test for indicated in the paragraph above on Detection of
mycoplasmas. The cells are maintained in culture for at least Specified Viruses.
15 days without antibiotics before carrying out the test. Tumorigenicity The risk of a cell IU1e for the target species
Absence of contaminating virus es The cells must not be must be evaluated and, if necessary, tests are carried out.
contaminated by viruses; suitably sensitive tests, including
PRIMARY CELLS
those prescribed below, are carried out.
For most mammalian vaccines, the use of primary cells is not
The monolayers tested shall have an area of at least 70 cm 2 , acceptable for the manufacture of vaccines since celllu1es can
and shall be prepared and maintained using medium and be used. If there is no altemative to the use of primary cells,
additives, and grown under similar conditions to those used the cells are obtained from a herd or fiod: free from specified
for the preparation of the vaccine. The monolayers are pathogens, with complete protection from introduction of
maintained in culture for a total of at least 28 days. diseases (for example, disease barriers, filters on air inlets,
Subcultures are made at 7-day intervals, unless the cells do suitable quarantine before introduction of animals). Chicken
not survive for this length of time, when the sub cultures are fiocks comply with the requirements prescribed in general
made on the latest day possible. Sufficient cells, in suitable chapter 5.2.2. Chicken Flocks Free fro112 Specified Pathogens for
containers, are produced for the final sub culture to carry out the Production and Quality Control of Vaccines. For all other
the tests specified below. species, the herd or fiock is shown to be free from relevant
The monolayers are examined regularIy throughout the specified pathogens. All the breeding stock in the herd or
incubation period for the possible presence of cytopathic fiod: intended to be used to produce primal)' cells for
effects and at the end of the observation period for cytopathic vaccine manufacture is subject to a suitable monitoring
effects, haemadsorbent viruses and specific viruses by procedure including regular serological checks carried out at
irnmuno-fiuorescence and other suitable tests as indicated least twice ayear and two supplementary serological
below. examinations perfonned in 15 per cent of the breeding stock
Detection of cytopathic virus es Two monolayers of at in the herd between the two checks mentioned above.
least 6 cm2 each are stained with an appropriate cytological Wherever possible, particularIy for mammalian cells, a seed-
stain. The entire area of each stained monolayer is examined lot system is used with, for example, a master cell seed
for any inclusion bodies, abnormal numbers of giant cells or formed after less than five passages, the working cell seed
any other lesion indicative of a cellular abnormality which being no more than five passages from the initial preparation
might be attributable to a contaminant. of the cell suspension from the animal tissues.
Detection of haemadsorbent virus es Monolayers totalling Each master cell seed, working cell seed and cells of the
at least 70 cm2 are washed several times with an appropriate highest passage of primary cells are checked in accordance
Vet-A14 Appendix XV JCVet)2 2016

with Table 5.2.4.-2 and the procedure des·cribed below. Detection of cytopathic vil'uses Two monolayers of at
The sample tested shall cover all the sources of cells used for least 6 cm 2 each are stained with an appropriate cytological
the manufacture of the batch. No batches of vaccine stain. Examine the entire area of each stained monolayer for
manufactured using the cells may be released if any one of any inclusion bodies, abnormal numbers of giant cel1s 01' any
the checks perfOlmed produces unsatisfactOlY results. other les ion indicative of a cel1ular abnOlmality that might be
attributable to a contaminant.
Table 5.2.4.-2. - Cel! culture stage at which tests are carried Detection of haemadsorbent viruses Monolayers totalling
out at least 70 cm2 are washed several times with a suitable
buffer solution and a sufficient volume of a suspension of
Master Working Highest passage
cell seed cell seed level suitable red blood cel1s added to cover the surface of the
+ + + monolayer evenly. After different incubation times, examine
General microscopy
cel1s for dle presence of haemadsorption.
Bacteria and fungi + +
Detection of specified virus es Tests are be carried out for
Mycoplasmas + + freedom of contaminants specific for the species of origin of
Viruses + + the cel1s and for the species for which dle product is
intended.
Identification of species +
Sufficient cel1s on suitable supports are prepared to carry out
tests for the agents specified. Suitable positive controls are
Characteristics of cultures The appearance of cel1 included in each test. The cells are subjected to suitable tests
monolayers, before and after histological staining, is using fiuorescein-conjugated antibodies or similar reagents.
described. Information, if possible numerical data, is Tests in other cell cultures Monolayers totalling at least
recorded, especially on the speed and rate of growth. 140 cm2 are required. The cells are frozen and thawed at
Similar1y, the presence or absence of contact inhibition, least dlree times and then centrifuged to remove cellular
polynucleated cells and any other cel1ular abnormalities are debris. Aliquots are inoculated onto the fol1owing cel1s at any
specified. time up to 70 per cent confiuency:
Identification of species It shall be demonstrated by one - prima1y cel1s of dle source species;
validated test that the master cel1 seed comes from the - cel1s sensitive to virus es pathogenic for the species for
specified species of origino which dle vaccine is intended;
When a fiuorescence test is carried out and the - cel1s sensitive to pestiviruses.
corresponding serum to the species óf origin of cel1s is used The inoculated cel1s are maintained in culture for at least
and shows that all the tested cel1s are fiuorescent, it is not 7 days, after which freeze-dlawed extracts are prepared as
necessary to carry out other tests with reagents able to detect above, and inoculated onto sufficient fresh cultures of dle
contamination by cel1s of other species. same cell types to allow for the testing as described below.
Bacterial and fungal sterility The cells comply with the The cel1s are incubated for at least a furdler 7 days.
test for sterility (2.6.1). The sample of cel1s to be examined All cultures are regularly examined for the presence of any
consists of not less than the number of cells in a monolayer cytopathic changes indicative of living organisms.
with an area of 70 cm 2 01' for cel1s grown in suspension an At the end of this period of 14 days, dle inoculated cel1s are
approximately equivalent number of cells. The cel1s are subjected to the fol1owing checks:
maintained in culture for at least 15 days without antibiotics freedom from cytopathic and haemadsorbent organisms is
before carrying out the test. demonstrated using the medl0ds specified in dle relevant
Mycoplasmas (2.6.7) The cells comply with the test for paragraphs above;
mycoplasmas. The cel1s are maintained in culture for at least - relevant substrates are tested for dle absence of
15 days without antibiotics before canying out the test. pestiviruses and other specific contaminants by
immunofiuorescence or other validated medl0ds as
Absence of contaminating virus es The cel1s must not be
indicated in dle paragraph above on Detection of
contaminated by viruses; suitably sensitive tests, including
Specified Viruses.
those prescribed below are carried out.
The monolayers tested shall be at least 70 cm2 , and shall be
prepared and maintained in culture using the same medium
and additives, and under similar conditions to those used for J (Vet) 2. Substancesof Animal Origin
the preparation of the vaccine.
The monolayers are maintained in culture for a total of at
for the Production of Immunological
least 28 days 01' for the longest period possible if culture for Veterinary Medicinal Products
28 days is impossible. Sub cultures are made at 7-day
intervals, unless the cells do not survive for this length of
time when the sub cultures .are made on dle latest day
possible. Sufficient cel1s, in suitable containers are produced Substances of Animal Origin for the
for the final subculture to carry out the tests specified below.
The monolayers are examined regularly throughout the
Production of Immunological Veterinary
incubation period for the possible presence of cytopathic Medicinal Products
effects and at the end of the observation period for cytopathic (Ph Bur general texts 5.2.5)
effects, haemadsorbent virus es and specific virus es by
1. SCOPE
irnmunofiuorescence and other suitable tests as indicated
Substances of animal origin (for example serum, uypsin and
below.
serum albumin) may be used during the manufacture of
irnmunological veterinalY medicinal products.
2016 Appendix XV J(Vet)2 Vet-A15

The requirements set out in this chapter apply to substances exu"aneous agents needs to be assessed. The risk of
of animal origin produced on a batch basis, for use at all contamination of d1e substm1ce and the resultant
stages of manufacture, for example in culture media 01' as üm11Unological veterinmy medicinal product with inactivated
added constituents of products during blending. These extraneous agents may also need to be taken into account.
requirements are not intended for the control of seed This would be the case if, for example, the contaminant was
materials 01' substrates of animal origin that are covered by one from which a European counuy is officially free and/or is
requirements in other pharmacopoeial texts such as the the subject of a specific disease control program in a
monograph Vaccines for veterinal)' use (0062) and chapter European counuy and where the presence of the inactivated
5.2.4. Gel! cultures for the productiol1 of veterinclI)' vaccines. agent could lead to the stimulation of a detectable immune
response in recipient animals.
2. GENERAL PRINCIPLES AND REQUIREMENTS
Substances of animal origin comply zuith the requirements of the As part of the risk assessment, the presence in the substance
European Pharmacopoeia (zuhere a relevant 1ll01l0graph exists). of antibodies d1at can intelfere with the detection and/or
inactivation of living exU'aneous agents must also be taken
Restrictions are placed on the use of substances of animal
into account.
origin because of safety concerns associated with pathogens
that may be present in them and epidemiological and/or The risk assessment may need to be repeated and the risk
regulatory concerns associated with the presence of particular management steps described below re-evaluated and revised
antigens (either live 01' inactivated). in order to take account of changes:
in the incidence of diseases occurring in the country or
General principIes:
countries of origin of animals used as the source for the
it is recommended to minimise, wherever practicable, the
substance, including emerging diseases (new pathogens);
use of substances of animal origin;
in the incidence of diseases and of disease conu"ol
- unless othelwise justified, the use of substances of animal
measures applied in the European countries in which
origin as constituents in the formulation of medicinal
immunological veterinalY medicinal products
products is not acceptable except where such substances
manufactured with the substance are used.
are subject to a treatment validated for the inactivation of
live extraneous agents. 3-2 RISK CONTROL
Generalrequirements: For each of the potential extraneous agents identified by the
- any batch of substance (after inactivation andlor risk assessment, and taking into account the proposed use of
processing, if relevant) found to contain 01' suspected of the substance, d1e risk must be conu"olled by the use of one
containing any living extraneous agent shall be discarded 01' a combination of the followings measures:
01' used only in exceptional and justified circumstances; placing restrictions on the source of d1e material and
to be accepted for use, further processing must be applied auditing this;
that will ensure elimination and/or inactivation of the using validated inactivation procedures;
extraneous agent, and it shall then be demonstrated d1at demonsu"ating the ability of a production step to remove
the elimination and/or inactivation has been satisfactory; or inactivate extraneous agents;
any batch of substance that, as concluded from the risk testing for extraneous agents.
assessment, may induce an unacceptable detectable 4. CONTROL MEASURES
immune response in the target species as a consequence 4-1 SOURCE
of contamination wid1 inactivated extraneous agents, must All substances of animal origin used in d1e manufacture
not be used for the manufacture of that particular (including blendü1g) of immunological veterinalY medicinal
immunological veterinary medicinal producto products must be from a known and documented source
3. RISK MANAGEMENT (including species of origin and counuy of origin of source
No single measure 01' combination of measures can gum"antee animals and tissues).
the safety of the use of substances of animal origin, but d1ey 4-2 PREPARATION
can reduce the risk from such use. It is therefore necessary Substances of animal origin are prepared from a
for the manufacturer of immunological veterinary medicinal homogeneous bulk designated wid1 a batch number. A batch
products to take account of this when choosing a substance may contain substances derived from as many animals as
of animal origin to use in manufacture, and to conduct a risk desired but once defined and given a batch number, the
assessment, taking into account the origin of d1e substance batch is not added to or contaminated in any way.
and the manufacturing steps applied to it.
The production med10d. used to prepare d1e substance of
In addition, risk management procedures must be applied. animal origin from the raw material may contribute to the
Any residual risk must be evaluate? in relation to d1e removal and/or inactivation of extraneous agents (see
potential benefits derived from the use of the substance for section 4-3).
the manufacture of d1e immunological veterinmy medicinal
product. 4-3 INACTIVATION AND/OR OTHER PROCESSING
STEPS FOR REMOVAL OF EXTRANEOUS AGENTS
3-1 RISK ASSESSMENT The inactivation procedure and/or other processü1g steps
The risk assessment must take account of d1e animal diseases chosen shall have been validated and shown to be capable of
occurring in the counuy of origin of the animals used as a reducing the titre of potential extraneous agents described
source of the substance, the potential infectious diseases below in the substance concerned by a factor of at least 10 6 .
occurring in the source species and d1e likely infectivity in the
If this l"eduction in titre cannot be shown experilnentally, a
source organ or tissue. From this information, as part of the
maxünum pre-treaunent titre of the exu'aneous agent must
risk assessment, a list can be prepared of the exu"aneous
be set, taking into account the reduction in titre afforded by
agents that may be present in the substance.
the inactivation/processing step and including a safety margin
The risk of contamination of d1e substance and d1e resultant factor of 100; each batch of substance must be tested to
immunological veterinary medicinal product wid1 living determine the pre-treatment starting titre and confillli it is no
Vet-A16 Appendix XV IZ (Vet) 1. 2016

greater than the specified limit, unless proper risk assessment,


based on valid and suitable data, shows that titres will always
K (Vet) 1. Evaluation of Safety of
be at least 100-fold below the titre that can effectively be Veterinary Vaccines and Immunosera
inactivated.
The validation of the procedure(s) is conducted with a
suitable representative range of viruses covering different
types and sizes (enveloped and non-enveloped, DNA and Safety Veterinary Vaccines and
RNA, single- and double-stranded, temperature- and
pH-resistant), including test viruses with different degrees of
Immunosera
resistance, taking into account the type of procedure(s) to be (Ph Bur general texts 5.2.6)
applied and the vil"uses that may be present in the material. The term 'product' means either a vaccine or an
The evidence for the efficacy of the procedure may take the immunosel"um throughout the texto
form of references to published literature and/or experimental During development, safety tests are carried out in the target
data generated by the manufacturer, but must be relevant to species to show the risks from use of the producto
the conditions that will be present during the production and Iml1lu11e status for tests 011 vaccines The immune status of
inactivation/processing of the substance. animals to be used for the safety test is specified in the
For inactivated irnmunological veterinaty medicinal products, specific monograph. For most monographs, 1 of the
the method used for inactivation of the active ingredient may 3 following categories is specified:
also be validated for inactivation of possible contaminants 1) the animals must be free from antibodies against the
from substances of animal origin used in the manufacture of virus/bacterium/toxin etc. contained in the vaccine;
this active ingredient.
2) the animals are preferably free from antibodies against the
4-4 TESTS virus/bacteriumltoxin etc. contained in ti1e vaccine, but
Depending on the outcome of the risk assessment and the animal s with a low level of antibody may be used as long as
validation data available for any procedure applied, tests for the animals have not been vaccinated and the administration
extraneous agents may be conducted on each batch before of the vaccine does not cause an anamnestic response;
and/or after the application of an inactivation/processing step. 3) the animals must not have been vaccinated against ti1e
For examination of the substance fol" freedom from disease ti1at the vaccine is intended to prevent.
extraneous agents, any solids are dissolved or suspended in a As a general rule, category 1 is specified for live vaccines.
suitable medium to provide a suitable preparation for testing.
For other vaccines, categ01y 2 is usually specified, but where
A sufficient quantity of the preparation is tested to give a
most animals available for use in tests would comply with
suitably sensitive test, as established in the validation studies.
categ01y 1, this may be specified for inactivated vaccines also.
As well as tests for living extraneous agents, tests may need Categ01y 3 is specified for sorne inactivated vaccines where
to be conducted for the presence of inactivated extraneous determination of antibodies prior to testing is unnecessaly or
agents, depending on the risks identified. impractical. For poulny vaccines, as a general rule ti1e use of
Freedom from living extraneous vil·uses specified-pathogen-free (SPF) birds is specified.
A sample from each batch of the substance is tested for For avian vaccines, the safety test is generally carried out
extraneous viruses by general and specific tests. These tests using SPF chickens (5.2.2), except ti1at for vaccines not
are validated with respect to sensitivity and specificity for recommended for use in chickens it is carried out using birds
detection of a suitable range of potential extraneous viruses. of one of the species for which ti1e vaccine is recommended,
Suitably sensitive cell cultures are used for the tests for the birds being free from antibodies against the disease agent
extraneous viruses, including primary cells from the same for which thevaccine is intended to provide protection.
species as the substance to be examined. Vaccines In laborat01Y tests, 'dose' means ti1at quantity of
General test The inoculated cell cultures are observed the product to be recommended for use and containing the
regularly for 21 days for cytopathic effects. At the end of maximum titre 01' potency likely to be contained in
each 7-day period, a proportion of the original cultures is production batches. Live vaccines are prepared only from
fixed, stained and examined for cytopathic effects, and a strains of organisms that have been shown to be safe. For live
proportion is tested for haemadsorbing agents. vaccines, use a batch 01' batches of vaccine containing
Specific tests A proportion of the cells available at the end vil"us/bacteria at the least attenuated passage level that will be
of the general test is tested for specific viruses. The specific present in a batch of vaccine.
vil"uses to be tested for are potential extraneous viruses that For combined vaccines, the safety shall be demonstrated;
are identified through the risk assessment and that would not for live components of combined vaccines, compliance with
be detected by the general test. A test for pestiviruses is the special requirements for live vaccines stated below shall
conducted if the source species is susceptible to these. be demonstrated separately for each vaccine strain.
Bacteria and fungi For inactivated vaccines, safety tests carried out on the
Before use, substances are tested for sterility (2.6.1), or combined vaccine may be regarded as sufficient to
sterilised to inactivate any bacterial 01' fungal contaminants. demonstrate the safety of the individual components.
Mycoplasma Immunosera In the tests, 'dose' means the maximum
Before use, substances are tested for freedom from quantity of the product to be recommended for use and
mycoplasma (2.6.7), or sterilised to inactivate any containing the maximum potency and maximum total
mycoplasmal contaminants. protein likely to be contained in production batches.
In addition, if appropriate, the dose tested also contains
maximum quantities of irnmunoglobulin or gammaglobulin.
The tests described below, modified or supplemented by
tests described in the Production section of a monograph,
2016 Appendix XV IZ (Vet) 1. Vet-A17

may be carried out as part of the tests necessary during general 8 birds per group are used unless od1erwise justified
development to demonstrate the safety of the product. or specified in a specific monograph. For vaccines intended
for use in birds younger than 3 weeks, in general 10 birds per
1 LABORATORY TESTS
group are used unless otherwise justified or specified in a
1-1 SAFETY OF THE ADMINISTRATION OF
specific monograph. The animals are observed and examined
1 DOSE
at least daily for signs of local and systemic reactions. Other
For each of the recommended routes of administration,
objective criteria are recorded, such as body temperature (for
administer 1 dose of product to animals of each species and
mammals) and perfonnance measurements. The animal s are
category for which use of the product is to be recommended.
observed and examined for at least 14 days after
This must include animals of the youngest recommended age
adminisu'ation.
and pregnant animals, if appropriate.
Unless otherwise prescribed in a specific monograph or, in
For vaccines intended for use in mammals, in general
the absence of a specific monograph, unless otherwise
8 animals per group are used unless otherwise justified or
justified and authorised, d1e vaccine complies with the test if
specified in a specific monograph.
no animal shows abnormal local or systemic reactions or
For fish vaccines administered by immersion, bathe the fish signs of disease, or dies from causes attlibutable to the
for twice the recommended time using a bath at twice the vaccine.
recommended concentration.
1-3 SAFETY OF THE REPEATED
For vaccines intended for use in fish, in general 50 fish per
ADMINISTRATION OF 1 DOSE
group are used unless otherwise justified or specified in a
Repeated adminisu'ation of 1 dose may be required to reveal
specific monograph.
any adverse effects induced by such adminisu'ation. These
For vaccines intended for use in birds older than 3 weeks, in tests are particularly important where the product, notably an
general 8 birds per group are used unless otherwise justified immunoserum, may be administered on several occasions
01' specified in a specific monograph. For vaccines intended
over a relatively short period of time. These tests are canied
for use in birds younger than 3 weeks, in general 10 birds per out on the most sensitive categOlies of the target species,
group are used unless od1erwise justified or specified in a using each recommended route of administration. If multiple
specific monograph. routes and methods of administration are specified for the
The animals are observed and examined at least daily for product concemed, administration by all routes is
signs of abnormallocal and systemic reactions. Where recommended. If 1 route of adminisu'ation has been shown
appropriate, these studies shall include detailed post-mortem to cause the most severe effects, d1is single route may be
macroscopic and microscopic examinations of the injection selected as the only one for use in the study. The number of
site. Other objective criteria are recorded, such as body adminisu'ations must be not less than the maximum number
temperature (for mammals) and performance measurements. recommended; for vaccines, this shall take account of d1e
The body temperatures are recorded on at least the day number of administrations for primary vaccination and the
before and at the time of administration of the product, 4 h 1st re-vaccination; for immunosera, it shall take account of
later and on the following 4 days. The animals are observed the number of administrations required for treatment.
and examined at least daily until reactions may no longer be The interval between administrations shall be suitable
expected but, in all cases, the observation and examination (e.g. period of risk or required for treatment) and appropriate
period extends at least until 14 days after administration. to the recommendations of use. Although, for convenience,
Unless otherwise prescribed in a specific monograph or, in as far as vaccines are concerned, a shorter interval may be
the absence of a specific monograph, unless otherwise used in the study d1an d1at recommended in the field, an
justified and authorised, the vaccine complies wid1 the test if interval of at least 14 days must be allowed between
no animal shows abnormal local or systemic reactions or administrations for the development of any hypersensitivity
signs of disease, or dies from causes attributable to the reaction. For immunosera, however, adminisu'ation shall
vaccine. follow the recommended schedule. For vaccines intended for
use in mammals, in general 8 animals per group are used
1-2 SAFETY OF 1 ADMINISTRATION OF AN
unless otherwise justified or specified in a specific
OVERDOSE
monograph. For vaccines intended for use in fish, in general
Overdose testing is required only for live vaccines.
50 fish per group are used unless od1erwise justified or
An overdose of the product is administered by each
specified in a specific monograph. For vaccines intended for
recommended route of administration to animal s of the
use in birds older than 3 weeks, in general 8 birds per group
categories of the target species that are expected to be the
are used unless otherwise justified or specified in a specific
most sensitive, such as animals of the youngest age.
monograph. For vaccines intended for use in birds younger
If multiple routes and methods of administration are
than 3 weeks, in general 10 birds per group are used unless
specified for the product concemed, administration by all
otherwise justified or specified in a specific monograph.
routes is recommended. If 1 route of administration has been
The animals are observed and examined at least daily for at
shown to cause the most severe effects, this single route may
least 14 days after the last administration for signs of
be selected as the only one for use in the study.
systemic and local reactions. Od1er objective clitelia are
The overdose normally consists of 10 doses of a live vaccine.
recorded, such as body temperature and perfoffi1ance
For freeze-dried live vaccines, the 10 doses shall be
measurements.
reconstituted in a suitable volume of diluent for d1e test.
For vaccines intended for use in mammals, in general Unless otherwise presclibed in a specific monograph or, in
8 animals per group are used unless otherwise justified or theabs~nce of a specific monograph, unless otherwise
specified in a specific monograph. For vaccines intended for justified and authorised, the product complies with the test if
use in fish, in general 50 fish per group are used unless no animal shows abnormal local or systemic reactions or
otherwise justified or specified in a specific monograph. signs of disease, or dies from causes attlibutable to the
For vaccines intended for use in birds older than 3 weeks, in producto
Vet-A18 Appendix XV !Z (Vet) 1. 2016

1-4 EXAMINATION OF REPRODUCTIVE recommended as part of a schedule of administration of


PERFORMANCE products within a short period of time.
When the vaccine is recommended for use or may be used in 1-8 SPECIAL REQillREMENTS FOR LIVE
pregnant animals or laying birds, carry out a test for safety in VACCINES
this categOly of animals. If the reproductive safety studies are
The following laboratOlY tests must also be carried out with
not perfOlmed, an exclusion statement appears on the label,
live vaccines.
unless a scientific justification for absence of risk is provided.
Examination of reproductive performance must also be For the following tests except for the test for increase in
considered when data suggest that the starting material from virulence (section 1-8-3), use the vaccine strain at the least
which dle product is derived may be a risk factor. Where attenuated passage level that will be present between the
appropriate, reproductive perfOlmance of males and females master seed lot and a batch of vaccine.
and harmful effects on the progeny, including teratogenic or 1-8-1 Spread of the vaccine strain Spread of the vaccine
abortifacient effects, are investigated by each of dle strain from vaccinated to unvaccinated target animal s is
recommended routes of administration. If multiple routes investigated using tlle recommended route of administration
and methods of administration are specified for the product most likely to result in spread. Moreover, it may be necessary
concemed, administration by all routes is recommended. If 1 to investigate tlle safety of spread to non-target species that
route of administration has been shown to cause dle most could be highly susceptible to a live vaccine strain.
severe effects, this single route may be selected as the only An assessment must be made of how many animal-to-animal
one for use in the study. passages are likely to be sustainable under normal
For vaccines intended for use in mammals, in general circumstances togedler with an assessment of the likely
8 animals per group are used unless otherwise justified or consequences.
specified in a specific monograph. Vaccines recoIhmended for 1-8-2 Dissemination in vaccinated animal Faeces, urine,
use or that may be used in pregnant animals, are tested in milk, eggs, and oral, nasal and other secretions shall be tested
each of the specific periods of gestation recommended for use for tlle presence of the organism as appropriate. Moreover,
on dle label. An exclusion statement will be required for studies may be required of the dissemination of the vaccine
those gestation periods not tested. strain in the body, with particular attention being paid to the
The observation period is extended to parturition, to examine predilection sites for replication of the organismo In the case
any harmful effects during gestation or on progeny, unless of live vaccines for well-established zoonotic diseases for
odlerwise justified or specified in a specific monograph. food-producing animals, these studies are obligatory and shall
particularly take into account the persistence of dle strain at
The following protocol is given as an example of an
the injection site.
appropriate test for vaccines.
1-8-3 Increase in virulence Unless otherwise prescribed in
SaJet), in pregnant anintals U se not fewer dlan 8 animals
a specific monograph or, in tlle absence of a specific
per group, at the recommended stage of gestation or at a
monograph, unless odlerwise justified and autllorised, the
range of stages of gestation according to the recommended
following applies. This test is carried out using the master
schedule. Not fewer dlan 8 animals are used for each stage of
seed loto If the quantity of dle master seed lot sufficient for
pregnancy (i.e. 24 animals for 3 trimesters of pregnancy in
performing the test is not available, tlle lowest passage
cattle). Administer to each animal a recommended dose of
material used for dle production that is available in sufficient
the vaccine. If the recommended schedule requires a
quantity may be used. At dle time of inoculation, the animals
2nd dose, administer anodler dose after an interval of at least
in all groups are of an age suitable for recovery of tlle st:rain.
14 days. Unless otherwise prescribed in a specific
Serial passages are carried out in target animals using
monograph, observe the animals at least daily until 1 day
5 groups of animals, lmless there is justification to cany out
after parturition. U nless odlerwise prescribed in a specific
more passages or unless the strain disappears from the test
monograph, or, in the absence of a specific monograph,
animal sooner. 171 vitro propagation may not be used to
unless otherwise justified and authorised, the vaccine
expand the passage inoculum.
complies with tlle test if no animal shows abnormallocal or
systemic reactions or signs of disease, or dies from causes The passages are carried out usillg animals most appropriate
attributable to the vaccine, and if no adverse effects on the to the potential risk being assessed.
pregnancy or the offspring are noted. The initial administration is carried out using the
recommended route of administ:ration most likely to lead to
1-5 RESIDUES
reversion to virulence, using an initial inoculum containing
In dle case of live vaccines for well-established zoonotic the maximum release titre. Afterthis, not fewer than
diseases, the determination of residual vaccine organisms at 4 furdler serial passages tlrrough animals of the target species
the injection site may be required, in addition to tlle studies are undertaken. The passages are undertaken by the route of
of dissemination described below. administ:ration most likely to lead to reversion to virulence.
1-6 ADVERSE EFFECTS ON IMMUNOLOGICAL If the properties of dle strain allow sequential passage via
FUNCTIONS natural spreading, tllis method may be used, otherwise
Where the product might adversely affect the immune passage as described in each specific monograph is carried
response of the animal to which the product is administered out and the micro-organismsthat have been recovered at the
or of its progeny, suitable tests on the immunological final passage are tested for increase in virulence. For the first
functions are carried out. 4 groups, a minimum of 2 animals is used for mammalian
vaccines, and a 'minimum of 5 birds is used for avían
1-7 ADVERSE EFFECTS FROM INTERACTIONS
vaccines. The last group consist of a minimum of
Studies are undertaken to show a lack of adverse effect on
8 mammals or 10 birds. At each passage, the presence of
the safety of the product when simultaneous administration is
living vaccine-derived micro-organisms in the material used
recommended or where administration of the product is
for passage is demonstrated. Care must be taken to avoid
contamination by micro-organisms from previous passages.
2016 Appendix XV IZ (Vet) 2. Vet-A19

When the micro-organism is not recovered from any grading and rejects at the processing planto For vaccines for
intelmediate in vivo passage, repeat the passage in 10 animals use in laying birds or in birds that may be maintained to lay,
using in vivo passaged material from the last passage in which the effect of the vaccine on laying performance and
the micro-organism was recovered. The micro-organism hatchability is investigated, as appropriate.
recovered is used as the inoculum for the next passage. If the
3 ECOTOXICITY
target micro-organism is not recovered, the experiment is
An assessment is made of tlle potential harrnful effects of the
considered to be completed with the conclusion that the
product for the environment and any necessary precautionary
target micro-organism does not show an increase in
measures to reduce such risks are identified. The likely
virulence.
degree of exposure of the environment to the product is
General clinical observations are made during the study. assessed, taking into account: the target species and mode of
Animals in the last group are observed for 21 days unless administration; excretion of tlle product; and disposal of
otherwise justified. These observations include all relevant unused producto If these factors indicate that there will be
parameters typical for the disease that could indicate increase significant exposure of the environment to the product, the
in virulence. Compare the clinical signs and other relevant potential ecotoxicity is evaluated, taking into account the
parameters with those observed in the animals used in the properties of the product.
test for safety of the administration of 1 dose (section 1-1).
If the last group of animals shows no evidence of an increase
in virulence, further testing is not required. Otherwise,
material used for the 1st passage and the microorganisms K (Vet) 2. Evaluation of Efficacy of
recovered at the final passage level are used in a separate
experiment using at least 8 animals per group for mammal
Veterinary Vaccines and Immunosera
vaccines and at least 10 birds per group for avian vaccines, to (Ph Bur general texts 5.2.7)
compare directly the clinical signs and other relevant The term 'product' means either a vaccine or an
parameters. This study is carried out using the route of immunoserum throughout the texto
administration that was used for previous passages. During development of the product, tests are carried out to
An alternative route of administration may be used if demonstrate that the product is efficacious when
justified. administered by each of the recommended routes and
Unless otherwise justified and authorised, the product methods of administration and using the recommended
complies with the test if no animal dies or shows signs schedule to animals of each species and category for which
attributable to the vaccine strain and no indication of use of the product is to be recommended. The type of
increased virulence is observed in the animals of the last efficacy testing to be carried out varies considerably
group. dependil1g on the particular type of producto
1-8-4 Biological properties of the vaccine strain Other As part of tests carried out during development to establish
tests may be necessmy to detelmine as precisely as possible efficacy, the tests described in the Production section of a
the intrinsic biological properties of tlle vaccine strain (for monograph may be carried out; the following must be taken
example, neurotropism). For vector vaccines, evaluation is into account.
made of the risk of changing the tropism or virulence of tlle The dose to be used is that quantity of tlle product to be
strain and where necessmy specific tests are carried out. Such recommended for use and containing the minimum titre or
tests are systematically carried out where tlle product of a potency expected at the end of the period of validity.
foreign gene is incorporated into the strain as a structural For live vaccines, use vaccine containing virus/bactetia at the
protein. most attenuated passage level tllat will be present in a batch
1-8-5 Recombination or genomic reassortment of strain ofvaccine.
The probability of recombination or genomic reassortment For immunosera, if appropriate, the dose tested also contains
with field or other strains shall be considered. minimum quantities of irnmunoglobulin or gammaglobulin
2 FIELD STUDIES and/or total protein.
Results from laboratory studies shall normally be The efficacy evidence must support all tlle claims being
supplemented with supportive data from field studies. made. For example, claims for protection against respiratory
Provided that laboratory tests have adequately assessed the disease must be suppOlted at least by evidence of protection
safety and efficacy of a product under experimental from clinical signs of respiratory disease. Where it is c1aimed
conditions using vaccines of maximum and minimum titre or that there is protection from infection tllis must be
potency respectively, a single batch of product may be used demonstrated using re-isolation techniques. If more than one
to assess both safety and efficacy under field conditions. c1aim is made, suppOlting evidence for each c1aim is
In these cases, a typical routine batch of intermediate titre 01' required.
potency may be used. Vaccines The infiuence of passively acquired and maternally
For food-producing mammals, the studies include derived antibodies on the efficacy of a vaccine is adequately
measurement of the body temperatures of a sufficient evaluated. Any c1aims, stated 01' implied, regarding onset and
number of animals, before and after administration of the duration of protectión shall be supported by data from trials.
product; for other mammals, such measurements are carried Claims related to duration of irnmunity are supported by
out if the laboratory studies indicate that there might be a evidence of protection. The test model described under
problem. The size and persistence of any local reaction and Irnmunogenicity and/or Potency is not necessarily used to
the proportion of animal s showing local or systemic reactions support c1aims regarding the_ duration of irnmunity afforded
are recorded. Performance measurements are made, where by a vaccine.
appropriate.
The efficacy of each of the components of multivalent and
Performance measures for broilers include weekly mortality, combined vaccines shall be demonstrated using the combined
feed conversion ratios, age at slaughter and weight, down vaccine.
Vet-A20 Appendix XV IZ (Vet) 3. 2016

lmmunosera Particular attention must be paid to providing and systemic reactions is then used as part of the operation
supporting data for the efficacy of the regime that is to be procedure for the batch safety test to evaluate acceptable and
recommended. For example, if it is recommended that the unacceptable reactions.
immunoserum needs only to be administered once to achieve Amount to be administered in the test In the tests, 'dose'
a prophylactic or therapeutic effect then this must be means the quantity of the immunoserum to be recommended
demonstrated. Any claims, stated or implied, regarding onset for use and containing the titre or potency within the limits
and duration of protection or therapeutic effect must be specified for production batches. The amount to be
supported by data from trials. For example, the duration of administered in the test is usually defined in a number of
the protection afforded by a prophylactic dose of an doses.
antiserum must be studied so that appropriate guidance for Route of administration The immunoserum is
the user can be given on the label. administered by a recommended route. In principIe,
Studies of immunological compatibility are undertaken when preference should be given to the application route with the
simultaneous administration is recommended or where it is a higher possibility to detect reactions.
part of a usual administration schedule. Wherever a product Target animal species and category of animals Use
is recommended as part of an administration scheme, the animal s of the most sensitive species and of the minimum
priming or booster effect or the contribution of the product age recommended for administration of the immunoserum,
to the efficacy of the scheme as a whole is demonstrated. unless otherwise justified and authorised.
LABORATORY TESTS Animal numbers The number of animals to be used for the
In principIe, demonstration of efficacy is undertaken under test is prescribed in the general monograph l11l111UnOSera for
well-controlled laboratory conditions by challenge of the vetenna1Y use (0030).
target animal under the recommended conditions of use. Identification of animals Unless otherwise justified and
In so far as possible, the conditions under which the authorised, all animal s are marked in a suitable way to ensure
challenge is carried out shall mimic the natural conditions for individual documentation of data for the whole observation
infection, for example with regard to the amount of challenge periodo
organism and the route of administration of the challenge. Observation period Where objective criteria such as body
Vaccines Unless otherwise justified, challenge is carried out temperature are to be recorded as described below, the
using a strain different from the one used in the production animals are examined ánd observed for at least 3 days prior
of the vaccine. to administration of the immunoserum. After adminisu"ation
If possible, me immune mechanism (cell-mediated/humoral, of the immunoserum, the animals are observed and
local/general, classes of immunoglobulin) that is initiated examined at least once every day for a period of at least
after the administration of the vaccine to target animal s shall 14 days for signs of local and systemic reactions. On the day
be determined. of administration of the immunoserum, at least 1 additional
bnmunosera Data are provided from measurements of the inspection is necessary after 4 h or at intervals as specified in
antibody levels achieved in the target species after the monograph. Where there is a 2nd administration of the
administration of the product, as recommended. Where immunoserum the period usually ends 14 days after the
. suitable published data exist, references are provided to 2nd administration .
relevant published literature on protective antibody levels and Local and systemic reactions Animals showing severe
challenge studies are avoided. abnormal local or systemic reactions are euthanised. All dead
Wher¡; challenges are required, these can be given before or animals undergo a post-mortem with macroscopic
after administration of the product, in accordance with the examination. Additional microscopic and microbiological
indications and specific claims to be made. investigations may be indicated.
The animals are observed and examined for signs of local
FIELD TRIALS
and systemic reactions. Where it is known to be a useful
In general, results from laboratory tests are supplemented
indicator, other criteria are recorded, such as body
with data from field trials, canied out, unless otherwise
temperature, body mass, other performance measurements
justified, with untreated control animals. Provided that
and food intake.
laboratory tests have adequately assessed the safety and
efficacy of a product under experimental conditions using Local reactions As far as appropriate and possible, the size
vaccines of maximum and minimum titre or potency and persistence of any local reaction (including incidence of
respectively, a single batch of product could be used to assess painful reactions) and the proportion of animals showing
both safety and efficacy under field conditions. In these local reactions are recorded.
cases, a typical routine batch of intelmediate titre or potency Systemic reactions Body temperature and, if appropriate,
may be used. Where laboratory trials cannot be supportive of body mas s are documented as general indicators of systemic
efficacy, the perfOlmance of field trials alone may be effects of administration of the immunoserum. In addition,
acceptable. all clinical signs are recorded.
Body temperature For mammals, the studies include
measurement of body temperature during the observation
periodo The body temperatures are recorded beginning at
K (Vet) 3. Evaluation of Safety of each least 3 days before administration of the immunoserum, at
Batch of Immunosera for Veterinary Use the time of administration, 4 h after and at suitable intervals.
The body temperature before administration of the
(Ph Bur general texts 5.2.9)
immunoseruin has to be within the physiological range.
Definition of abnormal reactions During development At least for immunosera where a significant increase in body
studies, the type and degree of reactions expected after temperature may be expected or where an increase in body
administration of the immunoserum are defined in the light temperature is specified in an individual monograph, it is
of safety testing. This definition of normal or abnormal local
2016 Appendix XV IZ (Vet) 3. Vet-A21

recommended to use the mean temperature of the days


before administration of the immunoserum (e.g. day -3 to
day O) as the baseline temperature to have clear guidance for
evaluation of the test.
Body mass and Jood intake Where it is known to be a
reliable and useful indicator of safety, for example in young
growing animals, the body mas s is measured and
documented shortly before administration of the
immunoserum and during the observation periodo The food
intake is monitored and documented as an indicator of the
effect of administering the immunoserum. In most cases, it
will be sufficient to record the daily ration has been
consumed or partly 01' wholly rejected but, in sorne cases it
may be necessary to record the actual weight of food
consumed, if this is a relevant indicator of the safety of the
immunoserum.
Clinical signs AH expected and unexpected clinical signs of
a general nature are recorded, including changes in health
status and behaviour changes.
Score sheets The score sheets are prepared for each
immunoserum in the light of expected signs. All parameters
and data are recorded in score sheets. The score sheets
contain general parameters but are also adapted for each kind
of immunoserum to list clinical signs that might be more
evident for a given immunoserum.
Criteria for repeating the test If an abnOlmal sign occurs,
the responsible veterinarian determines, based on post-
mortem examination if necessary, whether this was due to
the immunoserum 01' noto If it is not clear what caused the
abnormal sign 01' where an animal is withdrawn for reasons
unrelated to the immunoserum, the test may be repeated.
If in the 2nd test there is the same abnormal sign as in the
1sr test, the immunoserum does not comply with the test.
Any treatment administered to an animal during the
observation period is recorded. If tl1e treatment may interfere
with the test, the test is invalido
Vet-A22 Appendix XVI B (Vet) 3. 2016

INCUBATION CONDITIONS
Appendix XVI Incubate liquid media in tightly stoppered containers at
35-38 oc. Incubate solid media in microaerophilic conditions
(nitrogen containing 5-10 per cent of carbon dioxide and
8 (Vet) 3. Test tor Absence of sufficient humidity to prevent desiccation of the agar surface)
Mycoplasmas at 35-38 oc.
(Ph. Eur. method 2.6. 7 as applied to vaccines for human use) NUTRITIVE PROPERTIES
Cany out the test for nutritive propel1ies for each nezu batch of
Where the test for mycoplasmas is prescribed for a master
mediu11l. Inoculate the chosen media with the appropriate test
cell bank, for a working cell bank, for a virus seed lot or for
micro-organisms; use not more than 100 CFU (colony-
control cells, both the culture method and the indicator cell
forming units) per 60 mm di ame ter plate containing 9 mL of
culture method are used. Where the test for mycoplasmas is
solid medium and per 100 mL container of liquid medium;
prescribed for a virus harvest, for a bulk vaccine or for the
use a separate plate and container for each species of micro-
finallot (batch), the culture method is used. The indicator
organismo Incubate the media and make sub cultures from
cell culture method may also be used, where necessary, for
0.2 mL of liquid medium to solid medium at the specified
screening of media.
intervals (se e below under Test for mycoplasmas in the
Nucleic acid amplification techniques (NAT) may be used as product to be examined). The solid medium complíes with
an alternative to one or both of the other methods after the test if adequate growth is found for each test micro-
suitable validation. organism (growth obtained does not differ by a factor greater
Culture method than 5 from the value calculated with respect to the
inoculum). The liquid medium complies with the test if
CHOICE OF CULTURE MEDIA
growth on agar plates subcultured from the broth is found
The test is carried out using a sufficient number of both solid
for at ·least 1 sub culture for each test micro-organismo
and liquid media to ensure growth in the chosen incubation
conditions of small numbers of mycoplasmas that may be INHIBITORY SUBSTANCES
presentin the product to be examined. Liquid media must The test for inhibitory substances is carried out once for a
contain phenol red. The range of media chosen is shown to given product and is repeated whenever there is a change in
have satisfactory nutritive properties for at least the micro- production method that may affect the detection of
organisms shown below.The nutritive properties of each new mycoplasmas.
batch of medium are verified for the appropriate micro- To demonstrate absence of inhibitOly substances, carry out
organisms in the listo When testing for mycoplasmas in the the test for nutritive properties in the presence and absence
product to be examined, at least 1 of the following species of tlle product to be examined. If growth of a test micro-
will be included as a positive control: organism occurs more than 1 sub culture sooner in the
- Acholeplasma laidlazuii (vaccines for human and veterinary absence of the product to be examined than in its presence,
use where an antibiotic has been used during production); or if plates direct1y inoculated with the product to be
- lvIycoplas11la gallisepticum (where avian material has been examined have fewer tllan 1/5 of the number of colonies of
used during production or where the vaccine is intended those inoculated without the product to be examined,
for use in poultry); inhibitory substances are present and they must be
- Mycoplasma hyorhinis (non-avian veterinary vaccines); neutralised or their effect otherwise countered, for example
- lvIycoplasma orale (vaccines for human and veterinary use); by passage in substrates not containing inhibitors or dilution
- Mycoplasma pneumoniae (vaccines for human use) or other in a larger volume of medium before the test. If dilution is
suitable species of D-glucose fermenter such as used, larger medium volumes may be used or the inoculum
M ycoplasma fermentans; volume may be divided among several 100 mL fiasks.
- Mycoplasma synoviae (where avían matelial has been used The effectiveness of the neutralisation or other process is
during production or where the vaccine is intended for checked by repeating the test for inhibit01Y substances after
use in poultry). neutralisation.
The test strains are field isolates having undergone a limited TEST FOR MYCOPLASMAS IN THE PRODUCT TO BE
number of sub cultures (not more than 15), and are stored EXAMINED
frozen or freeze-dried. After cloning, the strains are identified Inoculate 10 mL of the product to be examined per 100 mL
as being of the required species by compmison with type of each liquid medium. If it has been found that a significant
cultures, for example: pH change occurs upon the addition of the product to be
examined, the liquid medium is restored to its Oliginal pH
A. laidla'luii NCTC10116 CIP 75.27 ATCC 23206
value by the addition of a solution of either sodium
M. gallisepticum NCTC10115 CIP 104967 ATCC 19610
hydroxide or hydrochlOlic acid. Inoculate 0.2 mL of the
M. fermentans NCTC 10117 CIP 105680 ATCC 19989
product to be examined on each plate of each solid medium.
lvI. hyorhinis NCTC10130 CIP 104968 ATCC 17981
Incubate liquid media for 20-21 days. Incubate solid media
M. orale NCTC 10112 CIP 104969 ATCC 23714
for not less than 14 days, except those corresponding to the
M. pneumoniae NCTC10119 CIP 103766 ATCC 15531
20-21 day sub culture, which ?re incubated for 7 days. At the
M. synoviae NCTC 10124 CIP 104970 ATCC 25204
same time incubate an uninoculated 100 mL portion of each
liquid medium and agar plates, as a negative control.
Acholeplasma laidlazuii BRP) lvIycoplasma fermemans BRP) On days 2-4 after inoculation, sub culture each liquid
Mycoplasma hyorhinis BRP) Mycoplasma orale BRP and medium byinoculating 0.2 mL on at least 1 plate of each
Mycoplas711a synoviae BRP are suitable for use as low-passage solid medium. Repeat the procedure between the 6th and 8th
reference strains. days, again between the 13 th and 15 th days and again
between the 19 th and 21 st days of the test. Observe the liquid
media every 2 or 3 days and if a colour change occurs,
subculture. If a liquid medium shows bactelial or fungal
2016 Appendix XVI B (Vet) 3. Vet-A23

contamination, the test is invalido The test is valid if at least Solid medium
1 plate per medium and per inoculation day can be read. Beef heart infusion broth (1) 90.0 mL
Include in the test positive controls prepared by inoculation Agar, purified (3) 1.4 g
of not more than 100 CFU of at least 1 test micro-organism
on agar medium or into broth medium. Where the test for
Adjust to pH 7.8, sterilise by autoclaving then add:
mycoplasmas is carried out regularly and where possible, it is
recommended to use the test micro-organisms in regular Essential vitamins (2) 0.025 mL
rotation. The test micro-organisms used are those listed Glucose monohydrate (500 gIL solution) 2.0 mL
under Choice of culture media. Swine serum (unheated) 12.0 mL
~-Nicotinamide adenine dinucleotide (lO gIL 1.0 mL
INTERPRETATION OF RESULTS
solution)
At the end of the prescribed incubation period, examine all
Cysteine hydrochloride (10 gIL solution) 1.0 mL
inoculated solid media microscopically for the presence of
Phenol red (0.6 giL solution) 5.0 mL
mycoplasma colonies. The product complies with the test if
Penicillin (20 000 IU/rnL) 0.25 mL
growth of typical mycoplasma colonies has not occurred.
The product does not comply with the test if growth of
typical mycoplasma colonies has occurred on any of the solid FRIIS MEDIA (RECOMMENDED FOR THE DETECTION OF
media. The test is invalid if 1 or more of the positive controls NON-A VIAN MYCOPLASMAS)
do not show growth of mycoplasmas on at least 1 sub culture
Liquid medium
plateo The test is invalid if 1 01' more of the negative controls
show growth of mycoplasmas. If suspect colonies are Hanks' balanced salt solution (modified) (4) 800 mL
observed, a suitable validated method may be used to Distilled water 67 rnL
Brain heart infusion (5) 135 mL
determine whether they are due to mycoplasmas.
PPLO Broth (6) 248 mL
The follozuing section is published for information.
Yeast extract (170 gIL) 60 mL
BacÍ1:racin 250 mg
Recommended media for the culture method
Meticillin 250 mg
The following media are recommended. Other media may be
Phenol red (5 giL) 4.5 mL
used, provided that their ability to sustain the growth of
Horse serum 165 mL
mycoplasmas has been demonstrated on each batch in the
Swine serum 165 mL
presence and absence of the product to be examined.
HA YFLICK MEDIA (RECOMMENDED FOR THE GENERAL
DETECTION OF MYCOPLASMAS)
Adjust to pH 7.40-7.45.
Liquid medium Solid medium
Beef heart infusion broth (1) 90.0 mL Hanks' balanced salt solution (modified) (4) 200 mL
Horse serum (unheated) 20.0 mL D EAE-dextran 200 mg
Yeast extract (250 giL) 10.0 mL Agar, purified (3) 15.65 g
Phenol red (0.6 giL solution) 5.0 mL
Penicillin (20 000 IU/rnL) 0.25 mL
Deoxyribonucleic acid (2 gIL solution) 1.2 mL
Mix well and sterilise by autoclaving. Cool to 100 ce. Add to
1740 mL of liquid medium as desclibed above.
(1) Beef heart infusion brotlz
Adjust to pH 7.8.
Beef heart (for preparation of the infusion) 500 g
Solid medium Peptone 10 g
Prepare as described above replacing beef heart infusion Sodium chloride 5 g
broth by beef heart infusion agar containing 15 gIL of agar. Distilled water to 1000 mL
FREY MEDIA (RECOMMENDED FOR THE DETECTION OF M.
SYNOVIAE) Sterilise by autoclaving.
(2) Essential vitamins
Liquid medium Biotin 100 mg
Beef heart infusion broth (1) 90.0 mL Calcium pantothenate 100 mg
Essential vitamins (2) 0.025 mL Choline chloride 100 mg
Glucose monohydrate (500 gIL solution) 2.0 mL Folic acid 100 mg
Swine serum (inactivated at 56 oC for 30 min) 12.0 mL i-Inositol 200 mg
~- Nicotinamide adenine dinucleotide (10 giL 1.0 mL Nicotinamide 100 mg
solution) Pyridoxal hydrochloride 100 mg
Cysteine hydrochloride (10 gIL solution) 1.0 mL Riboflavine 10 mg
Phenol red (0.6 giL solution) 5.0 mL Thiamine hydrochlOlide 100 mg
Penicillin (20 000 IU/rnL) 0.25 mL Distilled water to 1000 mL

Mix the solutions of ~-nicotinamide adenine dinucleotide and


cysteine hydrochloride and after 10 min add to the other
ingredients. Adjust to pH 7.8.
Vet-A24 Appendix XVI B (Vet) 3. 2016

(3) Agar, pur/fied NI. hyorhinis ATCC 29052


A highly refined agar for use in microbiology and M. orale NCTC 10112 CIP 104969 ATCC 23714
immunology, prepared by an ion-exchange procedure that
results in a product having superior purity, clarity and gel The cells are suitable if both reference strains are detected.
strength. It contains about:
The indicator cells must be subcultured without an antibiotic
Water 12.2 per cent before use in the test.
Ash 1.5 per cent
TEST METHOD
Acid-insoluble ash 0.2 per cent
Chlorine 1. Seed the indicator cell culture at a suitable density (for
O
Phosphate (calculated as P 2 0 S) 0.3 per cent example, 2 x 104 to 2 x 105 cells/mL, 4 x 10 3 to 2.5
Total nitro gen 0.3 per cent x 104 cells/cm2 ) that will yield confiuence after 3 days of
Copper 8ppm growth. Inoculate 1 mL of the product to be examÍned
Iron 170 ppm into the cell culture vessel and incubate at 35-38 oC.
Calcium 0.28 per cent 2. After at least 3 days of incubation, when the cells have
Magnesium 0.32 per cent grown to confiuence, make a sub culture on cover slips in
suitable containers 01' on sorne other surface (for example,
chambered slides) suitable for the test procedure. Seed the
(4) Hanks' balanced salt soludon (nzodified)
cells at low density so that they reach 50 per cent
Sodium chloride 6.4 g confiuence after 3-5 days of incubation. Complete
Potassium chloride 0.32 g confiuence impairs visualisation of mycoplasmas after
Magnesium sulfate heptahydrate 0.08 g staining and must be avoided.
Magnesium chloride hexahydrate 0.08 g
3. Remove the medium and rinse the indicator cells with
Calcium chloride, anhydrous 0.112 g
phosphate buffered saline pH 7.4 R, then add a suitable
Disodium hydrogen phosphate dihydrate 0.0596 g
fixing solution (a freshly prepared mixture of 1 volume of
Potassium dihydrogen phosphate, anhydrous 0.048 g
glacial acetic acid R and 3 volumes of methanol R is
Distilled water to 800 mL
suitable when bisbenzimide R is used for staining).
4. Remove the fixing solution and wash the cells with sterile
(5) Brain heart infllsio1Z 'Water R. Dry the slides completely if they are to be stained
Calf-brain infusion 200 g more than 1 h later (particular care is needed for staining
Beef-heart infusion 250 g of slides after drying owing to artefacts that may be
Proteos e peptone 10 g produced).
Glucose monohydrate 2g 5. Add a suitable DNA stain and allow to stand for a
Sodium chloride 5g suitable time (bisbenzimide 'Working solution R and a
Disodium hydrogen phosphate, anhydrous 2.5 g standing time of 10 min are suitable).
Distilled water to 1000 mL
6. Remove the stain and rinse the monolayer wid1 'Water R.
7. Mount each coverslip, where applicable (a mixture of
(6) PPLO broth
equal volumes of glycerol R and phosphate-citrate buffer
Beef-heart infusion 50 g solution pH 5.5 R is suitable for mounting). Examine by
Peptone 10 g fiuorescence (for bisbenzimide stain a 330 nm/380 nm
Sodium chloride 5g excitation filter and an LP 440 nm barrier filter are
Distilled water to 1000 mL suitable) at 400 x magnification .01' greater.
8. Compare the microscopic appearance of the test cultures
with that of the negative and positive controls, examining
Indicator cell culture method for extranuclear fiuorescence. Mycoplasmas produce
Cell cultures are stained with a fiuorescent dye that binds to pinpoints 01' filaments over the indicator cell cytoplasm.
DNA. Mycoplasmas are detected by their characteristic They may also produce pinpoints and filaments in the
particulate or filamentous pattern of fiuorescence on the cell intercellular spaces. Multiple microscopic fields are
surface and, if contamination is heavy, in surrounding areas. examined according to the protocol established during
Mitochondria in the cytoplasm may be stained but are readily validation.
distinguished from mycoplasmas.
INTERPRETATION OF RESULTS
If for viral suspensions the interpretation of results is affected The product to be examÍned complies with the test if
by marked cytopathic effects, the virus may be neutralised fiuorescence typical of mycoplasmas is not presento The test
using a specific antiserum that has no inhibitory effects on is invalid if the positive controls do not show fiuorescence
mycoplasmas or a cell culture substrate that does not allow typical of mycoplasmas. The test is invalid if the negative
growth of the virus may be used. To demonstrate the controls show fiuorescence typical of mycoplasmas.
absence of inhibitory effects of serum, carry out the positive
control tests in the presence and absence of the antiserum. Nucleic acid amplificatiori techniques (NAT)
VERIFICATION OF THE SUBSTRATE NAT (2.6.21) may be used for detection of mycoplasmas by
Use Vero cells 01' another cell culture (for example, the amplification of nucleic acids extracted from a test sample
production cellline) that is equivalent in effectiveness for with specificprimers that reveal the presence of the target
detecting mycoplasmas. Test the effectiveness of the cells to nucleic acid. NAT indicate the presence of a particular
be used by applying the procedure shown below and nucleic acid sequence and not necessarily the presence of
inoculating not more than 100 CFU 01' CFU -like micro- viable mycoplasmas. A number of different techniques are
organisms of suitable reference strains of M. hyorhinis and M. available. This general chapter does not prescribe a particular
orale. The following strains have been found to be suitable: method for the test. The procedure applied must be
2016 Appendix XVI B (Vet) 3. Vet-A25

validated as described, taking account of the guidelines The extemal negative control contains no target sequence
presented at the end of this section. Where a commercial kit but does not necessarily represent the same matrix as the test
is used, certain elements of the validation may be carried out article.
by the manufacturer and information provided to the user INTERPRETATION OF RESULTS
but it must be remembered that full information on the The primers used may also amplify non-mycoplasmal
primers may not be available and that production of the kit bacterial nucleic acid, leading to false positive results.
may be modified or discontinued. Procedures are established at the time of validation for
NATare applied where prescribed in a monograph. They dealing with confirmation of positive results, where necessaly.
may also be used instead of the culture method and the The follozuing section is published for information.
indicator cell culture method after suitable validation.
Direct NAT Can be applied in the presence of cytotoxic Validation of nucleic acid amplification techniques
material and where a rapid method is needed. (NAT) for the detection of mycoplasmas : guidelines
Cell-culture enrichment followed by NAT The test 1 SCOPE
sample and a suitable cell substrate (as described under the Nucleic acid amplification techniques (NAT) are either
indicator cell-culture method) are cultured together for a qualitative or quantitative tests for the presence of nucleic
suitable period; the nucleic acids are then extracted from acid. For the detection of mycoplasma contamination of
cells and supernatant and used for detection by NAT. various samples such as vaccines and cell substrates,
VALIDATION qualitative tests are adequate and may be considered to be
Reference standards are required at various stages during limit tests.
validation and for use as controls during routine application These guidelines describe methods to validate qualitative
of the test. The reference standards may be mycoplasmas or nucleic acid amplification analytical procedures for assessing
nucleic acids. mycoplasma contamination. They may also be applicable for
For validation of the limit of detection, the following species real-time NAT used as limit tests for the control of
represent an optimal selection in terms of the frequency of contaminants.
occurrence as contaminants and phylogenetic relationships: The 2 characteristics regarded as the most important for
- A. laidlazuii; validation of the analytical procedure are the specificity and
- M. fermentans)' the detection limito In addition, the robustness of the
- M. hyorhinis (where cell-culture enrichment is used, a analytical procedure should be evaluated.
fastidious strain such as ATCC 29052 is included); For the purpose of this document, an analytical procedure is
- NI. orale; defined as the complete procedure from extraction of nucleic
- M. pneumoniae or M. gallisepticum; acid to detection of the amplified products.
- M. synoviae (where there is use of or exposure to avian Where commercial kits are used for part or all of the
material during production); analytical procedure, documented validation points already
- Mycoplasma arginini; covered by the kit manufacturer can replace validation by the
- Spiroplasma citri (where there is use of or exposure to user. Nevertheless, rile performance of rile kit with respect to
insect or plant material during production). its intended use has to be demonstrated by rile user
Demonstration of specificity requires the use of a suitable (e.g. detection limit, robustness, cross-detection of other
range of bacterial species other than mycoplasmas. Bacterial classes of bacteria).
genera with close phylogenetic relation to mycoplasmas are NAT may be used as:
most appropriate for this validation; these include Clostridium, - a complementary test (for example, for cytotoxic viral
Lactobacillus and Streptococcus. suspensions) or for in-process control purposes;
Comparability studies for use of NAT as an alternative - an altemative method to replace an official method
method For each mycoplasma test species: (indicator cell culture meril0d or culture method).
- as an alternative to the culture method: the NAT test These guidelines will ~hus separate these 2 objectives by
system must be shown to detect 10 CFU/mL; presenting first a guideline for the validation of the NAT
- as an alternative to the indicator cell culture method: the themselves, and second, a guideline for a comparability study
NAT test system must be shown to detect 100 CFU/mL; between NAT and official methods.
or an equivalent limit of detection in terms of the number of 2 GUIDELINE FOR MYCOPLASMA NAT VALIDATION
copies of mycoplasma nucleic acid in the test sample (using 3 parameters should be evaluated: specificity, detection limit
suitable reference standards of mycoplasma nucleic acid). and robustness.
CONTROLS 2-1 Specificity Specificity is the ability to unequivocally
Internal controls Internal controls are necessary for routine assess target nucleic acid in the presence of components that
verification of absence of inhibition. The internal control may may be expected to be presento
contain the primer binding-site, or sorne other suitable
The specificity of NAT is dependent on the choice of
sequence may be used. It is preferably added to the test
primers, the choice of probe (for analysis of the final
material before isolating the nucleic acid and therefore acts as
product) and the stringency of the test conditions (for both
an overall control (extraction, reverse transcription,
the amplification and detection steps).
amplification, detection).
The ability of the NAT to detect a large panel of
External controls The external positive control contains a
mycoplasma species will depend on the choice of pri.mers,
defined number of target-sequence copies or CFUs from 1 or
probes and method parame~ers. This ability should be
more suitable species of mycoplasma chosen from those used
demonstrated using characterised reference panels
during validation of the test conditions. 1 of the positive
(e.g. reference strains provided by the EDQM). Since NAT
controls is set close to the positive cut-off point to
systems are usually based on a mix of primers, the theoretical
demonstrate that the expected sensitivity is achieved.
analysis of primers and probes by comparison with databases
Vet-A26 Appendix XVI B (Vet) 3. 2016

is not recommended, because interpretation of the results preliminary cut-off point. The concentration of mycoplasmas
may be quite complex and may not reflect the experimental (CFUs or copies) dlat can be detected in 95 per cent of test
results. runs can then be calculated using an appropriate statistical
Moreover, as it is likely that the primers will detect other evaluation.
bacterial species, the potential cross-detection should be These results may also serve to evaluate the variability of the
documented in the validation study. Bacterial genera such as analytical procedure.
gram-positive bacteria with close phylogenetic relation to 2-3 Robustness The robustness of an analytical procedure
mycoplasmas are most appropriate for this validation; these is a measure of its capacity to remain unaffected by small but
include Clostridiu11l) Lactobacillus and Streptococcus. However, deliberate variations in method parameters, and provides an
this is not an exhaustive list and species to be tested will indication of its reliability during nOlmal usage.
depend on the theoretical ability (based on primers/probes The evaluation of robustness should be considered during
sequences) of the NAT system to detect such other species.
the development phase. It should show the reliability of the
Based on the results from dus validation of the specificity, if analytical procedure with respect to deliberate variations in
a gap in the specificity of the method is identified (such as method parameters. Fol' NAT, small variations in the
detection of non-mycoplasmal bacterial nucleic acid), an method parameters can be crucial. However, the robustness
appropriate strategy must be proposed in the validation study of the method can be demonstrated during its development
to al10w interpretation of positive results on a routine basis. when smaH variations in the concentrations of reagents
For example, a second test may be perfonned using an (e.g. MgCI 2 , primel's 01' deoxyribonucleotides) are tested.
altemative method without this specificity gap or using an Modifications of extraction kits 01' extraction procedures as
official method. well as diffel'ent thermal cycler types may also be evaluated.
2-2 Detection limito The detection limit of an individual Finally, robustness of the method can be evaluated through
analytical procedure is the lowest amount of target nucleic collabol'ative studies.
acid in a sample that can be detected but not necessarily
3 GUIDELINE FOR COMPARABILITY STUDY
quantitated as an exact value.
NAT may be used instead of official methods (indicator cell
For establishment of the detection limit, a positive cut-off culture method and/ol' culture medlod). In this case a
point should be determined for the nucleic acid amplification compal'ability study should be carried out. This comparability
analytical procedure. The positive cut-off point (as defined in study should include mainly a comparison of the respective
general chapter 2.6.21) is the minimum number of target detection limits of the altemative method and official
sequence copies per volume of sample that can be detected methods. However, specificity (mycoplasma panel detected,
in 95 per cent of test runs. This positive cut-off point is putative false positive results) should also be considel'ed.
influenced by the distribution of mycoplasmal genomes in the
For the detection limit, acceptability criteria are defined as
individual samples being tested and by factors such as
follows:
enzyme efficiency, and can result in different 95 per cent cut-
- if the alternative method is proposed to replace the
off values for individual analytical test runs.
culture method, the NAT system must be shown to
To determine dle positive cut-off point, a dilution series of detect 10 CFU/mL for each mycoplasma test species
characterised and calibrated (either in CFUs or nucleic acid described in paragraph 2-2;
copies) in-house working strains or EDQM standards should - if the alternative method is proposed to replace the
he tested on different days to examine variation between test indicator cell culture ínethod, the NAT system must be
runs. shown to detect 100 CPU/mL for each mycoplasma test
For validation of the limit of detection, dle following species species described in paragraph 2-2
represent an optimal selection in terms of the frequency of For both cases, suitable standards calibl'ated for the numbel'
occurrence as contaminants and phylogenetic relationships: of nucleic acid copies and dle number of CPU s may be used
- A. laidlawii; for establishil1g that these acceptability criteria are reached.
- M. fermentans; The l'elation between CPU s and nucleic acid copies for the
- M. hyorhinis; refel'ence preparations should be pl~eviously established to
- M. orale; compare the pel'fOlmance of the altemative NAT method
- M. pneu1110niae or NI. gallisepticul1l; with the pel'fonnance of the official methods.
- M. synoviae (where there is use of or exposure to avian
1 of the following 2 strategies can be used to perfonn this
material during production);
comparability study:
- M. arginini;
- pel'fonn the NAT alternative method in parallel with the
- S. citri (where there is use of or exposure to insect or
official method(s) to evaluate simultaneously the detection
plant material during production).
limit of both methods using the same samples of
For each strain, at least 3 independent 10-fold dilution series calibrated strains;
should be tested, with a sufficient number of replicates at - compare the perfonnance of the NAT altemative method
each dilution to give a total number of 24 test results for using previously obtained data from official method
each dilution, to enable a statistical analysis of the results. validation. In this case, calibration of standal'ds used fol'
For example, a laboratory may test 3 dilution series on both validations as well as their stabilities should be
different days with 8 replicates for each dilution, 4 dilution documented cal'efully.
series on different days with 6 replicates for each dilution, or Comparability study reports should describe aH the validation
6 dilution series on different dáys with 4 replicates for each elements described in section 2 (specificity, limit of detection
dilution. In order to keep the number of dilutions at a and variability, as weH as robustness) in order to assess aH
manageable level, a preliminary test should be perfonned to the advantages and/or disadvantages of the altemative NAT
obtain a preliminary value for the positive cut-off point method compared to official methods.
(i.e. the highest dilution giving a positive signal). The range
of dilutions can then be chosen around the predetennined
2016 Appendix XVI B (Vet) 4. Vet-A27

valid unless at least 6 embryos in each group survive beyond


B (Vet) 4. Avian Viral Vaccines: Tests tor the first 24 h after inoculation. Examine macroscopically for
Extraneous Agents in Seed Lots abnormalities all embryos that die more than 24 h after
inoculation, 01' that survive the incubation periodo Examine
(Ph. Eur. method 2.6.24)
also the chorio-allantoic membranes of these eggs for any
GENERAL PROVISIONS abnormality and test the allantoic fluids for the presence of
a) In the following tests, chickens and/or chicken material haemagglutinating agents.
such as eggs and cell cultures shall be derived from Carry out a further embryo passage. Pool separately material
chicken flocks free from specified pathogens (SPF) from live and from the dead and abnormal embryos.
(5.2.2). Inoculate each pool into 10 eggs for each route as described
b) Cell cultures for the testing of extraneous agents comply above, chorio-allantoic membrane material being inoculated
with the requirements for the master cell seed of general onto chorio-allantoic membranes, allantoic fluids into the
chapter 5.2.4. Cell cultures for the production of veterinary allantoic cavity and embryo material into the yolk saco
vaccines, with the exception of the karyotype test and the For eggs inoculated by the allantoic and chorio-allantoic
tumorigenicity test, which do not have to be carried out. routes, candle the eggs daily for 7 days, proceeding and
c) In tests using cell cultures, precise specifications are given examining the material as described above. For eggs
for the number of replicates, monolayer surface areas and inoculated by the yolk sac route, candle the eggs daily for
minimum survival rate of the cultures. Alternative 12 days, proceeding and examining the material as described
numbers of replicates and cell surface are as are possible as above.
well, provided that a minimum of 2 replicates are used, The seed lot complies with the test if no test embryo shows
the total surface area and the total volume of test macroscopic abnormalities or dies from causes attributable to
substance applied are not less than that prescribed here the seed lot and if examination of the chorio-allantoic
and the survival rate requirements are adapted membranes and testing of the allantoic fluids show no
accordingly. evidence of the presence of any extraneous agent.
d) For a freeze-dried preparation, reconstitute using a 2 TEST IN CIllCKEN KIDNEY CELLS
suitable liquido Unless otherwise stated or justified, the Prepare 7 monolayers of chicken kidney cells, each
test substance must contain a quantity of virus equivalent monolayer having an area of about 25 cm2 . Maintain
to at least 10 doses of vaccine in 0.1 mL of inoculum. 2 monolayers as negative controls and treat these in the same
e) If the virus of the seed lot would interfere with the way as the 5 monolayers inoculated with the test substance,
conduct and sensitivity of the test, neutralise the virus in as described below. Remove the culture medium when the
the preparation· with a monospecific antiserum. cells reach confluence. Inoculate 0.1 mL of the test substance
f) Monospecific antiserum and serum of avian origin used onto each of the 5 monolayers. Allow adsorption for 1 h, add
for cell culture or any other purpose, in any of these tests, culture medium and incubate the cultures for a total of at
shall be free from antibodies against and free from least 21 days, subculturing at 4- to 7-day intervals. Each
inhibitory effects on the organisms listed hereafter under 7 passage is made with pooled cells and fluids from all
Antibody specifications for sera used in extraneous agents 5 monolayers after carrying out a freeze-thaw cyc1e. Inoculate
testing. 0.1 mL of pooled material onto each of 5 recently prepared
g) Where specified in a monograph or otherwise justified, if monolayers of about 25 cm2 each, at each passage. For the
neutralisation of the virus of the seed lot is required but last passage, grow the cells also on a suitable substrate so as
difficult to achieve, the tests described below are adapted, to obtain an area of about 10 cm2 of cells from each of the
as required, to provide the necessary guarantees of monolayers for test A. The test is not valid if les s than
freedom from contamination with an extraneous agent. 80 per cent of the monolayers survive after any passage.
h) Other types of tests than those indicated may be used, Examine microscopically all the cell cultures frequently
provided they are at least as sensitive as those indicated throughout the entire incubation period for any signs of
and of appropriate specificity. Nuc1eic acid amplification cytopathic effect 01' other evidence of the presence of
techniques (2.6.21) give specific detection for many contaminating agents in the test substance. At the end of the
agents and can be used after validation for sensitivity and total incubation period, carry out the following procedures.
specificity . A. Fix and stain (with Giemsa or haematoxylin and eosin)
about 10 cm2 of confluent cells from each of the
1 TEST FOR EXTRANEOUS AGENTS USING
5 monolayers. Examine the cells microscopically for any
EMBRYONATED HENS' EGGS
cytopathic effect, inc1usion bodies, syncytial formation, or
Use a test substance, diluted if necessary, containing a
other evidence of the presence of contaminating agents
quantity of neutralised virus equivalent to at least 10 doses of
from the test substance.
vaccine in 0.2 mL of inoculum. Suitable antibiotics may be
added. Inoculate the test substance into 3 groups of B. Drain and wash about 25 cm2 of cells from each of the
10 embryonated hens' eggs as follows: 5 monolayers. Cover these cells with a 0.5 per cent
- group 1: 0.2 mL into the allantoic cavity of each 9- to 11- suspension of washed chicken erythrocytes (using at least
day-old embryonated egg; 1 mL of suspension for each 5 cm2 of cells). Incubate the
- group 2: 0.2 mL onto the chorio-allantoic membrane of cells at 4 oC for 20 min and then wash gently in
each 9- to ll-day-old embryonated egg; phosphate buffered saline pH 7.4. Examine the cells
- group 3: 0.2 mL into the yolk sac of each 5- to 6-day-old microscopically for haemadsorption attributable to the
embryonated egg. presence of a haemadsorbing agent in the test substance.
Candle the eggs in groups 1 and 2 daily for 7 days and the C. Test individual samples -of the fluids from each cell
eggs in group 3 daily for 12 days. Discard embryos that die culture using chicken erythrocytes for haemagglutination
during the first 24 h as non-specific deaths; the test is not attributable to the presence of a haemagglutinating agent
in the test substance.
Vet-A28 Appendix XVI B (Vet) 4. 2016

The test is not valid if there are any signs of extraneous than 3 of the 4 positive controls or fewer than 4 of the 5 test
agents in the negative control cultures. The seed lot complies monolayers or neither of the 2 negative controls survive after
with the test if there is no evidence of the presence of any any passage.
extraneous agent. For the last sub culture, grow the fibroblasts on a suitable
3 TEST FOR AVIAN LEUCOSIS VIRUSES substrate so as to obtain an area of about 10 cm2 of
Prepare at least 13 replicate monolayers of either DF-1 cells confluent fibroblasts from each of the original 11 monolayers
or primary or secondary chick embryo fibroblasts from the for the subsequent test: test about 10 cm2 of confluent
tissues of 9- to 11-day-old embryos that are known to be fibroblasts derived from each of the original 11 monolayers
genetically susceptible to subgroups A, B and J of avian by irnmunostaining for the presence of avian
leucosis viruses and that support the growth of exogenous reticuloendotheliosis virus. The test is not valid if avian
but not endogenous avian leucosis virus es (cells from CIE reticuloendotheliosis virus is detected in fewer than 3 of the
strain chickens are suitable). Each replicate shall have an are a 4 positive control monolayers or in any of the negative
of about 50 cm2 . control monolayers, or if the results for both of the
2 negative control monolayers are inconclusive. If the results
Remove the culture medium when the cells reach confluence.
for more than 1 of the test monolayers are inconclusive then
Inoculate 0.1 mL of the test substance onto each of 5 of the
further subcultures of reserved portions of the fibroblast
replicate monolayers. A1low adsorption for 1 h, and add
monolayers shall be made and tested until an unequivocal
culture medium. Inoculate 2 of the replicate monolayers with
result is obtained.
subgroup A avian leucosis virus (not more than 10 CCID so
in 0.1 mL), 2 with subgroup B avian leucosis virus (not more The seed lot complies with the test if there is no evidence of
than 10 CCID so in 0.1 mL) and 2 with subgroup J avian the presence of avian reticuloendotheliosis virus.
leucosis virus (not more than 10 CCID so in 0.1 mL) as 5 TEST FOR CHICKEN ANAEMIA VIRUS
positive controls. Maintain not fewer than 2 non-inoculated Prepare eleven 20 mL suspensions of the MDCC-MSBI cell
replicate monolayers as negative ccintrols. line or another cellline of equivalent sensitivity in 25 cm2
Incubate the cells for a total of at least 9 days, subculturing cell culture flasks containing about 5 x 105 cells/mL.
at 3- to 4-day intervals. Retain cells from each passage level Inoculate 0.1 mL of the test substance into each of 5 flasks.
and harvest the cells at the end of the total incubation Inoculate 4 of the suspensions with 10 CCID so chicken
periodo Wash cells from each passage level from each anaemia virus as positive controls. Maintain not fewer than
replicate and resuspend the cells at 10 7 cells per millilitre in 2 non-inoculated suspensions. Maintain all the cell cultures
barbital-buffered saline for subsequent testing by a for a total of at least 24 days, subculturing 8 times at 3- to 4-
Complement Fixation for Avian Leucosis (COFAL) test or day intervals. During the subculturing the presence of
in phosphate buffered saline for testing by Enzyme-Linked chicken anaemia virus may be indicated by a metabolic
Immunosorbent Assay (ELISA). Then, carry out 3 cycles of colour change in the infected cultures, the culture fluids
freezing and thawing to rdease any group-specific antigen becoming red in comparison with the control cultures.
and perform a COFAL test or an EUSA test on each extract Examine the cells microscopically for cytopathic effect.
to detect group-specific avian leucosis antigen if presento At this time or at the end of the incubation period, centrifuge
The test is not valid if group-specific antigen is detected in the cells from each flask at low speed and resuspend at
fewer than 5 of the 6 positive control replicate monolayers or about 10 6 cells/mL and place 25 ~lL in each of 10 wells of a
if a positive result is obtained in any of the negative control multi-well slide.Examine the cells by immunostaining.
monolayers, or if the results for both of the 2 negative The test is not valid if chicken anaemia virus is detected in
control monolayers are inconclusive. If the results for more fewer than 3 of the 4 positive controls or in any of the non-
than 1 of the test replicate monolayers are inconclusive, then inoculated controls. If the results for more than 1 of the test
further sub cultures of reserved portions of the fibroblast suspensions are inconclusive, then further sub cultures of
monolayers shall be made and tested until an unequivocal reserved portions of the test suspensions shall be made and
result is obtained. Tf a positive result is obtained for any of tested until an unequivocal result is obtained.
the test monolayers, then the presence of avian leucosis virus The seed lot complies with the test if there is no evidence of
in the test substance has been detected. the presence of chicken anaemia virus.
The seed lot complies with the test if there is no evidence of 6 TEST FOR EXTRANEOUS AGENTS USING
the presence of any avian leucosis virus.
CHICKS
4 TEST FOR AVIAN RETICULOENDOTHELIOSIS Inoculate each of at least 10 chicks with the equivalent of
VIRUS 100 doses of vaccine by the intramuscular route and with the
Prepare 11 monolayers of primary or secondary chick embryo equivalent of 10 doses by eye-drop. Chicks that are 2 weeks
fibroblasts from the tissues of 9- to 11-day old chick embryos of age are used in the test except that if the seed virus is
or duck embryo fibroblasts from the tissues of 13- to 14-day- pathogenic for birds of this age, older birds may be used, if
old embryos, each monolayer having an area of about required and justified. In exceptional cases, for inactivated
25 cm2 . vaccines, the virus may be neutralised by specific antiserum if
Remove the culture medium when the cells reach confluence. the seed virus is pathogenic for birds at the age of
Inoculate 0.1 mL of the test substance onto each of 5 of the administration. Repeat these iÍloculations 2 weeks latero
monolayers. A1low adsorption for 1 h, and add culture Observe the chicks for a period of 5 weeks from the day of
medium. Inoculate 4 of the monolayers with avian the first inoculation. N o antimicrobial agents shall be
reticuloendotheliosis virus as positive controls (not more than administered to fue chicks during the test periodo The test is
10 CCID so in 0.1 mL). Maintain 2 non-inoculated not val id if fewer than 80 per cent of t!le chicks survive lo the
monolayers as negative controls. end of the test periodo
Incubate the cells for a total of at least 10 days, subculturing Collect serum from each chick at the end of the test periodo
twice at 3- to 4-day intervals. The test is not valid if fewer Test each serum sample for antibodies against each of the
2016 Appendix XVI B (Vet) 4. Vet-A29

agents listed below (with the exception of the virus type of microscopic lesions (other than those attributable to the seed
the seed lot) using one of the methods indicated for testing lot virus) and no poult dies from causes attributable to the
for the agent. seed loto
Clinical signs of disease in the chicks during the test period C. Additional tests for duck extraneous agents
(other than signs attributable to the virus of the seed lot) and If the seed virus is of duck origin or was propagated in duck
the detection of antibodies in the chicks after inoculation substrates, tests for antibodies against the following agents
(with the exception of antibodies to the virus of the seed lot), are also carried out.
are classed as evidence of the presence of an extraneous
agent in the seed loto
Agent Type oftest
It is recommended that sera from these birds is retained so
that additional testing may be carried out if requirements Chlamydia spp. EIA
change. Duck and goose parvoviruses SN, EIA
A. Standard tests
Duck enteritis virus SN

Duck hepatitis virus type I SN


Agent Type oftest

Avian adenoviruses, group 1 SN, EIA, AGP


The seed lot complies with the test if there is no evidence of
Avian encephalomyelitis virus AGP, EIA
the presence of any extraneous agent.
Avian infectious bronchitis virus EIA,HI D. Additional tests for goose extraneous agents
Avian infectious laryngotracheitis virus SN, EIA, IS If the seed virus is of goose origin or was prepared in goose
Avian leucosis viruses SN, EIA substrates, tests for the following agents are also carried out.

Avian nephritis virus IS


Agent Type oftest
Avian orthoreoviruses IS, EIA
Duck and goose parvovirus SN, EIA
Avian reticuloendotheliosis virus AGP, IS, EIA
Duck enteritis virus SN
Chicken anaemia virus IS, EIA, SN
Goose haemorrhagic polyomavirus test in goslings shown below
Egg drop syndrome virus HI, EIA or another suitable test

Avian infectious bursal disease virus Serotype 1: AGP, EIA, SN


Serotype 2: SN Inoculate subcutaneously the equivalent of at least 10 doses
Influenza A virus AGP, EIA, HI to each of ten l-day-old susceptible goslings. Observe the
goslings for 28 days. The test is not valid if more than
Marek's disease virus AGP
20 per cent of the goslings die from non-specific causes.
Newcastle disease virus HI, EIA The seed virus complies with the test if no gosling dies fram
EIA
causes attributable to the seed loto
Turkey rhinotracheitis virus
Agg
7 ANTIBODY SPECIFICATIONS FOR SERA USED
Salman ella pullarum
IN EXTRANEOUS AGENTS TESTING
Agg: agglutination All batches of serum to be used in extraneous agents testing,
AGP: agar gel precipitation either to neutralise the vaccine virus (seed lot or batch of
EIA: enzyme immunoassay (e.g. ELISA) finished product) 01' as a supplement for cell culture media,
HI: haemagglutination inhibition shall be shown by suitably sensitive tests to be free from
IS: immunostaining (e.g. fluorescent antibody) antibodies against and free from inhibitory effects on the
SN: serum neutralisation
following micro-organisms - except for one type, namely,
antibodies against the virus that they are supposed to
B. Additional tests for turkey extraneous agents neutralise.
If the seed virus is of turkey origin 01' was propagated in Avian adenoviruses
turkey substrates, tests for antibodies against the following
Avian encephalomyelitis virus
agents are also carried out.
Avian infectious bronchitis viruses
Agent Type oftest Avian infectious bursal disease virus types 1 and 2
Avian infectious haemorrhagic enteritis virus
Chlamydia spp. EIA
Avian infectious laryngotracheitis virus
Avian infectious haemorrhagic enteritis virus AGP
Avian leucosis viruses
Avian pararnyxovirus 3 HI Avian nephritis virus
Avian infectious bursal disease virus type 2 SN Avian paramyxoviruses 1 to 9
Avian orthoreoviruses
A test for freedom from turkey lympho-proliferative disease Avian reticuloendotheliosis virus
virus is carried out by intraperitoneal inoculation of twenty Chicken anaemia virus
4-week-old turkey poults. Observe the poults for 40 days. Duck enteritis virus
The test is not valid if more than 20 per cent of the poults
Duck hepatitis virus type I
die from non-specific causes. The seed lot complies with the
test if sections of spleen and thymus taken from 10 poults Egg drop syndrome virus
2 weeks after inoculation show no macroscopic or Fowl pox virus
Vet-A30 Appendix XVI B (Vet) 5. 2016

Influenza virus es to provide the necessary guarantees of freedom from


Marek' s disease virus contamination with an extraneous agent.
Turkey herpesvirus If the vaccine virus cannot be completely neutralised - using
Turkey rhinotracheitis virus monospecific antiserum - for the yolk sac inoculation test,
the following tests may be performed:
Non-irnmune serum for addition to culture media can be
- for enveloped virus es such as Newcastle disease virus:
assumed to be free from antibodies against any of these
perform the test for extraneous agents using
viruses if the agent is known not to infect the species of
embryonated hens' eggs by the yolk sac route with
origin of the serum and it is not necessary to test the serum
prior inactivation of enveloped vaccine vil'uses with
for such antibodies. Monospecific antisera for virus
lipid solvents,
neutralisation can be assumed to be free from the antibodies
- for avian encephalomyelitis virus and avian nephritis
against any of these viruses if it can be shown that the
viruses: pelfOlm appropriate tests to detect these
irnmunising antigen could not have been contaminated with
viruses. For avian nephritis virus, the test in chicken
antigens derived from that virus and if the virus is known not
kidney cells described in section 2 of general chapter
to infect the species of origin of the serum; it is not necessary
2.6.24. Avian viral vaccines: tests for extraneous agel1ts in
to test the serum for su eh antibodies. It is not necessary to
seed lots may be used.
retest sera obtained from birds from SPF chicken flocks
(5.2.2). If the vaccine virus cannot be completely neutralised - using
monospecific antiserum - for the other tests below,
Batches of sera prepared for neutralising the vaccine virus
altematively or in addition to in vitra tests conducted on the
must not be prepared from any passage level derived from
batch, a test for extraneous agents may be conducted on
the virus iso late used to prepare the master seed lot 01' from
chick sera obtained from testing the batch of vaccine, as
an isolate cultured in the same cellline.
described under 6 Test for extraneous agents using chicks in
general chapter 2.6.24. Avian viral vaccines: tests for extraneous
agents in seed lots.
h) Other types of tests than those indicated may be used,
B (Vet) 5. Avian live Virus Vaccines: provided they are at least as sensitive as th_ose indicated and
Tests tor Extraneous Agents in Batches of appropriate specificity. Nucleic acid amplification
techniques (2.6.21) give specific detection for many agents
ot Finished Product and can be used after validation for sensitivity and specificity.
(Ph. Eur. method 2.6.25)
1 TEST FOR EXTRANEOUS AGENTS USING
GENERAL PROVISIONS EMBRYONATED HENS' EGGS
a) In the following tests, chickens and/or chicken material Prepare the test vaccine, diluted if necessary, to contain
such as eggs ¡md cell cultures shall be derived from chicken neutralised virus equivalent to 10 doses of vaccine in 0.2 mL
flocks free from specified pathogens (SPF) (5.2.2). of inoculum. Suitable antibiotics may be added. Inoculate the
b) Cell cultures for the testing of extraneous agents comply test vaccine into 3 groups of 10 emblyonated hens' eggs as
with the requirements for the master cell seed of general follows:
chapter 5.2.4. Gel! cultures for the production of veterina7Y - group 1: 0.2 mL into the allantoic cavity of each 9-tG
vaccines, with the exception of the karyotype test and the 11-day-old embryonated egg,
tumorigenicity test, which do not have to be carried out. - group 2: 0.2 mL onto the chorio-allantoic membrane of
c) In tests using cell cultures, precise specifications are given each 9- to 11-day-old embryonated egg,
for the number of replicates, monolayer surface are as and - group 3: 0.2 mL into the yolk sac of each 5- to 6-day-old
minimum survival rate of the cultures. Altemative numbers embryonated egg.
of replicates and cell surface areas are possible as well, Candle the eggs in groups 1 and 2 daily for 7 days and the
provided that a minimum of 2 replicates are used, the total eggs in group 3 for 12 days. Discard embryos that die during
surface area and the total volume of vaccine test applied are the first 24 h as non-specific deaths; the test is not valid
not less than that prescribed here and the survival rate unless at least 6 emblyos in each group survive beyond the
requirements are adapted accordingly. first 24 h after inoculation. Examine macroscopically for
d) In these tests, use the liquid vaccine or reconstitute a abnormalities all emblyos which die more than 24 h after
quantity of the freeze-dried preparation to be tested with the inoculation, 01' which survive d1e incubation periodo Examine
liquid stated on the label or another suitable diluent such as also the chorio-allantoic membranes of these eggs for any
water for injections. Unless otherwise stated 01' justified, the abnormality and test the allantoic fluids for the presence of
test substance contains the equivalent of 10 doses in 0.1 mL haemagglutinating agents.
of inoculum. Cany out a further embryo passage. Pool separately material
e) If the vaccine virus would interfere with the conduct and from live and from the dead and abnOlmal embryos.
sensitivity of the test, neutralise the vir~s in the preparation Inoculate each pool into 10 eggs for each route as described
with a monospecific antiserum. above, chorio-allantoic membrane material being inoculated
onto chorio-allantoic membranes, allantoic fluids into the
f) Monospecific antiserum· and serum of avian origin used for
allantoic cavity and embryo material into the yolk saco
cell culture and any otherpurpose, in any of these tests, shall
For eggs inoculated by the allantoic and chorio-allantoic
be free of antibodies against and free from inhibitory effects
routes, candle the eggs daily for 7 days, proceeding and
on the organisms listed under 7 Antibody specifications for
examining the material as described above. For eggs
sera used in extraneous agents testing (2.6.24).
inoculated by the yolk sac route, candre the eggs daily for
g) Where specified in a monograph or otherwise justified, if 12 days, proceeding and examining the material as described
neutralisation of the vaccine virus is required but difficult to above.
achieve, the tests described below are adapted, as required,
2016 Appendix XVI B (Vet) 5. Vet-A31

The batch of vaccine complies with the test if no test embryo 3 TEST FOR EGG DROP SYNDROME VIRUS
shows macroscopic abnormalities 01' dies fram causes Prepare 11 monolayers of chicken embryo liver cells, from
attributable to the vaccine and if examination of the chorio- the tissues of 14- to 16-day-old embryos, each monolayer
allantoic membranes and testing of the allantoic fluids show having an area of about 25 cm2 . Remove the culture medium
no evidence of the presence of extraneous agents. when the cells reach confluence. Inoculate 0.1 mL of test
As previously mentioned under General provisions vaccine onto each of 5 of the monolayers (test monolayers).
(paragraph g), if neutralisation of the virus is not possible Allow adsorption for 1 h, add culture medium. Inoculate 4 of
and as a consequence the yolk sac inoculation cannot be the monolayers with a suitable strain of egg drap syndrome
evaluated, suitable tests other than those indicated may be virus (not more than 10 CCID so in 0.1 mL) to serve as
carried out to provide the necessary guarantees of freedom positive control monolayers. Maintain 2 non-inoculated
from contamination with an extraneous agent; in particular monolayers as negative control monolayers.
other tests to detect avian encephalomye1itis and avian Incubate the cells for a total of at least 21 days, subculturing
nephritis viruses may be carried out. In this case, justification every 4-5 days. Each passage is made as follows: carry out a
to use other tests must be provided, and the vaccine complies freeze-thaw cycle; prepare separate pools of the cells plus
if there is no evidence of the presence of avian fluid from the test monolayers, from the positive control
encephalomyelitis virus or avian nephritis virus. monolayers and from the negative control monolayers;
2 TEST IN CHICKEN EMBRYO FIBROBLAST inoculate 0.1 mL of the pooled material onto each of 5, 4
CELLS and 2 recent1y prepared monolayers of chicken embryo liver
Prepare 7 monolayers of primary or secondary chicken cells, each monolayer having an area of about 25 cm2 as
embryo fibroblasts, from the tissues of 9- to 11-day-old before. The test is not valid if fewer than 4 of the 5 test
embryos, each monolayer having an area of about 25 cm2 . monolayers 01' fewer than 3 of the 4 positive controls 01'
Maintain 2 monolayers as negative controls and treat these in neither of the 2 negative control monolayers survive after any
the same way as the 5 monolayers inoculated with the test passage.
vaccine, as described below. Remove the culture medium Examine microscopically all the cell cultures at frequent
when the cells reach confluence. Inoculate 0.1 mL of test intervals throughout the entire incubation period for any
vaccine onto each of 5 of the monolayers. Allow adsorption signs of cytopathic effect or other evidence of the presence of
for 1 h and add culture medium. Incubate the cultures for a a contaminating agent in the test vaccine. At the end of the
total of at least 21 days, subculturing at 4- to 5-day intervals. total incubation period, carry out the following procedure:
Each passage is made with pooled cells and fluids from all test separately, cell culture fluid from the test monolayers,
5 monolayers after carrymg out a freeze-thaw cycle. Inoculate positive control monolayers and negative control monolayers,
0.1 mL of pooled material onto each of 5 recently prepared using chicleen red blood cells, for haemagglutination
monolayers of chicken embryo fibroblast cells, each attributable to the presence of haemagglutinating agents.
monolayer having an area of about 25 cm2 each as before. The test is not valid if egg drop syndrome virus is detected in
For the last passage, grow the cells also on a suitable fewer tIlan 3 of t11e 4 positive control monolayers 01' in any of
subst:rate so as to obtain an area of about 10 cm 2 of cells the negative control monolayers, or if the results for both of
fram each of the monolayers, for test A. The test is not valid the 2 negative control monolayers are inconclusive. If the
if less than 80 per cent of the test monolayers, 01' neither of results for more than 1 of the test monolayers are
the 2 negative control monolayers survive after any passage. inconclusive then further sub cultures of reserved portions of
Examine microscopically all the cell cultures frequentIy the monolayers shall be made and tested until an
throughout the entire incubation period for any signs of unequivocal result is obtained.
cytopathic effect 01' other evidence of the presence of The batch of vaccine complies with the test if there is no
contaminating agents in the test vaccine. At the end of the evidence of the presence of egg drop syndrome virus 01' any
total incubation period, carry out the following procedures. other extraneous agent.
A. Fix and stain (with Giemsa 01' haematoxylin and eosin) 4 TEST FOR MAREK'S DISEASE VIRUS
about 10 cm 2 of confluent cells fram each of the 5 original Prepare 11 monolayers óf primary 01' secondary chicl, embryo
monolayers. Examine the cells microscopically for any fibroblasts fram the tissues of 9- to 11-day-old embryos, each
cytopathic effect, inclusion bodies, syncytial formation, 01' any monolayer having an area of about 25 cm2 . Remove the
other evidence of the presence of a contaminating agent from culture medium when the cells reach confluence. Inoculate
the test vaccine. 0.1 mL of test vaccine onto each of 5 of the monolayers (test
B. Drain and wash about 25 cm2 of cells from each of the monolayers). Allow adsorption for 1 h, and add culture
5 monolayers. Cover tIlese cells with a 0.5 per cent medium. Inoculate 4 of the monolayers with a suitable strain
suspension of washed chicken red blood cells (using at least of Marek' s disease virus (not more than 10 CCID so in
1 mL of suspension for each 5 cm2 of cells). Incubate the 0.1 mL) to serve as positive controls. Maintain 2 non-
cells at 4 oC for 20 min and then wash gently in phosphate inoculated monolayers as negative controls.
buffered saline pH 7.4. Examine the cells microscopically for Incubate the cultures for a total of at least 21 days,
haemadsorption attributable to the presence of a subculturing at 4- to 5-day intervals. Each passage is made as
haemadsorbing agent in the test vaccine. follows: trypsinise thé cells, prepare separate pools of the cells
C. Test individually samples of the fluid from each cell from the test monolayers, from the positive control
culture using chicken red blood cells for haemagglutination monolayers and from the negative control monolayers.
attributable to the presence of a haemagglutinating agent in Mix an' appropriate quantity of each with a suspension of
the test vaccine. freshly prepared primary or s~condary chic k embryo
The test is not valid if there are any signs of exttaneous fibroblasts and prepare 5, 4 and 2 monolayers, as before.
agents in tIle negative control cultures. The batch of vaccine The test is not valid if fewer than 4 of the 5 test monolayers
complies with the test if there is no evidence of the presence 01' fewer than 3 of the 4 positive controls 01' neither of the
of any extraneous agent. 2 negative control monolayers survive after any passage.
Vet-A32 Appendix XVI B (Vet) 5. 2016

Examine microscopically all the cell cultures frequently Prepare 11 monolayers of Vero cells, each monolayer having
throughout the entire incubation period for any signs of an area of about 25 cm2 . Remove the culture medium when
cytopathic effect or other evidence of the presence of a the cells reach confluence. Inoculate 0.1 mL oftest vaccine
contaminating agent in the test vaccine. onto each of 5 of tl1e monolayers (test monolayers). A1low
For the last sub culture, grow the cells on a suitable substrate adsorption for 1 h, and add culture medium. Inoculate 4 of
so as to obtain an area of about 10 cm2 of confluent cells the monolayers with a suitable strain of turkey rhinotracheitis
from each of the original 11 monolayers for the subsequent virus (not more than 10 CCID so in 0.1 mL) to serve as
test: test about 10 cm 2 of confluent cells derived from each positive controls. Maintain 2 non-inoculated monolayers as
of the original 11 monolayers by immunostaining for the negative controls.
presence of Marek's disease virus. The test is not valid if Incubate the cultures for a total of at least 21 days,
Marek' s disease virus is detected in fewer than 3 of the subculturing at 4- to 5-day intervals. Each passage is made as
4 positive control monolayers or in any of the negative follows: carry out a freeze-thaw cycle. Prepare separate pools
control monolayers, or if the results for both of the 2 of the cells plus fluid from the test monolayers, from the
negative control monolayers are inconclusive. positive control monolayers and from the negative control
The batch of vaccine complies with the test if there is no monolayers. Inoculate 0.1 mL of the pooled material onto
evidence of the presence of Marek' s disease virus or any each of 5, 4 and 2 recently prepared monolayers ofVero
other extraneous agent. cells, each monolayer having an area of about 25 cm2 as
before. The test is not valid if fewer than 4 of the 5 test
5 TESTS FOR TURKEY RmNOTRACHEITIS VIRUS monolayers or fewer than 3 of the 4 positive control s or
A. In chicken embryo fibroblasts neither of the 2 negative controls survive after any passage.
NOTE: this test can be combined zuith Test 2 by using the same For the last sub culture, grow the cells on a suitable substrate
test 112011olaye1's and negative controls, fo1' all stages up to the final so as to obtain an area of about 10 cm2 of confluent cells
specific test for turkey rhinotracheitis virus 011 cells prepared fl"o112 from each of the original 11 monolayers for the subsequent
the last sub culture. test: test about 10 cm2 of confluent cells derived from each
Prepare 11 monolayers of primal)' or secondal)' chick embryo of the original 11 monolayers by immunostairring for the
fibroblasts .from the tissues of 9- to 11-day-old embryos, each presence of turkey rhinotracheitis virus. The test is not valid
monolayer having an area of about 25 cm2 . Remove the if turkey rhinotracheitis virus is detected in fewer than 3 of
culture medium when the cells reach confluence. Inoculate the 4 positive control monolayers or in any of the negative
0.1 mL oftest vaccine onta each of 5 of the monolayers (test control monolayers, or if the results for both of the 2
monolayers). A1low adsorption for 1 h, and add culture negative control monolayers are inconclusive. If the results
medium. Inoculate 4 of tl1e monolayers with a suitable strain for more than 1 of the test monolayers are inconclusive then
of turkey rhinotracheitis virus as positive controls (not more further sub cultures of reserved portions of tl1e monolayers
than 10 CCID so in 0.1 mL). Maintain 2 non-inoculated shall be made and tested until an unequivocal result is
monolayers as negative controls. obtained.
Incubate the cultures for a total of at least 21 days, The batch of vaccine complies with the test if there is no
subculturing at 4- to 5-day intervals. Each passage is made as evidence of tl1e presence of turkey rhinotracheitis virus or any
follows: can)' out a freeze-thaw cycle; prepare separate pools other extraneous agent.
of the cells plus fluid from the test monolayers, from the
6 TEST FOR CHICKEN ANAEMIA VIRUS
positive control monolayers and from the negative control
monolayers; inoculate 0.1 mL of the pooled material onto Prepare eleven 20 mL suspensions of the MDCC-MSBI cell
each of 5, 4 and 2 recently prepared monolayers of chicken line or another cellline of equivalent sensitivity in 25 cm 2
embl)'o fibroblasts cells, each monolayer having an area of flasks containing about 5 x lOs cells/mL. Inoculate 0.1 mL
about 25 cm 2 as before. The test is not valid if fewer than 4 of test vaccine into each of 5 of these flasks. Inoculate
of the 5 test monolayers or fewer than 3 of the 4 positive 4 other suspensions with 10 CCID so chicken anaemia virus
controls or neither of the 2 negative control monolayers as positive controls. Maintain not fewer than 2 non-
survive after any passage. inoculated suspensions. Maintain all the cell cultures for a
total of at least 24 days, subculturing 8 times at 3- to 4-day
For the last sub culture, grow the cells on a suitable substrate intervals. During the subculturing the presence of chicken
so as to obtain an area of about 10 cm2 of confluent cells anaemia virus may be indicated by a metabolic colour change
from each of the original 11 monolayers for the subsequent in the infected cultures, the culture fluids becoming red in
test: test about 10 cm2 of confluent cells derived from each comparison with the control cultures. Examine the cells
of the original 11 monolayers by immunostaining for the microscopically for cytopathic effect. At this time or at the
presence of turkey rhinotracheitis virus. The test is not valid end of tl1e incubation period, centrifuge the cells fram each
if turkey rhinotracheitis virus is detected in fewer than 3 of flask at low speed, resuspend at about 10 6 cells per millilitre
the 4 positive control monolayers or in any of the negative and place 25 flL in each of 10 wells of a multi-well slide.
control monolayers, or if the results for both of the 2 Examine the cells by immunostaining.
negative control monolayers are inconclusive. If the results
for both of the 2 test monolayers are inconclusive then The test is not valid if chicken anaemia virus is detected in
further sub cultures of reserved portions of the fibroblasts fewer than 3 of the 4 positive controls or in any of the non-
shall be made and tested until an unequivocal result is inoculated controls. If the results for more than 1 of the test
obtained. suspensions are inconclusive then further sub cultures of
reserved portions of the test suspensions shall be made and
The batch of vaccine complíes with the test if there is no tested until an unequivocal result is obtained.
evidence of the presence of turkey rhinotracheitis virus or any
other extraneous agent. The batch of vaccine complies with the test if there is no
evidence of the presence of chicken anaemia virus.
E. In Vero cells
2016 Appendix XVI B (Vet) 5. Vet-A33

7 TEST FOR DUCK ENTERITIS VIRUS or fewer than 3 of the 4 positive controls or neither of the
This test is canied out for vaccines prepared on duck 01' 2 negative controls survive after any passage.
goose substrates. For the last sub culture, grow the cells on a suitable substrate
Prepare 11 monolayers of primary 01' secondary Muscovy so as to obtain an area of about 10 cm2 of confluent cells
duck embryo liver cells, from the tissues of 21- 01' 22-day-old from each of the original 11 monolayers for the subsequent
embryos, each monolayer having an area of about 25 cm2 . test: test about 10 cm2 of confluent cells derived from each
Remove the culture medium when the cells reach confluence. of the Oliginal 11 monolayers by immunostaining for the
Inoculate 0.1 mL of test vaccine onto each of 5 of the presence of duck 01' goose parvovirus. The test is not valid if
monolayers (test monolayers). Allow adsorption for 1 h and duck parvovirus is detected in fewer than 3 of the 4 positive
add culture medium. Inoculate 4 of the monolayers with a control monolayers 01' in any of the negative control
suitable strain of duck enteritis virus (not more than monolayers, or if the results for both of the 2 negative
10 CCID so in 0.1 mL) to serve as positive controls. Maintain control monolayers are inconclusive.
2 non-inoculated monolayers as negative controls. The batch of vaccine complies with the test if there is no
Incubate the cultures for a total of at least 21 days, evidence of the presence of duck (or goose) parvovirus or any
subcultuling at 4- to S-day intervals. Each passage is made as other extraneous agent.
follows: trypsinise the cells and prepare separate pools of the
cells from the test monolayers, from the positive control
monolayers and from the negative control monolayers. Mix a
portion of each with a suspension of freshly prepared primary
01' secondary Muscovy duck embryo liver cells to prepare 5,
4 and 2 monolayers, as before. The test is not valid if fewer
than 4 of the 5 test monolayers or fewer than 3 of the
4 positive controls 01' neither of the 2 negative controls
survive after any passage.
For the last sub culture, grow the cells on a suitable substrate
so as to obtain an area of about 10 cm2 of confluent cells
from each of the Oliginal 11 monolayers for the subsequent
test: test about 10 cm2 of confluent cells derived from each
of the original 11 monolayers by irnmunostaining for the
presence of duck enteritis virus. The test is not valid if duck
enteritis virus is detected in fewer than 3 of the 4 positive
control monolayers or in any of the negative control
monolayers, or if the results for both of the 2 negative
control monolayers are inconclusive. If the results for more
than 1 of the test monolayers are inconclusive then Íurther
sub cultures of reserved portions of the monolayers shall be
made and tested until an unequivocal result is obtained.
The batch of vaccine complies with the test if there is no
evidence of the presence of duck enteritis virus 01' any other
extraneous agent.
8 TEST FOR DUCK AND GOOSE PARVOVIRUSES
This test is carried out for vaccines prepared on duck 01'
goose substrates.
Prepare a suspension of sufficient plimary or secondary
Muscovy duck embryo fibroblasts from the tissues of 16- to
18-day-old embryos, to obtain not fewer than 11 monolayers,
each having an area of about 25 cm2 . Inoculate 0.5 mL of
test vaccine into an aliquot of cells for 5 monolayers and seed
into 5 replicate containers to form 5 test monolayers.
Inoculate 0.4 mL of a suitable strain of duck parvovirus (not
more than 10 CCID so in 0.1 mL) into an aliquot of cells for
4 monolayers and seed into 4 replicate containers to form
4 positive control monolayers. Prepare 2 non-inoculated
monolayers as negative controls.
Incubate the cultures for a total of at least 21 days,
subculturing at 4- to S-day intervals. Each passage is made as
follows: carry out a freeze-thaw cycle. Prepare separate pools
of the cells plus fluid from the test monolayers, from the
positive control monolayers and from the negative control
monolayers. Inoculate 0.5 mL, 0.4 mL and 0.2 mL of the
pooled materials into aliquots of a fresh suspension of
sufficient primat-y or secondary Muscovy duck embryo
fibroblast cells to prepare 5, 4 and 2 monolayers, as before.
The test is not val id if fewer than 4 of the 5 test monolayers
Vet-A34 Appendix XXI B 2016

Appendix XXI
B (Vet). Approved Synonyms
Where the English title at the head of a monograph in the European Pharmacopoeia is different from that at the head of the
text incorporated into the British Pharmacopoeia or the British Pharmacopoeia (Veterinary), an Approved Synonyrn (or
Approved Synonyms) is created on the recommendation of the British Pharmacopoeia Commission.
In accordance with the General Notice on Titles, the name or names given in the right-hand column of the list below are
Approved Synonyms for the name at the head of the monograph of the European Pharmacopoeia given in the left-hand
column. Where there is more than one entry in the right-hand column, the first entry is used as the title of the monograph in
the British Pharmacopoeia or the British Pharmacopoeia (Veterinary) and the remaining entries are included as subsidiary titles.
Approved Synonyrns and subsidiary titles have the same significance as the main title and are thus official titles.
Names made by changing the order of the words in an Approved Synonym, with the addition of a preposition when necessary,
are also Approved Synonyrns.
Where square brackets are used in a title these may be replaced by round brackets, and vice versa. The words 'per cent' may be
replaced by the syrnbol '%'.
Where the word 'Injection' appears in the title or synonyrn of a monograph in the European Pharmacopoeia, the abbreviation
'Inj.' is declared to be an Approved Synonym for that part ofthe title.
A consolidated list of all Approved Synonyms is included in Appendix XXI B of the British Pharmacopoeia.

EUROPEAN PHARMACOPOEIA TITLE APPROVED SYNONYM


Medicinal Substances and Formulated Preparations
Azaperone for Veterinary Use Azaperone
Carprofen for Veterinary U se Carprofen
Clazuril fo1' Vete1'inary U se Clazuril
Closantel Sodium Dihydrate fo1' Veterinary U se Closantel Sodium Dihydrate
Dembrexine Hyd1'ochloride Monohydrate for Vete1'inary Use Dembr-exine Hyd1'ochloride Monohydrate
Detomidine Hyd1'ochloride for Veterinary U se Detomidine Hyd1'ochloride
Diclazuril fo1' Veterinary U se Diclazuril
Difioxacin Hydrochloride Trihydrate fo1' Vete1'inary Use Difioxacin Hydrochloride Trihydrate
Dihydrostreptomycin Sulfate for Veterinary U se Dihydrostreptomycin Sulfate
Enilconazole for Veterinary U se Enilconazole
Enrofioxacin for Veterinary U se Enrofioxacin
Equine Serum Gonadotrophin for Vete1'inary Use Serum Gonadotrophin
Febantel for Veterinary Use Febantel
Fenbendazole for Veterinary Use Fenbendazole
Flunixin Meglumine for Veterinary U se Flunixin Meglumine
Levamisole for Veterinary Use Levamisole
Lufenuron (Anhydrous) for Veterinary Use Anhydrous Lufenuron
Marbofioxacin for Veterinay U se Marbofioxacin
Morantel Hydrogen Tartrate fo1' Vete1'inary U se Mo1'antel Tartrate
Moxidectin for Veterinary U se Moxidectin
Orbifioxacin for Veterinary U se Orbifioxacin
Oxfendazole fo1' Veterinary U se Oxfendazole
Selamectin fo1' Veterinary U se Selamectin
Spectinomycin Sulfate Tetrahydrate for Veterinary Use Spectinomycin Sulfate Tetrahydrate
Sulfadimethoxine Sodium for Veterinary U se Sulfadimethoxine Sodium
Sulfamethoxypyridazine for Veterinary U se Sulfameto:h.'Ypyridazine
Tiamulin fo1' Veterinary Use Tiamulin
Tiamulin Hydrogen Fumarate for Veterinary Use Tiamulin Hydrogen Fumarate
Triclabendazole for Veterinary U se Triclabendazole
2016 Appendix XXI B Vet-A35

EUROPEAN PHARMACOPOEIA TITLE APPROVED SYNONYM


Tylosin For Veterinary Use Tylosin
Tylosin Phosphate Bulk Solution for Veterinary U se Tylosin Phosphate
Tylosin Tartrate For Veterinary Use Tylosin Tartrate
Valnemulin Hydrochloride for Veterinary Use Valnemulin Hydroch1oride
Vedaprofen for Veterinary U se Vedaprofen
Xylazine Hydrochloride for Veterinary Use Xylazine Hydrochloride
Vet-A36 Appendix XXI B 2016

EUROPEAN PHARMACOPOEIA TITLE APPROVED SYNONYM


Immunological Products (Veterinary)
Anthrax Spore Vaccine (Live) for Veterinary Use Anthrax Vaccine, Living
Aujeszky's Disease Vaccine (Inactivated) for Pigs Aujeszky's Disease Vaccine, Inactivated
Aujeszky's Disease Vaccine (Live) for Pigs for Parenteral Aujeszky's Disease Vaccine, Living
Administration
Avian Infectious Bronchitis Vaccine (Live) Avian Infectious Bronchitis Vaccine, Living
Avian Infectious Bursal Disease Vaccine (Inactivated) Infectious Bursal Disease Vaccine, Inactivated
Gumboro Disease Vaccine, Inactivated
Avian Infectious Bursal Disease Vaccine (Live) Infectious Bursal Disease Vaccine, Living
Gumboro Disease Vaccine, Living
Avian Infectious Encephalomyelitis Vaccine (Live) Infectious Avian Encephalomyelitis Vaccine, Living Epidemic
Tremor Vaccine, Living
Avian Infectious Laryngotracheitis Vaccine (Live) Laryngotracheitis Vaccine, Living
Bovine Parainfiuenza Virus Vaccine (Live) Bovine Parainfiuenza Virus Vaccine, Living
Bovine Respiratory Syncytial Virus Vaccine (Live) Bovine Respiratory Syncytial Virus Vaccine, Living
Brucellosis Vaccine (Live) (Brucella Melitensis Rev. 1 Strain) BrL!cella Melitensis (Strain Rev. 1) Vaccine, Living
for Veterinary U se
Canine Adenovirus Vaccine (Inactivated) Canine Adenovirus Vaccine, Inactivated
Canine Adenovirus Vaccine (Live) Canine Adenovirus Vaccine, Living
Canine Distemper Vaccine (Live) Canine Distemper Vaccine, Living
Canine Parvovirosis Vaccine (Inactivated) Canine Parvovirus Vaccine, Inactivated
Canine Parvovirosis Vaccine (Live) Canine Parvovirus Vaccine, Living
Clostridillin Botulinum Vaccine for Veterinary Use Clostridium Botulinum Vaccine
Botulinum Vaccine
Clostridium Chauvoei Vaccine for Veterinary U se Clostridium Chauvoei Vaccine
Blacldeg Vaccine
ClostridiumNovyi (Type B) Vaccine for Veterinary Use Clostridium N ovyi Type B Vaccine
Black Disease Vaccine
Clostridium N ovyi Alpha Antitoxin for Veterinary U se Clostridium N ovyi Alpha Antitoxin
Clostridium Perfringens Beta Antitoxin for Veterinary Use Clostridium Perfringens Beta Antitoxin
Clostridium Perfringens Epsilon Antitoxin for Veterinary Use Clostridium Perfringens Epsilon Antitoxin
Clostridium Perfringens Type D Antitoxin
Clostridium Perfringens Vaccine for Veterinary U se Clostridium Perfringens Vaccines.
Type B Clostridium Perfringens Type B Vaccine
Type C Lamb Dysentery Vaccine
TypeD Clostridium Perfringens Type C Vaccine
Struck Vaccine
Clostridium Perfringens Type D Vaccine
Pulpy Kidney Vaccine
Clostridium Septicum Vaccine for Veterinary Use Clostridium Septicum Vaccine Braxy Vaccine
Distemper Vaccine (Live) for Mustelids Ferret and Mink Distemper Vaccine, Living
Egg Drop Syndrome '76 Vaccine (Inactivated) Egg Drop Syndrome 76 (Adenovirus) Vaccine
Equine Herpesvirus Vaccine (Inactivated) Equine Herpesvirus Vaccine, Inactivated
Equine Influenza Vaccine (Inactivated) Equine Influenza Vaccine, Inactivated
Feline Calicivirosis Vaccine (Inactivated) Feline Caliciyirus Vaccine, Inactivated
Feline Calicivirosis Vaccine (Live) Feline Calicivirus Vaccine, Living
Feline Infectious Enteritis (Feline Panleucopenia) Vaccine Feline Infectious Enteritis Vaccine, Inactivated
(Inactivated) Feline Panleucopenia Vaccine, Inactivated
2016 Appendix XXI B Vet-A37

EUROPEAN PHARMACOPOEIA TITLE APPROVED SYNONYM


Feline Infectious Enteritis (Feline Panleucopenia) Vaccine Feline Infectious Enteritis Vaccine, Living
(Live) Feline Panleucopenia Vaccine, Living
Feline Leukaemia Vaccine (Inactivated) Feline Leukaemia Vaccine, Inactivated
Feline Viral Rhinotracheitis Vaccine (Inactivated) Feline Viral Rhinotracheitis Vaccine, Inactivated
F eline Viral Rhinotracheitis Vaccine (Live) F eline Viral Rhinotracheitis Vaccine, Living
Foot-and-Mouth Disease (Ruminants) Vaccine (Inactivated) Foot and Mouth Disease (Ruminants) Vaccine
Fowl-pox Vaccine (Live) Fowl Pox Vaccine, Living
Furunculosis Vaccine (Inactivated, Oil-adjuvanted, Injectable) Furunculosis Vaccine for Salmonids, Inactivated
for Salmonids
Infectious Bovine Rhinotracheitis Vaccine (Live) Infectious Bovine Rhinotracheitis Vaccine, Living
Marek's Disease Vaccine (Live) Marek's Disease Vaccine, Living
Marek's Disease Vaccine (Turkey Herpes Virus)
Marek's Disease Vaccine, Living (HVT)
Neonatal Pigle! Colibacillosis Vaccine (Inactivated) Porcine E. Coli Vaccine, Inactivated
Porcine Escherichia Coli Vaccine, Inactivated
Neonatal Ruminant Colibacillosis Vaccine (Inactivated) Ruminant E. Coli Vaccine, Inactivated
Ruminant Escherichia Coli Vaccine, Inactivated
Newcastle Disease Vaccine (Inactivated) Newcastle Disease Vaccine, Inactivated
N ewcastle Disease Vaccine (Live) Newcastle Disease Vaccine, Living
Porcine Actinobacillosis Vaccine (Inactivated) Porcine Actinobacillosis Vaccine, Inactivated
Porcine Influenza Vaccine (Inactivated) Swine Influenza Vaccine, Inactivated
Porcine Parvovirosis Vaccine (Inactivated) Porcine Parvovirus Vaccine, Inactivated
Porcine Progressive Atrophic Rhinitis Vaccine (Inactivated) Porcine Progressive Atrophic Rhinitis Vaccine, Inactivated
Rabies Vaccine (Inactivated) for Veterinary U se Rabies Veterinary Vaccine, Inactivated
Swine Erysipelas Vaccine (Inactivated) Swine Erysipelas Vaccine, Inactivated
Tetanus Antitoxin for Veterinary Use Clostridium Tetani Antitoxin
Tetanus Antitoxin (Veterinary)
Tetanus Vaccine for Veterinary Use Clostridium Tetani Vaccines
Tetanus Toxoids (Veterinary)
(for vaccines 'luith an appropriate potency)
Clostridium Tetani Vaccine for Equidae
Tetanus Toxoid for Equidae
Tuberculin Purified Pro te in Derivative, Avian Avian Tuberculin Purified Protein Derivative
Avian Tuberculin P.P.D.
Tuberculin Purified Pro te in Derivative, Bovine Bovine Tuberculin Purified Protein Derivative
Bovine Tuberculin P.P.D.
Vibriosis (Cold-water) Vaccine (Inactivated) for Salmonids Cold-water Vibriosis Vaccine for Salmonids, Inactivated
Vibriosis Vaccine (Inactivated) for Salmonids Vibriosis Vaccine for Salmonids, Inactivated
Yersiniosis Vaccine (Inactivated) for Salmonids Enteric Redmouth Disease Vaccine for Rainbow Trout,
Inactivated
Supplementary Chapters
Supplementary Chapters contain no standards) tests or assays nor any other mandatory
specifications with respect to any Pharmacopoeial artide. They comprise explanatory and
other ancillary texts and are provided for the assistance and inloll1'lation 01 users 01 the
Phannacopoeia.
2016 Vet-A41

Contents of the Supplementary Chapters


SUPPLEMENTARY CHAPTER 1 A43
Phannacopoeial Organisation A43
A (Vet). Monograph Development: Mechanism A43
2016 Supplementary Chapter 1 A Vet-A43

in the United Kingdom; veterinary products identified by


Suppletnentary the members of the Panel of Experts for Veterinary
Medicines) .
Chapter 1 2. The innovator product is approaching or past its patent
expiry date (monographs will usually only be prepared in
the two years preceding patent expiry. However there may
A. Monograph Development: Mechanism be circumstances where it is justified to prepare a
The follO'luing Supple11lentG1JI Chapter provides an outline of the monograph at an earlier stage. These will be considered
mechanism by 'luhich monographs are selected and developed for by the British Pharmacopoeia Commission individually).
inclusion in the British Pharmacopoeia (Ve te rinG1J¡) . 3. There is a particular need based on the therapeutic
The British Pharmacopoeia Commission will not usually category and/or the importance of the material concemed;
develop monographs for drug substances or excipients. These the latter being particularly relevant to small patient
will usually be elaborated by the European Phannacopoeia populations.
Commission. 4. The product falls within a "family" of product
The British Pharmacopoeia Commission will consider a presentations-, of which there are already published
monograph for inc1usion in the British Pharmacopoeia monographs and/or monographs on the work programme.
(Veterinary) in the following circumstances: 5. Drug substances or excipients which are not on the
1. The formulation is widely used (for example: top 100 European Pharmacopoeia work programme-, but for which
products identified following a survey of wholesale dealers there is a specific UK need.

BP Laboratory
Practical assessment
Method development
Expert Advisory Group
Technical advice
Approve final draft

Final monograph BP Secretariat


~to,
Invite proposals
BP Commission Draft monograph

Draft monographs Feedback


Vet-A44 Supplementary Chapter 1 2016

6. A request is received from the Competent Authority


[Veterinary Medicines Directorate (VMD)].
7. A request is received from a manufacturer for one of their
own products.
8. Support fOl' relevant EC directives.
9. A request is received from official bodies [su eh as the
World Health Organization (WHO)].
10. Other circumstances considered on a case-by-case basis.
It should be noted that compliance with any of the above
criteria will not necessarily mean that a monograph will be
included in the British Pharmacopoeia (Veterinary).
The British Pharmacopoeia Commission may decide not to
elaborate a monograph for a number of reasons, including a
lack of interest from stakeholders, resource limitations 01'
other circumstances, decided on a case-by-case basis.
The diagram pro vides a simplifiedJ schematic representation of the
development of a monograph for a medicinal substance 01" an
associated formulated preparation.
page nUlnbers in bold type relate to monograph titles Index Vet-A45

Inde
Page numbers in bold type relate to monograph titles.

Pages - Vol VI: i - xxiv, (Preliminaries and Introduction)


Vet-l - 338, (General Notices and Monographs)
SI - S20, (Spectra)
Al - A44, (Appendices; Supplementary Chapters)
page numbers in bold type relate to monograph titles Index Vet-A47

A Aujeszky's Disease Vaccine, lnactivated,


Vet-204
Aujeszky's Disease Vaccine (lnactivated)
e
Abbreviated Titles, Status of, Vet-7 Calcium Borogluconate lnjection,
Abbreviations and symbols, Vet-29 for Pigs, see Aujeszk:y's Disease Vaccine, Vet-151
About, definition of, Vet-5 Inactivated Calcium Copperedetate, Vet-45
Acepromazine, Vet-S4 Aujeszky's Disease Vaccine, Living, Calcium Copperedetate lnjection,
Acepromazine lnjection, Vet-140 Vet-205 Vet-152
Acepromazine lnjection, Etorphine and, Aujeszky's Disease Vaccine (Live) for Calf Coronavirus Diarrhoea Vaccine
Vet-161 Pigs for Palenteral Administration, see (lnactivated), Vet-220
Acepromazine Maleate, Vet-39 Aujeszky's Disease Vaccine, Live Calf Paratyphoid Vaccine, Living, see
Acepromazine Tablets, Vet-140 Avian lnfectious Bronchitis Vaccine, Salmonella Dublin Vaccine, Living
Acknowledgements, Vet-xxiii Living, Vet-209 Calf Rotavirus Diarrhoea Vaccine
Action and Use, Status of, Vet-16 Avian lnfectious Bronchitis Vaccine, (lnactivated), Vet-222
Additions, List of Monographs, Vet-xxiv Living, N ewcastle Disease and, Candela, Definition of, Vet-31
Additions, List of, Vet-xx Vet-288 Canine Adenovirus Vaccine, lnactivated,
Albendazole Oral Suspension, Vet-141 Avian Live Virus Vaccines, Vet-223
Albendazole Oral Suspension with Tests for Extraneous Agents in Canine Adenovirus Vaccine, Living,
Minerals, Vet-142 Batches of Finished Product, Vet-224
Alfadolone Acetate, Vet-S5 Vet-A30 Canine Distemper Vaccine, Living,
Alfaxalone, Vet-39, Vet-S5 Avian Paramyxovirus 3 Vaccine for Vet-225
Amino Acid, Codes for, Vet-8 Turkeys, lnactivated, Vet-211 Canine Leptospirosis Vaccine
Amitraz, Vet-40, Vet-S5 Avian Tuberculin Purified Protein (lnactivated), Vet-278
Amitraz Dip Concentrate (Liquid), Derivative, Vet-328 Canine Parainfiuenza Virus Vaccine
Vet-143 Avian Viral Tenosynovitis Vaccine (Live), Vet-226
Amoxicillin Oily lnjections, Vet-144 (Live), Vet-2B Canine Parvovirus Vaccine, lnactivated,
Amoxicillin Tablets, Vet-146 Avian Viral Vaccines, Tests for Vet-227
Amoxicillin Trihydrate, Vet-S6 Extraneous Agents in Seed Lots, Canine Parvovirus Vaccine, Living,
Amoxicillin Veterinary Oral Powder, Vet-A27 Vet-228
Vet-145 Azaperone, Vet-44, Vet-S7 Capital lnitial Letters, Significance of,
Ampere, Definition of, Vet-31 Azaperone for Vetinary Use, see Vet-8
Ampicillin and Cloxacillin Azaperone Carprofen, Vet-46
lntramammary lnfusion (DC), Azaperone lnjection, Vet-151 Carprofen for Vetinary Use, see Cmprofen
Vet-148 Caution Statements, Vet-7
Ampicillin and Cloxacillin CClD so , Definition of, Vet-29
lntramammary lnfusion (LC), see B Cefalonium, Vet-47, Vet-S7
Ampicillin Sodi1l11l and Cloxacillin Cefalonium Intramammary Infusion
B (Vet) 4. Avian Viral Vaccines Tests for
SodizlJ71 Imramal7171lm)l Infllsion (Dry Cow), Vet-152
Extraneous Agents in Seed Lots,
(Lactating Cozo) Cell Cultures for the Production of
Vet-A27
Ampicillin Sodium and Cloxacillin Veterinary Vaccines, Vet-A12
Batch Release, Vet-9
Sodium lntramammary lnfusion Cell Cultures ev eterinary Vaccine
Biological Reference Preparations,
(Lactating Cow), Vet-146 Production) (5.2.4.), Vet-12
Vet-15
Ampicillin Trihydrate, Vet-S6 Ce1sius, Definition of, Vet-31
Bordetella Bronchiseptica Vaccine (live)
Ampicillin Trihydrate and Cloxacillin Centigrade, Definition of, Vet-31
for Dogs, Vet-214
Benzathine lntramammary lnfusion Centrifugation, Definition of, Vet-31
Bovine Leptospirosis Vaccine
(Dry Cow), Vet-148 Characteristics, Status of, Vet-ll
(lnactivated), Vet-276
Amprolium Hydrochloride, Vet-41, Chemical Abstracts Sel\Tice Registry
Bovine Parainfiuenza Virus Vaccine,
Vet-S6 Number, Status, Vet-4
Living, Vet-215
Anhydrous Lufenuron, Vet-78 Ghemical Formulae, Vet-4
Bovine Respiratory Syncytial Virus
Animals, Use of, Vet-15 Chemical Reference Substances, Vet-15
Vaccine, Living, Vet-217
Annex to Monographs, Status of, Vet-5 Chicken Flocks Free from Specified
Bovine Tuberculin Purified Protein
Anthrax Spore Vaccine (Live) for Pathogens for the Production and
Derivative, Vet-329
Veterinary Use, see Anthrax Vaccine, Quality Control of Vaccines, Vet-A9
Bovine Tuberculin P.P.D., see Bovine
Living Chloramphenicol, Vet-S7
Tuberculin Purified Protein Derivative
Anthrax Vaccine, Living, Vet-203 Chlortetracyc1ine Soluble Powder, see
Bovine Viral Diarrhoea Vaccine
Antibiotics in lntramammary lnfusions, Chlortetracycline VeterinGl)l Oral PO'luder
(lnactivated), Vet-218
Vet-17 Chlortetracyc1ine Tablets, Vet-154
BPCRS, Vet-15
Antibiotics, Potency of, Vet-14 Chlortetracyc1ine Veterinary Oral
Bpevet), Vet-3
Antimicrobial Preservative, Definition of Powder, Vet-153
British Pharmacopoeia Commission,
Suitable, Vet-ll c.I.P. - Collection de Bactéries de
Vet-viii
Approved Synonyms, Vet-A34 l'lnstitut Pasteur, address of, Vet-30
BRP, Vet-15
Apramycin Premix, Vet-149 Clazuril, Vet-48
Brucella Me1itensis (Strain Rev. 1)
Apramycin Sulfate, Vet-42 Clazuril for Vetinary Use, see Clazurz"l
Vaccine, Living, Vet-219
Apramycin Sulphate, see Apramycin Cloprostenol Injection, Vet-155
Brucellosis Vaccine (Live) (Brucella
Sulfate Cloprostenol Sodium, Vet-50, Vet-S8
Me1itensis Rev. 1 Strain), for
Apramycin Veterinary Oral Powder, Closantel Sodium Dihydrate, Vet-51
Veterinary Use, see Bntcella Melitensis
Vet-148 Closantel Sodium Dihydrate for Vetinary
(Strain Rev. 1) Vaccine, Living
Assays and Tests, Vet-12 U se, see Closantel Sodiu111 Dihydrate
ATCC - American Type Culture Clostridium Botulinum Vaccine, Vet-230
Collection, address of, Vet-30 Clostridium Chauvoei Vaccine, Vet-230
Atomic Weights, Vet-6
Vet-A48 Index

C10stridium Novyi A1pha Antitoxin, Cutaneous App1ication, Powders for, Equine Serum Gonadotrophin for
Vet-192 Vet-137 Vetinary U se, see Serum Gonadotrophin
C10stridium Novyi Type B Vaccine, Equiva1ent Texts, European
Vet-231 PhannacopoeiaJ Vet-A3
C10stridium Perfringens Antitoxins,
Vet-193
D Error, Fiducia1 Limits of, Vet-14
Estimated Potency, Vet-14
Da1mation Insect F10wers, see Pyrethr1l111
C10stridium Perfringens Beta Antitoxin Etamiphylline, Vet-Sll
Flower
Vet-193 ' Etamiphy1line Camsilate, Vet-69,
Date, Effective dates, Vet-vi
C10stridium Perfringens Epsi10n Vet-Sll
Dec!aration of Interests, Vet-xi
Antitoxin, Vet-195 Etamiphy1line Injection, Vet-160
Decoquinate, Vet-54, Vet-S9
C10stridium Perfringens Vaccines, Etamiphy1line Tab1ets, Vet-161
Decoquinate Premix, Vet-159
Vet-233 Ethopabate, Vet-70, Vet-Sll
Definition of Terms, Vet-5
C10stridium Septicum Vaccine, Vet-235 Etorphine and Acepromazine Injection,
Definition, Status of, Vet-8
C10stridium Tetani Antitoxin, Vet-196 Vet-161
Deltamethrin, Vet-54, Vet-S9
C10stridium Tetani Vaccines, Vet-237 Etorphine Hydroch10ride, Vet-71
Deltamed1rin Pour-on, Vet-159
C10xacillin Benzathine, Vet-53, Vet-S8 European Pharmacopoeia, Vet-3, Vet-xx
Dembrexine Hydrochloride
C10xacillin Benzathine Intramammary European Pharmacopoeia Equiva1ent
Monohydrate, Vet-56
Infusion (Dry Cow), Vet-156 Texts, Vet-A3
Dembrexine Hydroch10ride Monohydrate
C10xaci~lin Benzathine Intramammary European Pharmacopoeia General
for Vetinary Use, see Dembrexine
InfuslOn (Dry Cow), Ampicillin Med10ds, Vet-A3
HJldrochloride lvlonohydrate
Trihydrate and, Vet-148 European Pharmacopoeia, General
Detomidine Hydroch10ride, Vet-57
C10xacillin Intramammary Infusion Notices of the, Vet-19
Diagnostic Preparations, Vet-328
(DC), Ampicillin and, Vet-148 Eva1uation of Efficacy of Veterinary
Dic!azuri1, Vet-58
C10xacillin Intramammary Infusion Vaccines and Immunosera, Vet-19
Dic!azuri1 for Vetinary Use, see Diclazuril
(DC), see Cloxacillin Benzathine Evaluation of Safety of each Batch of
Difloxacin Hydroch10ride Trihydrate,
Imra71lanmzal)1 Infusion (D1Y Cow) Immunosera for Veterinary U se,
Vet-59
C10xacillin Intramammary Infusion Vet-A20
Difloxacin Hydroch10ride Trihydrate for
(LC), Ampicillin and, Vet-146 Safety Veterinary Vaccines and
Vetinary Use, see Dzfioxacin
C10xacillin Intramammary Infusion Immunosera, Vet-16
Hydrochloride TrihJldrate
(LC), see Cloxacillin SodiuJ1I Evaluation of Safety of Veterinary
Dihydrostreptomycin Sulfate, Vet-61
ImranzammaJ)1 Infusion (Lactating Cozu Vaccines and Immunosera (5.2.6.),
Dihydrostreptomycin Sulfate for Vetinary
C10xacillin Sodium, Vet-S8 Vet-16
Use, see Dihydrostreptomycin Sulfate
Cloxacillin Sodium Intramammary Excipients, Use of, Vet-11
Dimetridazole, Vet-S9
Infusion (Lactating Cow), Vet-156 Expert Advisory Groups, Vet-x
Dimpy1ate, Vet-63, Vet-SIO
C10xacillin Sodium Intramammary Expression of Standards, Vet-6
Dinitolmide, Vet-SIO
Infusion (Lactating Cow), Ampicillin Extemporaneous Preparation, Status of,
Dip Concentrates, Vet-130, Vet-131
Sodium and, Vet-146 Vet-9
Diprenorphine Hydroch10ride, Vet-65
Cobalt Oxide, Vet-53 Extracts, Vet-131
Diprenorphine Injection, Vet-160
Coccidiosis Vaccine (Live) for Chickens, Extraneous Agents in Batches of
Duck Plague Vaccine (Live), Vet-242
Vet-238 Finished Product, Avian Live Virus
Duck Viral Hepatitis Type I Vaccine
Code of Practice, Vet-xi, Vet-xxii Vaccines, Tests for, Vet-A30
(Live), Vet-244
Co1d-water Vibriosis Vaccine for Extraneous Agents in Seed Lots Avian
Salmonids, Inactivated, Vet-323 Viral Vaccines, Tests for, Vet~A27
Colouring Agents, Permitted Eye Ointments, Vet-131
Alternatives, Vet-11 E Eye Preparations, Vet-131
Competent aud10rity, Definition of, ED so , Definition of, Vet-29 Eye Preparations of the BP (Vet),
Vet-5 Effective date, Vet-vi, Vet-xx Vet-131
Comp1ementary Medicines, Crude Efficacy of Veterinary Vaccines and
Drugs; Traditiona1 Herba1 and, Status Immunosera, Eva1uation of, Vet-19
of, Vet-17 Egg Drop Syndrome 76 (Adenovirus) F
Concentrates for Injections 01' Infusions, Vaccine, Vet-245
Febante1, Vet-72
Vet-136 EID so , Definition of, Vet-29
Febantel for Vetinary Use, see Febantel
Constant Weight, Definition of, Vet-6 Enilconazole, Vet-66
Fe1ine Calicivirus Vaccine, Inactivated,
Contagious Pustu1ar Dermatitis Vaccine Enilconazo1e for Vetinary Use, see
Vet-250
Living, Vet-241 ' Enilconazole
Fe1ine Ca1icivirus Vaccine, Living,
Contents of the General Notices, Vet-2 Enrofloxacin, Vet-67
Vet-252
Corresponds, definition of, Vet-5 Enrofloxacin for Vetinary Use, see
Fe1ine Chlamydiosis Vaccine
Co-tlimazine Injection, Vet-157 Enrofioxacin
(Inactivated), Vet-253
Co-trimazine Tab1ets, Vet-158 Enteric Redmouth Disease Vaccine for
Fe1ine Infectious Enteritis Vaccine,
Co-trimazine Veterinary Oral Powder, Rainbow Trout (Inactivated), Vet-326
Inactivated, Vet-254
Vet-158 Epidemic Tremor Vaccine, Living, see
Fe1ine I11fectious Enteritis Vaccine,
CRS, Vet-15 Infectious A vian Encephalomyelitis
Living, Vet-255
Crude Drugs, Macroscopical Vaccine J Living
Fe1ine Leukaemia Vaccine, Inactivated,
characteristics of, Vet-17 EPBRP, Vet-15
Vet-256
Crude Drugs; Traditiona1 Herbal and EPCRS, Vet-15
Fe1ine Viral Rhinotracheitis Vaccine
Comp1ementary Medicines, Status of, Equine Herpesvirus Vaccine, Inactivated,
Inactivated, Vet-257 '
Vet-17 Vet-247
Feline Viral Rhinotracheitis Vaccine
Cut~ne?us Application of the BP (Vet) , Equine Influenza Vaccine, Inactivated,
Living, Vet-259 '
LlqUld Preparations for, Vet-131 Vet-248
F enbendazo1e, Vet-73
page numbers in bold type relate to monograph titles Index Vet-A49

Fenbendazole for Vetinary Use, see


Fenbendazole 1 IZ
Fenbendazole Granules, Vet-162 ID so , Definition of, Vet-29 K (Vet) 2. Evaluation of Efficacy of
Fenbendazole Oral Suspension, Vet-163 Identification, Vet-12 Veterinary Vaccines and Immunosera,
Fenbendazole Veterinary Oral Paste, IMI - International Mycological Institute, Vet-19
Vet-164 address of, Vet-30 K (Vet) 3. Evaluation of Safety of Each
Fenbendazole Veterinary Paste, see Immunological Veterinary Medicinal Batch of Immunosera for Veterinary
Fenbendazole VeterinaJ)1 Oral Paste Products, Substances of Animal Origin Use, Vet-20
Fenthion, Vet-xxiv, Vet-S12 for the Production of, Vet-A14 Kelvin, Definition of, Vet-3l
Ferret and Mink Distemper Vaccine, Immunosera and Vaccines, Veterinary, Kilogram, Definition of, Vet-3l
Living, Vet-260 Evaluation of Safety (5.2.6.), Vet-A16
Fiducial Limits of Error, Vet-14 Implants, Vet-137
Fluanisone, Vet-74, Vet-S12
Flunixin Meglumine, Vet-75, Vet-S12
Impurities, Limitation of Potential, Vet-4
Impurity Limits, Status of, Vet-14
L
Flunixin Meglumine for Vetinary Use, L+/lO dose, Definition of, Vet-29
Indicators, Use of Chemical, Vet-7
see Flunixin Meglumine Lamb Dysentery Antiserum, see
Infectious Avian Encephalomyelitis
Foot and Mouth Disease (Ruminants) Clostridium Pelji'ingens Antitoxins
Vaccine, Living, Vet-267
Vaccine, Vet-261 Laryngotracheitis Vaccine, Living,
Infectious Bovine Rhinotracheitis
Foot-and-Mouth Disease (Ruminants) Vet-275
Vaccine, Living, Vet-268
Vaccine (Inactivated), see Foot and LD so , Definition of, Vet-29
Infectious Bursal Disease Vaccine,
lvIouth Disease (Rumina11ts) Vaccine L+ dose, Definition of, Vet-29
Inactivated, Vet-269
Formulated Preparations, Vet-37, Levamisole, Vet-77
Infectious Bursal Disease Vaccine,
Vet-127, Vet-140 Levamisole for Vetinary Use, see
Living, Vet-271
General Monographs, Vet-37, Levamisole
Infectious Chicken Anaemia Vaccine
Vet-127 Levamisole Injection, Vet-170
(Live), Vet-273
Formulated Preparations, Manufacture Levamisole Oral Solution, Vet-170
Infrared Reference Spectra, Vet-xx
of, Vet-9 Levomepromazine, Vet-S 13
Infrared Reference Spectra, Preparation
Fowl Cholera Vaccine (Inactivated), Lf dose, Definition of, Vet-29
of, Vet-S2
Vet-263 Light, Protected from, Definition of,
Infusions, Vet-136
Fowl Pox Vaccine, Living, Vet-264 Vet-12
Injections, Vet-135
Freshly Prepared, Definition of, Vet-lO Light, Subdued, Definition of, Vet-12
Injections, Gels for, Vet-137
Furunculosis Vaccine for Salmonids, Limits, Application of, Vet-13
Injections or Infusions, Concentrates for,
Inactivated, Vet-265 Lincomycin Hydrochloride, Vet-S13
Vet-136
Furunculosis Vaccine (Inactivated, Oil- Lincomycin Premix, Vet-171
Injections or Infusions, Powders for,
Adjuvanted, Injectable) for Salmonids, Lincomycin Tablets, Vet-171
Vet-136
see FUrLl1ZCUlosis Vaccine for Salmonids, Liquid Preparations for Cutaneous
Insect Flowers, Dalmation, see Pyrethru771
lIza ctiva ted Application of the BP (Vet), Vet-131
Flower
Liquid Preparations for Oral Use,
International Reference Preparation,
Vet-134
Vet-14
Lo/lO dose, Definition of, Vet-29
G International Standard, Vet-14
Loss on drying, Definition of
Gels for Injections, Vet-137 International System Of Units, Vet-30
Temperature Range, Vet-12
General Chapters, Vet-A3 International Units, Definition of,
Louping-ill Vaccine, Vet-279
General Methods of the European Vet-14
Lp/lO dose, Definition of, Vet-29
Pharmacopoeia, Vet-A3 Intramammary Infusions, Vet-131
lr/1 00 dose, Definition of, Vet-29
Intramammary Infusions, Antibiotics in,
General Monographs for Formulated Lufenuron, Anhydrous, Vet-78
Vet-17
Preparations, Vet-3, Vet-5 Lufenuron, Anhydrous for Vetinaly U se,
General Monographs (Formulated Intramammary Infusions of the BP (Vet),
see Lufenuron, Allhydrous
Preparations), Vet-127 Vet-132
Lungworm (Dictyocaulus Viviparus) Oral
General Notices, Vet-3 Intraruminal Devices, Vet-132
Vaccine, Living, Vet-280
Intrauterine Preparations, Vet-133
General Notices of the European
Pharmacopoeia, Vet-19 Intrauterine Preparations, for Vetinary
Use, see lntrauterine Preparations,
Gonadotrophin Injection, Serum,
Vet-165 I.P. - Collection Nationale de Culture de M
Microorganismes (C.N.C.M.) Institut Macroscopical characteristics of Crude
Graduated Glassware, Requirements for,
Vet-6 Pasteur, address of, Vet-30 Drugs, Vet-17
Italic Type, Significance of, Vet-8 Mallein P.P.D., see Mallein Purified
Granules, Vet-131
IU, Definition of, Vet-14 Proteill Derivative
Graphic Formulae, Status of, Vet-8
Ivermectin Injection, Vet-166 Mallein Purified Protein Derivative,
Griseofulvin, Vet-S 13
Griseofulvin Premix, Vet-166 Ivermectin Oral Solution, Vet-167 Vet-329
Ivermectin Pour-on, Vet-169 Mannheimia Vaccine (Inactivated) for
Guinea-pigs, used in assays and tests,
Ivermectin Veterinary Oral Paste, Cattle, Vet-281
Vet-14
Vet-168 Mannheimia Vaccine (Inactivated) for
Ivermectin Veterinary Paste, see Sheep, Vet-282
lvermectin VeterinaJ)! Oral Paste Manufacture of Formulated
H Preparations, Vet-9
Herbal and Complementary Medicines, Marbofioxacin, Vet-80
Crude Drugs; Traditional, Status of,
Vet-17
J
Justified and Authorised, Definition of,
Marbofioxacin for Vetinary U se, see
Marbofloxacin
Homoeopathic Medicines, Status of, Marek's Disease Vaccine, Living,
Vet-18 Vet-5
Vet-284
Vet-ASO Index

Measures, Expression of Weights and, Oral Liquids, Vet-135, see also name of ppm, Definition of, Vet-7
Vet-6 substance Preface, Vet-vii
Mec10fenamic Acid, Vet-S14 Oral Powders of tlle BP (Vet), Vet-138 Premixes, Vet-138
Meloxicam Injection, Vet-172 Oral Powders (Vet), Definition of, Premixes for Vetinary U se, see Premixes
Meloxicam Oral Suspension, Vet-173 Vet-138 Premixes of tlle BP (Vet), Vet-138
Mepivacaine Injection, Vet-174 Oral Powder, Vet-138, see also naJ1le of Preparations for Oral Use, Liquid,
Methyltestosterone, Vet-S 14 substance Vet-134
Metre, Definition of, Vet-31 Oral U se, Veterinary Semi-solid Procaine Benzylpenicillin Injection,
Metronidazole, Vet-S 14 Preparations for, Vet-138 Vet-181
Mice, used in assays and tests, Vet-15 Orbifloxacin, Vet-86 Production, Status of, Vet-9
Microkatal, Definition of, Vet-31 Orbifloxacin for Veterinary Use, see Protected from Light, Vet-12
Mole, Definition of, Vet-31 Orbifloxacin Pustular Dermatitis Vaccine, Living,
Molecular Formula, Status of, Vet-8 Orf Vaccine, see Contagious Pustular Contagious, Vet-241
Molecular Weight, Status of, Vet-8 Dermatitis Vaccine J Living Pyrethrum Extract, Vet-183
Morantel Tartrate, Vet-81 Ovine Enzootic Abortion Vaccine, Pyrethrum Flower, Vet-92
Morantel Hyprogen Tartrate for Vetinary Inactivated, Vet-293
Use, see Morantel Tartrate Oxfendazole, Vet-88, Vet-S15
Moxidectin, Vet-82
Moxidectin for Vetinary U se, see
Oxfendazole for Vetinary Use, see
Oxfendazole
R
Rabbit Haemorrhagic Disease Vaccine
lvIoxidectin Oxfendazole Oral Suspension, Vet-178
(Inactivated), Vet-308
Moxidectin Injedion, Vet-174 Oxyc1ozanide, Vet-89, Vet-S16
Rabies Vaccine (Live, Oral) for Foxes
Moxidectin Oral Drench, see Moxidectin Oxyc1ozanide Oral Suspension, Vet;. . 178
and Raccoon Dogs, Vet-304
Oral Solution, Vet-175 Oxytetracyc1ine Veterinary Oral Powder,
Rabies Veterinary Vaccine, Inactivated,
Moxidectin Oral Gel, see Moxidectin Vet-179
Vet-305
Oro11lucosal Gel, Vet-176
Radian, Definition of, Vet-31
Moxidectin Oral Solution, Vet-175
Moxidectin Oromucosal Gel, Vet-176 p Reagents, Description of, Vet-7
Recently Prepared, Definition of, Vet-10
Moxidectin Pour-on, Vet-176
Panels of Experts, Vet-x Reference Materials and Spectra, Vet-12
Mycoplasma Gallisepticum Vaccine
Parametric Release, Vet-4 Reference Preparation, International,
(Inactivated), Vet-286
Parenteral Preparations, Vet-135 Vet-14
Mycoplasmas, Test for Absence of,
Parenteral Preparations of the BP (Vet), Reference spectra, Vet-12
Vet-A22
Vet-137 Reference spectra, concordance of,
Myxomatosis Vaccine (Live) for Rabbits,
Parenteral Preparations, Vet-137, see also Vet-12
Vet-287
name of substa1lce Reference Spectra, Preparation of
Pastes, Veterinary Oral, Vet-137 Infrared, Vet-S2
N Pasteurella Vaccine (Inactivated) for
Sheep, Vet-294
Reference Substances and Reference
Preparations, Vet-15
Nandrolone Laurate, Vet-85, Vet-S15 Patents, Vet-vi Requirements, Pharmacopoeial, Vet-xxi
Nandrolone Laurate Injection, Vet-177 PD so , Definition of, Vet-29 Requirements, Release, Vet-4
NCIMB - National Collection of RQnidazole, Vet-93, Vet-S17
Pentobarbital, Vet-S16
Industrial and Marine Bacteria, Pentobarbital Injection, Vet-180 Ruminant E. Coli Vaccine, Inactivated,
address of, Vet-30 Vet-309
PFU, Vet-29
NCPF - National Collection of Pharmaceutical Preparations, Vet-127
Pathogenic Fungi, address of, Vet-30 Phenylbutazone, Vet-S 16
NCTC - National Collection of Type
Cultures, address of, Vet-30
Phenylbutazone Tablets, Vet-180
Ph. Eur., Definition of, Vet-3
s
NCYC - National Collection of Yeast Safety of Veterinary Vaccines and
Piperazine Citrate Tablets, Vet-181
Cultures, address of, Vet-30 Immunosera, Evaluation of, Vet-16
Piperonyl Butoxide, Vet-90
N eonatal Piglet Colibeicillosis Vaccine Salmonelia Dublin Vaccine, Living,
Polystyrene, Vet-S3, Vet-S4
(Inactivated), see Porcine E. Coli Vet-310
Porcine Actinobacillosis Vaccine,
Vaccine J Inactivated Salmonella Enteritidis Vaccine
Inactiva ted, Vet-295
Newcastle Disease and Avian Infectious (Inactivated) for Chickens, Vet-311
Porcine E. Coli Vaccine, Inactivated,
Bronchitis Vaccine, Living, Vet-288 Salmonella Enteritidis Vaccine (Live,
Vet-298
N ewcastle Disease Vaccine, Inactivated, Oral) for Chickens, Vet-312
Porcine Enzootic Pneumonia Vaccine
Vet-289 Salmonella Typhimurium Vaccine
(Inactivated), Vet-297
Newcastle Disease Vaccine, Living, (Inactivated) for Chickens, Vet-314
Porcine Parvovirus Vaccine, Inactivated,
Vet-291 Salmonella Typhimurium Vaccine· (Live,
Vet-300
Nitroxinil, Vet-86, Vet-S15 Oral) for Chickens, Vet-315
Porcine Progressive Atrophic Rhinitis
Nitroxinil Injection, Vet-177 Second, Definition of, Vet-31
Vaccine, Inactivated, Vet-301
Notices, Vet-vi Selamectin, Vet-93
Potassium Selenate, Vet-91
Selamectin for Vetinary U se, see
Potency, Estimated, Vet-14
o Potency of Antibiotics, Vet-14
Potency, Stated, Vet-14
Selamecún
Semi-solid Preparations for Oral U se,
Official, Definition of, Vet-3 Veterinary, Vet-138
Pour-on Preparations, Vet-130
Official Standards, Vet-4 Serum Gonadotrophin, Vet-76
Powders for Cutaneous Application,
Official Titles, Vet-7 Serum Gonado1rophin for Injection,
Vet-137
Omissions, Vet-xx Vet-165
Powders for Injections or Infusions,
Omissions, List of, Vet-xxiv Serum Gonadotrophin Injection, Vet-165
Vet-136
Oral Liquids, Vet-134 SI Units, Vet-30
Powders, Topical, Vet-137
Oral Liquids of the BP (Vet), Vet-135 SI Units, Use of, Vet-6
page numbers in bold type relate to monograph titles Index Vet-A51

Similar, definition of, Vet-5 Sulfadoxine and Trimethoprim Injection, Trimethoprim and Sulfadiazine Tablets,
Sodium Calcium Edetate Concentrate Vet-185 see Co-trimazine Tablets
for Veterinary Use, Sterile, Vet-184 Sulfamerazine, Vet-104 Trimethoprim and Sulfadiazine
Sodium Calcium Edetate Intravenous Sulfametoxypyridazine, Vet-105, Veterinary Oral Powder, see Co-
Infusion for Veterinary Use, Vet-184 Vet-S18 trimazine VeterinaJY Oral Pozvder
Sodium Selenite, Vet-95 Sulfametoxypyridazine for Vetinary Use, Trimethoprim Injection, Sulfadoxine
Solubility, Definition of Terms Used, see Sulfametoxypyridazine and, Vet-185
Vet-ll Sulfanilamide, Vet-105 Turkey Infectious Rhinotracheitis
Solubility, Status of, Vet-ll Sulfaquinoxaline, Vet-106, Vet-S18 Vaccine (Live), Vet-322
Solvents for Pharmacopoeial Tests, Sulfathiazole Sodium, Vet-107, Vet-S18 Tylosin, Vet-1l4, Vet-S19
Vet-12 Supplementary Chapters, Contents of Tylosin for Vetinary Use, see Tylosin
Spectinomycin Sulfate Tetrahydrate, the, Vet-A4l Tylosin Injection, Vet-186
Vet-96 Sutures, Vet-333 Tylosin Phosphate, Vet-1l5
Spectinomycin Sulfate Tetrahydrate for Swine Erysipelas Vaccine, Inactivated, Tylosin Phosphate Bulle Solution for
Vetinary U se, see Spectino711ycin Sulfate Vet-318 Vetinary U se, see Tylosin Phosphate
Tetrahydrate Swine Influenza Vaccine, Inactivated, Tylosin Tartrate, Vet-1l7
SPF, Vet-29 Vet-321 Tylosin Tartrate for Vetinary Use, see
SPF, Chicken Flodes Free from Swine-Fever Vaccine (Live, Prepared in Tylosin Tartrate
Specified Pathogens for the Production Cell Cultures), Classical, Vet-319
and Quality Control of Vaccines, Synonyms, Approved, Vet-34
Vet-A9
Spiramycin, Vet-98
u
Udder-washes, Vet-l30
Spot-on Preparations, Vet-l30
Sprays, Veterinary, Vet-l30
T Units, International, Vet-14
Tablets, Vet-139 Units, International System Of, Vet-30
S.S.l. - Statens Serum Institut, address
Tablets of the BP (Vet), Vet-139 Units of Biological Activity, Definition
of, Vet-30
Tablets, Vet-139, see also nanze of of, Vet-14
Standard, International, Vet-14
substance
Standards,' Official, Vet-4
Tamper-evident Container, Definition of,
Stated Potency, Vet-14
Sterile Braided Silk Suture in
Vet-16
Tamper-proof Container, Definition ,of,
v
Distributor, Vet-337 Vaccine Strain, Choice of, Vet-9
Vet-16
Sterile Catgut in Distributor, Vet-333 Vaccines and Immunosera, Veterinary,
Teat Dips, Vet-l30
Sterile Linen Thread in Distributor, Evaluation of Safety (5.2.6.), Vet-A16
Teat Sprays, Vet-130
Vet-335 Vaccines, Veterinary, Cell Cultures for
Technical Changes to Monographs,
Sterile Non-absorbable Strands in the Production of (5.2.4.), Vet-A12
Vet-xxiv
Distributor, Vet-334 Valnemulin Hydrochloride, Vet-1l8
Temperature, Expression of, Vet-6
Sterile Polyamide 6 Suture in Valnemulin Hydrochloride for Vetinary
Terminology Used for Vaccines, Vet-9
Distributor, Vet-336 Use, see Valnemulin Hydrochloride
Terminology Used in Monographs on
Sterile Polyamide 6/6 Suture in Vedaprofen, Vet-120
Biological Products, Vet-9
Distributor, Vet-336 Vedaprofen for Vetinary U se, see
Terms, Definition of, Vet-5
Sterile Poly( ethylene terephthalate) Vedaprofen
Test for Absence of Mycoplasmas,
Suture in Distributor, Vet-336 Veterinary Immunosera, Vet-189
Vet-A22
Sterile Sodium Calcium Edetate Veterinary Liquid Preparations for
Testosterone Phenylpropionate, Vet-108,
Concentrate for Veterinary Use, Cutaneous Application, Vet-130
Vet-S19
Vet-184 Veterinary Oral Pastes, Vet-137
Tests and Assays, Vet-12
Sterilisation, Methods of, Vet-lO Veterinary Oral Powders, Vet-138
Tetanus Antitoxin, see Clostridium Tetani
Storage Statements, Status of, Vet-15 Veterinary Vaccines, Vet-9, Vet-197
Antitoxin
Subdued light, Vet-12 Terminology Used, Vet-9
Tetanus, see Clostridium Tetani Vaccine
Subdued Light, Definition of, Vet-12 Veterinary Vaccines and Immunosera,
Tiamulin, Vet-108
Subsidiary Titles, Status of, Vet-7 Vet-19
Tiamulin for Vetinary Use, see Tiamulin
Substances for Pharmaceutical Use, Evaluation of, Efficacy of, Vet-A19
Tiamulin Hydrogen Fumarate, Vet-110
Vet-37 Veterinary Vaccines and Immunosera,
Tiamulin Hydrogen Fumarate for
Substances of Animal Origin for the Evaluation of Safety of, Vet-A16
Vetinary U se, see Tiamulin Hydrogen
Production of Immunological Veterinary Vaccines, Cell Cultures for
Fumarate
Veterinary Medicinal Products, the Production of, Vet-A12
Titles, Abbreviated, Vet-7
Vet-A14 Vibriosis Vaccine for Salmonids,
Titles, Official, Vet-7
Suggested Methods, Status of, Vet-14 Inactivated, Vet-325
Titles, Subsidiary, Vet-7
Sulfadiazine Injection, Trimethoprim Visual Comparative Tests, Vet-12
Topical Powders, Vet-137
and, see Co-trimazine Injection
Topical Powders of the BP (Vet),
Sulfadiazine Tablets, Trimethoprim and,
see Co-trimazine Tablets
Sulfadiazine Veterinary Oral Powder,
Vet-137
Traditional Herbal and Complementary w
Medicines, Crude Drugs; Status of, Water Bath, Definition of, Vet-7
Trimethoprim and, see Co-trimazine
Vet-17 Water, Potable, Vet-lO
VeterinaJY Oral Pozvder
Tridabendazole, Vet-l13 Websites, Vet-xxii
Sulfadimethoxine Sodium, Vet-102
Tridabendazole for Veterinary Use, Weights and Measures, Expression of,
Sulfadimethoxine Sodium for Veterinary
Vet-l13 Vet:.6
Use, see Sulfadimethoxine Sodium
Trimethoprim, Vet-S 19 Weights, Atomic, Vet-6
Sulfadimidine, Vet-100, Vet-S17
Trimethoprim and Sulfadiazine Injection, Working Parties, Vet-x
Sulfadimidine Injection, Vet-184
see Co-trimazine Injection
Sulfadoxine, Vet-S 17
Vet-A52 Index

x
Xylazine Hydrochlo1'ide, Vet-121
Xylazine Hydrochloride for Veterinary
Use, see Xylazine Hydrochloride

y
Ye1'siniosis Vaccine (Inactivated) fo1'
Salmouids, see Enteric Redmouth
Disease Vaccine for Rainbow TroLtt
(Inactivated)

Вам также может понравиться