You are on page 1of 1484

H El

EIGHTH EDITION
Volume 1

Published in accordance with the


Convention on the Elaboration of a European Pharmacopoeia
(European Treaty Series No. 50)

COUNCIL OF EUROPE

~o¿tr!" O'"dom" ,,,.,


Quality 01 Medicines & HealthCare CONSE IL DE L EUROPE

Council of Europe
Strasbourg
The European Pharmacopoeia is published by the Directorate for the Quality ofMedicines & HealthCare
ofthe Council ofEurope (EDQM).

© Couneil ofEurope, 67075 Strasbourg Cedex, Franee - 2013


AH rights reserved. Apart from any fair dealing for the purposes of research or private study, this
publication may not be reproduced, sto red or transmitted in any form or by any means without the prior
permission in writing of the publisher.

ISBN: 978-92-871-7525-0
CONTENTS
VOLUME 1
1. PREFACE
II. INTRODUCTION v

III. EUROPEAN PHARMACOPOEIA COMMISSION ix


IV. CONTENTS OF THE EIGHTH EDITION xxi
GENERAL CHAPTERS
1. General notices
2. Methods of analysis 11
2.1. Apparatus 13
2.2. Physical and physicochemical methods 19
2.3. Identification 117
2.4. Limit tests 125
2.5. Assays 153
2.6. Biological tests 173
2.7. Biological assays 227
2.8. Methods in pharmacognosy 269
2.9. Pharmaceutical technical procedures 283
3. Materials for containers and containers 371
3.1. Materials used for the manufacture of containers 373
3.2. Containers 407
4. Reagents 423
5. General texts 551
GENERAL MONOGRAPHS 739
MONOGRAPHS ON DOSAGE FORMS 777
MONOGRAPHS ON VACCINES FOR HUMAN USE 815
MONOGRAPHS ON VACCINES FOR VETERINARY USE 919
MONOGRAPHS ON IMMUNOSERA FOR HUMAN USE 1027
MONOGRAPHS ON IMMUNOSERA FOR VETERINARY USE 1035
MONOGRAPHS ON RADIOPHARMACEUTICAL PREPARATIONS AND STARTING MATERIALS FOR
RADIOPHARMACEUTICAL PREPARATIONS 1043
MONOGRAPHS ON SUTURES FOR HUMAN USE 1115
MONOGRAPHS ON SUTURES FOR VETERINARY USE 1125
MONOGRAPHS ON HERBAL DRUGS AND HERBAL DRUG PREPARATIONS 1133
MONOGRAPHS ON HOMOEOPATHIC PREPARATIONS 1427

VOLUME2
MONOGRAPHS 1457
INDEX 3603

Note: on the first page of each chapter/section there is a list of contents.


1
1
1
1
1
1
1
1
1
1
1
1
1
1
1
1
1
1
1
1
1
1
1
1
1
1
1
1
1
1
1
1
1
1
1
1
1
1
1
1
1
1
1
1
1
1
1
1
1
1
1
1
1
1
1
11
11
11
11
11
11
1 1
1 1
1 1
1 1
1 1
1 1
1 1
1 1
1 1
1 1
1 1
1 1
1 1
1 1
1 1
1 1
i 1
i 1
I 1
1 1
I 1
I 1
I 1
I 1
I 1
I 1
I 1
I 1
I 1
I 1
I 1
I 1
I 1
I 1
I 1
I 1
I 1
I 1
I 1
I '
EUROPEAN PHARMACOPOEIA 8.0 Preface

I~ PREFACE
The European Pharmacopoeia was inaugurated in 1964 will now work on harmonising their policies and approaches
through the Convention on the Elaboration of a European towards monograph development by drafting what have been
Pharmacopoeia under the auspices of the Council of termed as a working title "Good Pharmacopoeial Practices".
Europe. The 8th Edition will be published just before the Convergence in policies, e.g. with regard to control of
50 th Anniversary of the European Pharmacopoeia. The impurities by applying ICH Q3 principIes, will facilitate future
work of the European Pharmacopoeia has gone through a collaboration and harmonisation.
remarkable development since the first difficult years to its The implementation date for the 8th Edition is 1 January 2014
current strong position. The 3-year cyde of publication with and this edition will, over the next 3 years, be augmented with
thrice-yearly supplements has proven to be an efficient way to 8 supplements containing the texts adopted at the sessions
publish and update the results of the work of the European of the European Pharmacopoeia Commission. As ever, it
PharmacopoeiaCommission and its Expert Groups and is published in the 2 officiallanguages of the Council of
Working Parties almost in real-time. Europe, Le. English and French, both as a printed version and
The monographs of the Pharmacopoeia, both specific and electronically (online and on a USB key). It is noteworthy
general, together with other texts made mandatory by virtue of that certain member states undertake national or regional
reference to them in monographs, are applicable throughout translations, which they incorporate into their own national
the 37 member states, and the European Union, which is pharmacopoeias.
also a signatory party to the European Pharmacopoeia The work programme of the European Pharmacopoeia is
Convention. This means that in addition to applicability decided by the European Pharmacopoeia Commission, the
in al! its member states, the European Pharmacopoeia has governing body of the Pharmacopoeia. Elaboration and
a special role in regulatory processes within the European approval of monographs and other texts proceed through an
Union. In addition to the 38 signatories of the European efficient and transparent process, which is based on scientif1c
Pharmacopoeia Convention, there are also a large number of co-operation between the members of the various Groups
observers, comprising the World Health Organization and of Experts and Working Parties set up by the European
23 countries, of which 16 are non-European. The quality Pharmacopoeia Commission. These experts give their time,
standards developed through the European Pharmacopoeia expertise and experience to produce public standards of the
therefore have an impact on the quality of medicinal products highest quality - standards that are continually revised in line
and substances far beyond the European region. Since the with scientif1c developments. The members of these groups
entry into force of the 7th edition, Ukraine has become a come from regulatory authorities, official medicines control
new member of the European Pharmacopoeia Convention laboratories, pharmaceutical and chemical manufacturers,
(in 2013), while the Republic of Guinea and Singapore have universities and research institutions. AH monographs are
become new observers (in 2012). experimentally verif1ed and submitted for public consultation
by online publication in Pharmeuropa, the forum of the
The 8 founder countries of the Convention realised in 1964 European Pharmacopoeia, before adoption and publication.
that manufacturing and quality control standards for medicinal
products on the European Market had to be harmonised for The growing number of monographs and the need to keep
reasons of public health and to facilitate free movement of them updated represents an increased workload and an
these products. Since then, the pharmaceutical world has increased need for experts with access to experimental
become globalised and international harmonisation among the facilities. The working procedures for the elaboration of
3 major pharmacopoeias (European Pharmacopoeia, Japanese monographs are:
Pharmacopoeia and United States Pharmacopeia) became a - Procedure 1: traditional elaboration by Groups of Experts
logical development. Harmonisation activities among these and Working Parties.
3 pharmacopoeias started in 1989 when the Pharmacopoeial - Procedure 2: adaptation of national monographs. (This
Discussion Group (PDG) was set up. The PDG has been procedure is no longer used since the work has been
working on monographs on widely-used excipients and 62 are completed.)
induded in its work programme. Soon after the PDG began - Procedure 3: elaboration of monographs on chemical
work, it was recognised that the absence of harmonised substances produced only by one manufacturer and
general methods represented a significant obstade. A wide typically close to patent expiry. The manufacturer and
range of general methods (35) have since been added to the national pharmacopoeia authority in the country where
work programme, induding those from the work of the the substance is produced elaborate preliminary drafts
International Conference on Harmonisation (ICH) and, in and check the requirements experimentally. This results
particular, its guideline on setting specifications (Q6A). To in a draft that is then reviewed by a Group of Experts
date, 28 of the 35 general methods and 43 of 62 excipient or Working Party and processed in the usual way by
monographs have been harmonised. Detailed information on public enquiry. (This procedure has been integrated into
the work programme of the PDG is published regularly in Procedure 4.)
Pharmeuropa and in General Chapter 5.8. Pharmacopoeial - Procedure 4 (P4): a modif1ed version of Procedure 3
harmonisation. for substances still under patent, which was introduced
However, it is evident that harmonisation between the by the European Pharmacopoeia Commission in 2002.
3 ICH regions is not enough in today's world, where a The P4 procedure involves collaboration between the
high percentage of Active Pharmaceutical Ingredients manufacturer of the substance and a Working Party
(APIs) come from outside Europe, Japan and the USA. In solely composed of representatives of authorities and
early 2012, the WHO took the initiative and convened the the EDQM. Together they prepare a draft monograph
pharmacopoeias of the world for their first international with experimental verification by the EDQM laboratory
meeting in Geneva. The discussions at this level dearly and/or by national pharmacopoeia authorities or Official
identified the need to strengthen col!aboration among Medicines Control Laboratories before publication for
pharmacopoeias worldwide. Based on the experience and public enquiry.
challenges with existing harmonisation initiatives such as - Procedure 5: applies to monographs on raw materials and
PDG that focus on retrospective harmonisation, it was stocks for homoeopathic preparations authorised for use
decided to take a different approach. World pharmacopoeias in the member states. The work is co-ordinated by the
Preface EUROPEAN PHARMACOPOEIA 8.0

EDQM and overseen by the HOM Working Party. This promote reduction and refinement of animal use, e.g. serology
procedure was introduced by the European Pharmacopoeia assays or single dilution assays for diphtheria, tetanus,
Commission in 2011. acellular pertussis and rabies (veterinary/human) vaccines.

Work under the P4 procedure has successfully continued A number of important European Pharmacopoeia activities
during the elaboration of the 7th Edition. Already have been initiated over the last few years, such as the
59 P4 monographs for chemical substances have been adopted establishment of a PAT (Process Analytical Technology)
Working Party based on a request from the EMA. PAT tools
by the European Pharmacopoeia Commission. Under the
P4 procedure for chemical substances, a pilot project on make it possible to use additional information collected
throughout the production process, e.g. use of NIR
bilateral prospective harmonisation of active substance
monographs with the USP was initiated and so far has resulted (near-infrared spectrophotometry) to determine tablet
in the adoption of4 harmonised monographs. As the P4 content. Chapter 2.2.40 Near Infrared Spectrophotometry was
revised to introduce PAT-related concepts such as in-line and
procedure for chemical substances has been such a success,
the European Pharmacopoeia Commission decided in 2009 on-line measurements. This was done in close consultation
to initiate a similar process for biological substances. The with the EMA's CVMP/CHMP Quality Working Party so that
so-called P4-BIO pro ce dure takes account of the increasing it would be aligned with the on-going revision ofthe EMA's
Note for guidance on NIR. The revised chapter was adopted by
number and importance of biologically-derived active
substances and biosimilars on the European market. Two the European Pharmacopoeia Commission at its November
monographs elaborated by the P4-BIO procedure have already session in 2012 and it will be complemented by the revised
been adopted by the European Pharmacopoeia Commission. EMA Guideline on the use of near infrared spectroscopy by the
pharmaceutical industry and the data requirements for new
The work on controlling impurities, a particular strength of submissions and variations, which is expected to be finalised
the European Pharmacopoeia, has continued. Monographs in 2013. The General Notices will be updated to take account
are evaluated and approved by the Competent Authorities of of real-time releas e testing, which will be done once the
member sta tes, and the impurity pro files covered by these EMA Guideline has been adopted. The alternative, opUonal
monographs reflect the existing, approved routes of synthesis. Chapter 2.9.47. Demonstration of Uniformity of Dosage
A revision mechanism is in place for newly-approved products Units (UDU) using large sample sizes that couId be used to
(e.g. new sources, new routes). The analytical methods in replace conventional UDU testing has also been adopted. The
monographs are robust and validated and are based on PAT Working Party is now reflecting on the need for new
coHaborative laboratory testing. The monographs reflect general chapters.
regulatory practice by applying ICH guideline Q3A R to the A Heavy Metals Working Party has been created to implement
pharmacopoeial substances. The guideline of the European the EMA's Guideline on metal catalysts and metal reagent
Medicines Agency (EMA) concerning the control of genotoxic residues and the future ICH Q3D guideline. The terms of
impurities, which came into force in 2007, has also been taken reference for this working party are to draft a general chapter
into account and has resulted in a revision of the general to implement the guideline, to assess the capability of the
monograph on Substances for Pharmaceutical use (2034) and current Chapter 2.4.8. Heavy metals to appropriately limit the
adoption of 3 general methods for genotoxic impurities. priority metals mentioned in the guideline and to consider
the introductiol1 of instrumental screening methods, whilst
The European Pharmacopoeia Commission is also continuing also allowing other means of ensuring compliance where
its efforts to reduce the number of animals needed to perform possible and justified. Since the ICH guideline has not yet
tests (implementation of the 3Rs principIe, i.e. replacing, been published, it was decided to introduce a new General
refining and reducing the use of animal s in tests). 1t has Chapter 5.20. Metal catalyst or metal reagent residues and a
aligned pharmacopoeial texts with VICH Guidelines 41 (test new General Method 2.4.20. Determination of metal catalyst
for reversion to virulence) and 44 (developmental safety or metal reagent residues. General Chapter 5.20 reproduces
tests), which came into force in 2008 and 2009, respectively, the EMA's guideline on the specification limits for residues
and with Directive 201O/63/EU of the European Parliament of metal catalysts or metal reagents. It is applicable to all
and of the Council of 22 September 2010 on the protection excipients and APIs, except those for veterinary use only, but
of animals used for scientific purposes. Furthermore, to not to starting materials or herbals. General Method 2.4.20
ensure consistency with European regulations the European describes the general approach for the determination of
Pharmacopoeia Commission has harmonised all the metal catalyst or metal reagent residues in substances for
veterinary vaccine monographs, including monographs on pharmaceutical use. As the chemical composition of the
vaccines intended for species that were outside the scope of the substances and the specification limits for the metal(s) of
VICH Guidelines. As a consequence, the safety tests and the interest vary considerably, it is not possible to describe al!
tests for increased virulence performed during development of suitable sample preparation and measurement methods.
the vaccines have been harmonised, which will greatly reduce Therefore, any method that fulfils the requirements described
the number of animals used for testing. in this chapter may be used. Both General Chapter 5.20 and
The European Pharmacopoeia Commission continuously General Method 2.4.20 have been published in European
revises general texts and monographs, re-evaluates Pharmacopoeia supplement 7.7. A cross-reference is to be
the relevan ce of animal tests mentioned in European introduced into the general monograph on Substances for
Pharmacopoeia texts and, if deemed appropriate, includes Pharmaceutical Use (2034) to make General Chapter 5.20
alternative methods. The general monograph on Vaccines for legally-binding.
veterinary use (0062) was revised to delete the TABST (target As a follow-up to the Workshop on thefuture ofmonographs
animal batch safety test), except in 'particular circumstances' in the field of biologicals organised by the EDQM in
to cover the need to perform, on an ad hoc basis, further February 2011, 2 new working parties have been created: (1)
testing and safety tests in particular. In the interest of the 3Rs, the Raw Materials for the Production of Cellular and Gene
the European Pharmacopoeia Commission also adopted the Transfer Products W orking Party, which will elaborate texts on
deletion of the TABST from the European Pharmacopoeia raw materials such as antibodies, basal media (for ceH culture),
for all veterinary vaccines. Currently, at the European serum/serum replacements, growth factors and cytokines, and
Pharmacopoeia leve!, animals are no longer used in the (2) the Host Cell Proteins Working Party, which will draft
testing of medicinal products derived from human blood and recommendations with regard to the development, validation
plasma. In many cases, in vivo testing has been replaced by and use of in-house or commercial kits or test methods
in vitro methods for human and veterinary vaccines. For the for the detection and quantification of host cell-derived
remaining in vivo assays, different strategies are being used to proteins. In addition, the scope of the P4-BIO pilot project

ii
EUROPEAN PHARMACOPOEIA 8.0 Preface

has been extended in order to elaborate monographs on and one multi-source product) allocated to it by the European
one monoelonal antibody, one hormone/enzyme and one Pharmacopoeia Commission, while addressing issues related
pegylated protein. The P4-BIO working party has also been to the elaboration of chemically-defined finished products
asked to elaborate one finished product monograph. The monographs in order to assess whether such monographs
terms of reference of the Cel! Therapy Products W orking Party should be elaborated by the European Pharmacopoeia in the
have also been extended in order to elaborate a general text future, and (2) the Second Identification Test Working Party,
dealing with microbiological control of organs and tissues for which will prepare a guidance document that defines the
human use, including preservation and other related media. criteria for inelusion of a second series of identification tests
As a consequence, the Working Party has been renamed the (solely intended to be carried out in pharmacies) in individual
Cell Therapy Products, Tissues and Organs Working Party. monographs and will review the methods and instrumentation
The production section of the monograph Human normal available in pharmacies for this purpose.
immunoglobulin for intravenous administration (0918) has Compliance with the EU REACH (Registration, Evaluation,
been revised due to experience with an immunoglobulin Authorisation and Restriction of Chemical substances)
preparation that caused an increased rate of thromboembolic Regulation has posed a significant challenge and this issue
complications. In light of concerns for public health associated has been high on the agenda of the current Presidium.
with these thromboembolic events, the revised monograph The European Pharmacopoeia Commission approved the
will be implemented by the accelerated procedure. request for the revision of215 monographs as a consequence
Due to the increasing number of fraudulent activities and cases of the EU REACH Regulation and already several revised
of adulteration, the European Pharmacopoeia Commission monographs have been adopted.
has decided to add a new section, Potential Adulteration, During the past 3 years 1 have had the honour, pIe asure and
under § 1.4. MONOGRAPHS of the General Notices. The privilege to serve the European Pharmacopoeia Commission
need to in elude this section in individual monographs will as its 16th elected Chair. The task has been challenging, but also
be decided by the European Pharmacopoeia Commission interesting and rewarding because of the insights it has given
on a case-by-case basis. The objective of this section is to me into the various aspects of the development work that goes
make relevant information available to users of the European into the drafting of the quality standards provided by the texts
Pharmacopoeia to ensure the proper quality of medicinal of the Pharmacopoeia. It has also given me an insight into the
products (i.e. active substances, excipients, intermediate many other important areas in which the EDQM is involved.
products, bulk products and finished products). The new 1 wish to thank all the members of the European
version of the General Notices was adopted by the European Pharmacopoeia Commission for the trust and support that
Pharmacopoeia Commission at its 140 th session. In relation allowed us to make substantial progre ss 011 a host of topics.
to this issue of adulteration, the Council of Europe and
its EDQM have adopted a multi-Ievel, anti-counterfeiting 1 would like to thank the Director of the EDQM, Dr Susanne
strategy comprising various aspects, such as legislative actions Keitel, my two vice-chairs, Prof. Jos Hoogmartens and
against pharmaceutical crime by means of the Medicrime Ms An Le, the Secretary to the European Pharmacopoeia
Convention. This Convention is the first international treaty Commission, Ms Cathie Vielle, and her two deputies,
against counterfeit medical products and similar crimes Dr Emmanuelle Charton and Dr Michael Wierer, for their
involving threats to public health. In addition, the EDQM is excellent work and support during my time as Chair. Together
developing eTACT; an anti-counterfeiting traceability service as the Presidium, we have managed to work very effectively to
for medicines. The aim of eTACT is to ensure the traceability guide the work of the European Pharmacopoeia Commission.
of individual packs of medicines using mass serialisation. It Finally, 1 would like to thank all the chairs and experts
would allow each pack of medicine to be traced and verified involved in the development of the European Pharmacopoeia
by the different stakeholders in the legal supply chain. Patients and the staff of the EDQM for their support. Their availability,
would also be allowed to verify the authenticity of their good advice and high quality input have made our work
medication. Governance of the eTACT system would be the possible and a pleasure to do.
responsibility of the EDQM as a public, inter-governmental
organisation that is able to ensure the confidentiality of the Dr Marianne Ek,
data handled by the system.
Chair of the European Pharmacopoeia Commission
Two additionalnew working parties have also recently been
created: (1) the Finished Product Monographs Working Party,
which aims to draft 2 monographs (i.e. on one single-source February 2013

iii
EUROPEAN PHARMACOPOEIA 8.0

iv
EUROPEAN PHARMACOPOEIA 8.0 Introduction

Ile INTRODUCTION
The European Pharmacopoeia is prepared under the auspices In accordance with the terms of the Convention, the
of the Council of Europe in accordance with the terms of the contracting parties undertake to take the necessary
Convention on the Elaboration of a European Pharmacopoeia measures to ensure that the monographs of the European
(European Treaty Series No. 50) ('the Convention') as amended Pharmacopoeia shall become the official standards applicable
by the protocol to the Convention (European Treaty Series within their respective territories.
No. 134), signed by the governments of 37 member states
(Austria, Belgium, Bosnia and Herzegovina, Bulgaria, Croatia, PURPOSE OF THE EUROPEAN PHARMACOPOEIA
Cyprus, Czech Republic, Denmark, Estonia, Finland, France, The purpose of the European Pharmacopoeia is to pro mote
Germany, Greece, Hungary, Iceland, Ireland, Italy, Latvia, public heaIth by the provision of recognised common
Lithuania, Luxembourg, Malta, Montenegro, Netherlands, standard s for the quality of medicines and their components.
Norway, Poland, Portugal, Romania, Serbia, Slovak Republic, Such standards are to be appropriate as a basis for the safe
Slovenia, Spain, Sweden, Switzerland, 'the former Yugoslav use of medicines by patients. In addition, their existence
Republic of Macedonia', Turkey, Ukraine and United facilitates the free movement of medicinal products in Europe
Kingdom) and by the European Union. and beyond.
The preparation of the European Pharmacopoeia is the European Pharmacopoeia monographs and other texts are
responsibility of the European Pharmacopoeia Commission designed to be appropriate to the needs of:
('the Commission'), appointed in accordance with article 5 - regulatory authorities;
of the Convention. It is composed of delegations appointed - those engaged in the quality control of medicinal products
by the contracting parties. Each delegation consists of not and their components;
more than 3 members chosen for their competence in matters
- manufacturers of medicinal products and their components.
within the functions of the Commission.
The European Pharmacopoeia is widely used internationally.
Observers from non-member states and international As globalisation and expansion in international trade present
organisations are admitted to sessions of the Commission a growing need to develop global quality standard s for
in accordance with the Rules of Procedure. Observers are at medicines, the Commission works closely with all users of the
present admitted from: Albania, AIgeria, Argentina, Armenia, Pharmacopoeia worldwide.
Australia, Brazil, Canada, China, Georgia, Israel, Madagascar,
Malaysia, Moldova, Morocco, Republic of Belarus, Republic SEAT OF THE EUROPEAN PHARMACOPOEIA
of Guinea, Republic of Kazakhstan, Republic of Singapore, COMMISSION
Russian Federation, Senegal, Syria, Tunisia, United States of The seat of the European Pharmacopoeia Commission is
America and the World Health Organization. situated in Strasbourg, the headquarters of the Council of
The Convention is open for signature by European countries Europe.
and observer status can serve to familiarise European
countries intending to become signatories with the working GENERAL PRINCIPLES
methods of the Commission. The Commission recognises that General rules for interpretation of the texts of the European
relations with countries outside Europe are essential in view Pharmacopoeia are given in the General Notices. The
of the globalisation of the suppIy chain for pharmaceuticals. following information should also be noted.
Observer status for non-European countries helps to foster The general principIes applied in the elaboration of
these relations by facilitating regulatory partnerships and the monographs of the European Pharmacopoeia are laid down
exchange of information and working documents. in procedures and in technical guides available on the EDQM
The functions of the Commission established by article 6 of website. The principIes applied are revised from time to
the Convention as amended by the protocol are: time without complete retrospective application so that
monographs already published may not always follow the
ArUcIe 6
latest recommendations, but wherever an issue with an impact
"Subject to the provision of Article 4 of the present on public health is identified, monographs are revised.
Convention, the functions of the Commission shall be: It is recognised that general chapters are also used elsewhere
(a) to determine the general principIes applicable to the than in the monographs of the European Pharmacopoeia; in
elaboration of the European Pharmacopoeia; these circumstances users are recommended to consult the
(b) to decide upon methods of analysis for that purpose; relevant technical guide, which gives extensive information on
the application of many of the methods.
(c) to arrange for the preparation of and to adopt monographs
General and individual monographs. The standards of the
to be included in the European Pharmacopoeia and;
European Pharmacopoeia are represented by general and
(d) to recommend the fixing of the time limits within which individual monographs. The use of general monographs has
its decisions of a technical character relating to the European developed in recent years to provide standards that best fulfil
Pharmacopoeia shall be implemented within the territories of the aims stated aboye and meet the needs of users. From the
the contracting parties:' 4 th Edition, the scope of general monographs was extended,
The European Directorate for the Quality of Medicines & except where otherwise stated, to cover products where there
HealthCare (EDQM) of the Council of Europe supports is no individual monograph. It is now usually necessary
the Commission in the elaboration and revision of texts of to apply one or more general monographs along with any
the European Pharmacopoeia by providing the scientific individual monograph. Where a substance is subject to the
secretariat. In addition, it is responsible for the establishment, provisions of both a general monograph and an individual
production, monitoring and distribution of reference monograph, the two are complementary. An individual
standard s needed when appIying the monographs. The monograph may, exceptionally, include an exemption from
EDQM is also active in a number of other are as related to one or more provisions of the general monograph.
the protection of public health, for exampIe in certifying the Since it is not practically possible to include in each individual
quality of active pharmaceutical ingredients from specific monograph a cross-reference to applicable or potentially
sources and in biological standardisation. applicable general monographs, cross-referencing has been

v
Introduction EUROPEAN PHARMACOPOEIA 8.0

discontinued except where it is necessary to avoid ambiguity. the older monographs elaborated before the establishment
A list of general monographs is ineluded in each new edition of this policy have been revised to introduce quantitative
and supplement to aid users in identifying those that are methods. Where a monograph does not conform to the
needed for use with an individual monograph. general policy, compliance with the general monograph
Use of anirnals. In accordance with the European Substances for pharmaceutical use (2034) implies that the
Convention for the Protection of Vertebrate Anímals used individual monograph requirements need to be supplemented
for Experimental and other Scientific Purposes (European accordingly.
Treaty Series No. 123) as amended by the protocol to the
convention (European Treaty Series No. 170), elaborated Except where required for the application of the monograph,
under the auspices of the Council of Europe, the Commission in which case the name is followed by 'CRS', impurities are
is committed to the reduction of animal usage wherever not provided as reference standards nor can they be provided
possible in pharmacopoeial testing, and encourages those for experimental purposes.
associated with its work to seek alternative procedures. An Chrornatographic columns. As an aid to users, information
animal test is ineluded in a monograph only if it has elearly is made available, via the website (see also Knowledge
been demonstrated that it is necessary to achieve satisfactory database, below), on chromatographic columns that have been
control for pharmacopoeial purposes. found to be satisfactory during development of monographs
Hydrates. Where applicable, the degree of hydration of a and general methods. Information is also given on other
substance is indicated in the monograph title. For exísting equipment and reagents where this is considered useful. This
monographs where this is not yet the case, the degree of information is given without warranty and does not imply that
hydration will be introduced into the title during the next other columns, equipment or reagents than those specified are
technical revision of the monograph (ineluding publication in not suitable.
Pharmeuropa Online). rf monographs are published for both Residual solvents. The requirements for residual solvents are
the anhydrous form and a hydrated form of a given substance, given in the general monograph Substances for pharmaceutical
'anhydrous' is ineluded in the title of the relevant monograph. use (2034) and general chapter 5.4. Residual solvents. Thus all
Chiral substances. Monographs on chiral substances that active substances and excipients are subject to relevant control
describe a particular enantiomer have a test to confirm of residual solvents, even where no test is specified in the
enantiomeric purity, usually by measurement of optical individual monograph. The requirements have been aligned
rotation. According to the current policy, a test for racemic
character using optical rotation is ineluded only if there is
information on the specific optical rotation of the enantiomers
that indicates that such a test would be discriminating in
terms of enantiomeric purity. lf other techniques, such as
with the ICH guideline on this topie.

Heavy metals. Limits for residues of metal catalysts or metal


reagents as defined in the respective guideline of the European
-
Medicines Agency are reproduced in general chapter 5.20.
circular dichroism, can serve the intended purpose, they will
The requirements laid down in this chapter are not legally
be prescribed instead of optical rotation.
binding for users of the European Pharmacopoeia as long as
Polyrnorphism. Where a substance shows polymorphism, the chapter is not cross-referenced in a monograph (e.g. in the
this is usually stated under Characters. In general, no general monograph Substances for pharmaceutical use (2034)).
particular crystalline form is required in monographs; The Commission plans to replace this chapter once the new
exceptionally, in a few monographs, the crystalline form ICH guideline for metal impurities, which is currently being
required is specified, for example, via an infrared absorption drafted, becomes available. Meanwhile, the Commission has
spectrophotometric identification test where the spectrum is decided not to devote further resources to revising existing
required to be recorded using the substance in the solid state tests (using method C or D) or creating new tests using general
without recrystallisation, the chemical reference substance chapter 2.4.8. Heavy Meta/s.
provided being of the required crystalline formo However,
for substances other than these exceptional cases, depending Homoeopathic preparations. A monograph on methods
on the use of a given substance in a dosage form, it may of preparation of homoeopathic stocks and potentisation,
be necessary for a manufacturer to ensure that a particular general monographs on homoeopathic preparations, mother
crystalline form is used. The information given under tinctures for homoeopathic preparations and herbal drugs for
Characters is intended to alert users to the need to evaluate this homoeopathic preparations, and individual monographs on
aspect during the development of a dosage formo The general raw materials and stocks for homoeopathic preparations are
monograph Substances for pharmaceutical use (2034) and ineluded in a separate section in Volume 1. It is understood
general chapter 5.9. Polymorphism should also be consulted. that when the same substance is used in both homoeopathic
and other preparations then the monograph in the main body
Spedfidty of assays. For the elaboration of monographs on of the European Pharmacopoeia applies.
chemical active substances, the approach generally preferred
by the Commission is to provide control of impurities Herbal drugs and herbal drug preparations (induding
(process-related impurities and degradation products) via a traditional Chinese medicines). Al! relevant monographs
well-designed Tests section, with stability-indicating methods, are grouped together in a separate section in Volume 1.
rather than by the inelusion of an assay that is specific for the Functionality-related charaderistics. Following a policy
active moiety. It is therefore the full set of requirements of a decision of the Commission to highlight the need for attention
monograph that is designed to ensure that the product is of to functionality-related characteristics of excipients and to
suitable quality throughout its period of use. foster harmonisation of methods for their evaluation, an
Impurities. Following a review of policy on control of informative section has been created in the monographs.
impurities, general chapter 5.10. Control of impurities in The contents of this section do not constitute mandatory
substances for pharmaceutical use was ineluded as of the requirements but the characteristics may be relevant for a
5 th Edition. Together with the general monograph Substances particular use of an excipient. The characteristics may be
for pharmaceutical use (2034), it describes the policy of presented in different ways:
controlling impurities in individual monographs and provides
explanations on how the limits in the related substances test - citing the name only;
should be understood.
The current general policy of the Commission is to inelude - citing the name and a suitable test method, preferably one
quantitative tests for impurities in monographs. Most of ineluded in the European Pharmacopoeia;

vi
EUROPEAN PHARMACOPOEIA 8.0 Introduction

- citing the name, a suitable test method and typical values the work programme. Changes to the work programme are
or tolerances on the stated value; these values or tolerances published on the EDQM website and in Pharmeuropa Online.
are used to define a suitable grade of an excipient for a Information is also provided to industry associations registered
particular use. with the secretariat and to manufacturers' liaison contacts.
In al! cases, the method and acceptance criteria are not Interested parties are invited to contact the secretariat for any
mandatory requirements but are given for guidance. The items where they wish to be involved in the work.
decision to control a functionality-related characteristic of an
CERTIFICATION PROCEDURE
excipient remains with the pharmaceutical manufacturer and
is taken with knowledge of the formulation of the product A procedure for the certification of suitability of monographs
in which it is to be used; the method of determination, of the European Pharmacopoeia with respect to quality
control of a product from a given source has been established

-
acceptance criteria and tolerances are determined on a
contractual basis by the user and the supplier of the excipient. (see Public Health Committee (Partial Agreement) Resolution
AP-CSP (07) 1 or any subsequent revision, available from the
EDQM and on its website) as an aid to the use of monographs
Patents. The description in the European Pharmacopoeia
in applications for marketing authorisation. The certification
of articles subject to protection by patent does not confer or
procedure also applies to herbal drugs, herbal drug
imply any right to the use of such patents by any person or
persons other than the proprietors of the patents concerned. preparations and transmissible spongiform encephalopathy
(TSE) risk. Certificates of suitability are issued by the EDQM
Chemical Absíracts Service (CAS) registry number. Since only for substances produced under a suitable quality system.
the 6th Edition, CAS registry numbers have been included Certificates are granted with respect to published monographs.
for information in monographs, where applicable, to provide Details of the operation of this scheme are available from the
convenient access to useful information for users. Previously secretariat and on the EDQM website. A daily updated list of
these numbers were given only for reagents, where they certificates granted is available online on the EDQM website,
are of use in locating suppliers. CAS Registry Number® is a including voided or suspended certificates.
registered trademark of the American Chemical Society.
PUBLICATIONS
Protecíed species. Monographs, notably those on herbal
drugs, may cover material obtained from protected species. The official version of the European Pharmacopoeia is
Inclusion of these monographs is without prejudice to the available in English and in French, in the form of a book with
provisions for protection of these species by national and 3 supplements per year, and in electronic format (online,
internationallaw. including a tablet version, and on USB stick).
Archives. The European Pharmacopoeia Archives contain the
MONOGRAPHS ON PHARMACEUTICAL PREPARATIONS
1st Edition to 7th Edition in PDF formato Theyare available to
Up to the 8th Edition, individual monographs on all European Pharmacopoeia subscribers with an up-to-date
pharmaceutical preparations have not been elaborated, with subscription (paper, online or USB stick) and a registered
a few exceptions, e.g. those on immunosera for human use, EPID codeo
immunosera for veterinary use, some biological preparations
Pharmeuropa, the European Pharmacopoeia forum, is
such as insulin preparations, radiopharmaceutical
published 4 times per year as an aid for the elaboration
preparations, vaccines for human use and vaccines for
of monographs and as a vehicle for information on
veterinary use.
pharmacopoeial and related matters. Pharmeuropa Bio &
The general monograph Pharmaceutical preparations (2619) Scientific Notes, a publication indexed by bibliographic
was introduced in the 7th Edition. This monograph is services, includes scientific papers related to the establishment
intended to be a reference source of standard s in the European ofbiological reference preparations and validation ofbiological
Pharmacopoeia on active substances, excipients and dosage methods within the Biological Standardisation Programme of
forms, which are to be applied in the manufacture/preparation the EDQM, and to various aspects of pharmaceutical analysis
of pharmaceuticals; it is not intended to be a guide on how to and other subjects relevant to the European Pharmacopoeia.
manufacture, as there is specific guidance available covering Since 2012, both publications are only available online, free
methods of manufacture and associated controls. of charge, and individual drafts and scientific papers are
Harmonisation and standardisation for pharmaceutical published on an ongoing basis.
preparations have so far been dealt with via the drafting Website. Information on activities and many other aspects of
of general dosage form monographs setting out elements the European Pharmacopoeia is to be found on the EDQM
common to al! preparations within the scope of the website.
monograph, and via the development of standard test
methods used for testing of finished products. The inclusion Knowledge database. The EDQM website provides access
of these general monographs and methods in the European to a database containing information of various sorts related
Pharmacopoeia gives a common basis for competent to monographs and intended to facilitate their proper use.
authorities and manufacturers in the preparation and Information is provided on:
evaluation of applications for marketing authorisation. - chromatography columns used in monograph development;
However, during its 143,d session, the Commission decided - suppliers of reagents and equipment that may be difficult
to revisit its policy and initiate a pilot phase on individual to find for sorne users;
pharmaceutical preparation monographs to investigate further - the status of monographs (in development, adopted,
their feasibility and usefulness. published, under revision);
Reference standards established for the assay of active - revisions of the monographs on a historical basis, beginning
substances and excipients may be suitable for use as assay from the 5th Edition;
standards for preparations when the conditions stated in
general chapter 5.12. Reference standards are fulfil!ed. - other useful information.
HelpDesk. Many technical and other enquiries are addressed
WORK PROGRAMME to the EDQM by users. They should be submitted via the
The work programme (elaboration of new monographs or HelpDesk on the EDQM website. The EDQM will deal with
general chapters or revision of existing texts) is decided enquiries that are related to the use of monographs of the
by the Commission at one of the three annual sessions. European Pharmacopoeia. The HelpDesk has a section of
In general, whenever 2 member states express a wish to Frequently Asked Questions that should be consulted by users
elaborate a monograph, the Commission adds the item to before submission of an enquiry.

vii
Introduction EUROPEAN PHARMACOPOEIA 8.0

Implementation. The date on which monographs are to INTERNATIONAL HARMONISATION


be implemented is fixed by a resolution of the European Globalisation and expansion in international trade present
Committee on Pharmaceuticals and Pharmaceutical Care a growing need to develop global quality standards for
(Partial Agreement) of the Council of Europe, following a medicines. Standards are a vital instrument for marketing
recommendation by the Commission. This date is usually authorisations, market surveillance, and free movement
1 year after adoption and about 6 months after publication. and trade of medicines between regions and countries.
Where a monograph is to be implemented at a date earlier The European Pharmacopoeia is engaged in a process of
than the next publication date of the European Pharmacopoeia harmonisation with the Japanese Pharmacopoeia and the
or a supplement, a resolution of the European Committee on United States Pharmacopeia, within an informal structure
Pharmaceuticals and Pharmaceutical Care gives the full text to referred to as the Pharmacopoeial Discussion Group (PDG).
be implemented. The text is also published in Pharmeuropa Information on the status of harmonised texts is given in
Online for information and posted 011 the EDQM website as general chapter 5.8. Pharmacopoeial harmonisation and on
part of the resolution. the PDG page of the EDQM website.
Revision programme. Monographs and other texts of the vVhere harmonisation of general chapters is carried out, the
European Pharmacopoeia are revised as necessary following a aim is to arrive at interchangeable methods or requirements
decision of the Commission. Revision proposals are published so that dernonstration of compliance using a general chapter
in Pharmeuropa Online. Proposals to revise monographs may from one of the 3 pharmacopoeias implies that the same
be submitted by a delegation, by the Chair of the Commission result would be obtained using the general chapter of either
or by the chair of a group of experts. Requests for revision of the other pharmacopoeias. When a formal declaration of
from other parties should be submitted via the natio11al interchangeability has been recommended by the International
pharmacopoeia authority of a member state or, where this is Conference on Harmonisation (ICH), it will be indicated
not possible, to the EDQM via the HelpDesk. Proposals to in general chapter 5.8. Pharmacopoeial harmonisation. If
revise monographs must be accompanied by sufficient data to residual differences remain in harmonised general chapters,
justify the need for revision. information is given in this general chapter.
Where harmonisation of monographs is carried out, the aim is
to arrive at identical requirements for all attributes of a producto
Informatio11 on any non-harmonised attributes is included in
COMBISTATS
general chapter 5.8. Pharmacopoeial harmonisation. Besides
Certain tests in monographs, particularly biological assays, the PDG, the European Pharmacopoeia is also actively
require statistical analysis of the results. The EDQM has involved in a number of other international harmonisation
developed a computer programme, CombiStats, that can be initiatives, such as the World Health Organization initiative of
used for statistical analysis of results of biological dilution drafting 'Good Pharmacopoeial Practices' that may serve as a
assays. Information 011 the programme, with conditions of basis for fnture work-sharing and collaboration between the
access and use, is available on the EDQM website. pharmacopoeias of the world.

viii
EUROPEAN PHARMACOPOEIA 8.0 European Pharmacopoeia Commission

111. EUROPEA PHARMACOPOEIA


COMMISSION
COMPOSITION OF THE COMMISSION, LIST OF EXPERTS
AND OF THE SECRETARIAT (2012)
CHAIR AND VICE-CHAIRS Hungary Goran BENKOVIC
OF THE COMMISSION Hilda KOSZEGI-SZALAI
Chair Marianne EK Jozsef J. LIPTAK
Iceland
Vice-chairs Jos HOOGMARTENS
An LE Ireland Michael MORRIS
Noreen QUINN
Mirza CATIBUSIC
MEMBERS OF THE COMMISSION
Austria Yvonne GASPAR Italy Elena BOSSU
Friedrich LACKNER Eugenia COGLIANDRO
Andreas MAYRHOFER Carlo PINI

Belgium Katrien VANLANDUYT Latvia Ilze BARENE


Inta KURAKINA
Bosnia and Alma KISO
Herzegovina
Lithuania Roma MOCKUTE

Bulgaria Ljuba KOSTOVA Luxembourg Jacqueline GENOUX-HAMES


Svetla BOGDANOVA Jean-Louis ROBERT
Svetoslav BRANCHEV
Malta Clint PACE
Croatia Goran BENKOVIC Montenegro
Mirela FILIPEC
Laila STEFANINIORESIC Netherlands Dries DE KASTE
Jan-Willem DORPEMA
Cyprus Louis PANAYI Josée M.HANSEN

Czech Republic Hana LOMSKA KOUBKOVA


Milos MACHACEK Norway Gunhild BRUGAARD

Poland Agnieszka JOZWIAK


Denmark Steen Honoré HANSEN Ewa LECIEJEWICZ ZIEMECKA
Eva SANDBERG Jan PACHECKA
Erik WOLTHERS

Estonia Signe LEITO Portugal José Manuel CORREIA NEVES SOUSA LOBO
Tuhan RUUT Domingos DE CARVALHO FERREIRA
Maria Joao PORTELA
Finland Marjo-Riitta HELLE
Eija PELKONEN Romania Anca CRUPARIU
Piia SALO Daniele ENACHE

France Alain NICOLAS Serbia Danica AGBABA


Marie-Lise MIGUERES Marija MALESEVIC

Germany U1rike HOLZGRABE Slovak Republic Marta BENKOVA


Jochen NORWIG Ruzena MARTINCOVA
Dietrich KRÜGER Jozef SLANY

Greece Michael A. KOUPPARIS Slovenia Tanja TEKAVCIC GLOVER


Alexandra TSOKA Uros URLEB

ix
European Pharmacopoeia Commission EUROPEAN PHARMACOPOEIA 8.0

Spain Franco FERNANDEZ GONZALEZ Ireland Susann BRADLEY


Carmen DE LA MORENA CRIADO Mirza CATIBUSIC

Sweden Lennart AKERBLOM Italy Alessandra DELL'UTRI


Torbjbrn ARVIDSSON Loredana NICOLETTI
Gert RAGNARSSON Christina VON HUNOLSTElN

Switzerland Lukas BRUCKNER Lithuania Valdemaras BRUSOKAS


Tobias GOSDSCHAN
Andreas H. PFENNINGER Luxembourg M. BACKES-LIES

Netherlands Ellen De ROOIJ-LAMME


"The former Aneta DIMITROVSKA Peter J.M. JONGEN
Yugoslav Tatjana PERUSEVSKA Yolanda VAN KOOIJ
Republic
of Macedonia"
Norway Svein Rune ANDERSEN
Turkey Eda AYPAR Valborg HOLTEN
Hanefi bZBEK
Bilge SENER Poland Jan LUDWICKI
Malgorzata SZNITOWSKA
United Samantha ATKINSON
Kingdom V'Iain FENTON-MAY Portugal Rui MORGADO

Serbia Lj ilj ana ZIVANOVIC


European Agnes MATHIEU
Commission Slovak Republic Daniel GRANCAI
Ladislav SOVIK
EMA Riccardo LUIGETTI
Slovenia Barbara RAZINGER-MIHOVEC
Ales ROTAR
ALTERNATE MEMBERS
Sweden My MOBERG
Austria Johann KURZ Eva LINDBERG
Christian NOE
Josef TRENKER
Switzerland Laure GIRARD
Karoline MATHYSBADERTSCHER
Belgium Luc ANGENOT Thomas SCHREITMÜLLER
René HANSELAER

Turkey Levent ALTUN


Bosnia and Alija UZUNOVIC Ebru CORA
Herzegovina H. Gül<¡:in SALTAN

Bulgaria Silvia DIMITONOVA United Alastair DAVIDSON


Kingdom Rodney HORDER
Czech Republic Hana BIZKOVA Matilda VALLENDER
Hana JUZOVA

Denmark Lone STENGELSHOEJ OLSEN


EXPERTS
Estonia Eveli KIKAS
H ABBOUD
Juhan RUUT
Valter ACQUATI
Finland Tom WIKBERG Joel AERTS

France Caroline VILAlN Neli AGAPOVA


Danica AGBABA
Germany Gerhard FRANZ
Siegfried GIESS Abdelaziz AGOUMI
Detlef MANNS Francisco AGUILAR PARRILLA
Maqbool AHMED
Greece loanna CHINOU Lennart AKERBLOM
Anastasios MAKRITIS
Arnoud AKKERMANS
Hungary Tamas L. PAAL Susanne ALBAN
Tamas NEMETH
Goran ALDERBORN

x
EUROPEAN PHARMACOPOEIA 8.0 European Pharmacopoeia Commission

Keith ALLEN GuInara BERDIMURATOVA


Mirandan ALMEIDA Joep BERGERS
Concepcion ALONSO VERDURAS Stéphane BERNARD
Levent ALTUN Margarita BERROCAL NAVAS
Ahmad AMINI Poul BERTELSEN
Oliver ANDERKA Agnes BERTOCCHI
Hans-Joachim ANDERS Serge BESSET
Svein Rune ANDERSEN Bohumir BIBA
Linda ANDERSON Anna Rita BILIA
Thomas ANDERSSON Jean Pierre BINDER
Murielle ANDRÉ Hanno BINDER
Luc ANGENOT Louise BISSET
Brigitte ANLIKER Hana BIZKOVA
Peter ANNEL Knud BJORNSON
Marie-Christine ANNEQUIN Philippe BLANCO
Gunnar ANTONI Marcelo BLANCO ROMIA
Peter Georgiev ANTONOV Elham BLOUET ABDELHAC
Sandra APERS Katja BOEGLI -STUBER
Jean Claude ARGOUD Svetla BOGDANOVA
Alp ARKAC Gerrit BORCHARD
Steve ARKLE Arantxa BORDAS CAMPANA
Sylvie ARMEL Elena BOSSU
Rolf ARNDT Karina BOSZKO
Beatriz ARTALEJO Yvonne BOUWMAN BOER
Torbjorn ARVIDSSON Susann BRADLEY
WiIfried ARZ Anna Maria BRADY
Nataliya Nikolaevna ASMOLOVA James Clifford BRAIN
Samantha ATKINSON Svetoslav Valentinov BRANCHEV
Sylvie AUDOLY Helena BRANDIN
Paolo AURELI Tomaz BRATKOVIC
Ofra AXELROD Susanne BREITNER RUDDOCK
Eda AYPAR Harald BREIVIK
Mariette BACKES LIES Charlotte BRENIER
Mamadou BADIANE Adrian Francis BRISTOW
Arlette BAILLET GUFFROY Neville BROAD
Ilze BARENE Kirsten BRONNUM HANSEN
Sandra BARNES Lukas BRUCKNER
Maria BARRETT Rene BRUEGGER
Dietmar BARTSCHAT Ines Maria BRUENTRUP
KHC BASER Gunhild BRUGAARD
Rudolf BAUER Peter BRÜGGER
Alain BAYOL Biancamaria BRUNO
Wolfgang BECK Valdemaras BRUSOKAS
Denis BELLENOT Christian J BUCHHOLZ
Silvana BELLINI Volker BÜHLER
Norbert BENDA Marian BUKOVSKY
Marta BENKOVA Rosario BULLIDO
Goran BENKOVIC Jorg BUND

xi
European Pharmacopoeia Commission EUROPEAN PHARMACOPOEIA 8.0

Heike BUNJES Gerard DAMIEN


Roger BURGENER Rolf DANIEL S
Robert BURMAN Teresa DANNERT ALSASUA
Christiane BUSCH David DARRAS
Jesus CABANAS Alastair DAVIDSON
Laurent CABURET Domingos DE CARVALHO FERREIRA
Salvador CANIGUERAL Josep M DE CIURANA 1 GAY
Anna Maria CAPPELLI Hendrik Jan DE JONG
Pilar CARRASCO SAINZ Dries DE KASTE
EZQUERRA
Carmen DE LA MORENA CRIADO
Monica CARVALHO SOARES Ellen DE ROOI] LAMME
Patricia CATALAO Berber DE VRIES
Mirza CATIBUSIC Paul DECLERCK
Adrian CAWS Clemens DECRISTOFORO
Richard CAWTHORNE Louis H T DEDEREN
Rajko CEBEDZIC Alexandre DELAVOLPILIERE
Marina CERQUETTI Alessandra DELL UTRI
Pierre CHAMINADE Isabelle DELNEUVILLE
Kelvin CHAN Joseph DEMEESTER
Denis CHAUVEY L. Omur DEMIREZER
Nuan Ping CHEAH Aysegul DEMIRTAS
CHEN YIN QING Stephen DENYER
Carlos A CHIALE Sven DEUTSCHMANN
Keith CHIDWICK Gines DIAZ
Ioanna CHINOU Michele DIBOUNE
Vivienne CHRIST Milen DIMITROV
Stefan CHRISTIANS Aneta DIMITROVSKA
Maurizio CIANGRIGLIA Papa Amadou DIOP
Romolo CICCIARELLI Anica DJAMIC
Patrik CINTHIO Johannes DODT
Mihai CIOBANU Eric DOELKER
Maija CIRKINA Milada DOLEZALOVA
Corinne CIVADE DELAY Jan Willem DORPEMA
Juan CLARAMUNT CAMPANA Pi erre DUEZ
Eugenia COGLIANDRO Alain DUGUET
Laurence COLLIERE Stephen EDGE
Harald S CONRADT Erling EHRIN
Anne COOK Marianne EK
Ebru CORA Lindsay ELMGREN
Stéphane CORNEN Jürgen EMIG
Jose Manuel CORREIA NEVES Daniela ENACHE
SOUSALOBO
Joerg ENGELBERGS
Desmond CORRIGAN
Lena ENGMAN
Yves CORTEZ
Ilkay ERDOGAN ORHAN
Martin CRNUGELJ
Kristina ERLANDSSON PERSSON
Anca CRUPARIU
Lina ERTLE
Klaus CUSSLER
Jean Pierre ETCHEGARAY
Martina CVELBAR
Adrian EVANS
Elisabeth DADOLE

xii
EUROPEAN PHARMACOPOEIA 8.0 European Pharmacopoeia Commission

Mareus EVANS Daniel GLAUSER


0ystein EVENSEN Kazimierz GLOWNIAK
Huguette FABRE Tanja GMEINER STOPAR
Gemma FEENSTRA BIELDERS Chris T GODDARD
Rainer FENDT Muge GOKDERE
V'Iain FENTON MAY Carine GOLAZ
Franco FERNANDEZ GONZALEZ Maria Jesus GOMEZ MIGUEL
Rosella FERRETTI Els GOOSSENS
Petra FESSER Tobias GOSDSCHAN
Mirela FILIPEC Gianaluea GOSTOLI
Luey FINDLAY Mareel GOVERDE
Ton FORCH Tor GRABERG
Lucien FOSSE Astrid GRAEFE
Isabelle FOURASTÉ Tatjana GRAFNETTEROVA
Peer Lyng FRANDSEN Daniel GRANCA!
Bruno FRANK Yvan GRANGE
Laura FRANQUEZA GARCIA Kevin GRANT
Gerhard FRANZ Norbert GREIDZIAK
Lars Vaelds FREDERIKSEN Gerhard GROHMANN
Urban FREY Kjell-Olov GRONVIK
Erika FRIEDL Oliver GROSCHE
Gloria FRUTOS CABANILLAS Nannette GROSS
Pascal FURRER Oleksandr GRYZODUB
Rose E GAINES DAS Emanuel GUADAGNINO
Maria Cristina GALLI Didier GUEDON
Igor GAON Thorsten GUMZ
Dominique GARCIA Sylvie GUYOMARD DEVANLAY
Gloria GARCIA LORENTE Bert GYSBERG
Maria GARD René GYSIN
Olivier GARINOT Klaus HABERER
Piotr GARNUSZEK Adrian HAEBERLI
Yvonne GAS PAR Geertrui HAEST
Uwe GASSER Lilian HAMILTON
Niek GATE Thomas HÁMMERLE
Matthias GAUTSCHI Vagn Neerup HANDLOS
Anne GAYOT Kay-Martin HANSCHMANN
Andrea GAZZANIGA Rene HANSELAER
Jaequeline GENOUX HAMES Steen Honoré HANS EN
Christoph GERBER Josee HANSEN
Philippe GERVAIS Bente Tange HARBO
Christian GEYER Sebnem HARPUT
Lilit GHAZARYAN Kaare HASLOV
Nicole GIBELIN Jostein HATLELID
Barbara GIESE Ivana HAUNEROVA
Siegfried GIESS Heribert HÁUSLER
Michel GIRARD Marjo-Riitta HELLE
Philippe GIRARD Keith HELLIWELL
Laure GIRARD Peter HENCKEN

xiii
European Pharmacopoeia Comrnission EUROPEAN PHARMACOPOEIA 8.0

Bilkis HENEKA Elena JELIAZKOVA


Martina HENK Anna JELINSKA
Peter HENRYS Jotgen Skov JENSEN
Harm HERMSEN Thomas G JENSEN
Rolf HESSELMANN Jana JERABKOVA
Tim HEWINS Edgar JOHN
Sven Erik HILLVER Christopher JONES
Thomas HINZ Allison JONES
Comelia HIPPCHEN Peter M J M JONGEN
Jouni HIRVONEN Juan Ignacio JORQUERA NIETO
Marjatta HIRVONEN Annelies JORRITSMA -SMIT
Kowid HO Mats JOSEFSON
Wolfgang Alexander HOEPPING Zarko JOVIC
Jéirg HOERNSCHEMEYER Agnieszka JOZWIAK
Patrick HOET Carmen ]UNGBACK
Andreas HOFMANN Hana JUZOVA
Oyvind HOLTE Mohamed KALLEL
Valborg HOLTEN Winfried KAMMER
Ulrike HOLZGRABE Elisabeth KAMPHUIS
Jos HOOGMARTENS Tarja KANKKUNEN
Andrew HOPKINS Mimi KAPLAN
Rodney HORDER lmre KAPUI
Konrad HORN Rami KARIV
Ottmar HORN LOHRENS Anders KARLSSON
Michael HORNIG Sture KARLSSON
Matej HORVAT Jan-Oliver KARO
Erno HORVATH Petrus KARSTEN
Thomas HOTTIGER Pasi KAUPPINEN
Christian G HOUGHTON Hans KEEGSTRA
Rolf HOVlK Christoph KEFEDER
Anthony R HUBBARD Birgit KEGEL
Comelia HUBER Naman KEITA
Ronny HUBINETTE Edwin KELLENBACH
Peter HUBRECHTS Ernst KELLER
Jana HUMHEJOVA Melanie KERST
Tiina HUOTARI Waltraud KESSLER
Lars J HUSAGER Michael KEUSGEN
Gérard HUYGHE Eveli KIKAS
Alexandra Maria Bagulho INVERNO SAFARA Alistair KIPPEN
Iliana IONKOVA Karin KIRCHDORFER
Canan ISKENDEROGLU Christian e KIRCHNER
Avi ISRAEL! Alma K1S0
Evert IZEBOUD Anne KJOELBY
Elke JAECKLE Robert KLASSON
Miia JAKAVA VILJANEN Caroline KLEBER
Planinka JAKSIC Peter KLEINEBUDDE
Armand JANSSEN Bemhard KL!ER
Renaud JANSSEN Hector KNIGHT CASTRO

xiv
EUROPEAN PHARMACOPOEIA 8.0 European Pharmacopoeia Commission

Filiz KOC Erich LEITNER


Hannelore KOPELENT FRANK Signe LEITO
Mireille KOPF Ulla LENNMARKER
Sabine KOPP Dmitro LEONTIEV
Valentina KOSENKO Valérie LIEVRE
Ljuba KOSTOVA Jobst LIMBERG
Eniko KOSZEGI Eva LINDBERG
Hilda KOSZEGI SZALAI Comelia LIPPERHEIDE
Andrey KOTOV JozseO LIPTAK
Michael A KOUPPARIS Lino LIVERANI
Renata KOVACOVA Anders LOFGREN
Emil KOZHUHAROV Philippe LOISEAU
Marija KOZLOVIC Hana LOMSKA
Beate KRÁMER Silvano LONARDI
Mateja KRAMER Karen LONGSTAFF
Adrian KRAUSS Susana LOPEZ HERNANDEZ
Aldona KRAWCZYK Sabina LORENZ
Katjusa KREFT Céline LORTEAU
Siegfried KRIMMER Patrick LOVIS
Thijs KROON Jan LUDWICKI
Dietrich KRUGER Hans LUNDBACK
Christof KRUMMEICH Ian LYNCH
Urs KUENZLE Milos MACHACEK
Harry V KUPPERS Suzana MACHADO DE AVILA
lnta KURAKINA Hans-Jürgen MACHULLA
Johann KURZ Lourdes MACUA PEQUENO
Herbert KVATERNIK Antonio MAGNI
Friedrich LACKNER Mohamed Abdelmoumen MAHLY
Reinhard LAENGER Anastasios MAKRITIS
Ellen LAMME Daniel MALARME
Stefan LAMOTTE Marija MALESEVIC
Andreas LANG Laurent MALLET
Reinhard LANGE Dave MALPAS
Thomas LANGE Paul MALTBY
Manuela LANGOS-MABBOUX Tamara MANDUSIC NAZOR
Mervi LANKINEN Detlef MANNS
Maria Grazia LAVAGGI MB MANSOURI
Dorothée LAY Siarhei MARCHANKA
Waldemar LAZIK Anca MARCOVICH
An LE Andreas MARTI
Ondrej LEBEDA Dierk MARTIN
Ewa LECIEJEWICZ ZIEMECKA Alessandro MARTINI
Gerard LEE Ana Paula MARTINS
Catherine LEFEBVRE Graca MATA
Thomas LEHMANN Walter MATHEIS
Kristina LEHMANN Karoline MATHYS BADERTSCHER
Ari LEHTOLA Jaroslav MAXA
Stefan LEINER Andreas MAYRHOFER

xv
European Pharmacopoeia Commission EUROPEAN PHARMACOPOEIA 8.0

Ian MCEWEN Anastasia NIKITINA


Roger MEIER Vittorio NISTRIO
Beat MEIER Christian NOE
Luis Miguel MEIRINHOS SOARES Kim NORDFJELD
Margarida MENEZES FERREIRA Darragh NORTON
Monica MENNET - VON EIFF Ute NORWIG
Geerd J MEYER Jochen NORWIG
Jacques MICHAUD Micha NÜBLING
JohnM MIDGLEY Adela NUNEZ VELAZQUEZ
Giovanni MIGLIACCIO Susanne ODGAARD HERR
Marie Lise MIGUERES Hok Liang OEI
Robertas MIKALAUSKAS Silvester OELZANT
Paola MINGHETTI Volker OEPPLING
Monica MIRANDA Martin O'KANE
Christine MIRAS Jaap OOSTENDORP
Bojan MITROVIC Samira OMEROVIC
My MOBERG Didier ORTELLI
Roma MOCKUTE Erik OSTERGAARD
Birthe MOESGAARD Rainer OTTER
MOHD ZIN CHE AWANG Hans Peter OTTIGER
Jürgen MÜLLER KEMSA Carsten OVERBALLE PETERSEN
Maria MORAIS Inge OVERBY JENSEN
Marta MORENO CUARTAS Hanefi OZBEK
Rui MORGADO Esin OZMENER SOZEN
Sylvie MORGEAUX Clint PACE
Michael MORRIS J. Jan PACHECKA
Saliha MOUSSAOUI Ana PADILLA MARRO QUIN
Marie Anne MOUYART Steffen PAHLICH
Cad MROZ Pilar PAIS
Zdenka MRVOVA Peter PALLAS
Heide MUCKENFUSS PANXUE TIAN
Marco Thomas MUELLER Louis PANAYI
Christel MÜLLER GOYMANN Béatrice PANTERNE
Robert MUENTZ Yiannos PAPADOPOULOS
Ezra MULUGETA Rosemary PASKHUGHES
Sibel MURATOGLU Paolo PASQUALI
Michael MUTZ Stefan PAUKER
Vera MYSLIVCOVA Hugo PEETERS
Eva Maria NADAL ELDUAYEN Eija PELKONEN
Paul NÁTSCHER Isabel PEREZ GONZALEZ
Heidi NEEF lean-Marc PERSON
Tamas NEMETH Tatjana PERUSEVSKA
Peter NEU Jasmin PESCHKE
Robin A J NICHOLAS Alain PETIT
Steven C NICHOLS Francesco PETRUCCI
André NICK Andreas H PFENNINGER
Alain NICOLAS Erik PHILIPP
Loredana NICOLETTI Gerson Antonio PIANETTI

xvi
EUROPEAN PHARMACOPOEIA 8.0 European Pharmacopoeia Commission

Roger D PICKETT Ales ROTAR


Carlo PINI Jacques ROTGER
Marta PIPIC KOSANOVIC Sol RUIZ
Melanie Eva PIRES Torgny RUNDLOF
Bojan PIRNAT Juhan RUUT
Anja PISCHTIAK Knud RYHL
Daniel PISCITELLO Christoph SAAL
Klaus PITTERTSCHATSCHER Sirkku SAARELA
Christelle PLANTARD Michael SAENGER
Wolfgang POHLER Elena Ivanovna SAKANJAN
Bertrand POIRIER Paula SALMIKANGAS
Elizabeth POLLITT Piia SALO
Frederic POMMARAT H. Gukin SALTAN
Markus PONGRATZ Piero A SALVADORI
Cécile PONSART Eva SANDBERG
Maria Joao PORTELA Sverre Arne SANDE
Agustin PORTELA MOREIRA Monica SANTIMARIA
Bernard PRILLEUX Hanife Ebru SARPAY
Stanislav PRIMOZIC Jeannot SCHELCHER
Martin PUNZENGRUBER Silke SCHEPELMANN
Joachim QUEIROGA Jürgen SCHERER
Jo elle QUETIN LECLERCQ Roger SCHIBLI
Jose Carlos QUINTELA FERNANDEZ Martin SCHIESTL
Gert RAGNARSSON Heidemarie SCHINDL
Alain RAGON Ernst SCHLÁFLI
Giuseppe RAMASCHI Dirk SCHMALZ
Isabel RAMIREZ Birgit SCHMAUSER
DE ARELLANO SERNA
Andreas SCHMID
Benoit RAMOND Stephan SCHMIDT
Paula Maria RAMOS MARTINHO Heinz SCHMITTER
FIGUEIREDO
Martin SCHOEFNAGL
Jean Rene RANDRIASAMIMANANA
Thomas SCHREITMUELLER
Jukka RANTANEN
Walter SCHUH
Barbara RAZINGER MIHOVEC
Frank SCHULER
Klaus REH
Dieter SCHULZ
Eike REICH
Harald SCHULZ
Holger REIMANN
Volker SCHULZE
Klaus REITHMAYER
Utta SCHURIG
Joaquim Jose RIBAS PERDIGAO
QUEIROGA Karolina SCHWARZ SZEWCZUL
Ruediger SCHWETZLER -RASCHKE
Nathalie RIZZO PADOIN
Patricia SCOGNAMIGLIO
Jean Louis ROBERT
Jean Marc SEIGNEURET
Helmut ROCKSTROH
Osman SENER
Isabel RODRIGO CASTRO
Dorothea SESARDIC
Jose Maria RODRIGUEZ PACHON
Karin SEWERIN
Jacqueline ROOS
Nicky SHIPTON -YATES
Tilmann ROOS
Irena SHUMELI
Judith ROS FUENTES
Caroline SIEBECKE
Michael ROSSLER

xvii
European Pharmacopoeia Commission EUROPEAN PHARMACOPOEIA 8.0

Heinz Wilhelm SIESLER Jürgen THUERK


Claudia SIGNORETTI René THÜRMER
Glenda SILVESTER Marcos TIMON JIMENEZ
Natercia SIMOES Heinz Gerhard TOLLE
Francois SIMONDET Katarzyna TOMASZEWSKA
Kaarina SINIVUO Carlo TOMBA
Marie-Louise SJOHOLM Gabriella TOROK
Wenche SKARE Giangiacomo TORRI
Helena SKOG Daniel TOUW
Mikael SKOOG Eranui TOVMASYAN
Jozef SLANY Josef TRENKER
Ineke SLAPER CORTENBACH Wilhelm TRENTMANN
Jan WH SMEETS Hans-Peter TRIERWEILER
Glenn SMITH Catherine TROUBAT
Dean SMITH Jean Hugues TROUVIN
Francesco SOLINAS Mathias TRUYENS
Therese SOLSTAD SAUNDERS Lia TSIKLAURI
Stein SOMMER Alexandra TSOKA
Delphine SORDAT FORNEROD Michael TUBBY
Robert SOUSSAIN Ardak U TULEGENOVA
Ladislav SOVIK Michael TÜRCK
Margot SPOHN Peter TURECEK
Ingo SPREITZER Zsofia TUSKE
Alexandra STAERK Jacqueline Léonore TUXEN WISSING
Axel STAHLBOM Stefan TYSKI
Lone STENGELSHOEJ OLSEN Michel ULMSCHNEIDER
Willem STEVENS Uros URLEB
Peter STJARNKVIST Ana Luisa URMALRIBEIRO
Erich Andreas STOEGER Alija UZUNOVIC
Metka STOJICEVIC Claudia VALDER
Rainer SUCHI Vladimir VALICA
Jasbinder SUMAL Matilda VALLENDER
Sibel SUZEN Jan VANDERNAT
Lennart SVENSSON Willem G VAN DER SLUIS
Filip SVETOSLAVOV Hans VAN DOORNE
Beata SZABADY Yolanda M.A.W VAN KOOIJ
Julianna SZEMAN Katrien VANLANDUYT
Malgorzata SZNITOWSKA Bernard VAN GENUGTEN
Karoline SZWARZ SZEWCZUL Jos VAN ROMPAY
Mathieu Jean-André TAFANI Ann VAN SCHEPDAEL
Herre TALSMA Paul VARLEY
Fatih TANIS Markus VEIT
Khalil TAOUBI Alfons VERBRUGGEN
Pierre Cyril TCHORELOFF Geert VERDONK
Tanja TEKAVCIC GLOVER Christopher H VERMAAT
Tomas TESAR Peep VESKI
Robin C THORPE Jaana VESTERINEN
Ingela THORSON KAIJA Francesco VILLA

xviii
EUROPEAN PHARMACOPOEIA 8.0 European Pharmacopoeia Commission

Philippe VILLATTE Romana ZELKO


Maria Rosa VIRTO GARCIA Jorg ZESSIN
Eva VITKOVA Jürgen ZIRKEL
Caroline VOLTZ-GIROLT Ljiljana ZIVANOVIC
Christina VON DER HEIDT Hans-Joachim ZOLLER
Christina VON HUNOLSTEIN Gijsbert ZOMER
Elvedina VRANJES Klaus ZÜCHNER
lmran VURAL Jorg ZUERCHER
Genevieve WAETERLOOS
Hermann WAETZIG SECRETARIAT OF THE EUROPEAN
Rik WAGENAAR PHARMACOPOEIA COMMISSION
Thierry WAGNER
Director (European Directorale for the Quality of
Frank WAIMER Medicines & HealthCare)
Anna WALLINDER MARKLUND Susanne KEITEL
Mei WANG
Scientific officers and assistants (Technical secretariat,
Shu-Yuan WANG-TSCHEN Laboratory and Biological standardisation)
Peter WASCHICZEK Cathie VIELLE (Secretary to the Commission)
Claude WASTIEL Michael WIERER
Marjolein WEDA Emmanuelle CHARTON
Mark WEINSTEIN Andrea LOm
Wigand WEIRICH Stefan ALMELING
Irmgard WERNER Jean-Marc SPIESER
Ingrid WERNER Karl Heinz BUCHHEIT
Michael WHALEY Béatrice ANDLAUER
Brian WHITE Susanne BAHRKE
Michael WIEDMANN Melanie BALD
Tom WIKBERG Aurélie BARTH
Markus WILD Maria Silvana BELLINI
Brigitte WILD Anne-Sophie BOUIN
Maria WILHELM Angele COSTANZO
ToreW WILHELMSEN Arnold DAAS
Klemens WINNA Philippe DURET
Maria WIRZ Anne GAREL
Vincent WISARD Anne GARNIER-POIDEVIN
Jacqueline WISSING Sylvina IOSSIPHOVA
Bengt WITTGREN Brigitte JACQUEL
Petra WLASAK Sylvie JORAJURIA
Juliane WOEGERER Franck JUNG
Bernhard WOLF Lynn KELSO
Uwe WOLLEIN Catherine LANG
Erik WOLTHERS Gisele LOUIS
A David WOOLFSON Julia MAIER
Kim WUTHOLD Isabelle MERCIER
Michel YERLY Catherine MILNE
Semra YILMAZ Robert NEUGEBAUER
Stephen YOUNG Jochen PAUWELS
Chun Ting YUEN Ellen PEL
Pilar ZAMORANO SANCHEZ John QUINN
Guy RAUTMANN

xix
European Pharmacopoeia Commission EUROPEAN PHARMACOPOEIA 8.0

UIrich ROSE Quality


Cristiana SABATINI -SAMORI Pierre LEVEAU
MicheIe SCHOTT Jonna SUNELL-HUET
Monica SORINAS JIMENO Reference standard s
Laure TACONET Vincent EGLOFF
Eriko TERAO Fanny MOUTIER-GAME
Marianne THEBAUT Maurice TENDERO
Eva VITKOVA
Translation
Matthias WEBER
Michelle BACQUE
Stephen WICKS
Benolt BERNARD
Brigitte WILD
Rex HUISH
Sally WOODWARD
Publicrelations
Publications
Caroline LARSEN -LE TARNEC
MiseI JEVTIC
Fiona GILCHRIST
Hans-Joachim BIGALKE
Isabelle BYLINSKI Expert consultants

David CROWE Marie-Thérese BALDACINI

Ioulia IANKOVA Armand BLOMMAERT

Christopher JARVIS Raymond BOUDET -DALBIN

Carole KNAUP Isabelle FOURASTE

Mathieu MARRE
Bao Thanh NGUYENVAN
Catherine NICOLAS
Laura O'TOOLE
Uwe PIOCH
Alice ROBERTS
Sabine SCHAEFFER
Lynne TOUMASSON
Isabelle VERNAY

xx
EUROPEAN PHARMACOPOEIA 8.0 Contents of the 8th Edition

I~ CO TE TS OF THE
8th EDITIO
The 8th Edition consists of all texts published in the 7th Edition, indications, which are not necessarily exhaustive, are given
which may subsequently have been revised or corrected, and for information and do not form an official part of the texts.
new texts. Editorial changes are not indicated.
For the information of the reader, lists are given below of Lines in the margin that were present in revised or corrected
monographs and general chapters that are new, or that have texts in the previous edition are deleted with each new edition.
been revised or corrected, and texts whose title has been
changed for the 8th Edition. Corrections that are indicated by the note 'corrected 8.0'
under the version date are to be taken into account from the
The version date (for example 0112014 for a text that is new or publication date of the volume.
revised for the 8th Edition), completed by 'corrected X.x' if a
corrected version of the text has subsequently been published For the 8th Edition, the following decisions and systematic
in Supplement X.X, and the reference number (4 digits for modifications to the texts of the European Pharmacopoeia
monographs and 5 digits for general chapters) are specified have been made.
aboye the title of each text (monographs and general chapters).
The version date, completed by 'corrected X.X if appropriate, - Common logarithms are now indicated by loglO'
makes it possible to identify the successive versions of texts in
- The wording used in the tests for chlorides, sulfates,
different editions.
calcium, iron and magnesium in monographs has been
From the 7th Edition, if a text has not been revised for a new updated according to the style guide.
edition, the version date published in the previous edition is
kept in order to improve traceability. The volume in which - Reagent names have been modified to improve the
this version has been published for the first time is stated in functioning of the hyperlinks in the electronic version of
the Knowledge database on the EDQM website. the European Pharmacopoeia.
A verticalline in the margin indica tes where part of a text has - The graphical representation of saccharides has been
been revised or corrected. A horizontalline in the margin harmonised; the graphical representation of insulins has
indicates where part of a text has been deleted. However, these been harmonised.

Individual copies of texts published in this edition will not be supplied.


Subscribers to the current version (printed or electronic) of the European Pharmacopoeia have access to an archive version of all
previous editions of the European Pharmacopoeia.

NEW TEXTS INCLUDED IN THE 8 th EDITION


The texts below appear for the first time in the European Pharmacopoeia. They will be implemented on 1 January 2014
at the latest.

GENERAL CHAPTERS Fraxinus rhynchophyIla bark (2452)


Mandarin epicarp and mesocarp (2430)
2.2.66. Detection and measurement of radioactivity
Saw palmetto extract (2579)
Monographs
MONOGRAPHS
Alimemazine hemitartrate (2650)
Radiopharmaceutical preparations and starting maíerials
for radiopharmaceutical preparations Atomoxetine hydrochloride (2640)

Alovudine C8F) injection (2460) Desloratadine (2570)


Diacerein (2409)
Fluoromisonidazole C8F) injection (2459)
Dutasteride (2641)
Herbal drugs and herbal drug preparations
Follitropin (2285)
Belamcanda chinensis rhizome (2561)
Follitropin concentrated solution (2286)
Eclipta herb (2564)
Human coagulation factor VIIa (rDNA) concentrated solution
Eucommia bark (2412) (2534)
Fleeceflower root (2433) Insulin glargine (2571)

xxi
Contents of the 8 th Edition EUROPEAN PHARMACOPOEIA 8.0

REVISED TEXTS IN THE 8 th EDITION


The texts below have been technically revised since their last publication. They will be implemented on 1 January 2014.
GENERAL CHAPTERS Monographs
Aciclovir (0968)
2.2.40. Near-infrared spectroscopy
Alfacalcidol (1286)
2.4.25. Ethylene oxide and dioxan Brompheniramine maleate (0977)
Cefradine (0814)
2.6.31. Microbiological examination of herbal medicinal Cysteine hydrochloride monohydrate (0895)
products for oral use and extracts used in their
preparation Dexamethasone (0388)
Dextranomer (2238)
3.2.9. Rubber closures for containers for aqueous Dipyridamole (1199)
parenteral preparations, for powders and for Doxapram hydrochloride (1201)
freeze-dried powders
Ethylcellulose (0822)
4. Reagents Fenbendazole for veterinary use (1208)
Finasteride (1615)
5.1.4. Microbiological quality of non-sterile
Flutamide (1423)
pharmaceutical preparations and substances
for pharmaceutical use Glipizide (0906)
Heparins, low-molecular-mass (0828)
5.1.8. Microbiological quality of herbal medicinal Human plasma for fractionation (0853)
products for oral use and extracts used in their Human a-l-proteinase inhibitor (2387)
preparation
Hydrocortisone acetate (0334)
5.8. Pharmacopoeial harmonisation Hypromellose (0348)
Levonorgestrel (0926)
MONOGRAPHS Lomustine (0928)
General monographs Maize starch (0344)
Radiopharmaceutical preparations (0125) Mannitol (0559)
Dosage forms Megestrol acetate (1593)
Parenteral preparations (0520) Metformin hydrochloride (0931)
Premixes for medicated feeding stuffs for veterinary use (1037) Methylcellulose (0345)
Tablets (0478) Methylprednisolone (0561)
Vacdnes for veterinary use Metoprolol succinate (1448)
Aujeszky's disease vaccine (live) for pigs for parenteral Metoprolol tartrate (1028)
administration (0745) Minoxidil (0937)
Rabies vaccine (live, oral) for foxes and raccoan dogs (0746) Nicotine ditartrate dihydrate (2599)
Radiopharmaceutical preparations and starting materials Oxfendazole for veterinary use (1458)
for radiopharmaceutical preparations Peritoneal dialysis, solutions far (0862)
Fludeoxyglucose (1sF) injection (1325) Phytomenadione (1036)
Herbal drugs and herbal drug preparations Potato starch (0355)
Bistort rhizome (2384) Proline (0785)
Capsicum (1859) Serine (0788)
Capsicum oleoresin, refined and standardised (2336) Sulfadiazine (0294)
Capsicum 50ft extract, standardised (2529) Threonine (1049)
Capsicum tincture, standardised (2337) Timolol maleate (0572)
Milk thistle dry extract, refined and standardised (2071) Valine (0796)
Milk thistle fruit (1860) Wheat starch (0359)

CORRECTED TEXTS IN THE 8th EDITION


The texts below from the 7'11 Edition have been corrected and specify 'corrected 8.0' above the title. These corrections are to be
taken ¡nto account from the publication date of the 8th Edition (15 July 2013).
GENERAL CHAPTERS MONOGRAPHS
General chapters
2.4.1. Ammonium
Substances for pharmaceutical use (2034)
2.4.3. Calcium Monographs
Alteplase for injection (1170)
2.4. 13. Sulfates
Amisulpride (1490)
2.5.11. Complexometric titrations Carmellose sodium (0472)
Carvedilol (1745)

xxii
EUROPEAN PHARMACOPOEIA 8.0 Contents of the 8 th Edition

Dacarbazine (1691) Mesalazine (1699)


Fludrocortisone acetate (0767) Nitrendipine (1246)
Human albumin solution (0255) Racecadotril (2171)
Hydroxypropylbetadex (1804) all-rac-a-Tocopherol (0692)
Kanamycin mono sulfate (0032) Urea (0743)
Lactulose, liquid (0924)

TEXTS WHOSE TITLE HAS CHANGED FOR THE 8th EDITION


The titles of the following texts have been changed in the st" Edition.

GENERAL CHAPTERS
2.8.14. Tannins in herbal drugs (previously Determination
2.2.40. Near-infrared spectroscopy (previously of tannins in herbal drugs)
Near-infrared spectrophotometry)
3.1.3. Polyolefins (previously Polyolefines)
2.6.31. Microbiological examination of herbal medicinal
products for oral use and extracts used in 5.1.8. Microbiological quality of herbal medicinal
their preparation (previously Microbiological products for oral use and extracts used in their
examination of herbal medicinal products for oral preparation (previously Microbiological quality of
use) herbal medicinal products for oral use)

2.8.12. Essential oils in herbal drugs (previously MONOGRAPHS


Determination of essential oi/s in herbal drugs) Vaccines for veterinary use
Rabies vaccine (live, oral) for foxes and raceoon dogs (0746)
(previously Rabies vaccine (live, oral) for foxes)

xxiii
EUROPEAN PHARMACOPOEIA 8.0

xxiv
EUROPEAN PHARMACOPOEIA 8.0

1 General notices
e

l. General notices.. ........................ ............................................. 3

General Notices (1) apply to all monographs and other texts


EUROPEAN PHARMACOPOEIA 8.0

2 See the information section on general monographs (caver pages)


EUROPEAN PHARMACOPOEIA 8.0 1. General notices

0112013:10000 QuaHty systems. The quality standards represented by


monographs are valid only where the articles in question are
produced within the framework of a suitable quality system.
1. GENERAL NOTICES
General monographs. Substances and preparations that are
1.1. GENERAL STATEMENTS the subject oí an individual monograph are also required
to comply with relevant, applicable general monographs.
The General Notices apply to al! monographs and other texts Cross-references to applicable general monographs are not
of the European Pharmacopoeia. normally given in individual monographs.
The oÍficial texts of the European Pharmacopoeia are General monographs apply to all substances and preparations
published in English and French. Translations in other within the scope of the Definition section of the general
languages may be prepared by the signatory States of the monograph, except where a preamble limits the application,
European Pharmacopoeia Convention. In case of doubt for example to substances and preparations that are the subject
or dispute, the English and French versions are alone of a monograph of the Pharmacopoeia.
authoritative.
General monographs on dosage forms apply to all preparations
In the texts of the European Pharmacopoeia, the word of the type defined. The requirements are not necessarily
'Pharmacopoeia' without qualification means the European comprehensive for a given specific preparation and
Pharmacopoeia. The official abbreviation Ph. Eur. may be requirements additional to those prescribed in the general
used to indicate the European Pharmacopoeia. monograph may be imposed by the competent authority.
The use of the title or the subtitle of a monograph implies General monographs and individual monographs are
that the article complies with the requirements of the relevant complementary. If the provisions of a general monograph do
monograph. Such references to monographs in the texts of not apply to a particular product, this is expressly stated in the
the Pharmacopoeia are shown using the monograph title and individual monograph.
reference number in italics.
Validation of pharmacopoeial methods. The test methods
A preparation must comply throughout its period of validity; given in monographs and general chapters have been validated
a distinct period of validity and/or specifications for opened in accordance with accepted scientific practice and current
or broached containers may be decided by the competent recommendations on analytical validation. Unless otherwise
authority. The subject of any other monograph must comply stated in the monograph or general chapter, validation of the
throughout its period of use. The period of validity that is test methods by the analyst is not required.
assigned to any given article and the time from which that
period is to be calculated are decided by the competent Implementation of pharmacopoeial methods. When
authority in light of experimental results of stability studies. implementing a pharmacopoeial method, the user must assess
whether and to what extent the suitability of the method
Unless otherwise indicated in the General Notices or in the under the actual conditions of use needs to be demonstrated
monographs, statements in monographs constitute mandatory according to relevant monographs, general chapters and
requirements. General chapters become mandatory when quality systems.
referred to in a monograph, unless such reference is made in a
way that indicates that it is not the intention to make the text Convel1tional terms. The term 'competent authority'
referred to mandatory but rather to cite it for information. means the national, supranational or international body or
organisation vested with the authority for making decisions
The active substances, excipients, pharmaceutical preparations concerning the issue in question. 1t may, for example, be a
and other articles described in the monographs are intended national pharmacopoeia authority, a licensing authority or
for human and veterinary use (unless explicitly restricted an official controllaboratory.
to one of these uses). An article is not of Pharmacopoeia
quality unless it complies with all the requirements stated The expression 'unless otherwise justified and authorised'
in the monograph. This does not imply that performance means that the requirements have to be met, unless the
of all the tests in a monograph is necessarily a prerequisite competent authority authorises a modification or an
for a manufacturer in assessing compliance with the exemption where justified in a particular case.
Pharmacopoeia before release of a product. The manufacturer Statements containing the word 'should' are informative or
may obtain assurance that a product is of Pharmacopoeia advisory.
quality from data derived, for example, from validation studies
of the manufacturing process and from in -process cOl1trols. In certain monographs or other texts, the terms 'suitable' and
Parametric release in circumstances deemed appropriate by 'appropriate' are used to describe a reagent, micro-organism,
the competent authority is thus not precluded by the need to test method etc.; if criteria for suitability are not described in
comply with the Pharmacopoeia. the monograph, suitability is demonstrated to the satisfaction
of the competent authority.
The tests and assays described are the official methods upon
which the standards of the Pharmacopoeia are based. With Medicinal producto (a) Any substance or combination of
the agreement of the competent authority, alternative methods substances presented as having properties for treating or
of analysis may be used for control purposes, provided that the preventing disease in human beings and/or animals; or (b)
methods used enable an unequivocal decision to be made as any substance or combination of substances that may be used
to whether compliance with the standards of the monographs in or administered to human beings and/or animal s with a
would be achieved if the official methods were used. In the view either to restoring, correcting or modifying physiological
event of doubt or dispute, the methods of analysis of the functions by exerting a pharmacological, immunological or
Pharmacopoeia are alone authoritative. metabolic action, or to making a medical diagnosis.

Certain materials that are the subject of a pharmacopoeial Herbal medicinal producto Any medicinal product, exclusively
monograph may exist in different grades suitable for different containing as active ingredients one or more herbal drugs or
purposes. Unless otherwise indicated in the monograph, one or more herbal drug preparations, or one or more such
the requirements apply to all grades of the material. In herbal drugs in combination with one or more such herbal
some monographs, particularly those on excipients, a list of drug preparations.
functionality-related characteristics that are relevant to the Active substance. Any substance intended to be used in
use of the substance may be appended to the monograph for the manufacture of a medicinal product and that, when so
information. Test methods for determination of one or more used, becomesan active ingredient of the medicinal producto
of these characteristics may be given, also for information. Such substances are intended to furnish a pharmacological

General Notices (1) apply to all monographs and other texts 3


l. General uotices EUROPEAN PHARMACOPOEIA 8.0

activity or other direct effect in the diagnosis, cure, mitigation, Any solvent required in a test or assay in which an indicator is
treatment or prevention of disease, or to affect the structure to be used is previously neutralised to the indicator, unless a
and function of the body. blank test is prescribed.
Excipient (auxiliary substance). Any constituent of a medicinal Water-bath. The term 'water-batn means a bath of boiling
product that is not an active substance. Adjuvants, stabilisers, water unless water at another temperature is indicated.
antimicrobial preservatives, diluents, antioxidants, for Other methods of heating may be substituted provided the
example, are excipients. temperature is near to but not higher than 100 oC or the
indicated temperature.
Interchangeable methods. Certain general chapters contain
a statement that the text in question is harmonised with Drying and ignition lo constant mass. The terms 'dried
the corresponding text of the Japanese Pharmacopoeia to constant mass' and 'ignited to constant mass' mean that
and/or the UnitedStates Pharmacopeia and that these texts 2 consecutive weighings do not differ by more than 0.5 mg,
are interchangeable. This implies that if a substance or the 2nd weighing following an additional period of drying or
preparation is found to comply with a requirement using an of ignition respectively appropriate to the nature and quantity
interchangeable method from one of these pharmacopoeias of the residue.
it complies with the requirements of the European
Where drying is prescribed using one of the expressions 'in a
Pharmacopoeia. In the event of doubt or dispute, the text of desiccator' or 'in vacuo', it is carried out using the conditions
the European Pharmacopoeia is alone authoritative.
described in chapter 2.2.32. Loss on drying.
References to regulatory documents. Monographs and
Reagents. The proper conduct of the analytical procedures
general chapters may contain references to documents
described in the Pharmacopoeia and the reliability of the
issued by regulatory authorities for medicines, for example
results depend, in part, upon the quality of the reagents used.
directives and notes for guidance of the European Union.
The reagents are described in general chapter 4. It is assumed
These references are provided for information for users for
that reagents of analytical grade are used; for sorne reagents,
the Pharmacopoeia. Inclusion of such a reference does not
tests to determine suitability are included in the specifications.
modify the status of the documents referred to, which may be
mandatory or for guidance. Solvents. Where the name of the solvent is not stated, the
term 'solution' implies a solution in water.
1.2. OTHER PROVISIONS APPLYING TO GENERAL Where the use of water is specified or implied in the
CHAPTERS AND MONOGRAPHS analytical procedures described in the Pharmacopoeia or
Quantities. In tests with numericallimits and assays, the for the preparation of reagents, water complying with the
quantity stated to be taken for examination is approximate. requirements of the monograph Purified water (0008) is
The amount actually used, which may deviate by not more used, except that for many purposes the requirements for
than 10 per cent from that stated, is accurately weighed or bacterial endotoxins (Purified water in bulk) and microbial
measured and the result is calculated from this exact quantity. contamination (Purified water in cantainers) are not relevant.
In tests where the limit is not numerical, but usually depends The term 'distilled water' indicates purified water prepared
upon comparison with the behaviour of a reference substance by distillation.
in the same conditions, the stated quantity is taken for The term 'ethanol' without qualification means anhydrous
examination. Reagents are used in the prescribed amounts. ethanol. The term 'alcohol' without qualification means
Quantities are weighed or measured with an accuracy ethanol (96 per cent). Other dilutions of ethanol are indicated
commensurate with the indicated degree of precision. For by the term 'ethanol' or 'alcohol' followed by a statement of the
weighings, the precision corresponds to plus or minus 5 units percentage by volume of ethanol (C 2H 6 0) required.
after the last figure stated (for example, 0.25 g is to be Expression of content In defining content, the expression
interpreted as 0.245 g to 0.255 g). For the measurement of 'per cent' is used according to circumstances with one of 2
volumes, if the figure after the decimal point is a zero or ends meanings:
in a zero (for example, 10.0 mL or 0.50 mL), the volume is
measured using a pipette, a volumetric flask or a burette, as - per cent m/m (percentage, mass in mass) expresses the
appropriate; otherwise, a graduated measuring cylinder or a number of grams of substance in 100 g of final product;
graduated pipette may be used. Volumes stated in microlitres - per cent V/V (percentage, volume in volume) expresses
are measured using a micropipette or microsyringe. the number of millilitres of substance in 100 mL of final
It is recognised, however, that in certain cases the precision product.
with which quantities are stated do es not correspond to the The expression 'parts per million' (or ppm) refers to mass in
number of significant figures stated in a specified numerical mass, unless otherwise specified.
limit. The weighings and measurements are then carried out
with a sufficiently improved accuracy. Temperature. Where an analytical procedure describes
temperature without a figure, the general terms used have the
Apparatus and procedures. Volumetric glassware complies following meaning:
with Class A requirements of the appropriate International
Standard issued by the International Organisation for - in a deep-freeze: below - 15 oC;
Standardisation. - in a refrigerator: 2 oC to 8 oC;
Unless otherwise prescribed, analytical pro ce dures are carried - cold or cool: 8 oC to 15 oC;
out at a temperature between 15 oC and 25 oc.
- room temperature: 15 oC to 25 oc.
Unless otherwise prescribed, comparative tests are carried out
using identical tubes of colourless, transparent, neutral glass
with a flat base; the volumes of liquid prescribed are for use 1.3. GENERAL CHAPTERS
with tubes having an internal diameter of 16 mm, but tubes Containers. Materials used for containers are described
with a larger internal diameter may be used provided the in general chapter 3.1. General names used for materials,
volume ofliquid used is adjusted (2.1.5). Equal volumes of particularly plastic materials, each cover a range of products
the liquids to be compared are examined down the vertical varying not only in the properties of the principal constituent
axis of the tubes against a white background, OI if necessary but also in the additives used. The test methods and limits
against a black background. The examination is carried out in for materials depend on the formulation and are therefore
diffuse light. applicable only for materials whose formulation is covered by

4 See the information section on general monographs (ca ver pages)


EUROPEAN PHARMACOPOEIA 8.0 l. General notices

the preamble to the specification. The use of materials with Choice of vacdne strain, Choice of vacdne composition.
different formulations, and the test methods and limits applied The Production section of a monograph may define the
to them, are subject to agreement by the competent authority. characteristics of a vaccine strain or vaccine composition.
Unless otherwise stated, test methods given for verification of
The specifications for containers in general chapter 3.2 these characteristics are provided for information as examples
have been developed for general application to container s of suitable methods. Subject to approval by the competent
of the stated category, but in view of the wide variety of authority, other test methods may be used without validation
containers available and possible new developments, the against the method shown in the monograph.
publication of a specification does not exclude the use, in
justified circumstances, of containers that comply with POTENTIAL ADULTERATION
other specifications, subject to agreement by the competent Due to the increasing number of fraudulent activities and
authority. cases of adulteration, information may be made available to
Ph. Eur. users to help detect adulterated materials (i.e. active
Reference may be made within the monographs of the substances, excipients, intermediate products, bulk products
Pharmacopoeia to the definitions and specifications for and finished products).
containers provided in chapter 3.2. Containers. The general To this purpose, a method for the detection of potential
monographs for pharmaceutical dosage forms may, under adulterants and relevant limits, together with a reminder that
the heading Definition/Production, require the use of certain all stages of production and sourcing are subjected to a suitable
types of container; certain other monographs may, under quality system, may be included in this section of monographs
the heading Storage, indicate the type of container that is on substances for which an incident has occurred or that
recommended for use. present a risk of deliberate contamination. The frequency of
testing by manufacturers or by users (e.g. manufacturers of
lA. MONOGRAPHS intermediate products, bulk products and finished products,
where relevant) depends on a risk assessment, taking into
TITLES account the level of knowledge of the whole suppIy chain and
Monograph titles are in English and French in the respective national requirements.
versions and there is a Latin subtitle. This section constitutes requirements for the whole supply
RELATIVE A TOMIC AND MOLECULAR MASSES chain, from manufacturers to users (e.g. manufacturers of
The relative atomic mass (A r ) or the relative molecular intermediate products, bulk products and finished products,
mass (M) is shown, as and where appropriate, at the beginning where relevant). The absence of this section does not imply
of each monograph. The relative atomic and molecular masses that attention to features such as those referred to aboye is
and the molecular and graphic formulae do not constitute not required.
analytical standards fOI the substances described. CHARACTERS
CHEMICAL ABSTRACTS SERVICE (CAS) REGISTRY The statements under the heading Characters are not to be
NUMBER interpreted in a strict sense and are not requirements.
CAS registry numbers are included for information in Solubility. In statements of solubility in the Characters
monographs, where applicable, to provide convenient access section, the terms used have the following significan ce,
to useful information for users. CAS Registry Number@ is a referred to a temperature between 15 oC and 25 oc.
registered trademark of the American Chemical Society. Descriptive term Approximate volume of solvent in millilitres
DEFINITION per gram of solute
Statements under the heading Definition constitute an official Very soluble less than 1
definition of the substance, preparation or other article that is to 10
Freely soluble from
the subject of the monograph.
Soluble from 10 to 30
Limits of content. Where limits of content are prescribed,
they are those determined by the method described under Sparingly soluble from 30 to 100
Assay. to
Slightly soluble from 100 1000
Herbal drugs. In monographs on herbal drugs, the definition
Very slightly soluble from 1000 to 10000
indicates whether the subject of the monograph is, for
example, the whole drug or the drug in powdered formo Practically insoluble more than 10000
Where a monograph applies to the drug in several sta tes, for
example both to the whole drug and the drug in powdered The term 'partly soluble' is used to describe a mixture where
form, the definition states this. only some of the components dissolve. The term 'miscible' is
used to describe a liquid that is miscible in all proportions
PRODUCTION
with the stated solvent.
Statements under the heading Production draw attention
to particular aspects of the manufacturing process but are IDENTIFICA TION
not necessarily comprehensive. They constitute mandatory Scope. The tests given in the Identification section are not
requirements for manufacturers, unless otherwise stated. designed to give a ful! confirmation of the chemical structure
They may relate, for example, to source materials; to the or composition of the product; they are intended to give
manufacturing process itself and its validation and control; to confirmation, with an acceptable degree of assurance, that the
in-process testing; or to testing that is to be carried out by the article conforms to the description on the labe!'
manufacturer on the final article, either on selected batches First and second identifications. Certain monographs
or on each batch prior to release. These statements cannot have subdivisions entitled 'First identification' and 'Second
necessarily be verified on a sample of the final article by an identification'. The test or tests that constitute the 'First
independent analyst. The competent authority may establish identification' may be used in all circumstances. The test or
that the instructions have been followed, for example, by tests that constitute the 'Second identification' may be used
examination of data received from the manufacturer, by in pharmacies provided it can be demonstrated that the
inspection of manufacture or by testing appropriate samples. substance or preparation is fully traceable to a batch certified
The absence of a Production section does not imply that to comply with all the other requirements of the monograph.
attention to features such as those referred to aboye is not Certain monographs give two or more sets of tests for the
required. purpose of the first identification, which are equivalent

General Notices (1) apply to all monographs and other texts 5


l. General notices EUROPEAN PHARMACOPOEIA 8.0

and may be used independently. Oue or more of these sets Culture media. The culture media described in monographs
usually contain a cross-reference to a test prescribed in the and general chapters have been found to be satisfactory for
Tests section of the monograph. It may be used to simplify the intended purpose. However, the components of media,
the work of the analyst carrying out the identification and particularly those of biological origin, are of variable quality,
the prescribed tests. For example, oue identification set and it may be necessary for optimal performance to modulate
cross-refers to a test for enantiomeric purity while the other the concentration of sorne ingredients, notably:
set gives a test for specific optical rotation: the intended - peptones and meat or yeast extracts, with respect to their
purpose of the two is the same, that is, verification that the nutritive properties;
correct enantiomer is present.
- buffering substances;
Powdered herbal drugs. Monographs on herbal drugs may
contain schematic drawings of the powdered drug. These - bile salts, bile extract, deoxycholate, and colouring matter,
drawings complement the description given in the relevant depending on their selective properties;
identification test. - antibiotics, with respect to their activity.
TESTS AND ASSA YS STORAGE
Scope. The requirements are not framed to take account of all The information and recommendations given under the
possible impurities. It is not to be presumed, for example, that heading Storage do not constitute a pharmacopoeial
an impurity that is not detectable by means of the prescribed requirement but the competent authority may specify
tests is tolerated if common sense and good pharmaceutical particular storage conditions that must be meto
practice require that it be absent. See also below under
The articles described in the Pharmacopoeia are stored
Impurities.
in such a way as to prevent contamination and, as far as
Calculation. Where the result of a test or assay is required possible, deterioration. Where special conditions of storage
to be calculated with reference to the dried or anhydrous are recommended, including the type of container (see section
substance or on sorne other specified basis, the determination 1.3. General chapters) and limits of temperature, they are
of loss on drying, water content or other property is carried stated in the monograph.
out by the method prescribed in the relevant test in the
The following expressions are used in monographs under
monograph. The words 'dried substance' or 'anhydrous
Storage with the meaning shown.
substance' etc. appear in parentheses after the resulto
Where a quantitative determination of a residual solvent is In an airtight container means that the product is sto red in an
carried out and a test for loss on drying is not carried out, airtight container (3.2). Care is to be taken when the container
the content of residual solvent is taken into account for the is opened in a damp atmosphere. A low moisture content
calculation of the assay content of the substance, the specific may be maintained, if necessary, by the use of a desiccant in
optical rotation and the specific absorbance. No further the container provided that direct contact with the product
indication is given in the specific monograph. is avoided.
Limits. The Iimits prescribed are based on data obtained Protected from light means that the product is stored either
in normal analytical practice; they take account of normal in a container made of a material that absorbs actinic light
analytical errors, of acceptable variations in manufacture and sufficiently to protect the contents from change induced by
compounding and of deterioration to an extent considered such light, or in a container enclosed in an outer cover that
acceptable. No further toleran ces are to be applied to the Iimits provides such protection, or is sto red in a place from which all
prescribed to determine whether the article being examined such light is excluded.
complies with the requirements of the monograph. LABELLING
In determining compliance with a numericallimit, the In general, labelling of medicines is subject to supranational
calculated result of a test or assay is first rounded to the and national regulation and to international agreements. The
number of significant figures stated, unless otherwise statements under the heading Labelling are not therefore
prescribed. The limits, regardless of whether the values are comprehensive and, moreover, for the purposes of the
expressed as percentages or as absolute values, are considered Pharmacopoeia only those statements that are necessary
significant to the last digit shown (for example 140 indicates 3 to demonstrate compliance or non -compliance with the
significant figures). The last figure of the result is increased by monograph are mandatory. Any other labelling statements are
one when the part rejected is equal to or exceeds one half-unit, included as recommendations. When the term 'labe!' is used
whereas it is not modified when the part rejected is less than a in the Pharmacopoeia, the labelling statements may appear
half-unit. on the container, the package, a leaflet accompanying the
Indication of permitted limit of impurities. The acceptance package, or a certificate of analysis accompanying the artide,
criteria for related substances are expressed in monographs as decided by the competent authority.
either in terms of comparison of peak are as (comparative tests) WARNINGS
or as numerical values. For comparative tests, the approximate Materials described in monographs and reagents specified
content of impurity tolerated, or the sum of impurities, may for use in the Pharmacopoeia may be injurious to health
be indicated in brackets for information only. Acceptance unless adequate precautions are taken. The principIes of
or rejection is determined on the basis of compliance or good quality controllaboratory practice and the provisions
non -compliance with the stated test. If the use of a reference of any appropriate regulations are to be observed at all
substance for the named impurity is not prescribed, this times. Attention is drawn to particular hazards in certain
content may be expressed as a nominal concentration of the monographs by means of a warning statement; absence of such
substance used to prepare the reference solution specified in a statement is not to be taken to mean that no hazard exists.
the monograph, unless otherwise described.
IMPURITIES
Herbal drugs. For herbal drugs, the sulfated ash, total ash, A list of all known and potential impurities that have been
water-soluble matter, alcohol-soluble matter, water content, shown to be detected by the tests in a monograph may be
content of essential oil and content of active principIe are given. See also chapter 5.10. Control of impurities in substances
calculated with reference to the drug that has not been for pharmaceutical use. The impurities are designated by a
specially dried, unless otherwise prescribed in the monograph. letter or letters of the alphabet. Where a letter appears to
Equivalents. Where an equivalent is given, for the purposes be missing, the impurity designated by this letter has been
of the Pharmacopoeia only the figures shown are to be used in deleted from the list during monograph development prior to
applying the requirements of the monograph. publication or during monograph revision.

6 See the information section on general monographs (cover pages)


EUROPEAN PHARMACOPOEIA 8.0 1. General notices

FUNCTIONALITY-RELATED CHARACTERISTICS OF L+/10 dose The smallest quantity of a toxin that, in the
EXCIPIENTS conditions of the test, when mixed with
Monographs on excipients may have a section on 0.1 IU of antitoxin and administered by the
functionality-related characteristics. The characteristics, any specified route, causes the death of the test
test methods for determination and any tolerances are not animals within a given period
mandatory requirements; they may nevertheless be relevant L+ dose The smallest quantity of a toxin that, in the
for use of the excipient and are given for information (see also conditions of the test, when mixed with
section 1.1. General statements). 1 IU of antitoxin and administered by the
REFERENCESTANDARDS specified route, causes the death of the test
Certain monographs require the use of reference standards animals within a given period
(chemical reference substances, herbal reference standards, Ir/lOO dose The smallest quantity of a toxin that, in
biological reference preparations, reference spectra). See the conditions of the test, when mixed
also chapter 5.12. Reference standards. The European with 0.01 IU of antitoxin and injected
Pharmacopoeia Commission establishes the official intracutaneously causes a characteristic
reference standards, which are alone authoritative in case reaction at the site of injection within a
of arbitration. These reference standards are available from given period
the European Directorate for the Quality of Medicines & Lp/lO dose The smallest quantity of toxin that, in the
HealthCare (EDQM). Information on the available reference conditions of the test, when mixed with
standards and a batch validity statement can be obtained via 0.1 IU of antitoxin and administered by the
the EDQM website. specified route, causes paralysis in the test
animals within a given period
Lol10 dose The largest quantity of a toxin that, in the
1.5. ABBREVIATIONS AND SYMBOLS conditions of the test, when mixed with
A Absorbance 0.1 IU of antitoxin and administered by the
specified route, does not cause symptoms of
Al per cent Specific absorbance toxicity in the test animals within a given
1cm
Relative atomic mass period
Ar
Lf dose The quantity of toxin or toxoid that
[al~O Specific optical rotation
flocculates in the shortest time with 1 IU of
bp Boiling point antitoxin
BRP Biological reference preparation CCID so The statistically determined quantity of
virus that may be expected to infect 50 per
CRS Chemical reference substance cent of the ceU cultures to which it is added
d 20
20 Relative density The statistically determined quantity of
virus that may be expected to infect 50 per
Wavelength
ceni of the fertilised eggs into which it is
HRS Herbal reference standard inoculated
ID International Unit The statistically determined quantity of
a virus that may be expected to infect
M Molarity 50 per cent of the animals into which it is
Mr Relative molecular mass inoculated
mp Melting point The statistically determined dose of a
vaccine that, in the conditions of the test,
Refractive index may be expected to protect 50 per cent of
Ph. Eur. U. European Pharmacopoeia Unit the animals against a challenge dose of the
micro-organisms or toxins against which it
ppb Parts per billion (micrograms per kilogram) is active
ppm Parts per million (milligrams per kilogram) The statistically determined dose of a
vaccine that, in the conditions of the
R Substance or solution defined under test, may be expected to induce specific
4. Reagents antibodies in 50 per cent of the animals for
Retardation factor (see chapter 2.2.46) the relevant vaccine antigens
Used in chromatography to indicate the PFU Pock-forming units or plaque-forming units
ratio of the distan ce travelled by a substance SPF Specified-pathogen -free
to the distance travelled by a reference
substance Collections of micro-organisms
RV Substance used as a primary standard in ATCC American Type Culture Collection
volumetric analysis (chapter 4.2.1)
10801 University Boulevard
Abbreviations used in the monographs on Manassas, Virginia 20110-2209, USA
immunoglobulins, immunosera and vaccines C.LP. Collection de Bactéries de !'Institut Pasteur
LDso The statistically determined quantity of a B.p. 52, 25 rue du Docteur Roux
substance that, when administered by the
specified route, may be expected to cause 75724 Paris Cedex 15, France
the death of 50 per cent of the test animals 1MI International Mycological Institute
within a given period Bakeham Lane
MLD Minimum lethal dose Surrey TW20 9TY, Great Britain

General Natices (1) apply ta all managraphs and ather texts 7


l. General notices EUROPEAN PHARMACOPOEIA 8.0

LP. Collection Nationale de Culture de 1.6. UNITS OF THE INTERNATIONAL SYSTEM (SI) USED
Microorganismes (C.N.C.M.) IN THE PHARMACOPOEIA AND EQUIVALENCE WITH
Institut Pasteur OTHER UNITS
25, rue du Docteur Roux INTERNA TIONAL SYSTEM OF UNITS (SI)
The International System of Units eomprises 3 classes of units,
75724 Paris Cedex 15, France
namely base units, derived units and supplementary unitsOJ.
NCIMB National Collection of Industrial and The base units and their definitions are set out in Table 1.6-l.
Marine Bacteria Ud
The derived units may be formed by combining the
23 St Machar Drive base units aceording to the algebraic relationships linking
Aberdeen AB2 lRY, Great Britain the corresponding quantities. Sorne of these derived units
NCPF National Collection of Pathogenic Fungí have speeial names and symbols. The SI units used in the
Pharmacopoeia are shown in Table 1.6-2.
London School of Hygiene and Tropical
Medicine Sorne important and widely used units outside the
International System are shown in Table 1.6-3.
Keppel Street
The prefixes shown in Table 1.6-4 are used to form the names
London WCIE 7HT, Great Britain
and symbols of the decimal multiples and submultiples of
NCTC National Collection of Type Cultures SI units.
Central Public Health Laboratory NOTES
Colindale Avenue 1. In the Pharmacopoeia, the Celsius temperature is used
London NW9 5HT, Great Britain (symbol t). This is defined by the following equation:
NCYC National Collection of Yeast Cultures
t=T-To
AFRC Food Research Institute
Colney Lane where To = 273.15 K by definition. The Celsius or eentigrade
Norwich NR4 7UA, Great Britain temperature is expressed in degrees Celsius (symbol OC).
The unit 'degree Celsius' is equal to the unit 'kelvin'.
NITE Biological Resource Center
2. The practical expressions of eoncentrations used in the
Department of Biotechnology Pharmaeopoeia are defined in the General Notices.
National Institute of Technology and 3. The radian is the plane angle between two radii of a circle
Evaluation that cut off on the cireumferenee an are equal in length
2-5-8 Kazusakamatari, Kisarazu-shi, Chiba, to the radius.
292-0818 4. In the Pharmacopoeia, conditions of eentrifugation are
Tapan defined by referenee to the aceeleration due to gravity (g) :
S.S.L Statens Serum Institut
g = 9.80665 m·s- 2
80 Amager Boulevard, Copenhagen,
Denmark 5. Certain quantities without dimensions are used in the
Pharmacopoeia: relative density (2.2.5), absorbance
(2.2.25), specific absorbance (2.2.25) and refractive index
(2.2.6).
6. The microkatal is defined as the enzymie aetivity that,
under defined conditions, produces the transformation
(e.g. hydrolysis) of 1 micromole of the substrate per second.
Table 1.6.-1. - SI base units
Quantity Unit Definition

Name Symbol Name Symbol

m The metre is the length of the path travelled by light in a vacuum during a time
Length 1 metre
interval of 1/299 792 458 of a second.
Mass m kilogram kg The kilogram is equal to the mass of the international prototype of the kilogramo

The second is the duration of9 192 631 770 periods ofthe radiation corresponding
Time t second s to the transition between the two hyperfine levels of the ground state of the
caesium-133 atom.
Electric current J ampere A The ampere is that constant current which, maintained in two straight parallel
conductors of infinite length, of negligible circular cross-section and placed 1 metre
apart in vacuum would produce between these conductors a force equal to 2 x 10- 7
newton per metre of length.
Thermodynamic T kelvin K The kelvin is the fraction 1/273.16 ofthe thermodynamic temperature ofthe triple
temperature point of water.
Amount of substance n mole mol The mole is the amount of substance of a system containing as many elementary
entities as there are atoms in 0.012 kilogram of carbon-12*.
Luminous intensity J" candela cd The candela is the luminous intensity in a given direction of a source emitting
monochromatic radiation with a frequency of 540 x 10 12 hertz and whose energy
intensity in that direction is 1/683 watt per steradian.
* When the mole is used, the elementary entities must be specified and may be atoms, molecules, ions, electrons, other partides or specified
groups of such particles.

(1) The definitions ofthe units used in the International System are given in the booklet 'Le Systeme International d'Unités (SI)', published bythe Bureau International des Poids
et Mesures, Pavillon de Breteuil, F-92310 Sevres.

8 See the information section on general monographs (cover pages)


EUROPEAN PHARMACOPOEIA 8.0 1. General notices

Table 1.6.-2. - SI units used in the European Pharmacopoeia and equivalence with other units
Quantity Unit

Expression in Conversion of olher units into SI units


Name Symbol Name Symbol Expression in SI
base units olher SI .mits
v one per metre m--¡
Wave number 11m

Wavelength Á lnicrOlnetre [un 1O- 6 m


nanometre nm 1O- 9 m

Area A, S square metre m' ln l

Volume V eubie metre 111 3 m3 1 mL = 1 cm 3 = 10- 6 m 3

Frequency v hertz Hz s I

Density P kilogram per eubie kg/m 3 kg.m-' 1 g/mL = 1 g/em' = 103 kg.m- 3
rnetre

Veloeity v metre per seeond mis Ill'S-l

Force F newton N m·kg·s 1 dyne = 1 g·C1n·s -2=10- 5 N


1 kp = 9.806 65 N
Pressure p pascal Pa m-'·kg·s N·rn- 2 1 dyne/em' = lO' I Pa = 10- l N'1l1-2

1 alm = 101 325 Pa = 101.325 kPa


1 bar = 105 Pa = 0.1 MPa
1 mm Hg = 133.322 387 Pa
1 Torr = 133.322 368 Pa
1 psi = 6.894 757 kPa
Dynamie r¡ pascal seeond Pa·s 111-l.lZg·S- 1 N·s·m" 1 P = 10- I Pa·s = 10- I N-s·m-'
viseosity 1 eP = 1 mPa·s
v 1112.S- 1
Kinenlatic square metre per m'/s Pa·s·m'·kg- 1 1 St = 1 em'·s- I = 10- 4 m'.s- I
viscosity seeond N'111'S.kg- 1

Energy W joule I ln2.kg·s N-m 1 erg = 1 em 2'g's-' = 1 dyne·em = 10 7)

1 cal = 4.1868)
Power P watt W m 2 ·kg·s- J N'1l1'S- 1
1 erg/s = 1 dyne·em·s- ' =
Radiant flux J-s-I 10- 7 W = 10- 7 N-m·s- I = 10' 7 ¡'S-I

Absorbed dose D gray Gy m 2 ·s- 2 )·kg- 1 ! rad = 10- 2 Gy


(of radiant
energy)
Electric U volt V m 2 . kg.s- 3.A- I W·A- '
potential,
electro motive
force
Eleclric R ohm n ln z kg·s J·A- 2 V·A- 1
resistan ce

Quantity of Q coulomb C As
eleetricity
s- ,
Activity of a A beequerel Bq 1 Ci = 37.10 9 Bq = 37.10 9 S'I
radionuclide
Concentratian e mole per eubie mol/m 3 lllohn- 3 1 mol/L = 1 M = lmol/dm' = 103 mol·m- 3
(of amount 111etre
of substanee),
molar
concentraban
Mass P kilogram per eubie kg/m' kg·m-' 1 giL = 1 g/dm 3 = 1 kg·m J
concentratian metre

Table 1 6.-3 - Units used with the International System Table l.6.-4. - Decimal multiples and sub-multiples of units
Quantity Unít Value in SI uníts
Factor Prefix Symbol Factor Prefix Symbol
Name Symbol
10" exa E 10-' deci d
Time minute min 1 min = 60 s
10 10 peta P 10- 3 centi e
hour h 1 h = 60 min = 3600 s
10" tera T 10- , milli m
day d 1 d = 24 h = 86 400 s
10 9 giga G 10- 6 micro f1
o
Plane angle degree 10 = (rr/180) rad
106 mega M 10- 9 nano n
Volume litre L 1 L = 1 dm' = 10- 3 m'
10 3 kilo k 10-" pico P
Mass tonne t 1 t = lO' kg
lO' hecto h 10- 15 femto f
Rotational revolution r/min 1 r/min = (l/60) S-I
lO' deca da 10-'" atto a
frequency per minute

General Notices (1) apply to all monographs and other texts 9


EUROPEAN PHARMACOPOEIA 8.0

10 See the information section on general monographs (cover pages)


EUROPEAN PHARMACOPOEIA 8.0

2. Methods of analysis

General Notices (1) apply to all monographs and other texts 11


EUROPEAN PHARMACOPOEIA 8.0

12 See the information section on general monographs (cover pages)


EUROPEAN PHARMACOPOEIA 8.0

2.1. Apparatus
2.1. Apparatus ............................................................................. 15 2.1.4. Sieves ................................................................................. 16
2.1.1. Droppers .......................................................................... 15 2.1.5. Tubes for comparative tests ............................................ 17
2.1.2. Comparative table of porosity of sintered-glass filters .. 15 2.1.6. Gas detector tubes ............................................................ 17
2.1.3. Ultraviolet ray lamps for analytical purposes ............... 15

General Natices (1) apply ta all managraphs and ather texts 13


EUROPEAN PHARMACOPOEIA 8.0

14 See the information section on general monographs (ca ver pages)


EUROPEAN PHARMACOPOEIA 8.0 2.1.3. Ultraviolet ray lamps for analytical purposes

2.1. APPARATUS 0112008:20102

2.1.2. COMPARATIVE TABLE OF


01/2008:20101
POROSITY OF SINTERED-GLASS
FILTERS(l)
2.1.1. DROPPERS Table 2.1.2.-1
Porosity number Maximum Gennany Franee United
The term 'drops' means standard drops delivered from a (Ph. Eur.)(2) diameter of pores Kingdom
standard dropper as described below. in micrometres
l.6 less than l.6 5f
Standard droppers (Figure 2.1.1-1) are constructed of
practically colourless glass. The lower extremity has a circular 1 - 2.5 5 5
orifice in a flat surface at right angles to the axis.
4 l.6 - 4

4-6 5

I
10 4 - 10 4f 4

! 16 10 - 16 4 4
I 40 16 - 40 3 3 3

I 40 - 50 2

100 40 - 100 2 2
I
100 - 120
I 160 100 - 160
I 150 - 200 O O

250 160 - 250

200 - 500 00

Special Uses
Dimneters in micrometres
o
l!)
< 2.5 Bacteriological filtration

4 - 10 Ultra-fine filtration, separabon af micro-organisms oflarge


diameter
10 - 40 Analytica! filtraban, very fine filtration of mercury, very fine
--;.
~ dispersian of gases
40 - 100 Fine filtration, filtration of mercury, fine dispersion of gases

100 - 160 Filtration of coarse materials, dispersion and washing of gases,


support for other filter materials
160 - 500 Filtration of very coarse materials, dispersion and washing of
gases.
/
0112008:20103
o
(0

~;\ 2.1.3. ULTRAVIOLET RAY LAMPS FOR


ANALYTICAL PURPOSES
1\ 1/ Mercury vapour in quartz lamps is used as the source of
o
~
1 ultraviolet light. A suitable filter may be fitted to eliminate the
visible part of the spectrum emitted by the lampo When the
Pharmacopoeia prescribes in a test the use of ultraviolet light
---..¡ It-- 2: 1.0 of wavelength 254 nm or 365 nm, an instrument consisting of
---->i +-- 3.0 0-3.05 a mercury vapour lamp and a filter which gives an emission
band with maximum intensity at about 254 nm or 365 nm is
Figure 2.1.1.-1. - Standard dropper used. The lamp used should be capable of revealing without
Dimensions in millimetres doubt a standard spot of sodium salicylate with a diameter
of about 5 mm on a support of silica gel G R, the spot being
Other droppers may be used provided they comply with the examined while in a position normal to the radiation.
following test.
For this purpose apply 5 flL of a 0.4 giL solution of sodium
20 drops of water R at 20 ± 1 oC flowing freely from the salicylate R in alcohol R(3) for lamps of maximum output at
dropper held in the vertical position at a constant rate of 254 nm and 5 flL of a 2 giL solution in alcohol R(3) for lamps of
1 drop per second weighs 1000 ± 50 mg. maximum output at 365 nm. The distan ce between the lamp
The dropper must be carefully cleaned before use. Carry out 3 and the chromatographic plate under examination used in a
determinations on any given dropper. No result may deviate by pharmacopoeial test should never exceed the distance used to
more than 5 per cent from the mean of the 3 determinations. carry out the aboye test.

(1) The given limils are only approximate.


(2) The European Pharmacopoeia has adopted the system proposed by the lnternational Organizatian [ar Standardization (ISO).
(3) The alcohol R used ll1ust be frce fro111 fluorescence.

General Notices (1) apply to all monographs and other texts 15


2.1.4. Sieves EUROPEAN PHARMACOPOEIA 8.0

01/2008:20104 Tolerance for mean aperture (± Y): the average aperture size
shall not depart from the nominal size by more than ± Y,
2.1.4. SIEVES where:
WO. 98
Sieves are constructed of suitable materials with square
meshes. For purposes other than analytical procedures, sieves y= ~ +1.6
with circular meshes may be used, the internal diameters of
which are 1.25 times the aperture of the square mesh of the Intermediary tolerance (+ Z): not more than 6 per cent of the
corresponding sieve size. There must be no reaction between total number of apertures shall have sizes between "nominal
the material of the sieve alld the substance being sifted. + x" and "nominal + Z", where:
Degree of comminution is prescribed in the monograph
using the sieve number, which is the size of the mesh in z= X+Y
micrometres, given in parenthesis after the name of the 2
substance (Table 2.1.4.-1).
Wire diameter d: the wire diameters given in Table 2.1.4.-1
Maximum tolerance(4) for an aperture (+ X): no aperture size apply to woven metal wire cloth mounted in a frame. The
shall exceed the nominal size by more than X, where: nominal sizes of the wire diameters may depart from these
2 (WO. 75 ) values within the limits d max and d min • The limits define a
X= +4 (WO 25) permissible range of choice ± 15 per cent of the recommended
3
nominal dimensions. The wires in a test sieve shall be of a
w = width of aperture. similar diameter in warp and weft directions.
Table 2.1.4.-1 (values in micrometers)
Sieve Tolerances for apertures Wire diameters
numbers
Maximum Tolerance for Intermediary Recommended Admissible Iimits
(Nominal
tolerance for mean aperture tolerance nominal
dimensions of
an aperture dimensions
apertures)
+X ±Y +Z d dmax dmin
11 200 770 350 560 2500 2900 2100
8000 600 250 430 2000 2300 1700
5600 470 180 320 1600 1900 1300
4000 370 130 250 1400 1700 1200
2800 290 90 190 1120 1300 950
2000 230 70 150 900 1040 770
1400 180 50 110 710 820 600
1000 140 30 90 560 640 480
710 112 25 69 450 520 380
500 89 18 54 315 360 270
355 72 13 43 224 260 190
250 58 9.9 34 160 190 130
180 47 7.6 27 125 150 106
125 38 5.8 22 90 104 77
90 32 4.6 18 63 72 54
63 26 3.7 15 45 52 38
45 22 3.1 13 32 37 27
38 - - -
30 35 24

(4) See lhe ¡ntemational Standard ISO 331011 (1975).

16 See the information section on general monographs (cover pages)


EUROPEAN PHARMACOPOEIA 8.0 2.1.6. Gas detector tubes

01/2008:20105 In view of the wide variety of available compressor oils, it is


necessary to verify the reactivity of the oil detector tubes for
the oil used. Information on the reactivity for various oils is
2.1.5. TUBES FOR COMPARATIVE given in the leaflet supplied with the tube. If the oil used is
TESTS not cited in the leaflet, the tube manufacturer must verify the
reactivity and if necessary provide a tube speeific for this oil.
Tubes used for comparative tests are matched tubes of
2 3 4
colourless glass with a uniform internal diameter. The base
is transparent and flato 1------,-------- 7
A column of the liquid is examined down the vertical axis of
the tube against a white background, or if necessary, against a
black background. The examination is carried out in diffused 5
light.
It is assumed that tubes with an internal diameter of 16 mm
will be used. Tubes with a larger internal diameter may be
6
used instead but the volume of liquid examined must then be
increased so that the depth of liquid in the tubes is not less
than where the prescribed volume ofliquid and tubes 16 mm
in internal diameter are used. l. Gas supply 5. Indicator tube

2. Pressure regulator 6. Indicator tube pump

3. Needle valve 7. End open to atmosphere


0112008:20106
4. "Y" -pie ce

2.1.6. GAS DETECTOR TUBES Figure 2.1.6.-1. - Apparatus for gas detector tubes
Carbon dioxide detector tube. Sealed glass tube containing
Gas detector tubes are cylindrical, sealed tubes consisting of adsorbent filters and suitable supports for hydrazine and
an inert transparent material and are constructed to allow crystal violet indicators. The minimum value indicated is
the passage of gas. They contain reagents adsorbed onto 100 ppm with a relative standard deviation of at most ± 15 per
inert substrates that are suitable for the visualisation of the cent.
substance to be detected and, if necessary, they also contain
preliminary layers and/or adsorbent filters to eliminate Sulfur dioxide detector tube. Sealed glass tube containing
substances that interfere with the substance to be detected. adsorbent filters and suitable supports for the iodine and
The layer of indicator contains either a single reagent for starch indicator. The minimum value indicated is 0.5 ppm
the detection of a given impurity or several reagents for the with a relative standard deviation of at most ± 15 per cent.
detection of several substances (monolayer tube or multilayer Oil detector tube. Sealed glass tube eontaining adsorbent
tube). filters and suitable supports for the sulfurie aeid indicator.
The test is carried out by passing the required volume of the The minimum value indicated is 0.1 mg/m J with a relative
gas to be examined through the indicator tube. The length standard deviation of at most ± 30 per eent.
of the coloured layer or the intensity of a colour change on a Nitrogen monoxide and nitro gen dioxide detector
graduated scale gives an indication of the impurities presento tube. Sealed glass tube containing adsorbent filters and
The calibration of the detector tubes is verified according to suitable supports for an oxidising layer (Cr(VI) salt) and the
the manufacturer's instructions. diphenylbenzidine indicator. The minimum value indicated is
0.5 ppm with a relative standard deviation of at most ± 15 per
Operating conditions. Examine according to the manufacturer's eent.
instructions or proceed as follows:
Carbon monoxide detector tube. Sealed glass tube
The gas supply is connected to a suitable pressure regulator containing adsorbent filters and suitable supports for di-iodine
and needle valve. Conneet the flexible tubing fitted with a pentoxide, selenium dioxide and fuming sulfuric acid
Y-piece to the valve and adjust the f10w of gas to be examined indicators. The minimum value indicated is 5 ppm or less,
to purge the tubing in order to obtain an appropriate f10w with a relative standard deviation of at most ± 15 per eent.
(Figure 2.1.6.-1). Prepare the indicator tube and fit to the
metering pump, following the manufacturer's instructions. Hydrogen sulfide detector tube. Sealed glass tube eontaining
Connect the open end of the indicator tube to the short leg of adsorbent filters and suitable supports for an appropriate lead
the tubing and operate the pump by the appropriate number salt indicator. The minimum value indicated is 1 ppm or less,
of strokes to pass a suitable volume of gas to be examined with a relative standard deviation of at most ± 10 per cent.
through the tube. Read the value corresponding to the length Water vapour detector tube. Sealed glass tube containing
of the coloured layer or the intensity of the colour on the adsorbent filters and suitable supports for the magnesium
graduated seale. If a negative result is achieved, indieator perchlorate indicator. The minimum value indicated is
tubes can be verified with a calibration gas containing the 67 ppm or less, with a relative standard deviation of at most
appropriate impurity. ± 20 per cent.

General Notices (1) apply lo all monographs and other texts 17


EUROPEAN PHARMACOPOEIA 8.0

18 See the information section on general monographs (cover pages)


EUROPEAN PHARMACOPOEIA 8.0

2.2. Physical and physicochemical


methods
2.2. Physical and physicochemical methods ........................... 21 2.2.32. Loss on drying ................................................................ 52
2.2.1. Clarity and degree of opalescence ofliquids ................ 21 2.2.33. Nuclear magnetic resonance spectrometry................. 52
2.2.2. Degree of coloration ofliquids ....................................... 22 2.2.34. Thermal analysis ............................................................ 55
2.2.3. Potentiometric determination of pH ............................. 24 2.2.35. Osmolality....................................................................... 57
2.2.4. Relationship between reaction of solution, 2.2.36. Potentiometric determination of ionic
approximate pH and colour of certain indicators ................ 25 concentration using ion-selective electrodes ........................ 58
2.2.5. Relative density................................................................. 25 2.2.37. X-ray fluorescence spectrometry.................................. 59
2.2.6. Refractive index ................................................................ 26 2.2.38. Conductivity................................................................... 59
2.2.7. Optical rotation ................................................................ 26 2.2.39. Molecular mass distribution in dextrans .................... 60
2.2.8. Viscosity............................................................................. 27 2.2.40. Near-infrared spectroscopy........................................... 62
2.2.9. Capillary viscometer method ......................................... 27 2.2.41. Circular dichroism ......................................................... 67
2.2.10. Viscosity - Rotating viscometer method ..................... 28 2.2.42. Density of solids ............................................................. 68
2.2.11. Distillation range ............................................................ 30 2.2.43. Mass spectrometry......................................................... 69
2.2.12. Boiling point.. ................................................................. 31 2.2.44. Total organic carbon in water for pharmaceutical
2.2.13. Determination of water by distillation ........................ 31 use ............................................................................................... 71
2.2.14. Melting point - capillary method ................................. 32 2.2.45. Supercritical fluid chromatography............................. 72
2.2.15. Melting point - open capillary method ....................... 32 2.2.46. Chromatographic separation techniques .................... 72
2.2.16. Melting point - instantaneous method ........................ 32 2.2.47. Capillary electrophoresis ............................................... 79
2.2.17. Drop point.. .................................................................... 32 2.2.48. Raman spectrometry...................................................... 84
2.2.18. Freezing point.. ............................................................... 34 2.2.49. Falling ball viscometer method .................................... 85
2.2.19. Amperometric titration ................................................. 34 2.2.54. Isoelectric focusing ........................................................ 85
2.2.20. Potentiometric titration ................................................ 34 2.2.55. Peptide mapping ............................................................ 87
2.2.21. Fluorimetry..................................................................... 35 2.2.56. Amino acid analysis ....................................................... 90
2.2.22. Atomic emission spectrometry..................................... 35 2.2.57. Inductively coupled plasma-atomic emission
2.2.23. Atomic absorption spectrometry................................. 36 spectrometry.............................................................................. 97
2.2.24. Absorption spectrophotometry, infrared .................... 38 2.2.58. Inductively coupled plasma-mass spectrometry........ 98
2.2.25. Absorption spectrophotometry, ultraviolet and 2.2.59. Glycan analysis of glycoproteins ................................ 100
visible ......................................................................................... 40 2.2.60. Melting point - instrumental method ....................... 105
2.2.26. Paper chromatography.................................................. 42 2.2.61. Characterisation of crystalline solids by
2.2.27. Thin-Iayer chromatography.......................................... 42 microcalorimetry and solution calorimetry........................ 106
2.2.28. Gas chromatography...................................................... 43 2.2.64. Peptide identification by nuclear magnetic resonance
2.2.29. Liquid chromatography................................................. 45 spectrometry. ........................................................................... 109
2.2.30. Size-exclusion chromatography.................................... 46 2.2.65. Voltametric titratiol1 .................................................... 109
2.2.31. Electrophoresis ............................................................... 47 2.2.66. Detectiol1 and measurement ofradioactivity........... 110

General Notices (1) apply to all monographs and other texts 19


EUROPEAN PHARMACOPOEIA 8.0

20 See the information section on general monographs (cover pages)


EUROPEAN PHARMACOPOEIA 8.0 2.2.1. Clarity and degree of opalescence of liquids

2.2. PHYSICAL AND characteristics and traceability, instrument calibration


algorithms and performance criteria are mostly based on this
PHYSICOCHEMICAL standard.
METHODS INSTRUMENTAL METHODS
INTRODUCTION
01/2008:20201 The degree of opalescence may also be determined by
instrumental measurement of the light absorbed or scattered
2.2.1. CLARITY AND DEGREE OF on account of submicroscopic optical density inhomogeneities
OPALESCENCE OF LIQUIDS of opalescent solutions and suspensions. 2 such techniques are
nephelometry and turbidimetry. For turbidity measurement
VISUAL METHOD of coloured samples, ratio turbidimetry and nephelometry
Using identical test-tubes of colourless, transparent, neutral with ratio selection are used.
glass with a flat base and an internal diameter of 15-25 mm, The light scattering effect of suspended particles can be
compare the liquid to be examined with a reference suspension measured by observatiol1 of either the transmitted light
freshly prepared as described below, the depth of the layer (turbidimetry) or the scattered light (nephelometry). Ratio
being 40 mm. Compare the solutions in diffused daylight turbidimetry combines the principIes of both nephelometry
5 min after preparation of the reference suspension, viewing and turbidimetry. Turbidimetry and nephelometry are useful
vertically against a black background. The diffusion of light for the measurement of slightly opalescent suspensions.
must be such that reference suspension I can readily be Reference suspensions produced under well-defined
distinguished from water R, and that reference suspension II conditions must be used. For quantitative measurements,
can readily be distinguished from reference suspension I. the construction of calibration curves is essential, since the
A liquid is considered clear if its clarity is the same as that relationship between the opticaI properties of the suspension
of water R or of the solvent used when examined under the and the concentration of the dispersed phase is at best
conditions described aboye, or if its opalescence is not more semi -empirica!.
pronounced than that of reference suspension I. The determination of opalescence of coloured liquids is done
Hydrazine sulfate soluticm. Dissolve 1.0 g of hydrazine with ratio turbidimeters or nephelometers with ratio selection,
sulfate R in water R and dilute to 100.0 mL with the same since colour provides a negative interference, attenuating both
solvent. Allow to stand for 4-6 h. incident and scattered light and lowering the turbidity value.
Hexamethylenetetramine solution. In a 100 mL The effect is so great for even moderately coIoured samples
ground-glass-stoppered flask, dissolve 2.5 g of that conventional nephelometers cannot be used.
hexamethylenetetramine R in 25.0 mL of water R. The instrumental assessment of clarity and opalescence
provides a more discriminatory test that does not depend on
Primary opalescent suspension (formazin suspension). To
the visual acuity of the anaIyst. Numerical results are more
the hexamethylenetetramine solution in the flask add 25.0 mL
useful for quality monitoring and process control, especially
of the hydrazine sulfate solution. Mix and allow to stand for
in stability studies. For example, previous numerical data 011
24 h. This suspension is stable for 2 months, provided it is
stability can be projected to determine whether a given batch
stored in a glass container free from surface defects. The
of dosage formuIation or active pharmaceutical ingredient will
suspension must not adhere to the glass and must be well
exceed shelf-life limits prior to the expiry date.
mixed before use.
NEPHELOMETRY
Standard of opalescence. Dilute 15.0 mL of the primary
opalescent suspension to 1000.0 mL with water R. This When a suspension is viewed at right angles to the direction
suspension is freshly prepared and may be stored for up to of the incident light, the system appears opalescent due to
24 h. the reflection of light from the particles of the suspension
(Tyndall effect). A certain portion of the light beam entering
Reference suspensions. Prepare the reference suspensions a turbid liquid is transmitted, another portion is absorbed
according to Table 2.2.1.-1. Mix and shake before use. and the remaining portion is scattered by the suspended
Table 2.2.1.-1 particles. If measurement is made at 90° to the light beam,
n m IV the light scattered by the suspended particles can be used
for the determination of their concentration, provided
Standard of opalescence 5.0 rnL 10.0 rnL 30.0 rnL 50.0 rnL the number and size of particles inÍ!uencing the scattering
Water R 95.0 rnL 90.0 rnL 70.0 111L 50.0 rnL
remain constant. The reference suspension must maintain
a constant degree of turbidity and the sample and reference
Turbidity standard. The formazin suspension prepared suspensions must be prepared under identical conditions. The
by mixing equal volumes of the hydrazine sulfate solution Tyndall effect depends upon both the l1umber of particles and
and the hexamethylenetetramine solution is defined as a their size. Nephelometric measurements are more reliable
4000 NTU (nephelometric turbidity units) primary reference in Iow turbidity ranges, where there is a linear relationship
standard. Reference suspensions 1, II, III and IV have values of between nephelometric turbidity unit (NTU) values and
3 NTU, 6 NTU, 18 NTU and 30 NTU respectively. Stabilised relative detector signals. As the degree of turbidity increases,
formazin suspensions that can be used to prepare stable, 110t aH the particles are exposed to the incident light and
diluted turbidity standards are available commercially and the scattered radiation of other partides is hindered on its
may be used after comparison with the standard s prepared way to the detector. The maximum nephelometric values at
as described. which reliable measurements can be made lie in the range
of 1750-2000 NTU. Linearity must be demonstrated by
Formazin has several desirable characteristics that make it an
constructing a calibration curve using at least 4 concentrations.
excellent turbidity standard. It can be reproducibly prepared
from assayed raw materials. The physical characteristics make TURBIDIMETRY
it a desirable light-scatter calibration standard. The formazin The opticaI property expressed as turbidity is the interaction
polymer consists of chains of different lengths, which fold between light and suspended particles in liquido This is an
into random configurations. This results in a wide assay of expression of the optical property that causes light to be
particle shapes and sizes, which analytically fits the possibility scattered and absorbed rather than transmitted in a straight
of different particle sizes and shapes that are found in the line through the sample. The quantity of solid material in
real samples. Due to formazin's reproducibility, scattering suspension can be determined by the measurement of the

General Notices (1) apply to all monographs and other texts 21


2.2.2. Degree oí coloration of Hquids EUROPEAN PHARMACOPOEIA 8.0

transmitted light. A linear relationship between turbidity and are also used, and are equivalent to NTU in low regions
concentration is obtained when the particle sizes are uniform (up to 40 NTU). These units are used in a1l3 instrumental
and homogeneous in the suspension. This is true only in methods (nephelometry, turbidimetry and ratio
very dilute suspensions containing small particles. Linearity turbidimetry) .
between turbidity and concentration must be established by - Measuring range: 0.01-1100 NTU.
constructing a calibration curve using at least 4 concentrations. - Resolution: 0.01 NTU within the range of 0-10 NTU,
RATIO TURBIDIMETRY 0.1 NTU within the range of 10-100 NTU, and 1 NTU for
In ratio turbidimetry the relationship of the transmission the range > 100 NTU. The instrument is calibrated and
measurement to the 90° scattered light measurement is controlled with reference standards of formazin.
determined. This procedure compensates for the light that - Accuracy: 0-10 NTU: ± (2 per cent ofreading + 0.01) NTU.
is diminished by the colour of the sample. The influence 10-1000 NTU: ± 5 per cent.
of the colour of the sample may also be eliminated by - Repeatability: 0-10 NTU: ± 0.01 NTU. 10-1000 NTU: ± 2 per
using an infrared light-emitting diode (IR LED) at 860 nm cent of the measured value.
as the light source of the instrument. The instrument's
- Calibration: with 4 reference suspensions of formazin in
photodiode detectors receive and measure scattered light at a
the range of interest. Reference suspensions described
90° angle from the sample as well as measuring the forward
in this chapter or suitable reference standards calibrated
scatter (light reflected) in front of the sample along with
against the primary reference suspensions may be used.
the measurement of light transmitted directly through the
sample. The measuring results are given in NTU(ratio) and - Stray light: this is a significant source of error in low level
are obtained by calculating the ratio of the 90° angle scattered turbidimetric measurement; stray light reaches the detector
light measured to the sum of the components of forward of an optical system, but do es not come from the sample;
scattered and transmitted light values. In ratio turbidimetry < 0.15 NTU for the range 0-10 NTU, < 0.5 NTU for the
the influence of stray light becomes negligible. Nephelometers range 10-1000 NTU.
are used for measurements of the degree of opalescence of Instruments complying with the aboye characteristics and
colourless liquids. verified using the reference suspensions described under
Measurements of reference suspensions 1-IV with a ratio Visual method may be used instead of visual examination for
turbidimeter show a linear relationship between the determination of compliance with monograph requirements.
concentrations and measured NTU values (see Table 2.2.1.-2). Instruments with range or resolution, accuracy and
Reference suspensions I-IV (Ph. Eur.) may be used as repeatability capabilities other than those mentioned aboye
calibrators for the instrument. may be used provided they are sufficiently validated and
are capable for the intended use. The test methodology for
Table 2.2.1.-2
the specific substance/product to be analysed must also
Formazin suspensious Opalescent values (NTU) be validated to demonstrate its analytical capability. The
Reference suspension I 3 instrument and methodology should be consistent with the
att1'ibutes of the product to be tested.
Reference suspension II 6

Reference suspension TII 18

Reference suspension IV 30
01/2008:20202
Standard of opalescence 60

Primary opalescent suspension 4000 2.2.2. DEGREE OF COLORATION OF


LIQUIDS
INSTRUMENTAL DETERMINATION OF OPALESCENCE
The examination of the degree of coloration of liquids in the
Requirements in monographs are expressed in terms of
range brown-yellow-red is carried out by one of the 2 methods
the visual examination method with the defined reference
below, as prescribed in the monograph.
suspensions. Instrumental methods may also be used for
determining compliance with monograph requirements once A solution is colourless if it has the appearance of water R or
the suitability of the instrument as described below has been the solvent 01' is 110t more intensely coloured than refe1'ence
established and calibration with reference suspensions 1-IV solution B9'
and with water R or the solvent used has been performed. METHOD I
Apparatus. Ratio turbidimeters or nephelometers with Using identical tubes of colourless, transparent, neutral glass
selectable ratio application use as light source a tungsten of 12 mm external diameter, compare 2.0 mL of the liquid
lamp with spectral sensitivity at about 550 nm operating at to be examined with 2.0 mL of water R or of the solvent or
a filament colour temperature of 2700 1(, or IR LED having of the reference solution (see Tables of reference solutions)
an emission maximum at 860 nm with a 60 nm spectral prescribed in the monograph. Compare the colours in diffused
bandwidth. Other suitable light sources may also be used. daylight, viewing horizontally against a white background.
Silicon photodiodes and photomultipliers are commonly
used as detectors and record changes in light scattered or METHOD II
transmitted by the sample. The light scattered at 90 ± 2.5 0 is Using identical tubes of colourless, transparent, neutral glass
detected by the primary detector. Other detector s are those to with a flat base and an internal diameter of 15 mm to 25 mm,
detect back and forward scatter as well as transmitted light. compare the liquid to be examined with water R or the solvent
The instruments used are calibrated against standards of or the reference solution (see Tables of reference solutions)
known turbidity and are capable of automatic determination prescribed in the monograph, the depth of the ¡ayer being
of turbidity. The test results expressed in NTU units are 40 mm. Compare the colours in diffused daylight, viewing
obtained directly from the instrument and compared to the vertically against a white background.
specifications in the individual monographs.
Instruments complying with the following specifications are REAGENTS
suitable. Primary solutions
- Measuring units: NTU. NTU is based on the turbidity of a Yellow solution. Dissolve 46 g of ferric chloride R in about
primary reference standard of formazin. FTU (Formazin 900 mL of a mixture of 25 mL of hydrochloric acid R and
Turbidity Units) or FNU (Formazin Nephelometry Units) 975 mL of water R and dilute to 1000.0 mL with the same

22 See the information section on general monographs (cover pages)


EUROPEAN PHARMACOPOEIA 8.0 2.2.2. Degree of coloration of Hquids

mixture. Titrate and adjust the solution to contain 45.0 mg of Table 2.2.2.-2. - Reference solutions B
FeC13 ,6HP per millilitre by adding the same acidic mixture.
Protect the solution from light. Volumes in millilitres

Titration. Place in a 250 mL conical flask fitted with a Reference Standard solution B Hydrochloric acid
solntion (lO giL He!)
ground-glass stopper, 10.0 mL ofthe solution, 15 mL of
B¡ 75.0 25.0
water R, 5 mL of hydrochloric acid R and 4 g of potassium
iodide R, close the flask, allow to stand in the dark for 15 min B2 50.0 50.0
and add 100 mL of water R. Titrate the liberated iodine with
B3 37.5 62.5
0.1 M sodium thiosulfate, using 0.5 mL of starch solution R,
added towards the end of the titration, as indicator. B, 25.0 75.0

1 mL of 0.1 M sodium thiosulfate is equivalent to 27.03 mg Bs 12.5 87.5


of FeCI3'6Hp. B6 5.0 95.0

Red solution. Dissolve 60 g of cobalt chloride R in about B7 2.5 97.5


900 mL of a mixture of 25 mL of hydrochloric acid R and B8 1.5 98.5
975 mL of water R and dilute to 1000.0 mL with the same
mixture. Titrate and adjust the solution to contain 59.5 mg of B, 1.0 99.0
CoC12 ,6HP per millilitre by adding the same acidic mixture.

Titration. Place in a 250 mL conical flask fitted with a Table 2.2.2.-3. - Reference solutions BY
ground-glass stopper, 5.0 mL of the solution, 5 mL of dilute
hydrogen peroxide solution R and 10 mL of a 300 giL solution
Volumes in millilitres
of sodium hydroxide R. Boil gently for 10 min, allow to cool
and add 60 mL of dilute sulfuric acid R and 2 g of potassium Reference Standard solution BY Hydrochloric acid
iodide R. Close the flask and dissolve the precipitate by solution (10 giL Hel)
shaking gently. Titrate the liberated iodine with 0.1 M sodium BY¡ 100.0 0.0
thiosulfate, using 0.5 mL of starch solution R, added towards BY, 75.0 25.0
the end of the titration, as indicator. The end -point is reached
when the solution turns pink. BY 3 50.0 50.0

BY, 25.0 75.0


1 mL of 0.1 M sodium thiosulfate is equivalent to 23.79 mg of
CoC12 ,6Hp. BYs 12.5 87.5

BY6 5.0 95.0


Blue primary solution. Dissolve 63 g of eopper sulfate R in
about 900 mL of a mixture of 25 mL of hydrochloric aeid R BY7 2.5 97.5
and 975 mL of water R and dilute to 1000.0 mL with the same
mixture. Titrate and adjust the solution to contain 62.4 mg of
CuS0 4 ,5HP per millilitre by adding the same acidic mixture.
Table 2.2.2.-4. - Referenee solutions Y
Titration. Place in a 250 mL conical flask fitted with a
ground-glass stopper, 10.0 mL ofthe solution, 50 mL of Volumes in millilitres
water R, 12 mL of dilute aeetie acid R and 3 g of potassium Reference Standard solution Y Hydrochloric acid
iodide R. Titrate the liberated iodine with 0.1 M sodium 80lution (10 giL HCl)
thiosulfate, using 0.5 mL of starch solution R, added towards Y¡ 100.0 0.0
the end of the titration, as indicator. The end -point is reached
when the solution shows a slight pale brown colour. Y, 75.0 25.0

Y3 50.0 50.0
1 mL of 0.1 M sodium thiosulfate is equivalent to 24.97 mg
of CuS0 4,5Hp. Y, 25.0 75.0

Standard solutions Ys 12.5 87.5

Y6 5.0 95.0
Using the 3 primary solutions, prepare the 5 standard solutions
as follows (Table 2.2.2.-1): Y7 2.5 97.5

Table 2.2.2.-1
Table 2.2.2.-5. - Referenee solutions GY
Volume in millilitres

Standard solution Yellow Red Blue Hydrochloric acid Volumes in millilitres


solution solution solution (lO giL Hel)
Reference Standard solution GY Hydrochloric acid
B (brown) 3.0 3.0 2.4 1.6 solution (10 giL Hel)
BY (brownish-yellow) 2.4 1.0 0.4 6.2 GY¡ 25.0 75.0

Y (yellow) 2.4 0.6 0.0 7.0 GY 2 15.0 85.0

GY (greenish-yellow) 9.6 0.2 0.2 0.0 GY, 8.5 91.5

R (red) 1.0 2.0 0.0 7.0 GY, 5.0 95.0

GY 5 3.0 97.0
Reference solutions for Methods 1 and n
GY6 1.5 98.5
Using the 5 standard solutions, prepare the following reference GY7 0.75 99.25
solutions.

General Notices (1) apply to all monographs and other texts 23


2.2.3. Poíentiometric determination of pH EUROPEAN PHARMACOPOEIA 8.0

Table 2.2.2.-6. - Reference solutions R Method. Unless otherwise prescribed in the monograph, aH
measurements are made at the same temperature (20-25 OC).
Volumes in millilitres
Table 2.2.3.-2 shows the variation of pH with respect to
Reference Standard solution R Hydrochloric "cid temperature of a number of reference buffer solutions used for
solution (lO gIL Hel) calibration. For the temperature correction, when necessary,
R¡ 100.0 0.0 follow the manufacturer's instructions. The apparatus is
calibrated with the buffer solution of potassium hydrogen
R, 75.0 25.0
phthalate (primary standard) and 1 other buffer solution of
R, 50.0 50.0 different pH (preferably one shown in Table 2.2.3.-2). The
pH of a third buffer solution of intermediate pH read off on
R, 37.5 62.5
the scale must not differ by more than 0.05 pH unit from the
Rs 25.0 75.0 value corresponding to this solution. Immerse the electro des
in the solution to be examined and take the reading in the
R6 12.5 87.5
same conditions as for the buffer solutions.
R7 5.0 95.0
When the apparatus is in frequent use, checks must be carried
Storage out regularly. lf not, such checks should be carried out before
each measurement.
For Method 1, the reference solutions may be sto red in sealed
tubes of colourless, transparent, neutral glass of 12 mm Al! solutions to be examined and the reference buffer solutions
external diameter, protected from light. must be prepared using carbon dioxide-free water R.
For Method II, prepare the reference solutions immediately
before use from the standard solutions.
PREPARATION OF REFERENCE BUFFER SOLUTIONS
Potassium tetraoxalate 0.05 M. Dissolve 12.61 g of
C4H3KOs,2H20 in carbon dioxide-free water R and dilute to
1000.0 mL with the same solvent.
0112008:20203 Potassium hydrogen tartrate, saturated at 25 oc. Shake an
excess of C4HsK06 vigorously with carbon dioxide-free water R
at 25 oc. Filter or decanto Prepare immediately before use.
2.23. POTENTIOMETRIC Potassium dihydrogen citrate 0.05 M. Dissolve 11.41 g
DETERMINATION OF pH of C6H7K07 in carbon dioxide-free water R and dilute to
1000.0 mL with the same solvento Prepare immediately before
The pH is a number which represents conventionally the use.
hydrogen ion concentration of an aqueous solution. For Potassium hydrogen phthalate 0.05 M. Dissolve 10.13 g of
practical purposes, its definition is an experimental one. The CsH sK0 4, previously dried for 1 h at 110 ± 2 oC, in carbon
pH of a solution to be examil1ed is related to that of a reference dioxide-free water R and dilute to 1000.0 mL with the same
solution (pHJ by the following equation: solvent.
E-Es Potassium dihydrogen phosphate 0.025 M + disodium
pH = pH s - --k-
hydrogen phosphate 0.025 M. Dissolve 3.39 g ofKH 2P0 4 and
3.53 g ofNa 2HP0 4, both previously dried for 2 h at 120 ± 2 oC,
in which E is the potential, expressed in volts, of the cel! in carbon dioxide-free water R and dilute to 1000.0 mL with
containing the solution to be examined and Es is the potential, the same solvent.
expressed in volts, of the ceH containing the solution of known Potassium dihydrogen phosphate 0.0087 M + disodium
pH (pHJ, k is the change in potel1tial per unit change in pH hydrogen phosphate 0.0303 M. Dissolve 1.18 g of KHzP04
expressed in volts, and calculated from the Nernst equation. and 4.30 g of Na 2 HP0 4 , both previously dried for 2 h at
120 ± 2 oC, in carbon dioxide-free water R and dilute to
Table 2.2.3.-1. - Values of k at different temperatures
1000.0 mL with the same solvento
Temperature (OC) k (V) Disodium tetraborate 0.01 M. Dissolve 3.80 g of
15 0.0572 NazB407,10HzO in carbon dioxide-free water R and dilute
to 1000.0 mL with the same solvent. Store protected from
20 0.0582 atmospheric carbon dioxide.
25 0.0592
Sodium carbonate 0.025 M + sodium hydrogen carbonate
30 0.0601 0.025 M. Dissolve 2.64 g of NaZC0 3 and 2.09 g of NaHC0 3
in carbon dioxide-free water R and dilute to 1000.0 mL with
35 0.0611 the same solvent. Store protected from atmospheric carbon
dioxide.
The potentiometric determination of pH is made by measuring
the potential difference between 2 appropriate electro des Caldum hydroxide, saturated at 25 oc. Shake an excess of
immersed in the solution to be examined: 1 of these electro des calcium hydroxide R with carbon dioxide-free water R and
is sensitive to hydrogen ions (usually a glass electro de ) and decant at 25 oc. 5tore protected from atmospheric carbon
the other is the reference electro de (for example, a saturated dioxide.
calomel electrode).
Apparatus. The measuring apparatus is a voltmeter with an
STORAGE
input resistance at least 100 times that of the electro des used.
It is normally graduated in pH units and has a sensitivity such 5tore buffer solutions in suitable chemically resistant, tight
that discrimination of at least 0.05 pH unit or at least 0.003 V containers, such as type 1 glass bottles or plastic containers
may be achieved. suitable for aqueous solutions.

24 See the information sectian on general monagraphs (cover pages)


EUROPEAN PHARMACOPOEIA 8.0 2.2.5. Relative density

Table 2.2.3.-2. - pH of reference buffer solutions at various temperatures


Temperature Potassium Potassium Potassium Potassium Potassium Potassium Disodium Sodium Calcium
(OC) tetraoxalate hydrogeu dihydrogen hydrogen dihydrogen dihydrogen tetraborate carbonate hydroxide,
0.05 M tartrate dtrate phthalate phosphate phosphate 0.01 M 0.025 M saturated
saturated at 0.05 M 0.05 M 0.025 M 0.0087 M + al 25°C
25 oC
+ + sodium
disodium disodium bicarbonate
hydrogen hydrogen 0.025 M
phosphate phosphate
0.025 M 0.0303 M

C4 H,KOs,2H 2 O C 4 H,K0 6 C 6 H 7 K0 7 C,H,K0 4 KH 2 P0 4 + KH 2 P0 4 + Na2 B.10 7, Na2 C0 3 + Ca(OH)2


Na2HP0 4 Na2 HPO, lOH 2 O NaHC0 3

15 1.67 3.80 4.00 6.90 7.45 9.28 10.12 12.81

20 1.68 3.79 4.00 6.88 7.43 9.23 10.06 12.63

25 1.68 3.56 3.78 4.01 6.87 7.41 9.18 10.01 12.45

30 1.68 3.55 3.77 4.02 6.85 7.40 9.14 9.97 12.29

35 1.69 3.55 3.76 4.02 6.84 7.39 9.10 9.93 12.13

b.pH(l)
+ 0.001 - 0.0014 - 0.0022 + 0.0012 - 0.0028 - 0.0028 - 0.0082 - 0.0096 - 0.034
b.t

(1) pH variation per degree Celsius.

0112008:20204 0112008:20205

2,2.5. RELATIVE DENSITY


2.2.4. RELATIONSHIP BETWEEN
REACTION OF SOLUTION, The relative density d;; of a substance is the ratio of the mass
of a certain volume ora substance at temperature t¡ to the
APPROXIMATE pH AND COLOUR OF mass of an equal volume of water at temperature t 2 •
CERTAIN INDICATORS Unless otherwise indicated, the relative density d~g is used.
Relative density is also commonly expressed as d~o. Density P2O'
To 10 mL ofthe solution to be examined, add 0.1 mL deflned as the mas s of a unit volume of the substance at 20 oC
of the indicator solution, unless otherwise prescribed in may also be used, expressed in kilograms per cubie metre or
Table 2.2.4.-1. grams per cubie centimetre (1 kg.m- 3 = 10- 3 g.cm- 3 ). These
quantities are related by the following equations where density
Table 2.2.4.-1 is expressed in grams per cubie centimetre:
Reaction pH Indicator Colour P20 = 0.998203 x d~g or d~g = 1.00180 >( P20

Alkaline >8 Red litmus paper R Blue


P20 = 0.999972 X d 240 or d~o = 1.00003 X P20
Thymol blue Grey or violet-blue
solution R (0.05 mL)
Slightly alkaline 8.0 - 10.0 Phenolphthalein Colourless or pink
solution R (0.05 mL)
Thymol blue Grey Relative density or density are measured with the precision
solution R (0.05 mL) to the number of decimals prescribed in the monograph
Stronglyalkaline >10 Phenolphthalein Red using a density bottle (solids or liquids), a hydrostatie balance
paper R (solids), a hydrometer (liquids) or a digital density meter
Thymol blue Violet-blue with an oscillating transducer (liquids and gases). When the
solution R (0.05 mL) determination is made by weighing, the buoyancy of air is
Neutral 6.0 - 8.0 Methyl red solution R Yellow disregarded, whieh may introduce an error of 1 unit in the
Phenol red solution R 3,d decimal place. When using a density meter, the buoyancy
(0.05 mL) of air has no influence.
Neutral to methyl 4.5 - 6.0 Methyl red solution R Orange-red Oscillating transducer density meter. The apparatus consists of:
red
- a U -shaped tube, usually of borosilieate glass, whieh
Neutral to < 8.0 Phenolphthalein Colourless; pink or red
phenolphtalein solution R (0.05 rnL) after adding 0.05 mL contains the liquid to be examined;
of 0.1 M base - a magneto-electrieal or piezo-electrieal excitation system
Acid <6 Methyl red solution R Orange or red that causes the tube to oscillate as a cantilever oscillator
Bromothymol blue Yellow
at a characteristie frequency depending on the density of
solution Rl the liquid to be examined;
Slightly acid 4.0 - 6.0 Methyl red solution R Orange - a means of measuring the oscillation period (T), whieh may
be converted by the apparatus to give a direct reading of
Bromocresol green Green or blue
solution R density, or used to calculate density using the constants A
Strongly acid <4 Congo red paper R Green or blue
and B described below.

General Natices (1) apply ta all monagraphs and other texts 25


2.2.6. Refractive index EUROPEAN PHARMACOPOEIA 8.0

The resonant frequency (j) is a function of the spring When white light is used, the refractometer is provided with a
constant (e) and the mass (m) of the system: compensating system. The apparatus gives readings accurate
to at least the third decimal place and is provided with a means
l = ;2 = ~ x 4:2 of operation at the temperature prescribed. The thermometer
is graduated at intervals of 0.5 oC or less.
Hence:

0112008:20207

M mass of the tube, 2.2.7. OPTICAL ROTATION


V inner volume of the tube. Optical rotation is the property displayed by chiral substances
? of rotating the plan e of polarisation of polarised light.
Introduction of 2 constants A = el (47r- x V) and B = M IV,
leads to the elassical equation for the oscillating transducer: Optical rotation is considered to be positive (+) for
dextrorotatory substances (Le. those that rotate the plane of
p = A X T2 - B polarisation in a elockwise direction) and negative (-) for
laevorotatory substances.
The constants A and B are determined by operating the The specific optical rotation [Ctrn 1~ is the rotation, expressed
instrument with the U-tube filled with 2 different samples in radians (rad), measured at the temperature t and at the
of known density, for example, degassed water R and airo wavelength A given by a 1 m thickness of liquid or a solution
Control measurements are made daily using degassed water R. containing 1 kg/m 3 of optically active substance. Por practical
The results displayed for the control measurement using
reasons the specific optical rotation [Ctml~ is normally
degassed water R shall not deviate from the reference value expressed in milliradians metre squared per kilogram
(P20 = 0.998203 g·cm- 3, d~g = 1.000000) by more than its (mrad.m 2 ·kg- 1).
specified error. Por example, an instrument specified to
± 0.0001 g·cm- 3 shall display 0.9982 ± 0.0001 g·cm- 3 in The Pharmacopoeia adopts the following conventional
order to be suitable for further measurement. Otherwise a definitions.
re-adjustment is necessary. Calibration with certified reference The angle of optical rotation of a neat liquid is the angle of
materials is carried out regularly. Measurements are made rotation a, expressed in degrees (O), of the plane of polarisation
using the same procedure as for calibration. The liquid to at the wavelength of the D-line of sodium (A = 589.3 nm)
be examined is equilibrated in a thermostat at 20 oC before measured at 20 oC using a layer of 1 dm; for a solution, the
introduction into the tube, if necessary, to avoid the formation method of preparation is prescribed in the monograph.
of bubbles and to reduce the time required for measurement. The speeific optieal rotation [Ctl~O of a liquid is the angle of
Pactors affecting accuracy inelude: rotation a, expressed in degrees (O), of the plane of polarisation
- temperature uniformity throughout the tube, at the wavelength of the D-line of sodium (A = 589.3 nm)
measured at 20 oC in the liquid substance to be examined,
- non-linearity over a range of density,
calculated with reference to a layer of 1 dm and divided by the
- parasitic resonant effects, density expressed in grams per cubic centimetre.
- viscosity, whereby solutions with a higher viscosity than The specific optical rotation [Ctl~O of a substance in solution
the calibrant have a density that is apparently higher than is the angle of rotation a, expressed in degrees (O), of the
the true value. plane of polarisation at the wavelength of the D-line of
The effects of non-linearity and viscosity may be avoided by sodium (A = 589.3 11m) measured at 20 oC in a solution of the
using calibrants that have density and viscosity close to those substance to be examined and calculated with reference to a
of the liquid to be examined (± 5 per cent for density, ± 50 per layer of 1 dm containing 1 g/mL of the substance. The specific
cent for viscosity). The density meter may have functions for optical rotation of a substance in solution is always expressed
automatic viscosity correction and for correction of error s with reference to a given solvent and concentration.
arising from temperature changes and non-linearity. In the conventional system adopted by the Pharmacopoeia the
Precision is a function of the repeatability and stability of the specific optical rotation is expressed by its value without units;
oscillator frequency, which is dependent on the stability of the the actual units, degree millilitres per decimetre gram
volume, mass and spring constant of the cell. [(0)·ml.dm-1.g- 1] are understood.
Density meters are able to achieve measurements with an The conversion factor from the International System to the
error of the order of 1 x 10- J g·cm- 3 to 1 x 10- 5 g·cm- 3 and a Pharmacopoeia system is the following:
repeatability of 1 x 10- 4 g·cm- J to 1 x 10- 6 g.cm- J.
[Ctrnl~ = [Ctl~ X 0.1745

In certain cases specified in the monograph the angle of


0112008:20206 rotation may be measured at temperatures other than 20 oC
and at other wavelengths.
2.2.6. REFRACTIVE INDEX The polarimeter must be capable of giving readings to the
nearest 0.01 0. The scale is usually checked by means of
The refractive index of a medium with reference to air is equal
certified quartz plates. The linearity of the scale may be
to the ratio of the sine of the angle of incidence of a beam of
checked by means of sucrose solutions.
light in air to the sine of the angle of refraction of the refracted
beam in the given medium. Method. Determine the zero of the polarimeter and the
angle of rotation of polarised light at the wavelength of
Unless otherwise prescribed, the refractive index is measured
the D-line of sodium (A = 589.3 nm) at 20 ± 0.5 oC, unless
at 20 ± 0.5 oC, with reference to the wavelength of the D-line
otherwise prescribed. Measurements may be carried out at
of sodium (A = 589.3 nm); the symbol is then n5°.
other temperatures only where the monograph indicates the
Refractometers normally determine the critical angle. In such temperature correction to be made to the measured optical
apparatus the essential part is a prism of known refractive rotation. Determine the zero of the apparatus with the tube
index in contact with the liquid to be examined. elosed; for liquids the zero is determined with the tube empty
Calibrate the apparatus using certified reference materials. and for solids filled with the prescribed solvent.

26 See the information sectiol1 011 general monographs (ca ver pages)
EUROPEAN PHARMACOPOEIA 8.0 2.2.9. Capillary viscometer method

Calculate the specific optical rotation using the following 01/2008:20209


formulae.

For neat liquids :


2.2,9. CAPILLARY VISCOMETER
METHOD

For substances in solution: The determination of viscosity using a suitable capillary


viscometer is carried out at a temperature of 20 ± 0.1 oC,
[a]20 = 1000a unless otherwise prescribed. The time required for the level
D l.e of the liquid to drop from one mark to the other is measured
with a stop-watch to the nearest one-fifth of a second. The
where e is the concentration of the solution in grams per litre. result is valid only if two consecutive readings do not differ
by more than 1 per cent. The average of not fewer than three
readings gives the flow time of the ¡iquid to be examined.
Calculate the content e in grams per litre or the content e'
in per cent rn/rn of a dissolved substance using the following
formulae: Calculate the dynamic viscosity rr (2.2.8) in millipascal
seconds using the formula:
1000a f 100a
e~---
e = --:on-- TI = kpt
~ l· [a]~o l· [a]~o 'P20

k constant of the viscometer, expressed in square


a angle of rotation in degrees read at 20 ± O.5°C; millimetres per second squared,
length in decimetres of the polarimeter tube; p density of the liquid to be examined expressed
density at 20 oC in grams per cubic centimetre. in milligrams per cubic millimetre, obtained by
For the purposes of the Pharmacopoeia, density is multiplying its relative density (d§g) by 0.9982,
replaced by relative density (2.2.5). flow time, in seconds, of the liquid to be examined.

The constant le is determined using a suitable viscometer


calibration liquido

To calculate the kinematic viscosity (mm 2·s- 1), use the


following formula: v = kt.

The determination may be carried out with an apparatus


01/2008:20208
(Figure 2.2.9.-1) having the specifications described in
Table 2.2.9.-1 (1):

2,2.8. VISCOSITY The minimum flow time should be 350 s for size no. 1 and
200 s for all other sizes.

The dynamic viscosity or viscosity coefficient /1 is the tangential


force per unit surface, known as shearing stress r and expressed Table 2.2.9.-1
in pascals, necessary to move, parallel to the sliding plane, a
Size Nominal Kinematic Internal Volume Internal
layer of liquid of 1 square metre at arate (v) of 1 metre per
second relative to a parallellayer at a distance (x) of 1 metre. number constant viscosity diameter of diameter
ofvisco- mnge oftube bulb oftube
The ratio dv/dx is a speed gradient giving the rate of shear D meter R e N
expressed in reciprocal seconds (5- 1), so that rr = r/D. 111ll12.S- 2 rnm 2·s-- 1 IllITI mL 111m

(± 2 %) (± 5 %)
The Ul1it of dynamic viscosity is the pascal second (Pa·s).
The most commonly used submultiple is the millipascal 0.01 3.5 to lO 0.64 5.6 2.8 to 3.2
second (mPa·s). lA 0.03 6 to 30 0.84 5.6 2.8 to 3.2

2 0.1 20 lo lOO 1.15 5.6 2.8 to 3.2


The kinernatic viscosity v, expressed in square metres per
second, is obtained by dividing the dynamic viscosity rr by the 2A 0.3 60 lo 300 1.51 5.6 2.8 to 3.2
density p expressed in kilograms per cubic metre, of the liquid
3 1.0 200 to 1000 2.06 5.6 3.7to 4.3
measured at the same temperature, i.e. v = rr/p. The kinematic
viscosity is usually expressed in square millimetres per second. 3A 3.0 600 to 3000 2.74 5.6 4.6 to 5.4

4 10 2000 to 3.70 5.6 4.6 to 5.4


A capillary viscometer may be used for determining the
lO 000
viscosity of Newtonian liquids and a rotating viscometer for
4A 30 6000 to 4.07 5.6 5.6 to 6.4
determining the viscosity of Newtonian and non-Newtonian
liquids. Other viscometers may be used provided that the 30000
accuracy and precision is not less than that obtained with the lOO 20000 to 6.76 5.6 6.8 to 7.5
viscometers described below. lOO 000

(1) The European Phannacopoeia describes the system proposed by the lnternationaJ Organisation for Standardisatioo (1S0).

General Notices (1) apply to all monographs and other texts 27


2.2.10. Viscosity - Rotating viscometer method EUROPEAN PHARMACOPOEIA 8.0

L M N
values, which cannot be compared with absolute values or
A other relative values if not determined by the same relative
I viscometer method.
I Different measuring systems are available for given viscosity

~j
ranges as well as several rotational speeds.

APPARATUS
The following types of instruments are most common.

~l
CONCENTRIC CYLINDER VISCOMETERS (ABSOLUTE
VIS COME TERS)
In the concentric cylinder viscometer (coaxial double cylinder

~r
viscometer or simply coaxial cylinder viscometer), the viscosity
is determined by placing the liquid in the gap between the
'If
inner cylinder and the outer cylinder. Viscosity measurement

J
mj
can be performed by rotating the inner cylinder (Searle type
viscometer) or the outer cylinder (Couette type viscometer),
as shown in Figures 2.2.10.-1 and 2.2.10.-2, respectively. For
laminar flow, the viscosity (or apparent viscosity) r¡ expressed
in pascal-seconds is given by the following formula:

~ = ~ (4~,) (~7 - ~~) = k ~


o,
NI
t
M torque in newton-metres acting on the cylinder
~
surface,
~t W angular velocity in radians per second,

~II.
h

Ri
height of immersion in metres of the inner cylinder
in the liquid medium,
radius in metres of the inner cylinder,
t
LOI R, radius in metres of the outer cylinder,
""..¡,
k constant of the apparatus, expressed in radians per
Figure 2.2.9.- 1. - Suspended level viscometer cubic metre.
Dimensíons in millimetres For non-Newtonian liquids it is indispensable to specify the
shear stress (T) or the shear rate (y) at which the viscosity is
Method. Fill the viscometer through tube (L) with a sufficient measured. Under narrow gap conditions (conditions satisfied
quantity of the liquid to be examined, previously brought in absolute viscometers), there is a proportional relationship
to 20 oC unless otherwise prescribed, to fill bulb (A) but between M and T and also between w and y:
ensuring that the level of liquid in bulb (B) is below the exit
to ventilation tube (M). Immerse the viscometer in the bath
of water at 20 ± 0.1 oC, unless otherwise prescribed, maintain T=AM ¡=Bw
it in the upright position and allow to stand for not less than
30 min to allow the temperature to reach equilibrium. Close where A and B are constants for the instrument and are
tube (M) and raise the leve! of the liquid in tube (N) up to a calculated from the following expressions:
level about 8 mm aboye mark (E). Keep the liquid at this level
by closing tube (N) and opening tube (M). Open tube (N) - for concentric surface:
and measure, with a stop-watch to the nearest one-fifth of a
second, the time required for the level of the liquid to drop
from mark (E) to (F).

- for cone-plates:

0112008:20210

2.2.10, VISCOSITY - ROTATING M torque in Newtol1-metres acting on the cone or


VISCOMETER METHOD cy1inder surface,
w angular velocity in radian s per second,
The principie of the method is to measure the force acting on
a rotor (torque) when it rotates at a constant angular velocity radius in metres of the inner cylinder,
(rotational speed) in a liquido Rotating viscometers are used radius in metres of the outer cylinder,
for measuring the viscosity of Newtonian (shear-independent
viscosity) or non-Newtonian liquids (shear dependent radius in metres of the cone,
viscosity or apparent viscosity). Rotating viscometers can be height of immersion in metres of the inner cylinder
divided in 2 groups, namely absolute and relative viscometers. in the liquid medium,
In absolute viscometers the flow in the measuring geometry is a angle in radian s between the flat disk and the cone,
well defined. The measurements result in absolute viscosity
T shear stress in pascals (Pa),
values, which can be compared with any other absolute values.
In relative viscometers the flow in the measuring geometry y shear rate in reciprocal seconds (51).
is not defined. The measurements result in relative viscosity

28 See the information section on general monographs (cover pages)


EUROPEAN PHARMACOPOEIA 8.0 2.2.10. Viscosity - Rotating viscometer method

Constants A, B of the apparatus (see under coneentric cylinder


viscometers) .

M
ro

Figure 2.2.10.-3

Figure 2.2.10.-1

/;/////////////
M

Figure 2.2.10.-4
h SPINDLE VISCOMETERS (RELATIVE VISCOMETERS)
In the spindle viscometer, the viscosity is determined by
rotating a spindle (for example, cylinder- or disc-shaped,
as shown in Figures 2.2.10.-5 and 2.2.10.-6, respectively)
immersed in the liquido Relative values of viscosity (or
apparent viscosity) can be directly ca!culated using conversion
factors from the scale reading at a given rotational speed.

Figure 2.2.10.-2
CONE-PLATE VISCOMETERS (ABSOLUTE VISCOMETERS)
In the cone-plate viscometer, the liquid is introduced into the
gap between a flat disc and a cone forming a define angle.
Viscosity measurement can be performed by rotating the cone
or the flat disc, as shown in Figures 2.2.10.-3 and 2.2.10.-4,
respectively. For laminar flow, the viscosity (or apparent
viscosity) r¡ expressed in pascal-seconds is given by the
following formula: /\
r¡= (M)
w
(~)
27fR3
=kM W

M torque in Newton-metres acting on the flat disc or


cone surface,
w angular velocity in radians per second,
a angle in radians between the flat disc and the cone,
R radius in metres of the cone,
k constant of the apparatus, expressed in radians per
eubie metre. Figure 2.2.10.-5

General Natices (1) apply ta all managraphs and ather texts 29


2,2.11. DistiJlation range EUROPEAN PHARMACOPOEIA 8.0

liquids determined from the previous formula has a relative


character, which depends on the type of spindle and the
angular velocity as well as the dimensions of the sample
container (0 = minimum 80 mm) and the depth of immersion
of the spindle. The values obtained are comparable only if
the method is carried out under experimental conditions that
are rigorously the same.

0112008:20211

r --, 2.2.1L DISTILLATION RANGE


I h7 I
I I I I
The distillation range is the temperature interval, corrected
for a pressure of 101.3 kPa (760 Torr), within which a liquid,
or a specified fraction of a liquid, distils in the following
conditions.
Figure 2.2.10.-6
Apparatus. The apparatus (see Figure 2.2.11.-1) consists of a
In a general way, the constant k of the apparatus may be
determined at various speeds of rotation using a certified distillation flask (A), a straight tube condenser (B) which fits
on to the si de arm of the flask and a plain-bend adaptor (e)
viscometer calibration liquid. The viscosity 11 then corresponds
attached to the end of the condenser. The lower end of the
to the formula:
condenser may, alternatively, be bent to replace the adaptor.
M A thermometer is inserted in the neck of the flask so that the
1) = k -
w upper end of the mercury reservoir is 5 mm lower than the
junction of the lower wall of the lateral tube. The thermometer
METHOD is graduated at 0.2 oC intervals and the scale covers a range of
Measure the viscosity (or apparent viscosity) according to the about 50 oc. During the determination, the flask, including its
instructions for the operation of the rotating visco meter. The neck, is protected from draughts by a suitable screen.
temperature for measuring the viscosity is indicated in the Methad. Place in the flask (A) 50.0 mL of the liquid to be
monograph. For non-Newtonian systems, the monograph examined and a few pie ces of porous material. Collect the
indicates the type of viscometer to be used and if absolute distillate in a 50 mL cylinder graduated in 1 mL. Cooling
viscometers are used the angular velocity or the shear rate at by circulating water is essential for liquids distilling below
which the measurement is made. If it is impossible to obtain 150 oc. Heat the flask so that boiling is rapidly achieved and
the indicated shear rate exactly, use a shear rate slightly higher note the temperature at which the first drop of distillate falls
and a shear rate slightly lower and interpolate. into the cylinder. Adjust the heating to give a regular rate of
With relative viscometers the shear rate is not the same distillation of 2-3 mL/min and note the temperature when
throughout the sample and therefore it cannot be defined. the whole or the prescribed fraction of the liquid, measured
Under these conditions, the viscosity of non-Newtonian at 20 oC, has distilled.

o
OC!

Figure 2.2.11.-1. - Apparatus far the determinatian af distillatian range


Dimensians in millimetres

30 See the infarmatian section an general manographs (cover pages)


EUROPEAN PHARMACOPOEIA 8.0 2.2.13. Determination of water by distillation

Correct the observed temperatures for barometric pressure by Method. Clean the receiving tube and the condenser of the
means of the formula: apparatus, thoroughly rinse with water, and dry.
Introduce 200 mL of toluene R and about 2 mL of water R into
h = t2 + k (101.3 - b) the dry flask. Distil for 2 h, then allow to cool for about 30 min
and read the water volume to the nearest 0.05 mL. Place in the
t¡ the corrected temperature, flask a quantity of the substance, weighed with an accuracy of
t2 the observed temperature, at the barometric 1 per cent, expected to give about 2 mL to 3 mL of water. If
pressure b, the substance has a pasty consistency, weigh it in a boat of
metal foil. Add a few pieces of porous material and heat the
le the correction factor taken from Table 2.2.11.-1
flask gently for 15 mino When the toluene begins to boil, distil
unless the factor is given,
at the rate of about two drops per second until most of the
b the barometric pressure, expressed in kilopascals, water has distilled over, then increase the rate of distillation to
during the distillation. about four drops per second. When the water has all distilled
over, rinse the in si de of the condenser tube with toluene R.
Table 2.2.11.-1. - Temperature correction in relation to the Continue the distillation for 5 min, remove the heat, allow the
pressure receiving tube to cool to room temperature and dislodge any
Distillation temperature Correction factor k droplets of water which adhere to the walls of the receiving
tube. When the water and toluene have completely separated,
up to 100 oC 0.30
read the volume of water and calculate the content present in
aboye lOO oC up to 140 oC 0.34 the substance as millilitres per kilogram, using the formula:
aboye 140 oC up to 190 oC 0.38 1000 (n2 - nl)
m
aboye 190 oC up to 240 oC 0.41

aboye 240 oC 0.45 m the mass in grams of the substance to be examined,


the number of millilitres of water obtained in the
first distillation,
n2 the total number of millilitres of water obtained in
01/2008:20212 the 2 distillations.

2.2.12. BOILING POINT


The boiling point is the corrected temperature at which the
vapour pressure of a liquid is equal to 1Ol.3 kPa.
Apparatus. The apparatus is that used for Distillation Range
(2.2.11) with the exception that the thermometer is inserted
in the neck of the flask so that the lower end of the mercury
reservoir is level with the lower end of the neck of the o
o
distillation flask and that the flask is placed on a plate of N
/\1
isolating material pierced by a hole 35 mm in diameter. e
Method. Place in the flask (A) 20 mL of the liquid to be
examined and a few pieces of porous material. Heat the flask
so that boiling is rapidly achieved and record the temperature
at which liquid runs from the side-arm into the condenser.
Correct the observed temperature for barometric pressure by
means of the formula:

tl = t2 + k (101.3 - b)

the corrected temperature, B 1.[)


N
the observed temperature at barometric pressure b,
k the correction factor as shown in Table 2.2.1l.-1
under Distillation Range,
b the barometric pressure, in kilopascals, at the time
of the determination. E

0112008:20213
~]
2.2.13. DETERMINATION OF WATER
BY DISTILLATION
500:mL A
The apparatus (see Figure 2.2.13.-1) consists of a glass flask (A)
connected by a tube (D) to a cylindrical tube (B) fitted with I
165
a graduated receiving tube (E) and re flux condenser (e). The
receiving tube (E) is graduated in 0.1 mL. The source of heat
is preferably an electric heater with rheostat control or an oil Figure 2.2.13.-1. - Apparatus for the determination of water
bath. The upper portion of the flask and the connecting tube by distillation
may be insulated. Dimensions in millimetres

General Notices (1) apply to all monographs and other texts 31


2.2.14. Melting point - capiHary method EUROPEAN PHARMACOPOEIA 8.0

01/2008:20214 Repeat the operation with the other 4 capillary tubes and
calculate the result as the mean of the 5 readings.
2.2.14. MELTING POINT - CAPILLARY
0112008:20216
METHOD
The melting point determined by the capillary method is 2.2.16. MELTING POINT -
the temperature at which the last solid particle of a compact
column of a substance in a tube passes into the liquid phase.
INSTANTANEOUS METHOD
When prescribed in the monograph, the same apparatus and The instantaneous melting point is calculated using the
method are used for the determination of other factors, such expression:
as meniscus formation or melting range, that characterise the tI + t2
melting behaviour of a substance. 2
Apparatus. The apparatus consists of:
in which t) is the first temperature and t 2 the second
- a suitable glass vessel containing a liquid bath (for example, temperature read under the conditions stated below.
water, liquid paraffin or silicone oil) and fitted with a
suitable means of heating, Apparatus. The apparatus consists of a metal block resistant
to the substance to be examined, of good heat-conducting
- a suitable means of stirring, ensuring uniformity of capacity, such as brass, with a carefully polished plane upper
temperature within the bath, surface. The block is uniformly heated throughout its mass by
- a suitable thermometer with graduation at not more than means of a micro-adjustable gas heater or an electric heating
0.5 oC intervals and provided with an immersion mark. device with fine adjustment. The block has a cylindrical cavity,
The range of the thermometer is not more than 100 oC, wide enough to accomodate a thermometer, which should be
- alkali-free hard-glass capillary tubes of internal di ame ter maintained with the mercury column in the same position
0.9 mm to 1.1 mm with a wall 0.10 mm to 0.15 mm thick during the calibration of the apparatus and the determination
and sealed at one end. of the melting point of the substance to be examined. The
Method. Unless otherwise prescribed, dry the finely powdered cylindrical cavity is parallel to the upper polished surface of
substance in vacuo and over anhydrous silica gel R for 24 h. the block and about 3 mm from it. The apparatus is calibrated
Introduce a sufficient quantity into a capillary tube to give using appropriate substances of known melting point.
a compact column 4 mm to 6 mm in height. Raise the Method. Heat the block at a suitably rapid rate to a temperature
temperature of the bath to about 10 oC below the presumed about 10 oC below the presumed melting temperature, then
melting point and then adjust the rate of heating to about adjust the heating rate to about 1 oC/mino At regular intervals
1 oC/mino When the temperature is 5 oC below the presumed drop a few particles of powdered and, where appropriate, dried
melting point, correctly introduce the capillary tube into the substance, prepared as for the capillary tube method, onto the
instrument. For the apparatus described aboye, immerse the block in the vicinity of the thermometer bulb, cleaning the
capillary tube so that the closed end is near the centre of the surface after each test. Record the temperature t¡ at which the
bulb of the thermometer, the immersion mark of which is at substance melts instantaneously for the first time in contact
the level of the surface of the liquido Record the temperature with the metal. Stop the heating. During cooling drop a few
at which the last particle passes into the liquid phase. particles of the substance at regular intervals on the block,
Calibration of the apparatus. The apparatus may be calibrated cleaning the surface after each test. Record the temperature t2
using melting point reference substances such as those of the at which the substance ceases to melt instantaneously when it
World Health Organization or other appropriate substances. comes in contact with the metal.
Calibration of the apparatus. The apparatus may be calibrated
using melting point reference substances such as those of the
0112008:20215 World Health Organization or other appropriate substances.

2.2.15. MELTING POINT - OPEN 01/2008:20217


CAPILLARY METHOD
For certain substances, the following method is used to
2.2.17. DROP POINT
determine the melting point (also referred to as slip point and The drop point is the temperature at which the first drop of
rising melting point when determined by this method). the melting substance to be examined falls from a cup under
Use glass capillary tubes open at both ends, about 80 mm defined conditions.
long, having an external diameter of 1.4 mm to 1.5 mm and When a monograph does not specify the method to be used,
an internal di ame ter of l.0 mm to 1.2 mm. method A is applied. Any change from method A to method B
Introduce into each of 5 capillary tubes a sufficient amount of is validated.
the substance, previously treated as described, to form in each
METHODA
tube a column about 10 mm high and allow the tubes to stand
for the appropriate time and at the prescribed temperature. Apparatus. The apparatus (see Figure 2.2.17.-1) consists of
2 metal sheaths (A and B) screwed together. Sheath A is fixed
Unless otherwise prescribed, substances with a waxy
to a mercury thermometer. A metal cup is loosely fixed to
consistency are carefully and completely melted on a the lower part of sheath B by means of 2 tightening bands.
water-bath before introduction into the capillary tubes. Allow Fixed supports 2 mm long determine the exact position of
the tubes to stand at 2-8 oC for 2 h.
the cup, and in addition are used to centre the thermometer.
Attach one of the tubes to a thermometer graduated in 0.5 oC A hole pierced in the wall of sheath B is used to balance the
so that the substance is close to the bulb of the thermometer. pressure. The draining surface of the cup must be flat and the
Introduce the thermometer with the attached tube into a edges of the outflow orifice must be at right angles to it. The
beaker so that the distance between the bottom of the beaker lower part of the mercury thermometer has the form and size
and the lower part of the bulb of the thermometer is 1 cm. shown in the figure; it covers a range from O oC to 110°C and
Fill the beaker with water to a depth of 5 cm. Increase the on its scale a distance of 1 mm represents a difference of 1 oc.
temperature of the water gradually at arate of 1 oC/mino The mercury reservoir of the thermometer has a diameter of
The temperature at which the substance begins to rise in the 3.5 ± 0.2 mm and a height of 6.0 ± 0.3 mm. The apparatus
capillary tube is regarded as the melting point. is placed in the axis of a test-tube about 200 mm long and

32 See the information section on general monographs (cover pages)


EUROPEAN PHARMACOPOEIA 8.0 2.2.17. Drop point

with an external diameter of about 40 mm. It is fixed to the There is another, narrower cylindrical vertical hole in which
test-tube by means of a lateraUy grooved stopper through a temperature sensor sits, This is positioned level with the
which the thermometer pass es. The opening of the cup is sample cup. The heating block is surrounded by an electrical
placed about 15 mm from the bottom of the test-tube. The heating element. Below the heating block a lamp is mounted
whole device is immersed in a beaker with a capacity of about such that a beam of light shines through the light slit in the
1 L, filled with water. The bottom of the test -tube is placed collector sleeve, and onto a photo-sensor mounted opposite.
about 25 mm from the bottom of the beaker. The water level The heating block is capable of being maintained at a precise,
reaches the upper part of sheath A. A stirrer is used to ensure pre-defined temperature by the heating element, and of being
that the temperature oí the water remains uniformo heated at a slow and steady, pre-defined rate after an initial
isothermal periodo

~
~
f
~
t tI

, 10
~

12.5
~ ~----

~I
1 --
- - -
I
- - -
A

¡ °1
--'--
I
I

--1--
1 I
I
.~._----
\ ( ~I
B
i
r-- ,
e
JJ~-
- 17.5 ± 0.05
N
-<p ~

1 ,1 1
LD e

~r
D

~ hl~ E ---_.-
_I~-
t '1 ~, F I
N J
11 ~
1 I~ t
11 I - 1 , 6 ± 0.05
,I 1 ~

1
A. cup holder F. heatillg element
5 , 3 ± 0.05
~
~I ~------
B. heating block G. sample cup

e light source H. photo-sensor


A upper metal sheath D. fixed supports
D. light slit ). collector sleeve
B, lower metal sheath E, tightening bands
E. cartridge assembly K. temperature probe
C. pressure-balancing hole E metal sample cup
Figure 2.2.17.-2. - Example of automated drop paint apparatus
Figure 2.2.17.-1. - Apparatusfar the determinatian Method, Melt the substance to be examined and introduce
af drap paint it into the sample cup according to the prescriptions of the
Dimensians ín millímetres monograph, then proceed as follows or according to the
Methad. Prepare the substance to be examined according manufacturer's instructions, Remove the excess substance at
to the prescriptions of the monograph. Fill the cup to the the 2 ends of the cup with a spatula. Condition the sample at
brim with the substance to be examined. Remove the excess the temperature and for the time prescribed in the monograph
substance at the 2 ends of the cup with a spatula. When before making the measurement. Press the cup into the cup
sheaths A and B have been assembled, press the cup into its holder, and then press the coUector sleeve onto the cup, Place
housing in sheath B until it touches the supports. Remove the cartridge assembly in the heating block Set the instrument
with a spatula the substance pushed out by the thermometer. to the initial isothermal conditions and rate for subsequent
Place the apparatus in the water-bath as described aboye. heating as described in the monograph of the substance to be
Heat the water-bath and, when the temperature is at about examined. Start the temperature programme. When the first
10 oC below the presumed drop point, adjust the heating rate drop of molten sample falls through the hole at the bottom of
to about 1 oC/mino Note the temperature at the faH of the the sample cup, interrupting the light beam, the signal from
first drop. Carry out at least 3 determinations, each time with the photo-sensor causes the temperature of the heating block
a fresh sample of the substance. The difference between the to be recorded automatically.
readings must not exceed 3 oc. The mean of 3 readings is the Calibratían. Use the apparatus according to the manufacturer's
drop point of the substance. instructions and carry out the prescribed calibrations and
system performance tests at regular intervals, depending on
METHOD B - AUTOMATED METHOD the use of the apparatus and the substances to be examined.
Apparatus. The apparatus (see Figure 2.2.17.-2) consists of Benzoic acid and benzophenone are usually used as certified
a cartridge assembly comprising a cup holder into which reference materials. Other materials may be used provided
the sample cup containing the sample is loosely fixed, and they show no polymorphism. Proceed as follows or according
a coUector sleeve with a horizontallight slit, which is fixed to the manufacturer's instructions, Prepare 3 sample cups for
below the cup, This assembly is placed in a heating block each of the 2 certified reference materials. Place the sample
The block is a metal cylinder with a cylindrical hole along CUpS 011 a cIean surface. 1nto each sample cup, introduce
its vertical axis into which the cartridge assembly is placed. a small quantity of the sample and press it down with a

General Natices (1) apply ta all managraphs and ather texts 33


2.2.18. Freezing point EUROPEAN PHARMACOPOEIA 8.0

rod (diameter about 4.5 mm). Check that the opening is Apparatus. The apparatus (see Figure 2.2.18.-1) consists ofa
completely filled. Fill the sample cup about half full and test-tube about 25 mm in diameter and 150 mm long placed
compact the sample with a rod (diameter about 9 mm). Fill inside a test-tube about 40 mm in diameter and 160 mm
the sample cup completely and compact, adding more sample long. The inner tube is closed by a stopper which carries a
and compacting again if necessary, until the sample cup is thermometer about 175 mm long and graduated in 0.2 oC
completely full. fixed so that the bulb is about 15 mm aboye the bottom of the
Temperature programme for benzoic acid: start tube. The stopper has a hole allowing the passage of the stem
temperature = 118.0 oC; heating rate = 0.2 °C/min; end of a stirrer made from a glass rod or other suitable material
temperature = 126.0 oc. After inserting the cup at 118 oC, a formed at one end into a loop of about 18 mm overall diameter
waiting time of 30 s is set before heating starts. at right angles to the rod. The inner tube with its jacket is
Temperature programme for benzophenone: start supported centrally in a 1 L beaker containing a suitable
temperature = 44.0 oC; heating rate = 0.2 °C/min; end cooling liquid to within 20 mm of the topo A thermometer is
temperature = 56.0 oc. After inserting the cup at 44 oC, a supported in the cooling bath.
waiting time of 30 s is set before heating starts. Method. Place in the inner tube sufficient quantity of the
Check the 3 single results: the test is valid if the 3 results are liquid or previously melted substance to be examined, to cover
within 0.3 oC of the mean value. the thermometer bulb and determine the approximate freezing
point by cooling rapidly. Place the inner tube in a bath about
Calculate the corrected mean temperature (T)2 using the 5 oC above the approximate freezing point until all but the last
following expression: traces of crystals are melted. Fill the beaker with water or a
saturated solution of sodium chloride, at a temperature about
5 oC lower than the expected freezing point, insert the inner
tube into the outer tube, ensuring that sorne seed crystals are
present, and stir thoroughly until solidification takes place.
Note the highest temperature observed during solidification.
T¡ mean drop point temperature of 3 samples, in oC;
F compensation for the difference in temperature 0112008:20219
between the sample and the point in the heating
block where the temperature is measured; this will 2.2.19. AMPEROMETRIC TITRATION
vary depending upon the design of the automatic
drop point instrument and is provided by the In amperometric titratiol1 the end-point is determined by
manufacturero following the variation of the current measured between
2 electro des (either one indicator electro de and one reference
Taking into account the drop point (Ta) of the certified electrode or 2 indicator electro des ) immersed in the solution
reference material, the accuracy of the temperature scale is to be examined and maintained at a constant potential
satisfactory if IT2 ~ Tol is not greater than 0.3 oc. difference as a function of the quantity of titrant added.
The potential of the measuring electro de is sufficient to ensure
01/2008:20218 a diffusion current for the electro active substance.
Apparatus. The apparatus comprises an adjustable voltage
2.2.18. FREEZING POINT source and a sensitive microammeter; the detection system
The freezing point is the maximum temperature occurring generally consists of an indicator electro de (for example,
during the solidification of a supercooled liquido a platinum electrode, a dropping-mercury electro de, a
rotating-disc electro de or a carbon electrode) and a reference
Q electrode (for example, a calomel electrode or a silver-silver
chloride electro de ).
A three-electrode apparatus is sometimes used, consisting of
an indicator electro de, a reference electrode and a polarised
auxiliary electro de.
A
Method. Set the potential of the indicator electro de as
~~ ~
prescribed and plot a graph of the initial current and the
values obtained during the titratio11 as functions of the
r- quantity of titrant added. Add the titrant in not fewer than
-
- -
~

3 successive quantities equal to a total of about 80 per cent


~

of the theoretical volume corresponding to the presumed


- - equivalence point. The 3 values must faH 011 a straight lineo
~
-
Continue adding the titrant beyond the presumed equivalence
- point in not fewer than 3 successive quantities. The values
- obtained must fal! on a straight lineo The point of intersection
o
"' - ~
of the 2lines represents the end-point of the titration.
For amperometric titration with 2 indicator electro des, the
-
~
whole titration curve is recorded and used to determine the
end-point.
~

-- -
-- --f
-
-r o 07/2013:20220
.~
- O)

I I
'l:..-~-..v "'t l 2.2.20. POTENTIOMETRIC TITRATION
'r-F"18/ 7
'"t
I 1+ 25 -+, In a potentiometric titration the end -point of the titration
+--- 40 ----+ is determined by following the variation of the potential
difference between 2 electro des (either one indicator electro de
Figure 2.2.18.-1. - Apparatusfor the determination offreezing and one reference electro de or 2 indicator electrodes)
point immersed in the solution to be examined as a function of the
Dimensions in millimetres quantity of titrant added.

34 See the information section on general monographs (caver pages)


EUROPEAN PHARMACOPOEIA 8.0 2.2.22. Atomic emission spectrometry

The potential is usually measured at zero or practically zero concentration of the solution to be examined,
current.
concentration of the standard solution,
Apparatus. The apparatus used (a simple potentiometer or
electronic device) comprises a voltmeter allowing readings intensity of the light emitted by the solution to be
to the nearest millivolt. examined,
1, intensity of the light emitted by the standard
The indicator electrode to be used depends on the substance
solution.
to be determined and may be a glass or metal electro de (for
example, platinum, gold, silver or mercury). The reference If the intensity of the fluorescence is not strictly proportional
electro de is generally a calomel or a silver-silver chloride to the concentration, the measurement may be effected using
electro de. a calibration curve.
For acid-base titrations and unless otherwise prescribed, In sorne cases, measurement can be made with reference
a glass-calomel or glass-silver-silver chloride electro de to a fixed standard (for example a fluorescent glass or a
combination is used. solution of another fluorescent substance). In such cases,
the concentration of the substance to be examined must be
Methad. In potentiometric titrations of weak acids or determined using a previously drawn calibration curve under
bases using non-aqueous solvents, either carry out a blank the same conditions.
determination or pre-neutralise the solvent mixture, if
necessary, before dissolution of the substance to be examined.
Where it is impracticable to use potentiometric detection for 01/2008:20222
this purpose, the solvent mixture can be pre-neutralised by
titration using a suitable indicator. Sorne examples are given 2.2.22. ATOMIC EMISSION
below:
SPECTROMETRY
Titrant Indicator GENERAL PRINCIPLE
Perchloric acid Oystal violet solution R Atomic emission is a process that occurs when electromagnetic
3 giL solution of thymol blue R radiation is emitted by excited atoms or ions. In atomic
Tetrabutylammonium hydroxide emission spectrometry the sample is subjected to temperatures
in methanol R
Ethanolic sodium hydroxide Thymolphthalein solution R high enough to cause not only dissociation into atoms, but
also to cause significant amounts of collisional excitation
Plot a graph of the variation of potential difference as a and ionisation of the sample atoms to take place. Once the
function of the quantity of the titrant added, continuing the atoms and ion s are in the excited states, they can decay to
addition of the titrant beyond the presumed equivalence lower states through thermal or radiative (emission) energy
point. The end-point corresponds to a sharp variation of transitions and electromagnetic radiation is emitted. An
potential difference. emission spectrum of an element contains several more lines
than the corresponding absorption spectrum.
Atomic emission spectrometry is a technique for determining
the concentration of an element in a sample by measuring the
intensity of one of the emission lines of the atomic vapour of
01/2008:20221 the element generated from the sample. The determination is
carried out at the wavelength corresponding to this emission
lineo
2.2.21. FLUORIMETRY In this chapter only atomisation in flame is dealt with. The
method of inductively coupled plasma-atomic emission
Fluorimetry is a procedure which uses the measurement of the
spectrometry (ICP-AES) is described in a different general
intensity of the fluorescent light emitted by the substance to
chapter.
be examined in relation to that emitted by a given standard.
Methad. Dissolve the substance to be examined in the solvent APPARATUS
or mixture of solvents prescribed in the monograph, transfer This consists essentially of:
the solution to the cell or the tube of the fluorimeter and - a sample introduction and nebulisation system;
illuminate it with an excitant light beam of the wavelength - a flame to generate the atoms to be determined;
prescribed in the monograph and as near as possible
monochromatic. - a monochromator;
- a detector;
Measure the intensity of the emitted light at an angle of 90°
to the excitant beam, after passing it through a filter which - a data -acquisition unit.
transmits predominantly light of the wavelength of the Oxygen, air and a combustible gas such as hydrogen, acetylene,
fluorescence. Other types of apparatus may be used provided propane or butane may be used in flames. The atomisation
that the results obtained are identical. source is critica!, since it must provide sufficient energy to
excite and atomise the atoms. The atomic spectra emitted
For quantitative determinations, first introduce into the from flames have the advantage of being simpler than those
apparatus the solvent or mixture of solvents used to dissolve emitted from other sources, the main limitation being that the
the substance to be examined and set the instrument to zero. flames are not powerful enough to cause emission for many
Introduce the standard solution and adjust the sensitivity elements allowing their determination. Acidified water is the
of the instrument so that the reading is greater than 50. If solvent of choice for preparing test and reference solutions,
the second adjustment is made by altering the width of the although organic solvents may also be used if precautions are
slits, a new zero setting must be made and the intensity of taken to ensure that the solvent do es not interfere with the
the standard must be measured again. Finally introduce the stability of the flameo
solution of unknown concentration and read the result on the
instrument. Calculate the concentration ex of the substance in INTERFERENCES
the solution to be examined, using the formula: Spectral interference is reduced or eliminated by choosing an
appropriate emission hne for measurement or by adjusting the
slit for spectral band-width. Physical interference is corrected

General Natices (1) apply ta all managraphs and ather texts 35


2.2.23. Atomic absorption spectrometry EUROPEAN PHARMACOPOEIA 8.0

by diluting the sample solution, by matching the matrix or by VALIDATION OF THE METHOD
using the method of standard additions. Chemical interference Satisfactory performance of methods prescribed in
is reduced by using chemical modifiers or ionisation buffers. monographs is verified at suitable time intervals.
MEMORY EFFECT LINEARITY
Prepare and analyse not fewer than 4 reference solutions over
The memory effect caused by deposit of analyte in the
the calibration range and a blank solution. Perform not fewer
apparatus may be limited by thoroughly rinsing between runs,
than 5 replicates.
diluting the solutions to be measured if possible and thus
reducing their salt content, and by aspirating the solutions The calibration curve is calculated by least-square regression
through as swiftly as possible. from all measured data. The regression curve, the means, the
measured data and the confidence interval of the calibration
METHOD curve are plotted. The operating method is valid when:
Use of plastic labware is recommended wherever possible. - the correlation coefficient is at least 0.99,
Operate an atomic emission spectrometer in accordance with - the residuals of each calibration level are randomly
the manufacturer's instructions at the prescribed wave!ength. distributed around the calibration curve.
Optimise the experimental conditions (flame temperature, Calculate the mean and re!ative standard deviation for the
burner adjustment, use of an ionic buffer, concentration of lowest and highest calibration leveL
solutions) for the specific element to be analysed and in respect When the ratio of the estimated standard deviation of the
of the sample matrix. Introduce a blank solution into the lowest and the highest calibration leve! is les s than 0.5 or
atomic generator and adjust the instrument reading to zero or greater than 2.0, a more precise estimation of the calibration
to its blank value. Introduce the most concentrated reference curve may be obtained using weighted linear regression. Both
solution and adjust the sensitivity to obtain a suitable reading. linear and quadratic weighting functions are applied to the
1t is preferable to use concentrations which faH within the data to find the most appropriate weighting function to be
linear part of the calibration curve. lf this is not possible, theemployed. If the means compared to the calibration curve
calibration plots may also be curved and are then to be applied show a deviation from linearity, two-dimensionallinear
with appropriate calibration software. regression is used.
Determinations are made by comparison with reference ACCURACY
solutions with known concentrations of the element to Verify the accuracy preferably by using a certified reference
be determined either by the method of direct calibration material (CRM). Where this is not possible, perform a test
(Method 1) or the method of standard additions (Method II). for recovery.
METHOD 1 - DIRECT CALIBRATION Recovery. For assay determinations a recovery of 90 per cent
For routine measurements 3 reference solutions of the element to 110 per cent is to be obtained. For other determinations,
to be determined and a blank are prepared and examined. for example for trace element determination, the test is not
Prepare the solution of the substance to be examined (test valid if recovery is outside of the range 80 per cent to 120 per
solution) as prescribed in the monograph. Prepare not fewer cent at the theoretical value. Recovery may be determined on
than 3 reference solutions of the element to be determined, a suitable reference solutiol1 (matrix solution) which ís spiked
the concentrations of which span the expected value in the with a lmown quantity of analyte (middle concentration of
test solution. For assay purposes, optimal calibration levels the calibration range).
are between 0.7 and 1.3 times the expected content of the REPEA TABILITY
element to be determined or the limit prescribed in the The repeatability is not greater than 3 per cent for an assay
monograph. For purity determination, calibration levels are and not greater than 5 per cent for an impurity test.
between the limit of detection and 1.2 times the limit specified
LIMIT OF QUANTIFICATION
for the e!ement to be determined. Any reagents used in the
preparation of the test solution are added to the reference Verify that the Iimit of quantification (for example, determíned
using the 10 (J approach) is below the value to be measured.
solutions and to the blank solution at the same concentration.
Introduce each of the solutions into the instrument using the
same number of replicates for each solution, to obtain a steady 0112008:20223
reading.
Calculation. Prepare a calibration curve from the mean of 2.2023. ATOMIC ABSORPTION
the readings obtained with the reference solutions by plotting
the means as a function of concentration. Determine the SPECTROMETRY
concentration of the element in the test solution from the
GENERAL PRINCIPLE
curve obtained.
Atomic absorption is a process that occurs when a ground
METHOD JI - STANDARD ADDITIONS state-atom absorbs electromagnetic radiation of a specific
Add to at least 3 similar volumetric flasks equal volumes of wave!ength and is elevated to an excited state. The atoms in
the solution of the substance to be examined (test solution) the ground state absorb energy at theír resonant frequency
prepared as prescribed. Add to al! but 1 of the flasks and the electromagnetic radiation is attenuated due to
progressively larger volumes of a reference solution containing resonance absorption. The energy absorption is virtually a
a known concentration of the element to be determined to direct function of the number of atoms present.
produce a series of solutions containing steadily increasing This chapter provides general information and defines
concentrations of that element known to give responses in the
the procedures used in e!ement determinations by atomic
linear part of the curve, if at al! possible. Dilute the contents
absorption spectrometry, either atomisation by flame, by
of each flask to volume with solvent. electrothermal vaporisation in a graphite furnace, by hydride
Introduce each of the solutions into the instrument using the gene ratio n or by cold vapour technique for mercury.
same number of replicates for each solution, to obtain a steady Atomic absorption spectrometry is a technique for
reading. determining the concentration of an e!ement in a sample by
Calculation. Calculate the linear equation of the graph using measuring the absorption of electromagnetic radiation by
a least -squares fit, and derive from it the concentration of the the atomic vapour of the element generated from the sample.
element to be determined in the test solution. The determination is carried out at the wavelength of one of

36 See the information section on general monographs (cover pages)


EUROPEAN PHARMACOPOEIA 8.0 2.2.23. Atomic absorption spectrometry

the absorption (resonance) lines of the element concerned. matching, physical interference due to high salt content or
The amount of radiation absorbed is, according to the viscosity is eliminated. Spectral interference results from the
Lambert-Beer law, proportional to the element concentration. overlapping of resonance lines and can be avoided by using
a different resonance line. The use of Zeeman background
APPARATUS correction also compensates for spectral interference and
This consists essentially of: interferences from molecular absorption, especially when
- a source of radiation; using the eIectrothermal atomisation technique. The use of
multi-element hollow-cathode lamps may also cause spectral
- a sample introduction device; interference. Specific or non-specific absorptiol1 is measured
- a sample atomiser; in a spectral range defined by the band-width selected by the
- a monochromator or polychromator; monochromator (0.2-2 nm).
- a detector; BACKGROUND CORRECTION
- a data -acquisition unit. Scatter and background in the flame or the electrothermal
The apparatus is usually equipped with a background atomisation technique increase the measured absorbance
correction system. Hollow-cathode lamps and electrodeless values. Background absorption covers a large range of
discharge lamps (EDL) are used as radiation source. The wavelengths, whereas atomic absorption takes place in a very
emission of such lamps consists of a spectrum showing very narrow wavelength range of about 0.005-0.02 nm. Background
narrow lines with half-width of about 0.002 nm of the element absorption can in principIe be corrected by using a blank
being determined. solution of exactly the same composition as the sample, but
There are 3 types of sample atomisers: without the speciflc element to be determined, although this
method is frequently impracticable. With the electrothermal
- Flame technique atomisation technique the pyrolysis temperature is to be
A flame atomiser is composed of a nebulisation system optimised to eliminate the matrix decomposition products
with a pneumatic aerosol production accessory, a gas-flow causing background absorption. Background correction
regulation and a burner. Fuel-oxidant mixtures are can also be made by using 2 different light sources, the
commonly used to produce a range of temperatures from hollow-cathode lamp that measures the total absorption
about 2000 K to 3000 K. Fuel gases include propane, (element + background) and a deuterium lamp with a
hydrogen and acetylene; air and nitrous oxide are used as continuum emission from which the background absorption
oxidants. The configuration of the burner is adapted to is measured. Background is corrected by subtracting the
the gases used and the gas flow is adjustable. Samples are deuterium Iamp signal from the hollow-cathode lamp signal.
nebulised, acidified water being the solvent of choice for This method is limited in the spectral range on account of
preparing test and reference solutions. Organic solvents the spectra emitted by a deuterium lamp from 190-400 nm.
may also be used if precautions are taken to ensure that the Background can also be measured by taking readings at
solvent does not interfere with the stability of the flameo a non-absorbil1g line near the resonance line and then
- Electrothermal atomisation technique subtracting the results from the measurement at the resonance
lineo Another method for the correction of background
An electrothermal atomiser is generally composed of
absorption is the Zeeman effect (based 011 the Zeeman
a graphite tube furnace and an electric power source.
splitting of the absorption line in a magnetic fieId). This is
Electrothermal atomisation in a graphite tube furnace
particularly useful when the background absorption shows
atomises the entire sample and retains the atomic vapour
fine structure. It permits an efficient background correction
in the light path for an extended periodo This improves
in the range of 185-900 nm.
the detection limit. Samples, liquid as well as solid,
are introduced directly into the graphite tube furnace, CHOICE OF THE OPERATING CONDITIONS
which is heated in a programmed series of steps to dry
After selecting the suitable wavelength and slit width for
the sample and remove major matrix components by
the specific element, the need for the following has to be
pyrolysis and to then atomise aU of the analyte. The
ascertained:
furnace is cleaned using a final temperature higher than the
atomisation temperature. The flow of an inert gas during - correction for non-specific background absorption,
the pyrolysis step in the graphite tube furnace allows a - chemical modifiers or ionisation buffers to be added to the
better performance of the subsequent atomisation process. sample as well as to blank and reference solutions,
- Cold vapour and hydride technique - dilution of the sampIe to minimise, for example, physical
The atomic vapour may also be generated outside the interferences,
spectrometer. This is notably the case for the cold-vapour - details of the temperature programme, preheating, drying,
method for mercury or for certain hydride-forming pyrolysis, atomisation, post-atomisation with ramp and
elements such as arsenic, antimony, bismuth, selenium hold times,
and tino For mercury, atoms are generated by chemical - inert gas flow,
reduction with stannous chIoride or sodium borohydride
- matrix modifiers for electrothermal atomisation (furnace),
and the atomic vapour is swept by a stream of an inert gas
into a cold quartz cel! mounted in the optical path of the - chemical reducing reagents for measurements of mercury
instrument. Hydrides thus generated are swept by an inert or other hydride-forming elements along with cold vapour
gas into a heated ceU in which they are dissociated into cel! or heating ceH temperature,
atoms. - specification of furnace design (tank, L'vov platform, etc).
INTERFERENCES METHOD
Chemical, physical, ionisation and spectral interferences are Use of plastic labware is recommended wherever possible.
encountered in atomic absorption measurements. Chemical The preparation of the sample may require a dissolution, a
interference is compensated by addition of matrix modifiers, digestion (mostly microwave-assisted), an ignition step or a
of releasing agents or by using high temperature produced by combination thereof in order to clear up the sample matrix
a nitrous oxide-acetylene flame; the use of specific ionisation and/or to remove carbon-containing material. If operating in
buffers (for example, lanthanum and caesium) compensates an open system, the ignition temperature should not exceed
for ionisation interference; by dilution of the sample, 600 oC, due to the volatility of some metals, unless otherwise
through the method of standard additions or by matrix stated in the monograph.

General Natices (1) apply ta all monagraphs and other texts 37


2.2.24. Absorption spectrophotometry, infrared EUROPEAN PHARMACOPOEIA 8.0

Operate an atomic absorption spectrometer in accordance The calibration curve is calculated by least -square regression
with the manufacturer's instructions at the prescribed from all measured data. The regression curve, the means, the
wavelength. Introduce a blank solution into the atomic measured data and the confidence interval of the calibration
generator and adjust the instrument reading so that it indicatescurve are plotted. The operating method is valid when:
maximum transmission. The blank value may be determined - the correlation coefficient is at least 0.99,
by using solvent to zero the apparatus. Introduce the most - the residuals of each calibration level are randomly
concentrated reference solution and adjust the sensitivity to distributed around the calibration curve.
obtain a maximum absorbance reading. Rinse in order to
avoid contamination and memory effects. After completing Calculate the mean and relative standard deviation for the
the analysis, rinse with water R or acidified water. lowest and highest calibration leve!.
When the ratio of the estimated standard deviation of the
lf a solid sampling technique is applied, full details of the lowest and the highest calibration level is less than 0.5 or
procedure are provided in the monograph. greater than 2.0, a more precise estimation of the calibration
Ensure that the concentrations to be determined faH preferably curve may be obtained using weighted linear regression. Both
within the linear part of the calibration curve. lf this is not linear and quadratic weighting functions are applied to the
possible, the calibration plots may also be curved and are then data to find the most appropriate weighting function to be
to be applied with appropriate calibration software. employed. If the means compared to the calibration curve
show a deviation from linearity, two-dimensionallinear
Deferminations are made by comparison with rderence
regression is used.
solutions with known concentrations of the element to
be determined either by the method of direct calibration ACCURACY
(Method I) OY the method of standard additions (Method II). Verify the accuracy preferably by using a certified reference
material (CRM). Where this is not possible, perform a test
METHOD I - DIRECT CALIBRATION
for recovery.
For routine measurements 3 reference solutions and a blank
solution are prepared and examined. Recovery. For assay determinations a recovery of 90 per cent
to 110 per cent is to be obtained. For other determinations,
Prepare the solution of the substance to be examined (test for example, for trace element determination the test is not
solution) as prescribed in the monograph. Prepare not fewer valid if recovery is outside of the range 80 per cent to 120 per
than 3 reference solutions of the element to be determined, cent at the theoretical value. Recovery may be determined on
the concentrations of which span the expected value in the test a suitable reference solution (matrix solution) which is spiked
solution. For assay purposes, optimal calibration levels are with a known quantity of ana1yte (middle concentration of
between 0.7 and 1.3 times the expected content ofthe element the calibration range).
to be determined or the limit prescribed in the monograph.
For purity determination, calibration levels are the limit of REPEATABILITY
detection and 1.2 times the limit specified for the element to The repeatability is not greater than 3 per cent for an assay
be determined. Any reagents used in the preparation of the and not greater than 5 per cent for an impurity test.
test solution are added to the reference and blank solutions at LIMIT OF QUANTIFICATION
the same concentration. Verify that the limit of quantification (for example, determined
Introduce each of the solutions into the instrument using the using the 10 (j approach) is below the value to be measured.
same number of replicates for each of the solutions to obtain
a steady reading. 0112008:20224
Cakulation. Prepare a calibration curve from the mean of
the readings obtained with the reference solutions by plotting 2.2.24. ABSORPTION
the means as a function of concentration. Determine the
concentration of the element in the test solution from the
SPECTROPHOTOMETRY, INFRARED
curve obtained. Infrared spectrophotometers are used for recording spectra in
METHOD JI - STANDARD ADDITIONS the region of 4000-650 cm- 1 (2.5-15.4 !lm) or in some cases
Add to at least 3 similar volumetric flasks equal volumes of down to 200 cm- 1 (50 !lm).
the solution of the substance to be examined (test solution) APPARATUS
prepared as prescribed. Add to al! but 1 of the flasks
Spectrophotometers for recording spectra consist of a suitable
progressively larger volumes of a reference solution containing
light source, monochromator or interferometer and detector.
a known concentration of the element to be determined to
produce a series of solutions containing steadily increasing Fourier transform spectrophotometers use polychromatic
concentrations of that element known to give responses in the radiation and calculate the spectrum in the frequency
linear part of the curve, if possible. Dilute the contents of each domain from the original data by Fourier transformation.
flask to volume with solvent. Spectrophotometers fitted with an optical system capable of
producing monochromatic radiation in the measurement
Introduce each of the solutions into the instrument, using the regio n may a1so be used. Normally the spectrum is given as
same number of replicates for each of the solutions, to obtain a function of transmittance, the quotient of the intensity of
a steady reading. the transmitted radiation and the incident radiation. It may
Calculation. Calculate the linear equation of the graph using also be given in absorban ce.
a least -squares fit and derive from it the concentration of the The absorbance (A) is defined as the logarithm to base 10 of
element to be determined in the test solution. the reciprocal of the transmittance (T):

VALIDATION OF THE METHOD A = 10glO (~ ) = 10glO ( ; )


Satisfactory performance of methods prescribed in
monographs is verified at suitable time intervals. 1
T
LINEARITY lo'
Prepare and analyse not fewer than 4 reference solutions over intensity of incident radiation,
the calibration range and a blank solution. Perform not fewer
than 5 replicates. intensity of transmitted radiation.

38 See the information section on general monographs (ca ver pages)


EUROPEAN PHARMACOPOEIA 8.0 2.2.24. Absorption spectrophotometry, infrared

PREPARATION OF THE SAMPLE FOR RECORDING BY DIFFUSE REFLECTANCE


FOR RECORDING BY TRANSMISSION OR ABSORPTION Solids. Triturate a mixture of the substance to be examined
Prepare the substance by one of the following methods. with finely powdered and dried potassium bromide R or
potassium chloride R. Use a mixture containing approximately
Liquicls. Examine a liquid either in the form of a film between
5 per cent of the substance, unless otherwise specified.
2 pIates transparent to infrared radiation, or in a cell of
Grind the mixture, place it in a sample cup and examine the
suitable path length, also transparent to infrared radiation.
reflectance spectrum.
Liquids or solids in solution. Prepare a solution in a suitable The spectrum of the sample in absorbance mode may be
solvent. Choose a concentration and a path length of the ceH obtained after mathematical treatment of the spectra by the
which give a satisfactory spectrum. Generally, good results are Kubelka-Munk function.
obtained with concentrations of 10-100 giL for a path length
of 0.5-0.1 mm. Absorption due to the solvent is compensated FOR RECORDING BY A TTENUATED TOTAL REFLECTION
by placing in the reference beam a similar cell containing the Attenuated total reflection (including multiple reflection)
solvent used. Jf an FT -IR instrument is used, the absorption is involves light being reflected internally by a transmitting
compensated by recording the spectra for the solvent and the medium, typically for a number of reflections. However,
sample successively. The solvent absorbance, corrected by a several accessories exist where only one reflection occurs.
compensation factor, is subtracted using calculation software. Prepare the substance as follows. Place the substance to be
examined in close COl1tact with an internal reflection element
Solids. Examine solids dispersed in a suitable liquid (mull) or (IRE) such as diamond, germanium, zinc selenide, thallium
in a solid (halide disc), as appropriate. If prescribed in the bromide-thallium iodide (KRS-5) or another suitable material
monograph, make a film of a molten mass between 2 plates of high refractive indexo Ensure close and uniform contact
transparent to infrared radiatíon. between the substance and the whole crystal surface of the
A. Mull internal reflection element, either by applying pressure 01'
Tríturate a small quantity of the substance to be examined by dissolving the substance in an appropriate solvent, then
with the mínimum quantityof ¡iquid paraffin R 01" other covering the IRE with the obtained solution and evaporating
suitable liquid; 5-10 mg ofthe substance to be examined is to dryness. Examine the attenuated total reflectance (ATR)
usually sufficient to make an adequate mull using one drop spectrum.
of ¡iquid paraffin R. Compress the mull between 2 plates
IDENTIFICATION USING REFERENCE SUBSTANCES
transparent to infrared radiation.
Prepare the substance to be examined and the reference
B. Disc
substance by the same procedure and record the spectra
Triturate 1-2 mg of the substance to be examined with between 4000-650 cm- 1 (2.5-15.4 flm) under the same
300-400 mg, unless otherwise specified, of finely powdered operational conditions. The transmission minima (absorption
and dried potassium bromide R or potassium chloride R. maxima) in the spectrum obtained with the substance to be
These quantities are usually sufficient to give a disc of examined correspond in position and relative size to those in
10-15 mm diameter and a spectrum of suitable intensity. If the spectrum obtained with the reference substance (CRS).
the substance is a hydrochloride, it is recommended to use
When the spectra recorded in the solid state show differences
potassium chloride R. Carefully grind the mixture, spread it
in the positions of the transmission minima (absorption
uniformly in a suitable die, and submit it to a pressure of maxima), treat the substance to be examined and the reference
about 800 MPa (8 t·cm- 2). For substances that are unstable
substance in the same manner so 'chat they crystallise or are
under normal atmospheric conditions or are hygroscopic, produced in the same form, or proceed as prescribed in the
the disc is pressed in vacuo. Several factors may cause the monograph, then record the spectra.
formation of faulty discs, such as insufficient or excessive
grinding, humidity or other impurities in the dispersion IDENTIFICATION USING REFERENCE SPECTRA
medium Ol" an insufficient reduction of particle size. A
Control of resolution performance. For instruments having
disc is rejected if visual examination shows lack of uniform
a monochromator, record the spectrum of a polystyrene film
transparency or when transmittance at about 2000 cm- 1
approximately 35 11m in thickness. The difference x (see
(5 11m) in the absence of a specific absorption band is less
Figure 2.2.24.-1) between the percentage transmittance at the
than 60 per cent without compensation, unless otherwise
transmission maximum A at 2870 cm- 1 (3.48 11m) and that at
prescribed.
the transmission minimum B at 2849.5 cm- 1 (3.51 flm) must
Gases. Examine gases in a cell transparent to infrared be greater than 18. The difference y between the percentage
radiation and having an optical path length of about 100 mm. transmittance at the transmission maximum C at 1589 cm- 1
Evacuate the ceH and fill to the desired pressure through a (6.29 flm) and that at the transmission minimum D at
stopcock or needle valve using a suitable gas transfer line 1583 cm- 1 (6.32 ~lm) must be greater than 10.
between the cel! and the container of the gas to be examined. For Fourier-transform instruments, use suitable instrument
If necessary adjust the pressure in the ceH to atmospheric resolution with the appropriate apodisation prescribed by the
pressure using a gas transparent to infrared radiation (for manufacturero The resolution is checked by suitable means,
example nitrogen R and argon R). To avoid absorption for example by recording the spectrum of a polystyrene film
interferences dne to water, carbon dioxide or other approximately 35 11m in thickness. The difference between the
atmospheric gases, place in the reference beam, if possible, absorbances at the absorption minimum at 2870 cm- 1 and
an identical cel! that is either evacuated or filled with the gas the absorption maximum at 2849.5 cm- 1 is greater than 0.33.
transparent to infrared radiation. The difference between the absorbances at the absorption

General Natices (1) apply ta all monographs and other texts 39


2.2.25. Absorption spectrophotometry, ultraviolet and visible EUROPEAN PHARMACOPOEIA 8.0

minimum at 1589 cm- 1 and the absorption maximum at acquired using suitable purged instruments or ensuring that
1583 cm- 1 is greater than 0.08. sample and background single beam spectra are acquired
under exactly the same conditions.

IMPURITIES IN GASES
For the analysis of impurities, use a cel! transparent to infrared
radiation and of suitable optical path length (for example,

" ~A
1-20 m). Fill the cell as prescribed under Gases. For detection
80 80 and quantification of the impurities, proceed as prescribed in
the monograph.
V N "/
60 60
e
0112008:20225
1
40 40 2.2.25. ABSORPTION
A
'. o SPECTROPHOTOMETRY,
ULTRAVIOLET AND VISIBLE
20
Determination of absorbance. The absorbance (A) of a
B solution is defilled as the logarithm to base 10 of the reciprocal
of the transmittance (T) for monochromatic radiation:
o'----"---"'-----' o
(~ )
3200 3000 2800 2600 1800 1600 1400
Wave number cm - 1 A = loglo = loglo ( ; )

T l/lo;
Figure 2.2.24.-1. - TYPical spectrum of polystyrene used to
verify the resolution performance intensity of incident monochromatic radiation;
Verification of the wave-number scale. The wave-number I intensity of transmitted monochromatic radiation.
scale may be verified using a polystyrene film, which has
transmission minima (absorption maxima) at the wave In the absence of other physico-chemical factors, the
numbers (in cm- 1) shown in Table 2.2.24.-1. absorbance (A) is proportional to the path length (b) through
which the radiation passes and to the concentration (e) of the
Table 2.2.24.-1. - Transmission minima and acceptable substance in solution in accordance with the equation:
tolerances of a polystyrene film
A = Ecb
Transmission Acceptable tolerance (cm- l )
mínima (cm- l ) molar absorptivity, if b is expressed in centimetres
and e in moles per litre.
Monochromaíor Fonrier-transform
instrnments insíruments The expression Ai
~: cent representing the specific absorbance
of a dissolved substance refers to the absorbance of a 10 giL
3060.0 ± 1.5 ± 1.0
solution in a 1 cm cell and measured at a defined wavelength
2849,5 ± 2.0 ± 1.0 so that:

1942.9 ± 1.5 ± 1.0


Al per cent _ 1010
1 cm - Mr
1601.2 ± 1.0 ± 1.0
Unless otherwise prescribed, measure the absorbance at the
1583.0 ± 1.0 ± 1.0 prescribed wavelength using a path length of 1 cm. Unless
otherwise prescribed, the measurements are carried out with
1154.5 ± 1.0 ± 1.0 reference to the same solvent or the same mixture of solvents.
1028.3 ± 1.0 ± 1.0
The absorbance of the solvent measured against air and at the
prescribed wavelength shall not exceed 0.4 and is preferably
Method. Prepare the substance to be examined less than 0.2. Plot the absorption spectrum with absorbance
according to the instructions accompanying the reference or function of absorbance as ordinate against wavelength or
spectrum/reference substance. Using the operating conditions function of wavelength as abscissa.
that were used to obtain the reference spectrum, which will Where a monograph gives a single value for the position of an
usually be the same as those for verifying the resolution absorption maximum, it is understood that the value obtained
performance, record the spectrum of the substance to be may differ by not more than ± 2 nm.
examined.
Apparatus. Spectrophotometers suitable for measuring in
The positions and the relative sizes of the bands in the the ultraviolet and visible range of the spectrum consist
spectrum of the substance to be examined and the reference of an optical system capable of producing monochromatic
spectrum are concordant in the 2 spectra. radiation in the range of 200-800 nm and a device suitable
Compensation for water vapour and atmospheric carbon for measuring the absorbance.
dioxide. For Fourier-transform instruments, spectral Control of wavelengths. Verify the wavelength scale using
interference from water vapour and carbon dioxide is the absorption maxima of holmium perchlorate solution R, the
compensated using suitable algorithms according to the hne of a hydrogen or deuterium discharge lamp or the lines of
manufacturer's instructions. Alternatively, spectra can be a mercury vapour are shown in Table 2.2.25,-1. The permitted

40 See the information section on general monographs (ca ver pages)


EUROPEAN PHARMACOPOEIA 8.0 2.2.25. Absorption spectrophotometry, ultraviolet and visible

tolerance is ± 1 nm for the ultraviolet range and ± 3 nm for censo The tolerance on the path length of the cells used is
the visible range. Suitable certified reference materials may ± 0.005 cm. When filled with the same solvent, the cells
also be used. intended to contain the solution to be examined and the
compensation liquid must have the same transmittance. lf this
is not the case, an appropriate correction must be applied.
Table 2.2.25.-1. - Absorption maxíma jor control oj wavelength
scale The cells must be cleaned and handled with careo

24l.l5 nm (Ha) 404.66 nm (Hg) DERlVATlVE SPECTROPHOTOMETRY


253.7 nm (Hg) 435.83 nm (Hg) Derivative spectrophotometry involves the transformation
of absorption spectra (zero-order) into first-, second- or
287.15nm (Ha) 486.0 nm (D~) higher-order-derivative spectra.
302.25 nm (Hg) 486.1 nm (H~) A jirst-order-derivatíve spectrum is a plot of the gradient of
536.3 nm (Ha)
the absorption curve (rate of change of the absorbance with
313.16 nm (Hg)
wavelength, dA/dA) against wavelength.
334.15 nm (Hg) 546.07 nm (Hg)
A second-order-derivative spectrum is a plot of the curvature
361.5 nm (Ha) 576.96 nm (Hg) ofthe absorption spectrum against wavelength (d 2AldA 2 ).
The second-order-derivative spectrum at any wavelength A is
365.48 nm (Hg) 579.07 nm (Hg)
related to concentration by the following equation:
Control of absorbance. Check the absorbance using d2A d 2 A 1p ercent c'b d 2 AE cb
1 crn X - == - - x _
suitable filters or a solution of potassium dichromate R at the d.\2 d.\2 10 d.\2 10
wavelengths indicated in Table 2.2.25.-2, which gives for each
wavelength the exact value and the permitted limits of the e/ concentration of the absorbing solute, in grams
specific absorbance. The table is based on a tolerance for the per litre.
absorbance of ± 0.01.
Apparatus. Use a spectrophotometer complying with the
For the control of absorbance, use solutions of potassium requirements prescribed aboye and equipped with an analogue
díchromate R that has been previously dried to constant mass resistance-capacitance differentiation module or a digital
at 130 oc. For the control of absorbance at 235 nm, 257 nm, differentiator or other means of producing derivative spectra.
313 nm and 350 nm, dissolve 57.0-63.0 mg of potassium Sorne methods of producing second-order-derivative spectra
dichromate R in 0.005 M sulfuric acíd and dilute to 1000.0 mL produce a wavelength shift relative to the zero-order spectrum
with the same acid. For the control of absorbance at 430 nm, and this is to be taken into account where applicable.
dissolve 57.0-63.0 mg of potassium dichromate R in 0.005 M
sulfuric acid and dilute to 100.0 mL with the same acid.
Suitable certified reference materials may also be used.

Table 2.2.25.-2

Wavelength Specific absorbance Maximum


(nm) Al per cent tolerance
1 cm

235 124.5 122.9 ta 126.2

257 144.5 142.8 ta 146.2


263
313 48.6 47.0 tú 50.3 - - -- - - - -

~
350 107.3 105.6 ta 109.0 + +
430 15.9 15.7 ta 16.1
/"'\ /1/\ a A
Limit of stray light. Stray light may be detected at a given
v V ~V \(1

wavelength with suitable filters or solutions: for example, the A


absorbance of a 12 giL solution of potassium chloride R in a - - - - -- - -
1 cm cel! increases steeply between 220 nm and 200 nm and 265
is greater than 2.0 at 198 nm when compared with water as
compensation liquido Suitable certified reference materials B
may also be used. - - - - - --
261
Resolution (for qllalitative analysis). When prescribed in
a monograph, measure the resolution of the apparatus as
follows: record the spectrum of a 0.02 per cent V/V solution of
toluene R in hexane R. The minimum ratio of the absorbance
at the maximum at 269 nm to that at the minimum at 266 nm 268
is stated in the monograph. Suitable certified reference I
materials may also be used. 240 260 280 290 nm
Spectral slit-width (for quantitative analysis). To avoid errors
due to spectral slit-width, when using an instrument on
which the slit-width is variable at the selected wavelength, the Figure 2.2.25.-1
slit-width must be small compared with the half-width of the Resolution power. When prescribed in a monograph, record
absorption band but it must be as large as possible to obtain the second-order-derivative spectrum of a 0.02 per cent V/V
a high value of ID. Therefore, a slit-width is chosen such that solution of toluene R in methanol R, using methanol R as the
further reduction does not result in a change in absorbance compensation liquido The spectrum shows a small negative
reading. extremum located between 2large negative extrema at 261 nm

General Notices (1) apply to all monographs and other texts 41


2.2.26. Paper chromatography EUROPEAN PHARMACOPOEIA 8.0

and 268 nm, respectively, as shown in Figure 2.2.25.-l. Unless portions, allowing each to dry before the next application.
otherwise prescribed in the monograph, the ratio AIB (see When more than one chromatogram is to be run on the same
Figure 2.2.25.-1) is not less than 0.2. strip of paper, space the solutions along the pencilline at
Procedure. Prepare the solution of the substance to be points not less than 3 cm apart. Insert the paper in the tank,
examined, adjust the various instrument settings according close the lid, and allow to stand for 1 h 30 mino Introduce into
to the manufacturer's instructions, and calculate the amount the solvent trough, through the hole in the lid, a sufficient
of the substance to be determined as prescribed in the quantity of the mobile phase, close the tank and allow elution
monograph. to proceed for the prescribed distance or time. Remove the
paper from the tank and allow to dry in airo The paper should
be protected from bright light during the elution process.
0112008:20226
0112008:20227
2.2.26. PAPER CHROMATOGRAPHY
ASCENDING PAPER CHROMATOGRAPHY
2.2.27. THIN-LAYER
Apparatus. The apparatus consists of a glass tank of suitable
CHROMATOGRAPHY
size for the chromatographic paper used, ground at the top Thin-Iayer chromatography is a separation technique in
to take a closely fitting lid. In the top of the tank is a device which a stationary phase consisting of an appropriate material
which suspends the chromatographic paper and is capable of is spread in a uniform thin layer on a support (plate) of
being lowered without opening the chamber. In the bottom of glass, metal or plastic. Solutions of analytes are deposited
the tank is a dish to contain the mobile phase into which the on the plate prior to development. The separation is based
paper may be lowered. The chromatographic paper consists on adsorption, partition, ion-exchange or on combinations
of suitable filter paper, cut into strips of sufficient length and of these mechanisms and is carried out by migration
not less than 2.5 cm wide; the paper is cut so that the mobile (development) of solutes (solutions of analytes) in a solvent
phase runs in the direction of the grain of the papero or a suitable mixture of solvents (mobile phase) through the
Method. Place in the dish a layer 2.5 cm deep of the mobile thin -layer (stationary phase).
phase prescribed in the monograph. If prescribed in the
monograph, pour the stationary phase between the walls of APPARATUS
the tank and the dish. Close the tank and allow to stand for PIates. The chromatography is carried out using pre-coated
24 h at 20 oC to 25 oc. Maintain the tank at this temperature plates as described under Reagents (4.1.1).
throughout the subsequent procedure. Draw a fine pencil Pre-treatment of the plates. It may be necessary to wash the
line horizontally across the paper 3 cm from one end. Using plates prior to separation. This can be done by migration of
a micro pipette, apply to a spot on the pencilline the volume an appropriate solvent. The plates may also be impregnated
of the solution prescribed in the monograph. lf the total by pro ce dures su eh as development, immersion or spraying.
volume to be applied would produce a spot more than 10 mm At the time of use, the pI ates may be activated, if necessary, by
in diameter, apply the solution in portions allowing each heating in an oven at 120 oC for 20 mino
to dry before the next application. When more than one
Chromatographic tank with a flat bottom or twil1 trough,
chromatogram is to be run on the same strip of paper, space
of inert, transparent material, of a size suitable for the plates
the solutions along the pencilline at points not less than 3 cm
used and provided with a tightly fitting lid. For horizontal
apart. Insert the paper into the tank, close the lid and allow to
development the tank is provided with a trough for the mobile
stand for 1 h 30 mino Lower the paper into the mobile phase
phase and it additionally contains a device for directing the
and allow elution to proceed for the prescribed distan ce or
mobile phase to the stationary phase.
time. Remove the paper from the tank and allow to dry in airo
Protect the paper from bright light during the elution process. Micropipettes, microsyringes, calibrated disposable
capmaries or other application devices suitable for the proper
DESCENDING PAPER CHROMATOGRAPHY application of the solutions.
Apparatus. The apparatus consists of a glass tank of suitable Fluorescence detection device to measure direet fluorescence
size for the chromatographic paper used, ground at the top or the inhibition of fluorescence.
to take a closely fitting glass lid. The lid has a central hole
about 1.5 cm in diameter closed by a heavy glass plate or a Visualisation devices and reagents. Suitable devices are used
stopper. In the upper part of the tank is suspended a solvent for derivatisation to transfer to the pI ate reagents by spraying,
trough with a device for holding the chromatographic papero immersion or exposure to vapour and, where applicable, to
On each si de of the trough, parallel to and slightly aboye its facilitate heating for visualisation of separated components.
upper edges, are two glass guide rods to support the paper Documentation. A device may be used to provide
in such a manner that no part of it is in contact with the documentation of the visualised chromatogram, for example a
walls of the tank. The chromatographic paper consists of photograph or a computer file.
suitable filter paper, cut into strip s of sufficient length, and of
any convenient width between 2.5 cm and the length of the METHOD
trough; the paper is cut so that the mobile phase runs in the Sample application. Apply the prescribed volume of the
direction of the gl'ain of the papero solutions at a suitable distan ce from the lower edge and
Method. Place in the bottom of the tank a layer 2.5 cm deep of from the sides of the plate and on a line parallel to the
the solvent prescribed in the monograph, close the tan k and lower edge; allow an interval of at least 10 mm (5 mm on
allow to stand fol' 24 h at 20 oC to 25 oc. Maintain the tank high-performance plates) between the centres of circular
at this temperature throughout the subsequent procedure. spots and 5 mm (2 mm on high-performance plates) between
Draw a fine pencilline horizontally across the paper at such the edges of bands. Apply the solutions in sufficiently small
a distance from one end that when this end is secured in the portions to obtain circular spots 2-5 mm in diameter (1-2 mm
solvent trough and the remainder of the paper is hanging 011 high -performance plates) or bands 10-20 mm (5-10 mm

freely over the guide rod, the line is a few centimetres below on high-performance plates) by 1-2 mm.
the guide rod and parallel with it. Using a micro-pipette, apply In a monograph, where both normal and high-performance
on the pencilline the volume of the solution prescribed in the plates may be used, the working conditions for
monograph. lf the total volume to be applied would produce high-performance plates are given in the brackets [ 1after
a spot more than 10 mm in diameter, apply the solution in those for normal plates.

42 See the informatíon sectíon on general monographs (ca ver pages)


EUROPEAN PHARMACOPOEIA 8.0 2.2.28. Gas chromatography

Vertical development. Line the walls of the chromatographic Substances separated by thin-layer chromatography and
tank with filter papero Pour into the chromatographic tank responding to UV -Vis irradiation can be determined directly
a sufficient quantity of the mobile phase for the size of the on the plate, using appropriate instrumentation. While
tank to give after impregnation of the filter paper a layer of moving the plate or the measuring device, examine the plate
appropriate depth related to the dimension of the plate to be by measuring the reÍlectance of the incident light. Similarly,
used. For saturation of the chromatographic tank, replace the fluorescence may be measured using an appropriate optical
lid and allow to stand at 20-25 oC for 1 h. Unless otherwise system. Substances containing radionuclides can be quantified
indicated in the monograph, the chromatographic separatíon in 3 ways: either directly by moving the plate alongside a
is performed in a saturated tank. Apply the prescribed suitable counter or vice versa (see Radiopharmaceutical
volume of solutions as described aboye. When the solvent preparations (0125)), by cutting the plates into strip s and
has evaporated from the applied solutions, place the plate measuring the radioactivity on each individual strip using
in the chromatographic tank, ensuring that the pIate is as a suitable counter or by scraping off the stationary phase,
vertical as possible and that the spots or bands are aboye the dissolving it in a suitable scintillation cocktail and measuring
surface of the mobile phase. Close the chromatographic tank, the radioactivity using a liquid scintillation counter.
maintain it at 20-25 oC and protect from sunlight. Remove the Apparatus. The apparatus for direct measurement on the
plate when the mobile phase has moved over the prescribed plate consists of:
distance, measured between the points of application and the
solvent front. Dry the plate and visualise the chromatograms - a device for exact positioning and reproducible dispensing
as prescribed. of the amount of substances onto the plate;
For two-dimensional chromatography, dry the pIates after the - a mechanical device to move the plate or the measuring
first development and carry out a second development in a device along the x-axis or the y-axis;
direction perpendicular to that of the first development. - a recorder and a suitable integrator or a computer;
Horizontal development. Apply the prescribed volume - for substances responding to UV- Vis irradiation: a
of the solutions as described aboye. When the solvent has photometer with a source of light, an optical device able to
evaporated from the applied solutions, introduce a sufficient generate monochromatic light and a photo cell of adequate
quantity of the mobile phase into the trough of the chamber sensitivity are used for the measurement of reflectance
using a syrínge or pipette, place the plate in the chamber or transmittance; if fluorescence is measured, a suitable
after verifying that the latter is horizontal and connect the filter is required to prevent light used for excitation from
mobile phase direction device according to the manufacturer's reaching the detector while permitting emitted light or a
instructions. rf prescribed, develop the plate starting specific portion thereof to pass;
simultaneously at both ends. Close the chamber and maintain - for substances containing radionuclides: a suitable counter
it at 20-25 oc. Remove the plate when the mobile phase has for radioactivity. The linearity range of the counting device
moved over the distance prescribed in the monograph. Dry is to be verified.
the plate and visualise the chromatograms as prescribed. Method. Prepare the solution of the substance to be examined
Por two-dimensional chromatography, dry the plates after the (test solution) as prescribed in the monograph and, if
first development and carry out a second development in a necessary, prepare the reference solutions of the substance to
direction perpendicular to that of the first development. be determined using the same solvent as in the test solution.
Apply the same volume of each solution to the plate and
VISUAL EVALUATION
develop.
Identiflcation. The principal spot in the chromatogram Substances responding to UV- Vis irradiation. Prepare and
obtained with the test solution is visualIy compared to the apply not fewer than 3 reference solutions of the substance to
corresponding spot in the chromatogram obtained with the be examined, the concentrations of which span the expected
reference solution by comparing the colour, the size and the value in the test solution (about 80 per cent, 100 per cent and
retardation factor (R p ) ofboth spots. 120 per cent). Treat with the prescribed reagent, if necessary,
The retardation factor (R p ) is defined as the ratio of the and record the reflectance, the transmittance or fluorescence
distance from the point of application to the centre of the spot in the chromatograms obtained with the test and reference
and the distance travelled by the solvent front from the point solutions. Use the measured results for the calculation of the
of application. amount of substance in the test solution.
Verificatíon of the separating power for identification. Substances containing radionuclides. Prepare and apply a
Normally the performance given by the suitability test test solution containing about 100 per cent of the expected
described in Reagents (4.1.1) is sufficient. Only in special value. Determine the radioactivity as a function of the path
cases an additional performance criterion is prescribed in the length and report the radioactivity in each resulting peak as a
monograph. percentage of the total amount of radioactivity.
Related substances test. The secondary spot(s) in the Criteria for assessing the suitability of the system are described
chromatogram obtained with the test solution is (are) in the chapter on Chromatographic separation techniques
visually compared to either the corresponding spot(s) in (2.2.46). The extent to which adjustments of parameters of the
the chromatogram obtained with the reference solution chromatographic system can be made to satisfy the criteria of
containing the impurity(ies) or the spot in the chromatogram system suitability are also given in this chapter.
obtained with the reference solution prepared from a dilution
of the test solution.
Verification of the separatíng power. The requirements for 0112008:20228
the verification of the separating power are prescribed in the
monographs con cerned. 2.2.28. GAS CHROMATOGRAPHY
Verification of the detecting power. The detecting power
is satisfactory if a spot or band is clearly visible in the Gas chromatography (GC) is a chromatographic separation
chromatogram obtained with the most dilute reference technique based on the difference in the distribution of species
solution. between two non-miscible phases in which the mobile phase
is a carrier gas moving through or passing the stationary
QUANTITATIVE MEASUREMENT phase contained in a column. It is applicable to substances OI
The requirements for resolution and separation are prescribed their derivatives which are volatilised under the temperatures
in the monographs concerned. employed.

General Notices (1) apply to all monographs and other texts 43


2.2.28. Gas chromatography EUROPEAN PHARMACOPOEIA 8.0

GC is based on mechanisms of adsorption, mass distribution Helium or nitrogen are usually employed as the carrier gas for
or size exdusion. packed columns, whereas commonly used carrier gases for
capillary columns are nitro gen, helium and hydrogen.
APPARATUS
DETECTORS
The apparatus consists of an injector, a chromatographic
column contained in an oven, a detector and a data acquisition Flame-ionisation detectors are usually employed but additional
system (or an integrator or a chart recorder). The carrier gas detectors which may be used indude: electron-capture,
flows through the column at a controlled rate or pressure and nitrogen -phosphorus, mass spectrometric, thermal
conductivity, Fourier transform infrared spectrophotometric,
then through the detector.
and others, depending on the purpose of the analysis.
The chromatography is carried out either at a constant
temperature or according to a given temperature programme. METHOD
INJECTORS Equilibrate the column, the injector and the detector at
Direct injections of solutions are the usual mode of injection, the temperatures and the gas flow rates specified in the
unless otherwise prescribed in the monograph. Injection may monograph until a stable baseline is achieved. Prepare the test
be carried out either directly at the head of the column using a solution(s) and the reference solution(s) as prescribed. The
syringe or an injection valve, or into a vaporisation chamber solutions must be free from solid particles.
which may be equipped with a stream splitter. Criteria for assessing the suitability of the system are described
Injections of vapour phase may be effected by static or dynamic in the chapter on Chromatographic separation techniques
head-space injection systems. (2.2.46). The extent to which adjustments of parameters of the
Dynamic head-space (purge and trap) injection systems chromatographic system can be made to satisfy the criteria of
indude a sparging device by which volatile substances in system suitability are also given in this chapter.
solution are swept into an absorbent column maintained at a
low temperature. Retained substances are then desorbed into Static head -space gas chromatography
the mobile phase by rapid heating of the absorbent column.
Static head-space gas chromatography is a technique
Static head-space injection systems in dude a thermostatically particularly suitable for separating and determining volatile
controlled sample heating chamber in which dosed vials compounds present in solid or liquid samples. The method
containing solid or liquid samples are placed for a fixed is based on the analysis of the vapour phase in equilibrium
period of time to allow the volatile components of the sample with the solid or liquid phase.
to reach equilibrium between the non-gaseous phase and
the vapour phase. After equilibrium has been established, a APPARATUS
predetermined amount of the head-space of the vial is flushed The apparatus consists of a gas chromatograph provided with
into the gas chromatograph. a device for introducing the sample that may be connected
STATIONARY PHASES to a module that automatically controls the pressure and the
Stationary phases are contained in columns which may be: temperature. lf necessary, a device for eliminating solvents
- a capillary column of fused-silica whose wall is coated with can be added.
the stationary phase, The sample to be analysed is introduced into a container fitted
- a column packed with inert partides impregnated with the with a suitable stopper and a valve-system which permits
stationary phase, the passage of the carrier gas. The container is placed in
a thermostatically controHed chamber at a temperature set
- a column packed with solid stationary phase. according to the substance to be examined.
Capillary columns are 0.1 mm to 0.53 mm in internal diameter The sample is held at this temperature long enough to allow
(0) and 5 m to 60 m in length. The liquid or stationary phase,
equilibrium to be established between the solid or liquid phase
which may be chemically bonded to the inner surface, is a and the vapour phase.
film 0.1 flm to 5.0 flm thick.
The carrier gas is introduced into the container and, after
Packed columns, made of glass or metal, are usually 1 m to the prescribed time, a suitable valve is opened so that the gas
3 m in length with an internal diameter (0) of 2 mm to 4 mm. expands towards the chromatographic column taking the
Stationary phases usually consist of porous polymers or solid volatilised compounds with it.
supports impregnated with liquid phase.
Instead of using a chromatograph specifically equipped for
Supports for analysis of polar compounds on columns packed
the introduction of samples, it is also possible to use airtight
with low-capacity, low-polarity stationary phase must be inert
syringes and a conventional chromatograph. Equilibration is
to avoid peak tailing. The reactivity of support materials can then carried out in a separate chamber and the vapour phase
be reduced by silanising prior to coating with liquid phase.
is carried onto the column, taking the precautions necessary
Acid-washed, flux-calcinated diatomaceous earth is often
to avoid any changes in the equilibrium.
used. Materials are available in various partide sizes, the
most commonly used partides are in the ranges of 150 flm to METHOD
180 ~lm and 125 flm to 150 flm. Using the reference preparations, determine suitable
MOBILE PHASES instrument settings to produce an adequate response.
Retention time and peak efficiency depend on the carrier gas DIRECT CALIBRATION
flow rate; retention time is directly proportional to column Separately introduce into identical container s the preparation
length and resolution is proportional to the square root of the to be examined and each of the reference preparations, as
column length. For packed columns, the carrier gas flow rate prescribed in the monograph, avoiding contact between the
is usually expressed in millilitres per minute at atmospheric sampling device and the samples.
pressure and room temperature. Flow rate is measured at the
Close the containers hermetically and place in the
detector outlet, either with a calibrated mechanical device
thermostatically controlled chamber set to the temperature
or with a bubble tube, while the column is at operating
and pressure prescribed in the monograph; after equilibration,
temperature. The linear velocity of the carrier gas through a
carry out the chromatography under the prescribed conditions.
packed column is inversely proportional to the square root of
the internal djameter of the column for a given flow volume. STANDARD ADDITIONS
Flow rates of 60 mLlmin in a 4 mm internal diameter column Add to a set of identical suitable containers equal volumes
and 15 mLlmin in a 2 mm internal diameter column, give of the preparation to be examined. Add to aH but one of
identicallinear velocities and thus similar retention times. the containers, suitable quantities of a reference preparation

44 See the information section on general monographs (cover pages)


EUROPEAN PHARMACOPOEIA 8.0 2.2.29. Líquid chromatography

containing a known concentration of the substance to - resins OI polymers with acid or basic groups, used in
be determined so as to produce a series of preparations ion-exchange chromatography, where separation is based
containing steadily increasing concentrations of the substance. on competition between the ions to be separated and those
Close the containers hermetically and place in the in the mobile phase,
thermostatically controlled chamber set to the temperature - porous silica or polymers, used in size-exclusion
and pressure prescribed in the monograph; after equilibration, chromatography, where separation is based on differences
carry out the chromatography under the prescribed conditions. between the volumes of the molecules, corresponding to
Calculate the linear equation of the graph using a least-squares steric exclusion,
fit, and derive from it the concentration of the substance to be - a variety of chemically modified supports prepared from
determined in the preparation to be examined. polymers, silica or porous graphite, used in reversed-phase
Alternatively, plot on a graph the mean of readings against the LC, where the separation is based principally on partition
added quantity of the substance to be determined. Extrapolate of the molecules between the mobile phase and the
the line joining the points on the graph until it meets the stationary phase,
concentration axis. The distance between this point and the - special chemically modified stationary phases, e.g.
intersection of the axes represents the concentration of the cellulose or amylose derivatives, proteins or peptides,
substance to be determined in the preparation to be examined. cyclodextrins etc., for the separation of enantiomers (chiral
SUCCESSIVE WITHDRAWALS (MULTIPLE HEAD-SPACE chromatography).
EXTRACTION) Most separations are based upon partition mechanisms
If prescribed, the successive withdrawal method is fully utilising chemically modified silica as the stationary phase
described in the monograph. and polar solvents as the mobile phase. The surface of the
support, e.g. the silanol groups of silica, is reacted with various
silane reagents to produce covalently bound silyl derivatives
01/2008:20229 covering a varying number of active sites on the surface of
the support. The nature of the bonded phase is an important
2.2.29. LIQUID CHROMATOGRAPHY parameter for determining the separation properties of the
chromatographic system.
Liquid chromatography (LC) is a method of chromatographic Commonly used bonded phases are shown below:
separation based on the difference in the distribution of octyl = Si-[CH 217 -CH 3 Cs
species between two non-miscible phases, in which the mobile
phase is a liquid which percolates through a stationary phase octadecyl = Si -[CH2L7-CH3
contained in a column. phenyl = Si-[CH 2 L-C 6 H s
LC is mainly based on mechanisms of adsorption, mass
distribution, ion exchange, size exclusion or stereochemical cyanopropyl = Si-[CH 2 kCN CN
interaction. aminopropyl = Si-[CH 2 kNH 2
APPARATUS diol = Si-[CH 2 k O -CH(OH)-CH 2 -
The apparatus consists of a pumping system, an injector, a OH
chromatographic column (a column temperature controller Unless otherwise stated by the manufacturer, silica based
may be used), a detector and a data acquisition system (or an reversed-phase columns are considered to be stable in mobile
integrator or a chart recorder). The mobile phase is supplied phases having an apparent pH in the range 2.0 to 8.0. Columns
from one or several reservoirs and flows through the column, containing porous graphite or particles of polymeric materials
usually at a constant rate, and then through the detector. such as styrene-divinylbenzene copolymer are stable over a
PUMPING SYSTEMS wider pH range.
LC pumping systems are required to deliver the mobile Analysis using normal-phase chromatography with
phase at a constant flow rateo Pressure fluctuations are to be unmodified silica, porous graphite or polar chemically
minimised, e.g. by passing the pressurised solvent through a modified silica, e.g. cyanopropyl or diol, as the stationary
pulse-dampening device. Tubing and connections are capable phase with a non-polar mobile phase is applicable in certain
of withstanding the pressures developed by the pumping cases.
system. LC pumps may be fitted with a facility for "bleeding" For analytical separations, the particle size of the most
the system of entrapped air bubbles. commonly used stationary phases varies between 3 11m
Microprocessor controlled systems are capable of accurately and 10 11m. The particles may be spherical or irregular, of
delivering a mobile phase of either constant (isocratic elution) varying porosity and specific surface area. These parameters
or varying composition (gradient elution), according to a contribute to the chromatographic behaviour of a particular
defined programme. In the case of gradient elution, pumping stationary phase. In the case of reversed phases, the nature
systems which deliver solvent(s) from several reservoirs are of the stationary phase, the extent of bonding, e.g. expressed
available and solvent mixing can be achieved on either the low as the carbon loading, and whether the stationary phase is
or high-pressure side of the pump(s). end-capped (i.e. residual silanol groups are silylated) are
INJECTORS
additional determining factors. Tailing of peaks, particularly
of basic substances, can occur when residual silanol groups
The sample solution is introduced into the flowing mobile
are present.
phase at or near the head of the column using an injection
system which can operate at high pressure. Fixed-loop Columns, made of stainless steel unless otherwise prescribed
and variable volume devices operated manually or by an in the monograph, of varying length and internal diameter
auto-sampler are used. Manual partial filling of loops may (0) are used for analytical chromatography. Columns with
lead to poorer injection volume precision. internal diameters of less than 2 mm are often referred to as
microbore columns. The temperature of the mobile phase and
STATIONARY PHASES the column must be kept constant during an analysis. Most
There are many types of stationary phases employed in LC, separations are performed at room temperature, but columns
including: may be heated to give higher efficiency. It is recommended that
- silica, alumina or porous graphite, used in normal-phase columns not be heated aboye 60 oC because of the potential
chromatography, where the separation is based on for stationary phase degradation or changes occurring to the
differences in adsorption and/or mass distribution, composition of the mobile phase.

General Notices (1) apply to all monographs and other texts 45


2.2.30. Size-exdusion chromatography EUROPEAN PHARMACOPOEIA 8.0

MOBILE PHASES Molecules small enough to penetrate all the pore spaces elute
For normal-phase chromatography, less polar solvents are at the total permeation volume (Vt ). 011 the other hand,
employed. The presence of water in the mobile phase is molecules apparently larger than the maximum pore size of
to be strictly controlled to obtain reproducible results. In the packing material migrate along the column only through
reversed-phase Le, aqueous mobile phases, with or without the spaces between the particles of the packing material
organic modifiers, are employed. without being retained al1d elute at the exclusion volume
Components of the mobile phase are usually filtered to remove (Va void volume). Separation according to molecular size
particles greater than 0.45 flm. Multicomponent mobile phases occurs between the exclusion volume and the total permeation
are prepared by measuring the required volumes (unless volume, with useful separation usually occurring in the first
masses are specified) of the individual components, followed two thirds of this range.
by mixing. Alternatively, the solvents may be delivered by
Appa ra tus. The apparatus consists essentially of a
individual pumps controlled by proportioning val ves by which
chromatographic column of varying length and internal
mixing is performed according to the desired proportion.
diameter (0), if necessary temperature-controlled, packed
Solvents are normally degassed before pumping by sparging
with a separation material that is capable of fractionation in
with helium, sonication or using on-line membrane/vacuum
the appropriate range of molecular sizes and through which
modules to avoid the creation of gas bubbles in the detector
the eluent is passed at a constant rateo One end of the column
cell.
is usually fitted with a suitable device for applying the sample
Solvents for the preparation of the mobile phase are normally such as a flow adapter, a syringe through a septum or an
free of stabilisers and are transparent at the wavelength of injection valve and may also be connected to a suitable pump
detection, if an ultraviolet detector is employed. Solvents and for controlling the flow of the eluent. Alternatively the sample
other components employed are to be of appropriate quality. may be applied directly to the drained bed surface Of, where
Adjustment of the pH, if necessary, is effected using only the the sample is denser than the eluent, it may be layered beneath
aqueous component of the mobile phase and not the mixture. the eluent. The outlet of the column is usually connected
If buffer solutions are used, adequate rinsing of the system is to a suitable detector fitted with an automatic recorder
carried out with a mixture of water and the organic modifier which enables the monitoring of the relative concentrations
of the mobile phase (5 per cent V/V) to prevent crystallisation of separated components of the sample. Detectors are
of salts after completion of the chromatography. usual!y based on photometric, refractometric or luminescent
Mobile phases may contain other components, e.g. a properties. An automatic fraction collector may be attached, if
counter-ion for ion-pair chromatography or a chiral selector necessary.
for chromatography using an achiral stationary phase.
DETECTORS The packing material may be a soft support such as a swollen
gel or a rigid support composed of a material such as glass,
Ultraviolet/visible (UVlVis) spectrophotometers, including
silica or a solvent-compatible, cross-linked organic polymer.
diode array detector s, are the most commonly employed
Rigid supports usually require pressurised systems giving
detectors. Fluorescence spectrophotometers, differential
faster separations. The mobile phase is chosen according to
refractometers, electrochemical detectors, mass spectrometers,
sample type, separation medium and method of detection.
light scattering detectors, radioactivity detectors or other
Before carrying out the separation, the packing material
special detector s may also be used.
is treated, and the column is packed, as described in the
METHOD monograph, or according to the manufacturer's instructions.
Equilibrate the column with the prescribed mobile phase
Criteria for assessing the suitability of the system are described
and flow rate, at room temperature or at the temperature in the chapter on Chramatographic separatian techniques
specified in the monograph, until a stable baseline is achieved. (2.2.46). The extent to which adjustments of parameters of the
Prepare the solution(s) of the substance to be examined and
chromatographic system can be made to satisfy the criteria of
the reference solution(s) required. The solutions must be free system suitability are also given in this chapter.
from solid particles.
Criteria for assessing the suitability of the system are described DETERMINATION OF RELATIVE COMPONENT
in the chapter on Chromatographic separation techniques COMPOSITION OF MIXTURES
(2.2.46). The extent to which adjustments of parameters of the Carry out the separatiol1 as stated in the monograph. If
chromatographic system can be made to satisfy the criteria of possible, monitor the elution of the components continuously
system suitability are also given in this chapter. and measure the corresponding peak areas. If the sample
is monitored by a physico-chemical property to which al!
the components of interest exhibit equivalent responses (for
0112008:20230 example if they have the same specific absorbance), calculate
the relative amount of each component by dividing the
respective peak are a by the sum of the peak areas of al! the
2.2.30. SIZE-EXCLUSION components of interest. If the responses to the property used
CHROMATOGRAPHY for detection of the components of interest are not equivalent,
calculate the content by means of calibration curves obtained
Size-exclusion chromatography is a chromatographic with the calibration standards prescribed in the monograph.
technique which separates molecules in solution according
to their size. With organic mobile phases, the technique is DETERMINATION OF MOLECULAR MASSES
known as gel-permeation chromatography and with aqueous Size-exclusion chromatography may be used to determine
mobile phases, the term gel-filtration chromatography has molecular mas ses by comparison with appropriate calibration
been used. The sample is introduced into a column, which is standards specified in the monograph. The retentiol1
filled with a gel or a porous particle packing material, and is volumes of the calibration standards may be plotted against
carried by the mobile phase through the column. The size the logarithm of their molecular masses. The plot usually
separation takes place by repeated exchange of the solute approximates a straight line within the exclusion and total
molecules between the solvent of the mobile phase and the permeation limits for the separation medium used. From the
same solvent in the stagnant liquid phase (stationary phase) calibration curve, molecular masses may be estimated. The
within the pores of the packing material. The pore-size range molecular-mass calibration is valid only for the particular
of the packing material determines the molecular-size range macromolecular solute/solvent system used under the
within which separation can occur. specified experimental conditions.

46 See the information section 011 general manographs (ca ver pages)
EUROPEAN PHARMACOPOEIA 8.0 2.231. Electrophoresis

DETERMINATION OF MOLECULAR SIZE DISTRIBUTION - an electrophoresis chamber. This is usually rectangular


OF POLYMERS and made of glass or rigid plastic, with two separate
Size-exclusion chromatography may be used to determine compartments, the anodic and the cathodic, containing the
the distribution of the molecular size of polymers. However, electrolyte solution. In each compartment is immersed an
sample camparison may be valid anly far results obtained electro de, for example of platinum or graphite. These are
under the same experimental conditions. The reference connected by means of an appropriately isolated circuit to
substances used for the calibration and the methods for the corresponding terminal of the power suppIy to form
determination of the distribution of molecular sizes of the anode and the cathode. The level of the liquid in the
polymers are specified in the monograph. two compartments is kept equal to prevent siphoning.
The electrophoresis chamber is fitted with an airtight lid
which maintains a moisture-saturated atmosphere during
operation and reduces evaporation of the solvent. A safety
0112010:20231
device may be used to cut off the power when the lid is
removed. lf the electrical power measured across the strip
2.2.31. ELECTROPHORESIS(2) exceeds 10 W, it is preferable to cool the support.
- a support-carrying device:
• GENERAL PRINCIPLE
Under the influence of an electrical field, charged particles Strip electrophoresis. The supporting strip, previously
dissolved or dispersed in an electrolyte solution migrate in wetted with the same conducting solution and dipped at
the direction of the electrode bearing the opposite polarity. each end into an electrode compartment is appropriately
In gel electrophoresis, the movements of the particles are tightened and fixed on to a suitable carrier designed to
retarded by interactions with the surrounding gel matrix, prevent diffusion of the conducting electrolyte, such as a
which acts as a molecular sieve. The opposing interactions of horizontal frame, inverted -V stand or a uniform surface
the electrical force and molecular sieving result in differential with contact points at suitable intervals.
migration rates according to sizes, shapes and charges Gel electrophoresis. The device consists essentially of a
of particles. Because of their different physico-chemical glass plate (for example, a microscope slide) over the whole
properties, different macromolecules of a mixture will migrate surface of which is deposited a firmly adhering layer of
at different speeds during electrophoresis and will thus be gel of uniform thickness. The connection between the gel
separated into discrete fractions. Electrophoretic separations and the conducting solution is effected in various ways
can be conducted in systems without support phases (e.g. according to the type of apparatus used. Precautions must
free solution separation in capillary electrophoresis) and in be taken to avoid condensation of moisture OI drying of
stabilising media such as thin -layer plates, film s or gels. the solid layer.
- measuring device or means of deteetion.
FREE OR MOVING BOUNDARY ELECTROPHORESIS
Method. Introduce the electrolyte solution into the electrode
This method is mainly used for the determination of mobility, compartments. Place the support suitably impregnated with
the experimental characteristics being directly measurable and electrolyte solution in the chamber under the conditions
reproducible. It is chiefly employed with substances ofhigh prescribed for the type of apparatus used. Locate the starting
relative molecular mass and low diffusibility. The boundaries line and apply the sample. Apply the electric current for the
are initially located by a physical process such as refractometry prescribed time. After the current has been switched off,
or conductimetry. After applying a given electric field for remove the support from the chamber, dry and visualise.
an accurately measured time, the new boundaries and their
respective positions are observed. The operating conditions POLYACRYLAMIDE ROD GEL ELECTROPHORESIS
must be such as to make it possible to determine as many In polyacrylamide rod gel electrophoresis, the stationary
boundaries as there are components. phase is a gel which is prepared from a mixture of acrylamide
ZONE ELECTROPHORESIS USING A SUPPORTING and N,N"-methylenebisacrylamide. Rod gels are prepared
MEDIUM in tubes 7.5 cm long and 0.5 cm in internal diameter, one
solution being applied to each rod.
This method requires the use of small samples only.
Apparatus. This consists of two buffer solution reservoirs
The nature of the support, such as paper, agar gel, cellulose made of suitable material such as poly(methyl methacrylate)
acetate, starch, agarose, methacrylamide, mixed gel, introduces and mounted vertically one aboye the other. Each reservoir is
a number of additional factors modifying the mobility: fitted with a platinum electro de. The electro des are connected
a) owing to channelling in the supporting medium, the to a power supply allowing operation either at constant
apparent distance covered is less than the real distance, current or at constant voItage. The apparatus has in the base
b) some supporting media are not electrically neutral. As the of the upper reservoir a number of holder s equidistant from
medium is a stationary phase it may sometimes give rise to the electro de.
a considerable electro-endosmotic flow, Method. The solutions should usually be degassed before
e) any heating due to the joule effect may cause so me polymerisation and the gels used immediately after
evaporation of the liquid from the supporting medium preparation. Prepare the gel mixture as prescribed and pour
which, by cap ill arity, causes the solution to move from the into suitable glass tubes, stoppered at the bottom, to an equal
ends towards the centre. The ionic strength therefore tends height in each tube and to about 1 cm from the top, taking
to increase gradually. care to ensure that no air bubbles are trapped in the tubes.
Cover the gel mixture with a layer of water R to exclude air
The rate of migration then depends on four main factors:
and allow to set. Gel formation usually takes about 30 min
the mobility of the charged particle, the electro-endosmotic
and is complete when a sharp interface appears between the
flow, the evaporation flow, and the field strength. Hence it
gel and the water layer. Remove the water layer. Fill the lower
is necessary to operate under clearly defined experimental
reservoir with the prescribed buffer solution and remove the
conditions and to use, wherever possible, reference substances.
stoppers from the tubes. Fit the tubes into the holders of the
An apparatus for electrophoresis consists of: upper reservoir and adjust so that the bottom of the tubes
- a generator supplying direct curren! whose voltage can be are immersed in the buffer solution in the lower reservo ir.
controlled and, preferably, stabilised, Carefully fill the tubes with the prescribed buffer solution.

(2) This chapter has undergone pharmacopoeial harmonis3.tion. See chapter 5.8. Pharmacopoeial harmonisation.

General Notices (1) apply to all monographs and other texts 47


2.2.3 L Electrophoresis EUROPEAN PHARMACOPOEIA 8.0

Prepare the test and reference solutions containing the subunits and that minimise aggregation. Most commonly, the
prescribed marker dye and make them dense by dissolving strongly anionic detergent sodium dodecyl sulfate (SDS) is
in them sucrose R, for example. AppIy the solutions to the used in combination with heat to dissociate the proteins before
surface of a gel using a different tube for each solution. Add they are loaded on the gel. The denatured polypeptides bind
the same buffer to the upper reservoir. Connect the electro des to SDS, become negatively charged and exhibit a consistent
to the power suppIy and allow electrophoresis to proceed charge-to-mass ratio regardless of protein type. Because the
at the prescribed temperature and using the prescribed amount of SDS bound is almost always proportional to the
constant voltage or current. Switch off the power suppIy molecular mass of the polypeptide and is independent of
when the marker dye has migrated almost into the lower its sequence, SDS-polypeptide complexes migrate through
reservoir. Immediately remove each tube from the apparatus polyacrylamide gels with mobilities dependent 011 the size of
and extrude the gel. Locate the position of the bands in the the polypeptide.
electropherogram· as prescribed. + The electrophoretic mobilities of the resultant detergent-
SODIUM DODECYL SULFATE POLYACRYLAMIDE GEL polypeptide complexes all assume the same functional
ELECTROPHORESIS (SDS-PAGE) relationship to their molecular masses. Migration of SDS
complexes is toward the anode in a predictabIe manner, with
Scope. Polyacrylamide gel electrophoresis is used for Iow-molecular-mass complexes migrating faster than larger
the qualitative characterisation of proteins in biological ones. The molecular mass of a protein can therefore be
preparations, for control of purity and quantitative estimated from its relative mobility in calibrated SDS-PAGE
determinations. and the occurrence of a single band in such a gel is a criterion
Purpose. Analytical gel electrophoresis is an appropriate ofpurity.
method with which to identify and to assess the homogeneity Modifications to the polypeptide backbone, such as N- or
of proteins in pharmaceutical preparations. The method is O-linked glycosylation, however, have a significant impact
routinely used for the estimation of protein subunit molecular on the apparent molecular mass of a protein since SDS does
masses and for determining the subunit compositions of not bind to a carbohydrate moiety in a manner similar to a
purified proteins. polypeptide. Thus, a consistent charge-to-mass ratio is not
Ready-to-use gels and reagents are widely available on the maintained. The apparent molecular mass of proteins having
market and can be used instead of those described in this undergone post -translational modifications is 110t a true
text, provided that they give equivalent results and that they reflection of the mass of the polypeptide chain.
meet the validity requirements given below under Validation
of the test. Reducing conditions. Polypeptide subunits and
three-dimensional structure is often maintained in proteins
CHARACTERISTICS OF POLYACRYLAMIDE GELS by the presence of disulfide bonds. A goal of SDS-PAGE
The sieving properties of polyacrylamide geIs are established analysis under reducing conditions is to disrupt this structure
by the three-dimensional network of fibres and pores which is by reducing disulfide bonds. Complete denaturatiol1 and
formed as the bifunctional bisacrylamide cross-links adjacent dissociation of proteins by treatment with 2-mercaptoethanol
polyacrylamide chains. Polymerisation is catalysed by a free or dithiothreitol (DTT) will result in unfolding of the
radical-generating system composed of ammonium persulfate polypeptide backbone and subsequent complexatiol1 with SDS.
and tetramethylethylenediamine. In these conditions, the molecular mass of the polypeptide
As the acrylamide concentration of a gel increases, its subunits can be calculated by linear regression in the presence
effective pore size decreases. The effective pore size of a gel of suitable molecular-mass standards.
is operationally defined by its sieving properties; that is, by Non-reducing conditions. For some analyses, complete
the resistance it imparts to the migration of macromolecules. dissociation of the proteia into subunit peptides is not
There are limits on the acrylamide concentrations that can be desirable. In the absence of treatment with reducing agents
used. At high acrylamide concentrations, gels break much such as 2-mercaptoethanol or DTT, disulfide covalent bonds
more easily and are difficult to handle. As the pore size of remain intact, preserving the oligomeric form of the protein.
a gel de creases, the migration rate of a protein through the Oligomeric SDS-protein complexes migrate more slowly than
gel decreases. By adjusting the pore size of a gel, through their SDS-polypeptide subunits. In addition, non-reduced
manipulating the acrylamide concentration, the resolution proteins may not be completely saturated with SDS and,
of the method can be optimised for a given protein product. hence, may not bind the detergent in a constant mas s
Thus, a given gel is physically characterised by its respective ratio. This makes molecular-mass determinations of these
composition in acrylamide and bisacrylamide. molecules by SDS-PAGE less straightforward than anaIyses of
In addition to the composition of the gel, the state of the fully denatured polypeptides, sin ce it is necessary that both
protein is an important component to the electrophoretic standard s and unknown proteins be in similar configurations
mobility. In the case of proteins, the electrophoretic mobility for valid comparisons. However, the staining of a single band
is dependent on the pK value of the charged groups and the in such a gel is a criterion of purity.
size of the molecule. It is influenced by the type, concentration
CHARACTERISTICS OF DISCONTINUOUS BUFFER
and pH of the buffer, by the temperature and the field strength
SYSTEM GEL ELECTROPHORESIS
as well as by the nature of the support material.
The most popular electrophoretic method for the
DENATURING POLYACRYLAMIDE GEL ELECTROPHO- characterisation of complex mixtures of proteins involves
RESIS the use of a discontinuous buffer system consisting of two
The method cited as an example is limited to the analysis contiguous, but distinct gel s : a resolving or separating (lower)
of monomeric polypeptides with a mass range of 14000 gel and a stacking (upper) gel. The two gel s are cast with
to 100000 daltons. It is possible to extend this mass range different porosities, pH, and ionic strengths. In addition,
by various techniques (e.g. gradient gels, particular buffer different mobile ions are used in the gel and electro de buffers.
system) but those techniques are not discussed in this chapter. The buffer discontinuity acts to concentrate large volume .
Denaturing polyacrylamide gel electrophoresis using sodium samples in the stacking gel, resulting in improved resolution.
dodecyl sulfate (SDS-PAGE) is the most common mode of When power is applied, a voltage drop develops across the
electrophoresis used in assessing the pharmaceutical quality of sample solution which drives the proteins into the stacking gel.
protein products and will be the focus of the example method. Glycinate ions from the electrode buffer follow the proteins
Typically, analytical electrophoresis of proteins is carried into the stacking gel. A moving boundary region is rapidly
out in polyacrylamide gels under conditions that ensure formed with the highly mobile chloride ions in the front
dissociation of the proteins into their individual polypeptide and the relatively slow glycinate ions in the rearo A localised

48 See the information section on general monographs (cover pages)


EUROPEAN PHARMACOPOEIA 8.0 2.231. Electrophoresis

high-voltage gradient forms between the leading and trailing the desired concentration of acrylamide for the resolving
ion ffonts, causing the SDS-protein complexes to form into geL using the values given in Table 2.2.31.-1. Mix the
a thin zone (stack) and migrate between the chloride and components in the order shown. Where appropriate,
glycinate phases. Within broad limits, regardless of the height before adding the ammonium persulfate solution and the
of the applied sample, aH SDS-proteins condense into a very tetramethylethylenediamine (TEMED), filter the solution
narrow region and enter the resolving gel as a weH-defined, if necessary under vacuum through a cellulose acetate
thin zone of high protein density. The large-pore stacking gel membrane (pore diameter 0.45 ¡.un); keep the solution under
do es not retard the migration of most proteins and serves vacuum by swirling the filtration unit untilno more bubbles
mainly as an anticonvective medium. At the interface of the are formed in the solution. Add appropriate amounts of
stacking and resolving gels, the proteins experience a sharp ammonium persulfate so!ution and TEMED as indicated
in crease in retardation due to the restrictive pore size of the in Table 2.2.31.-1, swirl and pour immediately into the gap
resolving gel. Once in the resolving gel, proteins continue to between the two glass plates of the mould. Leave sufficient
be slowed by the sieving of the matrix. The glycinate ions space for the stacking gel (the length of the teeth of the comb
overtake the proteins, which then move in a space of uniform plus 1 cm). Using a tapered glass pipette, carefully overIay the
pH formed by the tris(hydroxymethyl)aminomethane and solution with water-saturated isobutanol. Leave the gel in a
glycine. Molecular sieving causes the SDS-polypeptide vertical position at room temperature to allow polymerisation.
complexes to separate on the basis of their molecular masses. Preparation of the stacking gel. After polymerisation is
PREPARING VERTICAL DISCONTINUOUS BUFFER SDS complete (about 30 min), pour off the isobutanol and wash
POLYACRYLAMIDE GELS the top of the gel several times with water to remove the
Assembling of the gel moulding cassette. Clean the two glass isobutanol overlay and any unpolymerised acrylamide. Drain
plates (size: e.g. 10 cm x 8 cm), the polytetrafluoroethylene as much fluid as possible from the top of the gel, and then
comb, the two spacers and the silicone rubber tubing remove any remaining water with the edge of a paper towel.
(diameter e.g. 0.6 mm x 35 cm) with mild detergent and In a conica! flask, prepare the appropriate volume of solution
rinse extensively with water. Dry all the items with a paper containing the desired concentration of acrylamide, using
towel or tissue. Lubricate the spacers and the tubing with the values given in Table 2.2.31.-2. Mix the components
non-silicone grease. Apply the spacers along each of the two in the order shown. Where appropriate, before adding the
short sides of the glass plate 2 mm away from the edges and ammonium persulfate solution and the TEMED, filter the
2 mm away from the long side corresponding to the bottom of solution if necessary under vacuum through a cellulose acetate
the gel. Begin to lay the tubing on the glass plate by using one membrane (pore diameter: 0.45 flm); keep the solution under
spacer as a guide. Carefully twist the tubing at the bottom of vacuum by swirling the filtration unit untilno more bubbles
the spacer and follow the long side of the glass plateo While are formed in the solution. Add appropriate amounts of
holding the tubing with one finger along the long side twist ammonium persulfate solution and TEMED as indicated
again the tubing and lay it on the second short side of the glass in Table 2.2.31.-2, swirl and pour immediately into the gap
plate, using the spacer as a guide. Place the second glass plate between the two glass plates of the mould directly onto the
in perfect alignment and hold the mould together by hand surface of the polymerised resolving gel. Immediately insert
pressure. Apply two clamps on each of the two short sides a clean polytetrafluoroethylene comb into the stacking gel
of the mould. Carefully apply four clamps on the longer si de solution, being careful to avoid trapping air bubbles. Add
of the gel mould thus forming the bottol11 of the gel mould. more stacking gel solution to fill the spaces of the comb
Verify that the tubing is running along the edge of the glass completely. Leave the gel in a vertical position and allow to
plates and has not been extruded while placing the clamps. polymerise at room temperature.
The gel mould is now ready for pouring the gel.
Preparation oí the gel In a discontinuous buffer SDS
polyacrylamide gel, it is recommended to pour the resolving
gel, let the gel set, and then pour the stacking gel since the
composition of the two gels in acrylamide-bisacrylamide,
buffer and pH are different.
Preparation of the resolving gel. In a conical flask,
prepare the appropriate volume of solution containing
Table 2.2.31.-1. - Preparation of resolving gel
Solution components Component volumes (mL) per gel mould volume of
5mL SOmL

6 per cent acrylamide

Water R 2.6 5.3 7.9 10.6 13.2 15.9 21.2 26.5

Acrylamide solution(J) l.0 2.0 3.0 4.0 5.0 6.0 8.0 10.0

l.5 Iv! Tris (pH 8.8)12) 1.3 2.5 3.8 5.0 6.3 7.5 10.0 12.5

100 giL SDS(3) 0.05 0.1 0.15 0.2 0.25 0.3 0.4 0.5

100 giL APS(4) 0.05 0.1 0.15 0.2 0.25 0.3 0.4 0.5

TEMED(5) 0.004 0.008 0.012 0.016 0.02 0.024 0.032 0.04

General Notices (1) apply to al! monographs and other texts 49


2.2.31. Electrophoresis EUROPEAN PHARMACOPOEIA 8.0

Solution componen!s Component volumes (mL) per gel mould volume of

5mL 50 mL

8 per cen! acrylamide

Water R 2.3 4.6 6.9 9.3 11.5 13.9 18.5 23.2

Acrylamide solution(J) 1.3 2.7 4.0 5.3 6.7 8.0 10.7 13.3

l.5 M Tris (pH 8.8)12) 1.3 2.5 3.8 5.0 6.3 7.5 10.0 12.5

100 giL SDS (3) 0.05 0.1 0.15 0.2 0.25 0.3 0.4 0.5

100 giL APS(4) 0.05 0.1 0.15 0.2 0.25 0.3 0.4 0.5

TEMED'S) 0.003 0.006 0.009 0.012 0.015 0.018 0.024 0.03

10 per (ent acrylamide

Water R 1.9 4.0 5.9 7.9 9.9 11.9 15.9 19.8

Acrylamide solution(l) 1.7 3.3 5.0 6.7 8.3 10.0 13.3 16.7

l.5 M Tris (pH 8.8)(2) 1.3 2.5 3.8 5.0 6.3 7.5 10.0 12.5

100 giL SDSIJ) 0.05 0.1 0.15 0.2 0.25 0.3 0.4 0.5

100 giL APS(4) 0.05 0.1 0.15 0.2 0.25 0.3 0.4 0.5

TEMED(5) 0.002 0.004 0.006 0.008 0.01 0.012 0.016 0.02

12 per cent acrylamide

Water R 1.6 3.3 4.9 6.6 8.2 9.9 13.2 16.5

Acrylamide solutionil ) 2.0 4.0 6.0 8.0 10.0 12.0 16.0 20.0

1.5 M Tris (pH 8.8)(2) 1.3 2.5 3.8 5.0 6.3 7.5 10.0 12.5

100 giL SDS (3) 0.05 0.1 0.15 0.2 0.25 0.3 0.4 0.5

100 giL APSI 41 0.05 0.1 0.15 0.2 0.25 0.3 0.4 0.5

TEMED(5) 0.002 0.004 0.006 0.008 0.01 0.012 0.016 0.02

14 per (ent acrylamide

Water R 1.4 2.7 3.9 5.3 6.6 8.0 10.6 13.8

Acrylamide solution(() 2.3 4.6 7.0 9.3 11.6 13.9 18.6 23.2

1.5 M Tris (pH 8.8)(2) 1.2 2.5 3.6 5.0 6.3 7.5 10.0 12.5

100 giL SDS 13 ) 0.05 0.1 0.15 0.2 0.25 0.3 0.4 0.5

100 giL APS (4) 0.05 0.1 0.15 0.2 0.25 0.3 0.4 0.5

TEMED (5 ) 0.002 0.004 0.006 0.008 0.01 0.012 0.016 0.02

15 per cent acrylamide

Water R 1.1 2.3 3.4 4.6 5.7 6.9 9.2 11.5

Acrylamide solution(1) 2.5 5.0 7.5 10.0 12.5 15.0 20.0 25.0

l.5 M Tris (pH 8.8)1 2) 1.3 2.5 3.8 5.0 6.3 7.5 10.0 12.5

100 giL SDSIJ) 0.05 0.1 0.15 0.2 0.25 0.3 0.4 0.5

100 giL APS(4) 0.05 0.1 0.15 0.2 0.25 0.3 0.4 0.5

TEMED(5) 0.002 0.004 0.006 0.008 0.01 0.012 0.016 0.02

(1) Acrylamide solution: 30 pe, cent acrylamide/bisacrylamide (29:1) solution R.


(2) 1.5 M Tris (pH 8.8): 1.5 M tris-hydrochloride buffer solution pH 8.8 R.
(3) 100 giL SDS: a 100 giL solution of sodium dodecyl sulfate R.
(4) 100 giL APS: a 100 giL solution of ammonium persulfate R. Ammonium persulfate provides the free radical s that drive polymerisation of
acrylamide and bisacrylamide. Sin ce ammonium persulfate solution decomposes slowly, fresh solutions must be prepared weekly.
(5) TEMED: tetral11ethylethylenediamine R.

Mounting the gel in the electrophoresis apparatus and Remove the tubing from the bottom part of the geL Mount the
electrophoretic separation. After polymerisation is complete gel in the electrophoresis apparatus. Add the electrophoresis
(about 30 min), remove the polytetrafluoroethylene comb buffers to the top and bottom reservoirs. Remove any bubbles
carefully. Rinse the wells immediately with water or with the that become trapped at the bottom of the gel between the
SDS-PAGE running buffer R to remove any unpolymerised glass plates. This is best done with a bent hypodermic needle
acrylamide. If necessary, straighten the teeth of the stacking gel attached to a syringe. Never pre-run the gel before loading the
with a blunt hypodermic needle attached to a syringe. Remove samples, since this will destroy the discontinuity of the buffer
the clamp s on one short side, carefully pull out the tubing and systems. Before loading the sample carefully rinse the slot with
replace the clamps. Proceed similarly on the other short side. SDS-PAGE running buffer R. Prepare the test and reference

50 See the information section on general monographs (caver pages)


EUROPEAN PHARMACOPOEIA 8.0 2.2.3 L Electl.'ophoresis

Table 2.2.31.-2. - Preparation of stacking gel


Solntion components Component volnmes (mL) per gel monld volume of

1 mL 2mL 3mL 4mL 5mL 6mL 8mL 10 mL

Water R 0.68 lA 2.1 2.7 3A 4.1 5.5 6.8

Acrylamide solution(1) 0.17 0.33 0.5 0.67 0.83 l.0 1.3 l.7

l.0 M Tris (pH 6.8)(2) 0.13 0.25 0.38 0.5 0.63 0.75 l.0 l.25

100 giL SDS(J) 0.01 0.02 0.03 0.04 0.05 0.06 0.08 0.1

100 giL APSI'J 0.01 0.02 0.03 0.04 0.05 0.06 0.08 0.1
TEMED(5) 0.001 0.002 0.003 0.004 0.005 0.006 0.008 0.01

(1) Acrylamide solution: 30 per cent acrylamide/bisacrylamide (29: 1) solution R.


(2) 1.0 M Tris (pH 6.8): 1 M tris-hydrochloride buffer solution pH 6.8 R.
(3) 100 giL SDS: a 100 giL solution of sodium dodecyl sulfate R.
(4) 100 giL APS: a 100 giL solution of ammonium persulfate R. Ammonium persulfate provides the free radicals that drive polymerisation of
acrylamide and bisacrylamide. Since ammonium persulfate solution decomposes slowly, fresh solutions must be prepared weekly.
(5) TEMED: tetramethylethylenediamine R.

solutions in the recommended sample buffer and treat as in a large excess of water R. Immerse the gel for about 1 min
specified in the individual monograph. Apply the appropriate in developer solution R until satisfactory staining has been
volume of each solution to the stacking gel wel!s. Start the obtained. Stop the development by incubation in the blocking
electrophoresis using the conditions recommended by the solution R for 15 mino Rinse the gel with water R.
manufacturer of the equipment. Manufacturers of SDS-PAGE DRYING OF STAINED SDS POLYACRYLAMIDE GELS
equipment may provide gels of different surface are a and
Depending 011 the staining method used, gels are treated in
thickness. Electrophoresis running time and current/voltage
a slightly different way. For Coomassie staining, after the
may need to vary as described by the manufacturer of the
destaining step, allow the gel to stand in a 100 giL solution of
apparatus in order to achieve optimum separation. Check that
glyeerol R for at least 2 h (overnight incubation is possible).
the dye front is moving into the resolving gel. When the dye
For silver staining, add to the final rinsing a step of 5 min in a
is reaching the bottom of the gel, stop the electrophoresis.
20 giL solution of glyeerol R.
Remove the gel assembly from the apparatus and separate
the glass plates. Remove the spacers, cut off and discard the Immerse two sheets of porous cellulose film in water R and
stacking gel and immediately proceed with staining. incubate for 5 min to 10 mino Place Ol1e of the sheets on
a drying frame. Carefully lift the gel and place it on the
DETECTION OF PROTEINS IN GELS cellulose film. Remove any trapped air bubbles and pour a
Coomassie staining is the most common protein staining few millilitres of water R around the edges of the gel. Place
method with a detection level of the order of 1 [lg to 10 [lg of the second sheet 011 top a11d remove any trapped air bubbles.
protein per bando Silver staining is the most sensitive method Complete the assembly of the drying frame. Place in an oven
for staining proteins in gels and a band containing 10 ng to or leave at room temperature until dry.
100 ng can be detected.
MOLECULAR-MASS DETERMINATION
Al! of the steps in gel staining are done at room temperature
Molecular masses of proteins are determined by comparison
with gentle shaking (e.g. on an orbital shaker platform) in any
of their mobilities with those of several marker proteins of
convenient container. Gloves must be worn when staining
known molecular weight. Mixtures of proteins with precisely
gels, since fingerprints will stain.
known molecular masses blended for uniform staining
Coomassie staining. Immerse the gel in a large excess of are available for calibrating gels. They are obtainable in
Coomassie staining solution R and allow to stand for at least various molecular mass ranges. Concentrated stock solutions
1 h. Remove the staining solution. of proteins of k110wn molecular mass are diluted in the
Destain the gel with a large excess of destaining solution R. appropriate sample buffer and loaded 011 the same gel as the
Change the destaining solution several times, until the protein sample to be studied.
stained protein bands are clearly distinguishable 011 a clear Immediately after the gel has been run, the position of the
background. The more thoroughly the gel is destained, the bromophenol blue tracking dye is marked to identify the
smaller is the amount of protein that can be detected by leading edge of the electrophoretic ion front. This can be done
the method. Destaining can be speeded up by including a by cutting notches in the edges of the gel or by inserting a
few grams of anion-exchange resin or a smal! sponge in the needle soaked in India ink into the gel at the dye front. After
destaining solution R. staining, measure the migration distan ces of each protein band
NOTE: the acid-a/cohol solutions used in this procedure do not (markers and unknowns) from the top of the resolving gel.
completely fix proteins in the gel. This can lead to losses of some Divide the migration distance of each protein by the distance
low-molecular-mass proteins during the staining and destaining travelled by the tracking dye. The normalised migration
of thin gels. Permanent fixation is obtainable by allowing the distances so obtained are called the relative mobilities of
gel to stand in a mixture of 1 volume of trichloroacetic acid R, the proteins (relative to the dye front) and conventional!y
4 volumes of methanol R and 5 volumes of water R for 1 h denoted as Rp. COl1struct a plot of the logarithm of the relative
before it is immersed in the Coomassie staining solution R. molecular masses (Mr ) of the protein standards as a function
of the Rp values. Note that the graphs are slightly sigmoid.
Silvel.' staining. Immerse the gel in a large excess of fixing
Unknown molecular masses can be estimated by linear
solution R and al!ow to stand for 1 h. Remove the fixing
regression analysis or interpolation from the curves of log
solution, add fresh fixing solution and incubate either for
M r against Rp as long as the values obtained for the unknown
at least 1 h or overnight, if convenient. Discard the fixing
samples are positioned along the linear part of the graph.
solution and wash the gel in a large excess of water R for
1 h. Soak the gel for 15 min in a 1 per cent V/V solution of VALIDATION OF THE TEST
glutaraldehyde R. Wash the gel twice for 15 min in a large The test is 110t valid unless the proteins of the molecular
excess of water R. Soak the gel in fresh si/ver nitrate reagent R mass marker are distributed along 80 per cent of the length
for 15 min, in darkness. Wash the gel three times for 5 min of the gel and over the required separation range (e.g. the

General Notices (1) apply to all monographs and other iexts 51


2.2.32. Loss on drying EUROPEAN PHARMACOPOEIA 8.0

range covering the product and its dimer or the product GENERAL PRINCIPLES
and its related impurities) the separation obtained for the Placing an ensemble of nuclei with angular momentum
relevant protein bands shows a linear relationship between and a magnetic moment in a static magnetic field (B o)
the logarithm of the molecular mass and the Rp Additional causes the nuclei to arrange themselves in different,
validation requirements with respect to the solution under test quantum-mechanically controlled orientations in relation
may be specified in individual monographs. to the axis of the magnetic field. These orientations are
QUANTIFICATION OF IMPURITIES different in energy. An oscillating high-frequency magnetic
Where the impurity limit is specifled in the individual field (B l ), perpendicular to Bo' will cause transitions between
monograph, a reference solution corresponding to that level these orientations with net energy absorption. According to
of impurity should be prepared by diluting the test solution. the resonance condition úl o = yEo (y = gyromagnetic ratio,
For example, where the limit is 5 per cent, a reference solution úl o = Larmor frequency), either the Bu magnetic field or
would be a 1:20 dilution of the test solution. No impurity the frequency (úl l ) of the E¡ field may be varied to achieve
(any band other than the main band) in the electropherogram a spectrum (continuous wave (CW) method). Nowadays the
obtained with the test solution may be more intense than the E¡ irradiation is achieved by the use of a radiofrequency (RF)
main band obtained with the reference solution. pulse (Fourier transform (FT) method). The coherent
Under validated conditions impurities may be quantified radiation emitted during the return to the initial state is
by normalisation to the main band using an integrating observed in the form of a decay curve, called the free induction
densitometer. In this case, the responses must be validated decay (FID). Subsequent Fourier transformation gives the
for linearity. spectrum in the frequency domain, providing information
about the molecular structure. Additional radiofrequency
fields may be applied during acquisition of the FID signal to
01/2008:20232 suppress scalar (through-bond) interactions between nuclei
(called 'decoupling'). One- and multi-dimensional techniques
can be applied for qualitative and quantitative purposes, on
2.2.32. LOSS ON DRYING samples in either the liquid or the solid state.
Loss on drying is the loss of mass expressed as per cent mlm. Important structural information is derived from the following
Method. Place the prescribed quantity of the substance to spectroscopic features:
be examined in a weighing bottle previously dried under
resonance frequency kind of nuelei observed
the conditions prescribed for the substance to be examined.
Dry the substance to constant mass or for the prescribed number of resonance signals (singlets, number of chemically distinct groups
time by one of the following procedures. Where the drying multiplets) of nuclei
temperature is indicated by a single value rather than a range, chemical shift i5 (ppm) chemical nature and environment of
drying is carried out at the prescribed temperature ± 2 oc. the structural "roup observed

al "in a desiccator": the drying is carried out over intensity of resonance signals relative number of resonant nuelei
diphosphorus pentoxide R at atmospheric pressure and at
Iper chemically distinct group
room temperature; multiplicity of coupling pattern number of nuclei that are scalar
coupled to the observed nueleus
b) "in vacuo": the drying is carried out over diphosphorus
coupling constant "J (Hz) number of bonds in the cOLlpling
pentoxide R, at a pressure of 1.5 kPa to 2.5 kPa at room pathway, and its geometry
temperature;
c) "in vacuo within a specified temperature range": the Correlations of different spectral parameters (e.g. chemical
drying is carried out over diphosphorus pentoxide R, at a shift and coupling constant, or chemical shifts of different
pressure of 1.5 kPa to 2.5 kPa within the temperature range nuclei within one molecular system) can be performed by
prescribed in the monograph; homo- and hetero-nuclear two- and higher-dimensional
methods. Information about the relaxation times T¡ and
d) "in an oven within a specifled temperature range": the T2 , nuclear Overhauser effects (NOEs) and the kinetics of
drying is carried out in an oven within the temperature time-dependent processes are also accessible from appropriate
range prescribed in the monograph; experiments.
e) "under high vacuum": the drying is carried out over
diphosphorus pentoxide R at a pressure not exceeding APPARATUS
0.1 kPa, at the temperature prescribed in the monograph. A high-resolution NMR spectrometer consists of at least the
rf other conditions are prescribed, the procedure to be used is following parts:
described in ful! in the monograph.
- a magnet to deliver the constant magnetic field Eo;
- a temperature-controlled probe to contain the sample, to
0112009:20233 deliver the radiofrequency pulse and to detect radiation
emitted by the sample;
2.2.33. NUCLEAR MAGNETIC - an electronic console to generate high-power
radiofrequency pulses and to collect and digitise the
RESONANCE SPECTROMETRY FID signal; this unit also maintains the stability of the
INTRODUCTION instrument electronics;
Nuclear magnetic resonance (NMR) spectrometry is an - a data acquisition and processing unit (computer);
analytical method in particular suitable for the elucidation and may also include:
of the chemical structure of organic molecules by means
of interpretation of their NMR spectra, arising from, for - a continuous flow ceH for coupled liquid
example, IH or the X-nuclei l3C, 19F, 15N, 31p' The spectra can chromatographic-NMR or flow injection analysis;
be used for qualitative and quantitative purposes. - a system for pulsed field gradient NMR.
Under suitable experimental conditions, the integrated NMR The high magnetic fieId is generated by a superconducting coil
intensities of the signals are directly proportional to the in a Dewar flask filled with liquid helium. The probe typically
number of nuclear spins of the molecular group responsible contains the sample in a 5 mm-outer-diameter sample tube
for the signa!. These integrals can be used for quantitative or in a flow cell, and is connected to the electronics cabinet
analysis. by RF cables carrying lock, ¡H-, and X-nucleus frequencies.

52 See the information section on general monographs (caver pages)


EUROPEAN PHARMACOPOEIA 8,0 2.2.33. Nuclear magnetic resonance spectrometry

Additional devices fol' tuning and matching the electronic Repetition time {t,}, The spin-lattice relaxation (T¡) governs
circuits are essential, and sample temperature control is often the time required for the spin system to l'eturn to equilibrium
used, after a pulse, Relaxation can be reduced by the use of special
reagents, For quantitative purposes, the repetition time used
The NMR spectrometer should be demonstrated to be should be set relative to T¡ and 8 to avoid saturation effects,
operating correctly, Appropriate tests to demonstrate this are,
Receiver gain, The analogue signal detected by the probe is
typically, measurement of linewidths at half height for defined
amplified prior to digitisation and storage, The amplification,
peaks under defined acquisition conditions, signal-to-noise
or receiver gain, should be set, either automatically or
ratios (SIN) for standard mixtures, pulse power (measured as
manually, so that the signal does 110t overload the ADC, which
a 90° pulse width), and pulse reproducibility, AH instrument
causes signal distortion, but allows random noise generated in
manufacturers publish specifications and measurement
the probe to be digitised (Le, is non-zero),
protocols for these parameters for specific instrument/probe
combinations, and compliance with these specifications OPTIMISATION OF ACQUISITION AND PROCESSING
should be demonstrated, PARAMETERS FOR QUANTITATIVE PURPOSES
Besides the acquisition parameters, signal intensity is also
influenced by several processing parameters, After collecting
FOURIER TRANSFORM NMR (FT-NMR) a sufficient number of scans, the resulting FrD is Fourier
transformed, For reliable quantitative purposes the fo11owing
Contempol'ary spectrometers generally operate according to parameters have to be optimised,
the Fourier transform (FT) principIe: after exciting the sample
Digital resolution, The digital resolution is the frequency
with a radiofrequency pulse of appropriate frequency (v),
separation between data points, The processed signal should
amplitude (B I ) and duration (T p ) and a succeeding short dead
have at least 5 digital points aboye half-height of the signals
time (td) (to enable the electronics to recover), the amplified
to be integrated, To improve the digital resolution additional
analogue FID signal is sampled during the acquisition time
points of zero intensity may be added to the end of the
(taJ and digitised with an analogue-to-digital converter
experimental FrD before transformation ('zero fi11ing'),
(ADC), and the results are sto red in the spectrometer memory,
The receiver output is amplified prior to digitisation to Signal-to-noise ratio (SIN), This is the ratio between the
maximise sensitivity without saturating the ADC In case of intensities (as peak height) of a specified signal in the NMR
observation of X-nudei, the standard experiment indudes, if spectrum and the random fluctuations in that signal, which
necessary, broadband IH decoupling, Le, irradiation of all the is usually measured in a region of the spectrum that contains
protons during the experiment To increase the SIN, multiple no signals fmm the analyte, A poor signal-to-noise ratio
FrD signals may be accumulated cohel'ently and summed, (SIN) limits the accuracy of peak integrations and quantitative
Fourier transformation of this time-domain data gives the analyses, An SIN equal to or greater than 150: 1 a110ws peak
frequency-domain spectrum, integrations with a standard deviation of less than 1 per cent
Contemporary spectrometel's have software algorithms to
PARAMETERS
report the SIN of appropriate peaks, A sufficiently high SIN
The following acquisition parameters influence the result of can be difficult to obtain when analysing dilute solutions, and
an FT experiment, and should be adjusted and controlled, especia11y when detecting nuclei other than ¡R, Methods to
Pulse width (1), The excitation pulse is directed along the enhance the SIN indude:
x -axis of the so-called rotating frame, its duration (or 'width', - increasing the number of accumulated scans (n), as SIN
T,) determines the flip angle (8) and thus the intensity (1) of increases with Vn;
the resonance signal:
- use of exponentional multiplication on the FID signal
e= ¡' x El X T p
before Fourier transformation; the exponentional
(1) multiplication factor should be in the order of the peak full
width at half-height (jwhh);
My = 1\IIo x sin e - use of spectrometers with a higher magnetic field Bo' since
(2)
SIN is proportional to B o3/ 2 ;
The observed magnetisation My is maximum at 8 = 90°, The - use of digital filtering to reduce noise;
pulse duration should be short to guarantee that a11 signals in - use of pro bes that maximise the filling factor;
the spectral width (SW) are excited to a similar degree, The
magnetisation decays due to relaxation processes, - use of cryoprobes that reduce thermal noise,
Dead time (td ), The dead time is the time between the end Integration region, The intensity of the NMR signals is
of the pulse and start of the acquisition, it is necessary for obtained by a quasi-analogue signal integration either by
technical reasons and care should be taken as it may influence a stepped -line plot or, more accurately, by separate line
signal intensities and peak phase, Rapidly decaying signals integration and digital data presentation, In liquid state, NMR
(giving rise to broad spectrallines) are reduced in intensity by signals have Lorentzianline shape, Unless otherwise specified
more than slowly decaying signals (which give rise to narrow in the monograph or when peak overlap occurs, the same
spectrallines) , integration range, expressed as a multiple of the peak fwhh,
should be used for the monitored peak and the reference peak
Acquisition time (ta), The acquisition time (t"cl is related to
the spectral width (Le, the whole observed region) and the Dynamic mnge, The dynamic range of the analogue-to-digital
number of digital data points (DP) collected during signal converter (ADC) determines the minimum intensity line
acquisition, that can be observed or quantified when integrating 2 signals
with the same linewidth in a spectrum, A 16-bit ADC allows
t -~~
DP identification of a signal of 0,003 per cent intensity relative to a
oc - 2SW (3) strong signal completely filling the dynamic range of the ADC
NMR OF SAMPLESIN SOLUTION
Maximal signal intensity and signal-to-noise ratio will be Most NMR experiments are performed on dilute solutions
achieved if tae '" L2/(nv 2), where v1/2 is the fu11 width at
j/ (about 1 per cent) of the analyte in an appropriate solvent,
half-height (jwhh), but it should be set to greater than 5/(nv1/2) which can be spiked with a suitable standard for chemical
to minimise signal distortion, shift calibration,

General Notices (1) apply to all monographs and other texts 53


2.2.33. Nuclear magnetic resonance spectrometry EUROPEAN PHARMACOPOEIA 8.0

Solvents. The solvent should be able to dissolve the analyte of reference and test samples should be acquired using the
without further interaction if not otherwise intended. To same procedure and operational conditions. The peaks
minimise the intense solvent signals, fully deuterated solvents in the 2 spectra, or characteristic regions of the spectra,
(deuterium oxide R, deuterated chloroform R, deuterated should correspond in position, intensity and multiplicity.
dimethyl sulfoxide R, deuterated acetone R, deuterated In appropriate cases, mathematical comparison, such as
methanol R, etc.) should be used. The solvent atoms give calculation of a correlation coefficient, may be appropriate. In
signals that are easily identified by their chemical shift and the absence of a reference standard, an in-house reference may
can be used to calibrate the chemical shift axis (secondary be used, whose identity has been demonstrated by alternative
reference) . methods, or the demonstration that the NMR spectrum is
fully consistent with the reported structure fOI that material.
Referencing. The spectral feature most dependent on the
chemical environment of the atom in the molecule is the QUANTITA TIVE ANALYSlS
chemical shift, designated as O and reported in parts per Signal intensity in the basic NMR experiment is the integrated
million. The chemical shift between the resonance for an are a under the signal curve measured. Only when 2 signals
NMR active nucleus X (Ox.samplc) is measured in parts per have equal fwhh and the same multiplicity may signal height
million as the ditTerence between the resonance frequency of serve as a measure of intensity. Under conditions of essentially
that nu.cleus (Vx,s8ln,ple) and that of an internal shift ref~rence complete relaxation between scans, the signal intensity (IA) is
standard (VX,referenceJ, both 111 hertz, dlvlded by the baslc a true measure of the number (NA) of nuclei responsible for
spectrometer operating frequency (VX,reference)' in megahertz, the respective signal:
at a given Bo:
(6)
oX,san~ple ==
(VX,sample - VX,reference)
The constant K5 includes fundamental constants, properties
Vx 1 reference (4) of the sample and receiver parameters, and can be omitted in
cases where signal intensities are compared, giving the direct
relation between the numbers of nuclei in the 2 compared
By convention, the standard for exact chemical shift
structure groups A and B;
referencing is the lH resonance of tetramethylsilane R (TMS),
setting 0TMS = O ppm. Pormally, once the lH shift scale has lA NA
been referenced relative to TMS, the exact frequency of any (7)
other X resonance can be calculated and its chemical shift scale
calibrated. The frequency of a (secondary) reference standard
at Ox = O ppm (vX,cefccence) is calculated from the lH frequency The numbers (N) of nuclei belonging to different structure
of TMS (VH,TMS) and a tabulated value of the ratio (3 X,rdmnce) of groups of 1 molecule are small integers. The values measured
the isotope-specific frequency in relation to that of lH in TMS: are rounded to the closest integer numbers. However,
the proper operation of acquisition and processing of the
spectrometer is easily checked by comparing exact intensities
VX,reference = VH,TMS X -lOOOOjilx,reference
within a spectrum of any suitable organic compound of
(5) known structure.

In additiol1 to the fact that the intensities of signals arising


Reference standards at = O ppm and corresponding 3 X,referencc from each component in a mixture are related to each
values are shown below: other by small integer numbers, the relative molar amounts
of these components can be measured by comparison of
Other the normalised intensities of resonances from different
Water"
Nucleus
'"
"""'X,rcferencc
solvents '"
""'X,reference
components. The molar ratio of 2 components of a mixture is
IH DSS b LOOOOOOoo TMS LOOOOOOOO calculated according to the following equation:
l3C DSS" 0.25144953 TMS 0.25145020
nA lA NB
15N
-=-x
NH 3 0.10132912 CH,NO, 0.10136767 nB lB NA (8)
19p CP3COOH nol reporled CCl,P 0.94094011
The determination is only valid in cases where the structure
Jlp
H,PO¡ 0.40480742 (CHP)}PO 0.40480864
(85 per cenl) of the molecules for which lA and lE are determined are
known (01' at least the values of N for the monitored groups).
"chemical shifl depends on pH
Determinations are made using either an internal standard
bDSS = sodium 2,2-dimelhyl-2-silapenlane-5-sultonate method or a peak-normalisation procedure.
Interna! standard method. The mass (m A ) of an analyte (A)
In practice, X chemical shifts are referenced directly using an can be determined if a known mass (mE) of a substance (E)
appropriate standard. In lH and l3C NMR, internal referencing with a known percentage content (P B ) is added to the solution
is mainly used, where the reference is added directly to as an intensity standard. Equation (8) can be converted to
the system under study. In lSN, 19p and 31p NMR, external equation (9):
referencing is often suitable, involving sample and reference
contained separately in coaxial cylindrical sample tubes.
Lock. In order to prevent drifting of the spectrum over time, (9)
a stabilising procedure, called field -frequency locking, is
performed. The 2H (deuterium) signal arising from deuterated
solvents is used to achieve this, unless otherwise specified in Here, Mi are the molecular masses.
the monograph.
The intensity standard has to be carefully chosen; it should be
QUALITA TIVE ANALYSlS completely soluble in the solvent used fol' the analyte, should
The principal use for qualitative NMR spectra is as an identity produce only a small number of signals, and the 'monitor
test, in which the 1H or llC spectrum of a test sample is group' should have a signal in an empty spectral region. A
compared to the spectrum of a reference sample or, less compound of high purity and with a relatively high molecular
commonly, with a published reference spectrum. Spectra mass is recommended for this purpose.

54 See the information section on general monographs (ca ver pages)


EUROPEAN PHARMACOPOEIA 8.0 2.2.34. Thel'mal analysis

Normalisation pl'Ocedure. The relative proportions of THERMOGRAVIMETRY


components in a mixture, the degree of substitution in a
Thermogravimetry is a technique in which the mass of a
structurally modified polymer, or the amount of a contaminant
sample of a substance is recorded as a function of temperature
can be determined by comparison of the relative intensities
according to a controlled temperature programme.
of resonances present.
The experimental method should be validated to ensure Apparatus. The essential components of a thermobalance are
that there is no overlap of the relevant signals. When the a device for heating or cooling the substance according to a
contaminant is of poorly defined structure or molecular given temperature program, a sample holder in a controlled
mass (e.g. an emulsifier), addition ofknown amounts of that atmosphere, an electrobalance and a recorder. In sorne cases
material to the NMR tube will allow a calibration curve to be the instrument may be coupled to a device permitting the
constructed. analysis of volatile products.
Temperatul'e verincation. Check the temperature scale
METHOD
using a suitable material according to the manufacturer's
Sample handling. Dissolve the sample in the solvent to which instructions.
the appropriate reference material may have been added to
calibrate chemical shift, as prescribed in the monograph. For Verincation of the electrobalance. Place a suitable quantity
quantitative analysis, the solutions must be free from solid of a suitable certifled reference material (for example, calcium
particles. Some quantitative analyses may require an internal oxalate monohydrate CRS) in the sample holder and record
standard to be included, so that integrations of resonances the mass. Set the heating rate according to the manufacturer's
from the test sample and the reference material can be instructions and start the temperature increase. Record the
compared. Appropriate references and concentrations are thermogravimetric curve as a graph with temperature, or
indicated in the speciflc monographs. In other quantitative time, on the abscissa, increasing from left to right, and mass
analyses, the result is obtained by comparing the relative on the ordinate, increasing upwards. Stop the temperature
intensities of 2 or all of the resonances that arise from the test increase at about 230 oc. Measure the difference on the graph
sample. After loading the sample into a tube and capping, the between the initial and final mass-temperature plateaux, or
sample is introduced into the NMR magnet, the experimental mass-time plateaux, which corresponds to the 108s of mass.
parameters are loaded and the experiment is executed. Key The declared loss of mass for the certified reference material is
experimental parameters are indicated in the monograph. stated on the label.

The meaSUl'ement pl'ocedul'e. Equilibrate the sample in Method. Apply the same procedure to the substance to be
the probe, and optimise the instrument to achieve best examined, using the conditions prescribed in the monograph.
resonance conditions and to maximise the SIN by tuning Calculate the 10ss of mass of the substance to be examined
and matching the probe, and make adjustments to maximise from the difference measured in the graph obtained. Express
magnetic field homogeneity over the sample volume (called the 10ss of mass as per cent /'<,. mlm.
'shimming'). Record, or save to computer, the parameter lf the apparatus is in frequent use, carry out temperature
settings. An experiment may be composed of multiple verification and calibration regularly. Otherwise, carry out
pulse-acquisition-delay sequences, and the individual FIDs are such checks before each measurement.
summed in the computer memory, with random noise being
averaged out. When an appropriate SIN has been achieved, Since the test atmosphere is critical, the following parameters
the FID is stored and the frequency-domain spectrum is are noted for each measurement: pressure or flow rate,
generated by Fourier transformation of the summed FIDs. composition of the gas.

NMR IN THE SOLID STATE DIFFERENTIAL SCANNING CALORIMETRY


Samples in the solid state can be analysed using NMR
spectrometers specially equipped for that purpose. Certain Differential Scanning Calorimetry (DSC) is a technique that
technical procedures make observable individuallines for can be used to demonstrate the energy phenomena produced
individual atomic sites with a valuable extension of the during heating (or cooling) of a substance (or a mixture of
applicability of NMR to inorganic materials as well. substances) and to determine the changes in enthalpyand
specific heat and the temperatures at which these occur.
One technique is the rapid rotation (4-30 kHz) of the
powdered sample in a rotor (about 4 mm outer diameter) The technique is used to determine the difference in the
inclined at an angle of 54.7° (the 'magic angle') to the direction flow of heat (with reference to the temperature) evolved or
of the Bo magnetic field axis. This technique is named magic absorbed by the test sample compared with the reference
angle spinning (MAS). Another effective tool is high-power ceH, as a function of the temperature. Two types of DSC
decoupling and a 3,d method is the transfer of polarisation apparatuses are available, those using power compensation to
from easily excitable nuclei towards less-polarisable nuclei, i.e. maintain a null temperature difference between sample and
cross polarisation (CP). The combination of these techniques reference and those that apply a constant rate of heating and
makes available high-resolution spectra containing much detect temperature differential as a difference in heat flow
information about chemical and structural details in solid between sample and reference.
glassy, amorphous, and crystalline materials of ceramic, Apparatus. The apparatus for the power compensation
polymeric or mineralogical origino DSC consists of a furnace containing a sample holder with
rf NMR is applied to a solid, full details of the procedure are a reference cell and a test cell. The apparatus for the heat
provided in the monograph. flow DSC consists of a fumace containing a single cel! with a
sample holder for the reference crucible and the test crucible.
01/2008:20234 A temperature-programming device, thermal detector(s) and
corl'ected 6.1 a recording system which can be connected to a computer
are attached. The measurements are carried out under a
2.2.34. THERMAL ANALYSIS controlled atmosphere.
Thermal analysis is a group of techniques in which the Calibration of the apparatus. Calibrate the apparatus for
variation of a physical property of a substance is measured temperature and enthalpy change, using indium of high
as a function of temperature. The most commonly used purity or any other suitable certified material, according to
techniques are those which measure changes of mass or the manufacturer's instructionso A combination of 2 metals,
changes in energy of a sample of a substance. e.g. indium and zinc may be used to controllinearity.

General Notices (1) apply to all monographs and other texts 55


2.2.34. Thermal analysis EUROPEAN PHARMACOPOEIA 8.0

Endolherme
~
1°C
___________________ ~
_lemperature

Figure 2.2.34.-1. - Thermogram


Operating procedure. Weigh in a suitable crucible an diagram, requiring the use of only a few milligrams of sample
appropriate quantity of the substance to be examined; place with no need for repeated accurate measurements of the true
it in the sample holder. Set the initial and final temperatures, temperature.
and the heating rate according to the operating conditions In theory, the melting of an entirely crystalline, pure
prescribed in the monograph. substance at constant pressure is characterised by a heat of
Begin the analysis and record the differential thermal analysis fusion IlH¡ in an infinitely narrow range, corresponding
curve, with the temperature or time on the abscissa (values to the melting point T o. A broadening of this range is a
increasing from left to right) and the energy change on the sensitive indicator of impurities. Hence, samples of the same
ordinate (specify whether the change is endothermic or substance, whose impurity contents vary by a few tenths of
exothermic) . a per cent, give thermal diagrams that are visually distinct
The temperature at which the phenomenon occurs (see Figure 2.2.34.-2).
(the onset temperature) corresponds to the intersection The determination of the molar purity by DSC is based on the
(A) of the extension of the baseline with the tangent at use of a mathematical approximation of the integrated form of
the point of greatest slope (inflexion point) of the curve the Van't Hoff equation applied to the concentrations (not the
(see Figure 2.2.34.-1). The end of the thermal phenomenon is activities) in a binary system [In (1 - X2) = -X2 and
indicated by the peak of the curve. T x T o = T51:
The enthalpy of the phenomenon is proportional to the area
under the curve limited by the baseline; the proportionality RT5 (1)
T= T o - - - X X2
factor is determined from the measurement of the heat of t:"Hf
fusion of a known substance (e.g., indium) under the same
Xl mole fraction of the impurity i.e. the number of
operating conditions.
molecules of the impurity divided by the total
Each thermogram may be accompanied by the following number of molecules in the Iiquid phase (or molten
data: conditions employed, record of last calibration, sample phase) at temperature T (expressed in kelvins),
size and identification (including thermal history), container,
atmosphere (identity, flow rate, pressure), direction and rate To melting point of the chemically pure substance, in
of temperature change, instrument and recorder sensitivity. kelvins,
Applications molar heat of fusion of the substance, in joules,
Phase changes. Determination of the temperature, heat gas constant for ideal gases,
capacity change and enthalpy of phase changes undergone by in joules·kelvin- '.mole- l.
a substance as a function of temperature. Hence, the determination of purity by DSC is limited to the
solid - solid transition: allotropy - polymorphism detection of impurities forming a eutectic mixture with the
glass transition principal compound and present at a mole fraction of less
desolvation than 2 per cent in the substance to be examined.
amorphous-crystalline This method cannot be applied to:
salid - liquid transition : melting
- amorphous substances,
- solvates or polymorphic compounds that are unstable
solid - gas transition: sublimation within the experimental temperature range,
liquid - solid transition: freezing - impurities forming solid solutions with the principal
recrystallisation
substance,
- impurities that are insoluble in the liquid phase or in the
liquid - gas transition: evaporation melt of the principal substance.
Changes in chemical composition. Measurement of heat and During the heating of the substance to be examined, the
temperatures of reaction under given experimental conditions, impurity melts completely at the temperature of the eutectic
so that, for example, the kinetics of decomposition or of mixture. Above this temperature, the solid phase contains only
desolvation can be determined. the pure substance. As the temperature increases progressively
from the temperature of the eutectic mixture to the melting
Application to phase diagrams. Establishment of phase
diagrams for solid mixtures. The establishment of a phase point of the pure substance, the mole fraction of impurity in
diagram may be an important step in the preformulation and the liquid decreases constantly, since the quantity of liquified
pure substance increases constantly. For al! temperatures
optimisatioll of the freeze-drying process.
aboye the eutectic point:
Determination of purity. The measurement of the heat of
fusion and the melting point by DSC enables the impurity 1 (2)
content of a substance to be determined from a single thermal
X2 = F x

56 See the information section on general monographs (ca ver pages)


EUROPEAN PHARMACOPOEIA 8.0 2.2.35. Osmolality

¡'iIII 1°C ~I.. ....... Temperature


./
/ /
/ (
..... I
I I
I
I
I
llH I
I
+o
TI
I
W
e I
99.50% ;;../

99.80%

Figure 2.2.34.-2. - Thermal diagrams according to purity

F molten fraction of the analysed sample, An acceptable approximation for the osmolality ~m of a given
aqueous solution is given by:
mole fraction of the impurity in the analysed
sample. ~m = vm<P
When the entire sample has melted, F = 1 and X2 = x~.
If the solute is not ionised, v = 1; otherwise v is the total
If equation (2) is combined with equation (1), the following number of ions already present 01' formed by solvolysis from
equation is obtained: 1 molecule of solute.
m molality of the solution, that is the number of
moles of solute per kilogram of solvent;
<D molal osmotic coefficient which takes account of
The value of the heat of fusion is obtained by integrating the the interactions between ions of opposite charge in
melting peak. the solution. It is dependent on the value of m. As
The melting point T o of the pure substance is extrapolated the complexity of solutions increases, <D becomes
from the plot of llF versus the temperature expressed in difficult to me asure.
kelvins. The slope a of the curve, obtained after linearisation, The unit of oSl110lality is osmole per kilogram (osmol/kg),
if necessary, corresponding to RT5 ttJ
f allows to be but the submultiple milliosmole per kilogram (mosmol/kg) is
evaluated. usually used.
The fraction X2' multiplied by 100 gives the mole fraction Unless otherwise prescribed, osmolality is determined
in per cent for the total eutectic impurities. by measurement of the depression of freezing point. The
following relationship exists between the osmolality and the
THERMOMICROSCOPY depression of freezing point j'., T:

Phase changes may be visualised by thermomicroscopy, a !'Y.T


~m = - x 1000 mosmol/kg
method which enables a sample subjected to a programmed 1.86
temperature change to be examined, in polarised light, under Apparatus. The apparatus (osmometer) consists of:
a microscope.
- a means of cooling the container used for the measurement;
The observations made in thermomicroscopy allow the nature
of the phenomena detected using thermogravimetry and - a system fo1' measuring temperature consisting of a resistor
differential thermal analysis to be clearly identified. sensitive to temperature (thermistor), with an appropriate
current or potential-difference measurement device that
Apparatus. The apparatus consists of a microscope fitted may be graduated in temperature depression or directly
with a light polariser, a hot plate, a temperature and heating in osmolality;
rate and/or cooling rate programmer and a recording system
for the transition temperatures. A video camera and video - a means of mixing the sample is usually included.
recorder may be added. Methad. Prepare reference solutions as described in
Table 2.2.35.-1, as required. Determine the zero of the
apparatus using water R. Calibrate the apparatus using the
reference solutions: introduce a suitable volul11e of sal11ple inta
the measurement cell, as indicated by the equipment supplier,
01/2012:20235 and start the cooling system. Usually, the mixing device is
programmed to operate at a temperature below that expected
through cryoscopic depression to prevent supercoaling. A
2.2.35. OSMOLALITY suitable device indicates attainment of equilib1'ium. Before
Osmolality is a practical means of giving an overall measure each measurement, rinse the measurement ceH with the
of the contribution of the various solutes present in a solutiol1 solution to be examined.
to the osmotic pressure of the solution.

General Natices (1) apply to all monographs and ather texts 57


2.2.36. Ionie concentration (ion-selective electrodes) EUROPEAN PHARMACOPOEIA 8.0

Table 2.2.35.-1. - Reference solutions for osmometer calibration Apparatus. Use a voltmeter allowing measurements to the
nearest 0.1 millivolt and whose input impedance is at least one
Mass in grams of Real Ideal Mola! Cryoscopic
sodium chloride R osmolality osmolality osmotic depression hundred times greater than that of the electro des used.
per Idlogram of (mosmol/kg) (mosmol/kg) coefficient (oC)
Ion-selective electrodes may be primary electrodes with a
water R
crystal or non-crystal membl'ane or with a rigid matrix (for
3.087 100 105.67 0.9463 0.186
example, glass electro des), or electro des with charged (positive
6.260 200 214.20 0.9337 0.372 or negative) or uncharged mobile carriers, ar sensitised
electro des (enzymatic-substrate electro des, gas-indicator
9.463 300 323.83 0.9264 0.558
electrodes). The reference electro de is generally a silvel'-silver
12.684 400 434.07 0.9215 0.744 chloride electrode or a calomel electro de, with suitable
junction liquids producing no interfel'ence.
15.916 500 544.66 0.9180 0.930
Procedure. Carry out each measurement at a temperature
19.147 600 655.24 0.9157 1.116
constant to ± 0.5 oC, taking into account the val'iation of the
22.380 700 765.86 0.9140 1.302 slope ofthe electro de with temperature (see Table 2.2.36.-1).
Adjust the ionic strength and possibly the pH of the solution
Carry out the same operations with the test sample. Read to be analysed using the buffer reagent described in the
directly the osmolality or calculate it from the measured monograph and equilibrate the electro de by immersing it in
depression of Íreezing point. The test is not valid unless the the solution to be analysed, undel' slow and uniform stirring,
value found is within 2 values of the calibration scale. until a constant reading is obtained.

Table 2.2.36.-1. - Values ofk at different temperatures


Temperature ("e) k

20 0.0582
0112008:20236
25 0.0592

30 0.0602
2.2.36. POTENTIOMETRIC
DETERMINATION OF IONIC If the electro de system is used frequently, check regularly the
repeatability and the stability of responses, and the linearity
CONCENTRATION USING of the calibration curve or the calculation algorithm in the
ION -SELECTIVE ELECTRODES range of cOl1centrations of the test solution; if not, carry out
the test beÍore each set of measul'ements. The response of
Ideally, the potential E of an ion -selective electro de varies the electrode may be regarded as linear if the slope S of the
linearly with the logarithm of the activity a¡ of a given ion, as calibration curve is approximately equal to klz" per unit of pC¡.
expressed by the Nernst equation:
RT METHOD 1 (DIRECT CALIBRATION)
E = Ea + 2.303-loga¡
z¡F Measure at least three times in succession the potential of
at least three reference solutions spanning the expected
part of the constant potential due to the apparatus concentration of the test solution. Calculate the calibration
used, curve, or plot on a chart the mean potential E obtained against
R gas constant, the concentratiol1 of the ion to be determined expressed as
~ 10glO C¡ or pC¡.
T absolute temperature,
Prepare the test solution as prescribed in the monograph;
F Faraday's number, measure the potential three times and, from the mean
charge number of the ion including its signo potential, calculate the concentl'ation of the ion to be
determined using the calibration curve.
At a constant ionic stl'ength, the following holds:
k METHOD II (MULTIPLE STANDARD ADDITIONS)
E = Ea + -logfC¡
z¡ Prepare the test solution as prescribed in the monograph.
Measure the potential at equilibrium ET of a volume V r of
C¡ molar concentration of the ion, this solution of unknown concentration CT of the ion to be
determined. Make at least three consecutive additions of
f the activity coeÍficient (a¡ = fC¡), a volume V s negligible compared to VT (Vs ::; 0.01 VI) of a
k RT reference solution oí a concentratiol1 Cs known to be within
F
the linear part of the calibratiol1 curve. After each addition,
measure the potential and calculate the difference of potential
1f : Ea
k
+ -logf I
= Ea an
d
S = -
k ¿,;E between the measured potential and Er' ¿,;E is related to
Zi Zi the concentl'ation of the ion to be determined by the equation:
s = slope of the calibration curve oí the electro de,

the following holds: E = Eb + S log Ci !lE = Slog


es Vs )
( 1 + CTV T
and Íol' -logC¡ = pCi : E = Eb - SpCi .
The potentiometric determination of the ion concentration is or
carried out by measuring the poten ti al difference between two L!.E Cs \Ás
suitable electro des immersed in the solution to be examined; lOS 1+
= CTVT
the indicator electro de is selective for the ion to be determined
and the other is a reference electro de.

58 See the information section on general monographs (cava pages)


EUROPEAN PHARMACOPOEIA 8.0 2.2.38. Condl1ctivity

volume of the test solution, wavelength, is proportional to the concentration of this


element and inversely proportional to the mass absorption
CT concentration of the ion to be determined in the coefficient of the matrix at this wavelength.
test solution,
Method. Set and use the instrument in accordance with
Vs added volume of the reÍerence solution, the instructions given by the manufacturero Liquid samples
Cs concentration of the ion to be determined in the are placed directly in the instrument; solid samples are first
reference solution, compressed into pellets, sometimes after mixing with a
S 510pe of the electro de determined experimentally, suitable binder.
at constant temperature, by measuring the To determine the concentration of an element in a sample, it
difference between the potentials obtained with is necessary to measure the net impulse rate produced by one
two reference solutions whose concentrations or several standard preparations containing known amounts
differ by a factor of ten and are situated within the of this element in given matrices and to calculate or measure
range where the calibration curve is linear. the mass absorption coefficient of the matrix of the sample
fO.E to be analysed.
Plot on a graph lOs (y-axis) against Vs (x-axis) and Calibration. From a calibration solution 01' a series of dilutions
extrapolate the line obtained until it intersects the x-axis. At of the element to be analysed in various matrices, determine
the intersection, the concentration Cr of the test solution in the slope of the calibration curve bu from the following
the ion to be determined is given by the equation: equation:
C T = CsVs
VT bo _ 1_ = I{f
{LM C
METHOD III (SINGLE STANDARD ADDITION)
fAM absorption coefficient of the matrix M, calculated
To a volume VT of the test solution prepared as prescribed
or measured,
in the monograph, add a volume Vs of a reference solution
containing an amount of the ion to be determined known to I{f net impulse rate,
give a response situated in the linear part of the calibration C concentration of the element to be assayed in the
curve. Prepare a blank solution in the same conditions. standard preparation.
Measure at least three times the potentials of the test solution
and the blank solution, before and after adding the reference Mass absorption coefficent of the matrix of the sample. If the
solution. Calculate the concentration CT of the ion to be empirical formula of the sample to be analysed is known,
analysed using the following equation and making the calculate its mass absorption coefficient from the known
necessary corrections for the blank: elemental composition and the tabulated elemental mass
absorption coefficients. rf the elemental composition is
GsVs unknown, determine the mas s absorption coefficient of the
CT = --~fO.~E~--~~------
lOs (VT + Vs) - VT sample matrix by measuring the intensity of the scattered
X-radiation fu (Compton scattering) from the following
equation:
Vr volume of the test solution or the blank,
1
CT concentration of the ion to be determined in the -- = a+ bIu
{LMP
test solution,
added volume of the reference solution, fAMP mass absorption coefficient of the sample,
concentration of the ion to be determined in the fu scattered X-radiation.
reference solution,
f',.E difference between the average potentials measured Determinatian of the net pulse rate of the element to be
before and after adding Vs' determined in the sample. Calculate the net impulse rate IJip
of the element to be determined from the measured intensity
S slope of the electro de determined experimentally,
of the fluorescence line and the intensity of the background
at constant temperature, by measuring the
line(s), allowing for any tube contaminants present.
difference between the potentials obtained from
two reference solutions whose concentrations Calculation of the trace contento If the concentration of the
differ by a factor of ten and are situated within the element is in the linear part of the calibration curve, it can be
range where the calibration curve is linear. calculated using the following equation:

C =
b
I:
p
1
xf
0112008:20237 °t-LMP

2.2.37. X-RAY FLUORESCENCE f dilution factor.


SPECTROMETRY(3)
Wavelength dispersive X-ray fluorescence spectrometry is a 0112008:20238
procedure that uses the measurement of the intensity of the
fluorescent radiation emitted by an element having an atomic 2.2.38. CONDUCTIVITY
number between 11 and 92 excited by a continuous primary
X -ray radiation. The intensity of the fluorescence produced by The current f (in amperes) flowing in a conductor is directly
a given element depends on the concentration of this element proportional to the applied electromotive force E (in volts)
in the sample but also on the absorption by the matrix of the and inversely proportional to the resistan ce R (in ohms) of
incident and fluorescent radiation. At trace levels, where the the conductor:
calibration curve is linear, the intensity of the fluorescent
radiation emitted by an element in a given matrix, at a given I= ~
R
(3) G. Andermann & M.W Kemp, An,alytical Chemistry 301306 (1958). Z.I-I. Kalman & 1. I-Ieller, AnaJ;.rtical Chemistry 34 946 (1962). R.C Reyoolds, Jr., The American Mineralogist46
1133 (1963). R.O. Müller, Spectrochirmca Acta 20 143 (1964). R.O. Müller, Spectrochemische Analyse mit Rontgení1uoreszenz, R. Oldenburg MÚllchen- V'lien (1967).

General Notices (1) apply to all monographs and other texts 59


2.2.39. Molecular mass distribution in dextrans EUROPEAN PHARMACOPOElA 8.0

The conductivity (formerly called specific conductance) of If the determination of the ceH constant is carried out at a
a solution (K) is, by definition, the reciprocal of resistivity different temperature than that indicated for the certified
(p). Resistivity is defined as the quotient of the electric field reference material, the conductivity value may be calculated
and the density of the current. The resistance R (in O) of a from the following expression:
conductor of cross-section S (in cm 2 ) and length L (in cm) is
given by the expression: K,T = "'TCRM x [1 + oc (T ~ TCRM)]
L KT vallle of conductivity at the different temperature,
R=p~
S
KTCRiVI = value of condllctivity of the certified reference
material,
1 L 1 L T temperature set fol' calibration,
thus: R = -;¿ x S or r;, = R x S
temperature indicated for the certified reference
LIS corresponds to the ideal cell constant. material,
The unit of conductivity in the International System is a temperature coefficient for the conductivity value
the siemens per metre (S·m- 1). In practice, the electrical of the certified reference material; for potassium
conductivity of a solution is expressed in siemens per chloride a = 0.021.
centimetre (S·cm- l ) or in microsiemens per centimetre
Determi.nation of the conductivity of the solution to be
(flS·cm- l ). The unit of resistivity in the International System is
examined
the ohm-metre (Üm). The resistivity of a solution is generally
expressed in ohm-centimetres (ücm). Unless otherwise After calibrating the apparatus with a certified reference
prescribed, the reference temperature for the expression of material solution, rinse the conductivity ceU several times with
conductivity or resistivity is 25 oc. distilled water R and at least twice with the aqueous solution to
be examined. Carry out successive measurements as described
The apparatus and operating procedure described below are in the monograph.
applicable to laboratory measurement of conductivity greater
than 10 flS.cm- l . The measurement of conductivity of water is 01/2008:20239
dealt with in the relevant monographs.
2.2.39. MOLECULAR MASS
APPARATUS
DISTRIBUTION IN DEXTRANS
The apparatus used (conductivity meter or resistivity meter)
measures the resistan ce of the column of liquid between the Examine by size-exclusion chromatography (2.2.30).
electro des of the immersed measuring device (conductivity Test solution. Dissolve 0.200 g of the substance to be examined
cell). The apparatus is supplied with alternating current to in the mobile phase and dilute to 10 mL with the mobile phase.
avoid the effects of electrode polarisation. It is equipped with Marker solution. Dissolve 5 mg of glucose R and 2 mg of
a temperature probe and a temperature compensation device. dextran V o CRS in 1 mL of the mobile phase.
The conductivity ceH contains 2 parallel platinum electro des Calibration solutions. Dissolve separate!y in 1 mL of the
coated with platinum black, each with a surface are a S, and mobile phase 15 mg of dextran 4 for calibration CRS, 15 mg
separated from the other by a distan ce L. Both are generally of dextran 10 for calibratíon CRS, 20 mg of dextran 40 for
protected by a glass tube. Other types of cells may also be used. calibratíon CRS, 20 mg of dextran 70 for calibration CRS and
20 mg of dextran 250 for calibration CRS.
OPERATING PROCEDURE System suitability so/utíon. Dissolve either 20 mg of dextran 40
for performance test CRS (for dextran 40) or 20 mg of
Determination of the ceH constant dextran 60/70 for performance test CRS (for dextran 60 and
Choose a conductivity cell that is appropriate fol' the dextran 70) in 1 mL of the mobile phase.
properties and conductivity of the solution to be examined. The chromatographic procedure may be carried out using:
The higher the expected conductivity, the higher the cel! - a column 0.3 m long and 10 mm in internal diameter,
constant that must be chosen (low p). Commonly used packed with cross-linked agarose for chromatography R or
conductivity cells have cel! constants of the order of 0.1 cm - 1, a series of columns, 0.3 m long and 10 mm in internal
1 cm- l and 10 cm- l. Use a certified reference material, for diameter, packed with polyether hydroxylated gel for
example a solution of potassium chloride, that is appropriate chromatography R,
for the measurement. The conductivity value of the certified
- as the mobile phase, at a flow rate of 0.5-1 mL/min, kept
reference material, should be near the expected conductivity
constant to ± 1 per cent per hour, a solution containing 7 g
value of the solution to be examined. Other certified reference
of anhydrous sodium sulfate R and 1 g of chlorobutanol R
materials may be used especially for cells having a constant of
in 1 L of water R,
0.1 cm - l. Rinse the ceH several times with distilled water R
and at least twice with the certified reference material used - as detector a differential refractometer,
for the determination of the ceH constant of the conductivity - a 100 I1L to 200 flL loop injector,
cell. Measure the resistance of the conductivity cell using the maintaining the system at a constant temperature (± 0.1 OC).
certified reference material at 25 ± 1 oc. The ceH constant Kcdl
(in cm- 1) depends on the geometry of the condllctivity ceU CALIBRATION OF THE CHROMATOGRAPHIC SYSTEM
and is given by tbe expression: Carry out replicate injections of the chosen volume of the
marker solution. The chromatogram shows 2 peaks, the first
Kcell = RCRM X K,CRM of which corresponds to dextran V o CRS and the second of
which corresponds to glucose R. From the elution volume
RCRi',f measured resistance, expressed in mega-ohms, of the peak corresponding to dextran Vo' calculate the void
volume V o and from the peak corresponding to dextros e,
KCRM conductivity of the certified reference material calculate the total volume V"~
solution used, expressed in microsiemens per
centimetre. Inject the chosen volume of each of the calibration solutions.
Draw carefully the baseline of each of the chromatograms.
The measured constant Kccli of the condllctivity cel! must be Divide each chromatogram into p (at least 60) equa! vertical
within 5 per cent of the value indicated. sections (corresponding to equa! e1ution volumes). In each

60 See the information section on general monographs (cover pages)


EUROPEAN PHARMACOPOElA 8.0 2.2.39. Molecular mass distribution i.n dextrans

section i, corresponding to an elution volume V¡ measure the


M
height (y¡) of the chromatogram Hne aboye the baseline and
calculate the coefficient of distribution K¡ using the expression: ,,
,,
(Vi - Vol ,,
(Vt - Vol (1) 106
,,
Vo void volume of the column, determined using ,
the peak corresponding to dextran V o CRS in the ,
chromatogram obtained with the marker solution, ,,
Vt total volume of the column, determined using
,,
,
the peak corresponding to glucose in the '11\. Dextran 250
chromatogram obtained with the marker solution, 105

V¡ elution volume of section i in the chromatogram IIiiI. Dextran 70


obtained with each of the calibration solutions.
Carry out the calibration using either of the following
methods.
" '\ Dextran 40

Calibration by plotting of the curve. For each of the dextrans


for calibration calculate the coefficient of distribution Km""
corresponding to the maximum height of the chromatographic
104 \ Oextran 10 ==
line, using expression (1). Plot on semilogarithmic paper the
values of Km", (on the x-axis) against the declared molecular
mass at the maximum height of the chromatographic line \. Dextran 4

(MmaJ of each of the dextrans for calibration and glucose. \


Draw a first calibration curve through the points obtained,
extrapolating it from the point Kmax obtained with dextran 250
\
103
for calibration CRS to the lowest K value obtained for this CRS
(Figure 2.2.39.-1). Using this first calibration curve, transform,

1\
for each chromatogram, al! K¡ values into the corresponding
molecular mass Mi' thus obtaining the molecular mass
distribution. Calculate for each dextran for calibration the
250
average molecular mass Mw using equation (3) below. If the Glucose/dextrose
calculated values for Mw do not differ by more than 5 per cent 7J 4C
1P 4
from those declared for each of the dextrans for calibration
and the mean difference is within ± 3 per cent, the calibration Ql Q2 Q3 0.4 Q5 Q6 Q7 Q8 Q9 1.0 K
curve is approved. If not, move the calibration curve along the
y-axis and repeat the procedure aboye until the calculated and
Figure 2.2.39.-1. - Example of a calibratíon curve.
the declared values for Mw do not differ by more than 5 per
cent.
The dotted line corresponds to the part of the curve that is
Calibration by calculation of the curve. Calculate from extrapolated. Horizontallines at the bottom of the figure
equations (2) and (3) below, using a suitable method(4), values represent the width and the position of the chromatographic
for bl' b2, b3, b4 and bs that give values of Mw within 5 per cent line obtained with each of the dextrans for calibratían.
of the declared values of each of the dextrans for calibration
and 180 ± 2 for glucose:

SYSTEM SUITABILITY
(2)
Inject the chosen volume of the appropriate system suitability
solution.
Average molecular mas s of dextran for performance
test CRS. Calculate the average molecular mass Mw as
(3)
indicated under Calibration of the chromatographic system,
using either the plotted calibration curve or the values
obtained aboye for bl' b2 , b3 , b 4 and bs' The test is not valid
p number of sections dividing the chromatograms, unless MIV is:
height of the chromatographic line aboye the
baseline in section i, - 41 000 to 47 000 (dextran 40 for performance test CRS),
molecular mass in section i. 67000 to 75 000 (dextran 60/70 for performance test CRS).

(4) An iterative method such as the Gauss-Newton method modified by Hartley is suitable (see O. Hartley, Tecnometrics, 3 (1961) and G. Nilsson and K. Nilsson, J. Chromat. 101, 137
(1974)). A curve-fitting prograrnme for microcomputers, capable afnan-linear regression, may be used.

General Notíces (1) apply to all monographs and other texts 61


2.2.40. Near-infrared spectl'Oscopy EUROPEAN PHARMACOPOEIA 8.0

Average molecular mass of the 10 per cent high-fraction 01/2014:20240


dextran. Calculate Mw for the 10 per cent high-fraction
dextran eluted through section n using the equation: 2.2.40. NEAR-INFRARED
SPECTROSCOPY
AJw == '--i=--'l=-cnc--_ _ Near-infrared (NIR) spectroscopy is a technique with wide
¿Yi and varied applications in pharmaceutical analysis. The
i=l (4) NIR spectral range extends from 780 nm to 2500 11m (from
12800 cm- I to 4000 cm- l ). NIR spectra are dominated
by C-H, N-H, O-H and S-H overtones and combinations
of fundamental mid-infrared (MIR) vibrations. They
in which n is denned by the expressions: contain composite chemical and physical information
and in most cases this information can be extracted by
suitable mathematical data treatment. NIR bands are much
weaker than the fundamental MIR vibrations from which
(5)
they originate. Because absorptivities in the NIR range are
low, radiation can penetrate up to several millimetres into
n+l
~Yi > 0.1 ~Yi
(P) materials, including solids. Furthermore, many materials such
as glass are relatively transparent in this region.
(6)
Measurements can be made directly in situ, in addition to
standard sampling and testing procedures. NIR measurements
P number of sections dividing the chromatograms, can be performed off-line, and also at-line or in-line, and
Yi height of the chromatographic line aboye the on-line for process analytical technology (PAT). Suitable
baseline in section i, chemometric methods may be required for identification.
molecular mass in section i. However, when the specificity criteria for a qualitative method
are met, chemical identincation or solid-state characterisation
The test is not valid unless Mw of the 10 per cent high fraction is possible by direct comparison of the untreated or pre-treated
dextran is: spectra obtained with the chemical substance being examined
with a spectrum of a reference substance.
- llO 000 to 130000 (dextran 40 for performance test CRS), NIR spectroscopy has a wide variety of applications for
- 190000 to 230 000 (dextran 60/70 for performance test CRS). chemical, physical and process analysis, for example:
Chemical analysis:
Average molecular mass of the 10 per cent low-fraction
- identification of active substances, excipients, dosage forms,
dextran. Calculate Mw for the 10 per cent low-fraction dextran
manufacturing intermediates, chemical materials and
eluted in and after section m using the expression:
packaging materials;
- qualification of active substances, excipients, dosage forms,
]v[w == _i=_'m--::_ __
manufacturing intermediates and packaging materials,
p including batch-to-batch spectral comparison and supplier
¿ Yi change assessment;
i=m (7)
- quantification of active substances in a sample matrix,
determination of chemical values such as hydroxyl value,
determination of absolute water content, determination of
in which m is denned by the expressions: degree of hydroxylation and control of solvent content.
Physical analysis:
- crystalline form and crystallinity, polymorphism, solvates,
particle size;
(8)
- disintegration, hardness;
- film properties.
Process analysis:
(9) - monitoring of unit operations such as synthesis,
crystallisation, blending, drying, granulation and coating,
P number of sections dividing the chromatograms, for the purpose of process control;
Yi height of the chromatographic line aboye the - control and endpoint detection.
baseline in section i, Measurements in the NIR region are influenced by many
Mi molecular mass in section i. chemical and physical factors as described below; the
reproducibility and relevan ce of results depend on control of
The test is not valid unless Mw of the 10 per cent low-fraction these factors and measurements are usually valid only for a
dextran is: defined calibration model.

- 6000 to 8500 (dextran 40 for performance test CRS), APPARATUS


AH NIR measurements are based on passing light through
- 7000 to 11 000 (dextran 60/70 for performance test CRS). or into a sample and measuring the attenuation of the
emerging (transmitted or reflected) beam. Spectrometers for
MOLECULAR MASS DISTRIBUTION OF THE DEXTRAN measurement in the NIR regio n consist of a suitable light
TO BE ANALYSED source (such as a highly-stable quartz-tungsten lamp), a
monochromator or interferometer, and a detector. Common
Inject the chosen volume of the test solution and calculate Mw monochromators are acousto-optic tunable nlters (AOTF),
of the total molecular mass distribution, Mw of the 10 per cent gratings or prisms. Traditionally, many NIR instruments
high-fraction dextran and Mw of the 10 per cent low-fraction have a single-beam design, though some process instruments
dextran as indicated under System suitability. use internal referencing and can therefore be dual-beam

62 See the information section on general monographs (ca ver pages)


EUROPEAN PHARMACOPOEIA 8.0 2.2.40. Near-infrared spectroscopy

(for example in dio de array instruments). Silicon, lead The resulting spectrum can be presented directly in terms of
sulfide, and indium gallium arsenide are examples of detector transflectance (T*) and/or absorbance (A *) (y-axis) versus the
materials. Conventional cuvette sample holders, fibre-optic wavelength or wavenumber (x-axis).
probes, transmission dip cells, neutral borosilicate vials and
spinning or traversing sample holders are a few examples of T* =.i
sampling devices. The selection is based on the intended h
application, payil1g particular attention to the suitability of
the sampling system for the type of sample to be analysed. 1 intensity of transflected radiation measured from
Suitable data processing and evaluation units (e.g. software the sample;
and computer) are usually part of the system. IT intensity of transflected radiation of the reference
It is common to express the wavelength (71.) in nanometres material as background;
and the wavenumber (v) in reciprocal centimetres (cm- l ),
depending on the measurement technique and apparatus.
Conversion between nm and cm- l is performed according
A* = 10glO ( ;* ) = 10glO ( 1; )
to the following expression:
SAMPLE PREPARATION/PRESENTATION
Sample preparation and presentation may vary according
to the measurement mode. The following requirements are
necessary for al! sampling techniques :
MEASUREMENT METHODS - optimise the measuring time and number of scans to
optimise the signal-to- noise ratio;
Transmíssion mode. Transmittance (T) is a measure of the
decrease in radiation intensity at given wavelengths when - find the best suitable measurement mode for the
radiation is passed through the sample. The sample is placed intended application (transmission, diffuse reflection or
in the optical beam between the source and the detector. transflection) ;
The arrangement is analogous to that in many conventional - find the best orientation of the sample (e.g. to minimise the
spectrometers. The resulting spectrum can be presented impact of debossing on tablets);
directly in terms of transmittance (T) and/or absorbance (A) - find the best suitable accessory (e.g. transmission ceU or
(y-axis) versus the wavelength or wavenumber (x-axis). immersion probe);
- optimise pathlength in transmission and transflection
T= i modes;
lo
- find a suitable spectroscopic background reference material;
lo intensity of incident radiation; - show that the background reference material is reliable
over time and that the measurement of the background is
1 intensity of transmitted radiation; reproducible and stable over time;
- when measuring moving materials or samples (for
(~ ) = 10glO ( ~ )
process-related measurements) it is important to obtain a
A =- 10glO T = 10glO representative spectrum (e.g. by adjusting the measuring
time, the number of scans, co-adding individual spectra, or
Diffuse reflection mode. The diffuse reflection mode gives increasing the beam size);
a measure of reflectance (R), the ratio of the intensity of - ensure there is no fouling of the sensor, for example with
light reflected from the sample (I) to that reflected from a build-up of material or contamination;
background or reference reflective surface (IJ Depending on - the measuring conditions (measuring time, beam size) in
the chemical composition and physical characteristics of the relation to the minimal sample size should be justified.
sample, NIR radiation can penetrate a more or less substantial In some process analysis situations it may be impossible to
distan ce into the samp!e, where it can be absorbed by the remove a probe for reference background data collection;
overtones and combinations of the fundamental vibrations various options are therefore to be considered, induding
of the analyte species present in the sample. Non -absorbed internal referencing, measurement of a background reference
radiation is partially reflected back from the sample to the using a 2nd detector, etc. Only spectra measured against a
detector. NIR reflectance spectra are typically obtained background possessing the same optical properties can be
by calculating and plotting loglo (l/R) (y-axis) versus the directly compared with one another.
wavelength or wavenumber (x-axis).
Transmission mode. The measurement of transmittance (T)
R= i is dependent on a background transmittance spectrum for its
calculation. Examples ofbackground references in dude air,
Ir
a polymeric disc, an empty cell, a solvent blank or in special
1 intensity of light diffusively reflected from the cases a reference sample. The method generally applies to
sample; liquids (diluted or undiluted), dispersions, solutions and
solids (induding tablets and capsules). For transmittance
1, intensity of light reflected from the background or measurements of solids, a suitable sample accessory is used.
reference reflective surface; Liquid samples are examined in a cell of suitable pathlength
(typically 0.5-4 mm), transparent to NIR radiation, or
alternatively by immersion of a fibre-optic probe of a suitable
configuration.
Diffuse reflection mode. This mode generally applies to
Transflection mode. This mode is a combinabon of solids. The sample is examined directly, or in a suitable device
transmittance and reflectance. In the measurement of (for example a sample holder), or in direct contact with a
transflectance (1'*), a mirror or a difEuse reflectance surface fibre-optic probe. For process monitoring, material can be
is used to reflect the transmitted radiation back through analysed through a polished window interface (e.g. sapphire),
the sample, thus doubling the pathlength. Non-absorbed or using an in-line fibre-optic probe. Care must be taken to
radiation is reflected back from the sample to the detector. ensure that the measuring conditions are as reproducible as

General Notices (1) apply to all monographs and other texts 63


2.2.40. Near-infrared spectroscopy EUROPEAN PHARMACOPOEIA 8.0

possible from one sample to another. The reflected radiation calibration model. The aim can be, for example, to reduce
of a background reference is scanl1ed to obtain the baseline, baseline variations, to reduce the impact of known variations
and then the reflectance of one 01' more analytical samples is that are interfering in the subsequent mathematical models,
measured. Common reflectance references indude ceramic, or to simplify data before use. In some cases spectra may
thermoplastic resins and gold. Other suitable materials may also be normalised or corrected for scatter, for example
be used. using standard normal variate (SNV) transformation.
Transflection rnoue. This mode generally applies to liquids, Spectral pre-treatment techniques may indude, for example,
suspel1sions and dear plastic materials. A reflector is placed windowing and noise reduction and the numerical calculation
behind the sample so as to double the pathlength. This of the first- or second-order derivative of the spectrum.
configuration can be adopted to share the same instrument Higher-order derivatives are 110t recommended because of
geometry with reflectance and fibre-optic probe systems increased spectral noise.
where the source and the detector are on the same side of the CONTROL OF INSTRUMENT PERFORMANCE
sample. The sample is examined through a cell with a mirror
Use the apparatus according to the manufacturer's instructions
or a suitable diffusive reflector, made either of metal or of
and carry out the prescribed verification at regular intervals,
an inert substance (for example, dried titanium dioxide) not
according to the use of the apparatus and the application. For
absorbing in the NIR region. Liquids can also be measured
in-line and on-line applications, the use of alternative means
using in-line transflectance probes.
of control of instrument performance must be scientifically
FACTORS AFFECTING SPECTRAL RESPONSE justified. For example, utilise the standards built into the
instrument or separate channels/probes to demonstrate
Environment. The environment temperature and humidity instrument performance (pending practicality).
must be taken into consideration before carrying out
measurements. System suitability tests may be required prior to sample
scanning, and the instrument attributes with potential
Sample presentation area. The sample presentation area or impact on suitability of the final measurement (typically
probe end must be clean of residue prior to measurement. photometric noise and wavelength accuracy) must be tested.
Similarly, the in-line or on-line interface to the sample should The frequency at which each performance test is carried out
not have significant product or contamination build-up, which must be risk-assessed depending on the instrument type and
would interfere with the desired measurement. its environment. For example, instruments placed in harsh
Sample temperature. This parameter is important for environments with variations in temperature and humidity
aqueous solutions and many liquids, where a difference of a may need frequent performance testing. Cases where the
few degrees can result in measurable spectral changes which measurement system cannot be removed such as an in-line
may have a significant effect on the analysis. Temperature probe or flow cell are also to be considered.
is also an important parameter for solids and powders Some accessories are custom designed and therefore require
containing water. adequate performance testing.
Moisture and solvent residues. Moisture and solvent residues Verification and ealibration of the wavelength or wavenumber
present in the samples will contribute significant absorption scale (exeept for fi/ter apparatus). Verify the wavelength
bands in the NIR region. scale employed, generally in the region between 780 nm and
Sample thickness. Sample thickness is a known source of 2500 nm ( 12800 cm- 1 to 4000 cm- 1) or in the intended
spectral variability and must be assessed and/or controlled, spectral range using one or more suitable wavelength
particularly for tablet and capsule analysis in t1'ansmittance standards which have characteristic maxima or minima within
mode. For the measurement of compressed powders, an the wavelength range to be used. For example, methylene
infinite thickness is typically reached after 5 mm of sample chloride R, tale R, wavelength reference lamps or a mixture
depth (e.g. in a vial). of rare-earth oxides are suitable reference materials. Other
suitable standards may also be used. Obtain a spectrum and
Sample optical properties. In solids, both surface and measure the position of at least 3 peaks distributed over the
bulk scattering properties of samples must be taken into range used. For rare-earth oxides, the National Institute of
account. Spectra of physically, chemically 01' optically Standards and Technology (NIST) provides suitable reference
heterogeneous samples may require increasing the beam materials. Fourier transform (FT) instruments have a linear
size, or examining multiple samples or spinning the sample frequency range, therefore wavelength certification at a single
to obtain a representative spectrum of the sample. Certain frequency is sufficient.
factors such as differing degree of compaction or particle size
Verification and calibration of photometric linearity.
in powdered materials and surface finish can cause significant
The photometric linearity is demonstrated with a set of
spectral differences.
transmittance or reflectance standards with known percentage
Solid-state forms. Variations in solid-state forms values of transmittance or reflectance. For reflectance
(polymorphs, hydrates, solvates and amorphous forms) measurements, carbon-doped polymer standards are
influence vibrational spectra. Hence, different crystalline available. Ensure that the absorbance of the materials used is
forms as well as the amorphous form of a solid may be relevant to the intended linear working range of the method.
distinguished from one another on the basis of their NIR Subsequent verifications of photometric linearity can use the
spectra. Where multiple crystalline forms are present, care initial observed absorbance values as the reference values.
must be taken to ensure that the calibration samples have a Non-linear calibration models and hence non-linear responses
distribution of forms relevant to the intended application. are acceptable with understanding demonstrated by the user.
Age of samples. Samples may exhibit changes in their Spectra obtained from reflectance and transmittance
chemical, physical or optical properties over time. Depending standards are subject to variability due to the differences
on the storage conditions, solid samples may either absorb between the experimental conditions under which they
or desorb water, and portions of amorphous material may were factory-calibrated and those under which they are
crystallise. Materials used for NIR calibration should be subsequently put to use. Hence, the percentage reflectance
representative of future samples and their matrix variables. values supplied with a set of calibration standards may not
be useful in the attempt to establish an 'absolute' calibration
PRE-TREATMENT OF NIR SPECTRAL DATA for a given instrument. As long as the standards do not
In many cases, and particularly for reflection mode spectra, change chemically or physically and the same reference
some form of mathematical pre-treatment of the spectrum background is used as was used to obtain the certified values,
may be useful prior to the development of a classification or subsequent measurements of the same standards under

64 See the information section on general monographs (cover pages)


EUROPEAN PHARMACOPOEIA 8.0 2.2.40. Near-infrared spectroscopy

identical conditions, including precise sample positioning, Recommendations for the conditions used to control
give information on long-term stability of the photometric instrument performance for the various measurement modes
response. A toleran ce of ± 2 per cent of the absorbance value are summarised in Table 2.2.40.-1.
is acceptable for long -term stability; this verification is only
necessary if the spectra are used without pre-treatment.
Table 2.2.40.-1 - Control of instrument performance
Measurement mode Reflection Transflection Tl'ansmission

Verification of Typical toleran ces for agreement with standard values are:
wavelength seale ± l.0 nm at 780 nm (± 16 cm- I at 12800 cm- I );
(except for filter
± l.0 nm at 1200 nm (± 8 cm- I at 8300 cm- I );
apparatus)
± l.0 nm at 1600 nm (± 6 cm- 1 at 6250 cm- I );
± l.5 nm at 2000 nm (± 4 cm- I at 5000 cm- I );
± l.5 nm at 2500 nm (± 2 cm- I at 4000 cm- I ).
For the reference material used, apply the tolerance for the neaJ'est wavelength or wavenumber for each peak used. For
dio de array instruments, most often the pixel resolution (wavelength belween pixels) can be as large as 10 nm. The pixel
resolution must be adapted to match the spectral resolution. The peak-finding algorithms are critical to wavelength accuracy.
PracticaUy, ± 2 nm is appropriate for peak wavelength accuracy using sueh instrumentation. Alternatively, refer to manufacturer's
specifications fol' acceptance.

Bench/mobile Measure tale R via a suitable medium A suspension of 1.2 g of dry titanium lvIethylene ehloride R may be used and has
instrument 01' by fibre-optic probe. Tale R has dioxide R in about 4 mL of methylene characteristic sharp bands at 1155 nm,
characteristie peaks at 948 nm, 1391 nm ehloride R is used directly with a 1366 nm, 1417 nm, 1690 nm, 1838 nm,
and 2312 nm suitable fol' ealibration. ceU or a probe. Titanium dioxide 1894 nm, 2068 nm and 2245 nm. Choose
Alternatively, other suitable standards has no absorption in the NIR l'ange. 3 peaks across the wavelength range for
may also be used that ensure wavelength Speetra are reeorded with a maximum calibration. Other suitable standards may
accuraey in the region of wOl'king nominal instrument bandwidth also be used.
methodology. For example, measure of 10 nm at 2500 nm (16 em- 1 at
an internal polystyrene standard if 4000 cm- I ). Methylene chlOl'ide has
built -in, 01' measure a NIST standard characteristie shal'p bands at 1155 nm,
01' other traeeable material, and assess
1366 nm, 1417 nm, 1690 nm, 1838 nm,
3 peaks across the wavelength range for 1894 nm, 2068 nm and 2245 nm.
calibration. Choose 3 peaks across the wavelength
range for calibration. Other suitable
standards may also be used, such as
a liquid transflection standard mixed
with titanium dioxide 01' some other
reflective medium.

Process instrument If it is nol practicaUy possible lo measure a traceable referenee material at lhe point al' sample measurement, use internal material
such as polystyrene, fibreglass 01' salvent and/or water vapour. Alternatively, adopt a second external channel/probe.
For FT instruments, the calibration of the wavenumber scale may be performed using a narrow, isolated water-vapour line, for
example, the line at 7306.74 cm- 1, or 7299.45 cm- I , 01' 7299.81 cm- I 01' a narrow line from a eertified reference material.

Verification The standard deviation of the wavelength is consistent with the specifieations of the instrument manufacturer, 01' otherwise
of wavelength scientifieaUy justified.
repeatability (exeept
for jilter apparatus)

Beneh/mobile Verify the wavelength l'epeatability using a suitable external or internal standard.
instrulnent

Proeess instrument Verify the wavelength repeatability using a suitable external 01' internal standard.

Verification of Measure 4 photometric standards across lhe working method absorbance range.
photometrie linearity
and response stability{!)

Beneh/mobile Analyse 4 reflectance standards, for Transflection measurenlents can Analyse 4 transmittance standards to cover
instrument example in the range of 10-99 per use appropriate reflectan ce or the absorbance values ayer the worldng
cent, including 10 per eent, 20 per transmittanee standards and eriteria. absOl'bance range of tbe modelled data.
eent, 40 per cent and 80 per cent. Evaluate the observed absorbance values
In some circumstances 2 per eent against the referenee absorbance values,
may be appropriate. Evaluate the for example perform a linear regression.
observed absorbanee values against the Aeceptable tolerances are l.00 ± 0.05 fOl'
referenee absorbanee values, for example the slope and 0.00 ± 0.05 fOl' the intereept
perfOl'm a linear regression. Acceptable for the 1" verifieation of photometrie
toleran ces are l.00 ± 0.05 for the slope linearity 01' an instrumento Subsequent
and 0.00 ± 0.05 fOl' the intereept for the verifications af photometric linearity can
1,1 verifieation of photometric linearity of use the initial observed absorbance values
an instrument. Subsequent verifications as the reference values.
of photometric linearity can use the
initial observed absOl'bance values as the
referenee values.

General Notices (1) apply to all monographs and other texts 65


2.2.40. Near-infrared spectroscopy EUROPEAN PHARMACOPOEIA 8.0

Measurement mode Reflection Transflection Transmission


I I
Process instru111ent If pbotometric reflectance and transmittance standards cannot be measured at tbe point of sample measurement, use the
photometric standards built into the instrument.
Process instruments can use internal photometric standards for pbotometric linearity. Follow the manufacturer's verifled
tolerances in such cases.

Verification af Determine the photometric l10ise at a relevant photometric region of the spectrum using a suitable reflectan ce standard, for
photametric noise(l) example, wbite reí1ective ceramic tiles or carbon-doped polymer standards. Follow the manufacturer's methodology and
speciflcations.

Bench/mobile Scan the reflectance low flux standard (e.g. Sor 10 per cent, carbon-doped polymer Scan the transmittance high flux standard
instrument standard) over a suitable wavelength range in accordance with the manufacturer's (e.g. 90 or 99 per cent, carbon-doped
recommendation and ca!eulate the photometric noise as peak-to-peak noise. polymer standard) over a suitable
wavelength/wavenumber range in
accordance with the ll1anufactllrer's
recommendation and ca!eulate the
photometric noise as peak-to-peak noise.

Process instru111ent As above, or if not practically possible, use the standard built into the instrument As above, or if not practically possible,
for noise testing and 111anufacturer speciflcations. use the standard built jnto the instrument
for noise testing and manufacturer
speciflcations.

(IJVerijication of photometric linearity and Verification of photometric noise are not reqnired for instruments using methods to perfonn simple
identiflcatiol1s which do not use the photometric absorbances as part of model strategy (for example, simple correlation with absorbing wavelengths).

QUALITATIVE ANALYSIS (IDENTIFICATION AND database on the basis of their mathematical correlation or
CHARACTERISATION) other suitable algorithms. A set of knowl1 reference mean
Establishment of a spectral reference library. Record speetra and the variability around this mean can be used
the spectra of a suitable number of representative samples with an algorithm for classification; alternatively, this can
of the substance which have known, traceable identities, be achieved visually by overlaying spectral data if speciflcity
and that exhibit the variation typical for the substance to is inherent. There are different techniques available, such as
be analysed (for example, solid-state form, particle size, principal component analysis (PCA), cluster analysis, and
50ft independent modelling by class analogy (SIMCA). The
etc.). Libraries are built using representative samples under
appropriate environmental conditions. The set of spectra reliability of the technique chosen for a particular application
obtained represents the information which can be used for has to be validated aecording to the following:
identification of the sample to be analysed. Validation of the modelo Identifieation methods using direct
The collection of spectra in the library may be represented in spectral comparison must be validated in aecordanee with
different ways defined by the mathematical technique used for identification method validation procedures.
identification. These may be: The validation parameters for qualitative methods are
- all individual spectra representing the substance; robustness and specificity.
- a mean spectrum of the measured batches for each chemical LIMIT ANALYSIS
substance;
Relative comparison of spectra. A calibration is not required
- if necessary, a description of the variability within the when comparing a set of spectra for limit analysis purposes,
substance spectra. such as the maximum or minimum absorbance at which an
The number of substances in the library depends on the analyte absorbs. AIso, dryer end point control may use a
specific application. All spectra in the library used have the qualitative approach around a specific absorbing wavelength.
same: Appropriate spectral ranges and pre-treatments (if used) must
- spectral range and number of data points; be shown to be fit for purpose.
- technique of measurement; Spedfidty. The relative discriminatory power for a limit test
must be demonstrated. The extent of specificity testing is
- data pre-treatment.
dependent 011 the applieation and the risks being controlled.
If sub-groups (sub-libraries) are created, the aboye criteria Variations in matrix coneentrations within the operating
are applied independently for each group. Sub-libraries range of the method must not affeet the measurement.
are individually validated. Original speetral data for the
preparation of the spectrallibrary must be archived. Caution TREND ANALYSIS
must be exercised when performing any mathematical Relative comparison of spectra. A calibration is not
transformation, as artefacts can be introduced or essential l1ecessarily required when comparing a set of spectra for
information (important with qualifieation methods) can trend analysis purposes, such as the moving block approach
be lost. The suitability of the algorithm used should be to estimate statistical parameters such as mean, median and
demonstrated by successful method validation and in all cases standard deviation. For example, blend uniformity monitoring
the rationale for the use of transform must be documented. using NIR spectroscopy has adopted such data analysis
Direct comparÍson of substance and reference spectra. approaches. Appropriate spectral ranges and algorithms must
Direet comparison of representative spectra of the substanee be used for trel1d analyses.
to be examined and of a reference substance for qualitative Specificity. The relative discriminatory power for trend
chemical or physical identification purposes may not require analysis must be demonstrated. The extent of specificity
use of a reference spectrallibrary where specificity permits. testing is dependent on the application and the risks being
Data evaluation. Direct comparison of the representative controlled. Variations in matrix concentrations within the
spectrum of the substance to be examined is made with the operating range of the method must not affect the trend
individual or mean reference spectra of all substances in the analysis.

66 See the information section on general monographs (cover pages)


EUROPEA N PHARMACOPOEIA 8.0 2.2.41. Circular diduoism

QUANTITATIVE ANALYSIS Circular dichroism is calculated using the equation:


Establishment of a spectral reference library for a .6. A
.6.e = eL - eR = - -
calibration model. Calibration is the process of constructing exl
a mathematical model to relate the response from a sample
scanned using an analytical instrument to the properties of molar circular dichroism or molar differential
the samples. Any calibration model that can be clearly defined dichroic absorptivity expressed in litre·mole-I·cm- I,
in a mathematical expression and gives suitable results can be
used. Record the spectra of a suitable number of representative
EL molar absorptivity (2.2.25) of left circularly
samples with known or future-established values of the polarised light,
attribute of interest throughout the range to be measured molar absorptivity of right circularly polarised
(for example, content of water). The number of samples light,
for calibration will depend on the complexity of the sample c concentration of the test solution in mole.litre- 1,
matrix and interferences (e.g. temperature, particle size, etc.).
AH samples must give quantitative results within a calibration optical path oí the ceH in centimetres.
interval as defined by the intended purpose of the method. The following units may also be used to characterise circular
Multiple linear regression (MLR), principal component dichroism:
regression (PCR) and partialleast squares regression (PLS) are
commonly used algorithms. For PLS or PCR calibrations, the Dissymmetry factor:
regression coefficients and/or the loadings should be plotted .6. e
g=-
and the regions of large coefficients or loadings compared e
with the spectrum of the analyte. Predicted residual error sum
of squares (PRESS) plots (or similar) are useful to facilitate the E molar absorptivity (2.2.25).
optimising of the number of PCR or PLS factors.
Molar ellipticity:
Pre-treatment of data. Wavelength selection or excluding
Certain types of instruments display directly the value of
certain wavelength ranges may enhance the accuracy and
robustness of calibration models. Wavelength compression ellipticity e, expressed in degrees. When such instruments
are used, the molar ellipticity [e] may be calculated using the
(wavelength averaging) techniques may be applied to the data.
following equation:
Model validation parameters. Analytical performance
exM
characteristics to be considered for demonstrating the
[e] = e x 1 x 10
validation of NIR methods are similar to those required for
any analytical procedure. Specific acceptance criteria for each
validation parameter must be consistent with the intended [e] molar ellipticity, expressed in
use of the method. Validation parameters for quantitative degrees.cm2.decimole- 1,
methods are accuracy, linearity, precision (repeatability and e value of ellipticity given by the instrument,
intermediate precision), robustness and specificity.
M relative molecular mas s of the substance to be
examined,
ONGOING MODEL EVALUATION e concentration of the solution to be examined
NIR models validated for use are subjected to ongoing in g/mL,
performance evaluation and monitoring of validation optical path of the cell in centimetres.
parameters.
Molar ellipticity is also related to molar circular dichroism by
the following equation:
TRANSFER OF DATABASES
4500
When databases are transferred to another instrument, [e] = 2.303.6.e- :=:::: 3300.6.e
'ir
spectral range, number of data points, spectral resolution and
other parameters have to be taken into consideration. Further Molar ellipticity is often used in the analysis of proteins and
procedures and criteria must be applied to demonstrate nucleic acids. In this case, molar concentration is expressed in
that the model remains valid with the new database or new terms of monomeric residue, calculated using the expression:
instrument. molecular mass
number of amino acids
The mean relative molecular mass of the monomeric residue
is 100 to 120 (generally 115) fOI proteins and about 330 for
nucleic acids (as the sodium salt).
0112008:20241
Apparatus. The light source (S) is a xenon lamp
(Figure 2.2.41.-1); the light passes through a double
2.2.41. CIRCULAR DICHROISM monochromator (M) equipped with quartz prisms (Pl, P2).
The linear beam from the first monochromator is split
The difference in absorbance of optically active substances into 2 components polarised at right angles in the second
within an absorption band for left and right circularly monochromator. The exit slit of the monochromator
polarised light is referred to as circular dichroism. eliminates the extraordinary beam.
The polarised and monochromatic light passes through a
Difect measurement gives a mean algebraic value: birefringent modulator (Cr): the result is alternating circularly
polarised light.
The beam then pass es through the sample to be examined (C)
and reaches a photomultiplier (PM) followed by an amplifier
!lA circular dichroic absorbance, circuit which produces 2 electrical signals: one is a direet
AL absorbance of left circularly polarised light, current Ve and the other is an alternating current at the
modulation frequency Vac characteristic of the sample to be
AR absorbance of right circularly polarised light. examined. The phase gives the sign of the circular dichroism.

General Notices (1) apply to all monographs and other texts 67


2.2.42. Density of solids EUROPEAN PHARMACOPOEIA 8.0

Figure 2.2.41.-l. - Optieal scheme of a diehrograph

The ratio VaJV e is proportional to the differential absorption - the particle density, which also includes the volume due to
L'.A which created the signal. The region of wavelengths intraparticulate pores;
normally covered by a dichrograph is 170 nm to 800 nm. - the bulk density, which further includes the interparticulate
Calibration of the apparatus void volume formed in the powder bed.
Aceuraey of absorbanee seale. Dissolve 10.0 mg of
isoandrosterone R in dioxan R and dilute to 10.0 mL with the TRUE DENS1TY
same solvento Record the circular dichroism spectrum of The true del1sity of a substance is the ratio of the mass to the
the solution between 280 nm and 360 nm. Measured at the volume of the unit ceH, exclusive of all voids that are not a
maximum at 304 nm, L'.f: is + 3.3. fundamental part of the molecular packil1g arrangement. It
The solution of (15)-(+)-1O-camphorsulfonic acid R may also is an intril1Sic property of the specified crystal structure of
be used. substance, and hence should be independent of the method of
determination. The true density is determined by calculatiol1.
Linearity of modulation. Dissolve 10.0 mg of
(15)-(+ )-lO-camphorsulfonic acid R in water R and It is obtained using crystallographic data (volume and
dilute to 10.0 mL with the same solvento Determine the exact composition of the unit cell) from, for example, X-ray
concentration of camphorsulfonic acid in the solution by diffraction data, either 011 a single crystal or by refinement of
ultraviolet spectrophotometry (2.2.25), taking the specific the crystalline structure from X-ray powder diffraction data.
absorbance to be 1.49 at 285 nm.
PARTICLE DENSITY
Record the circular dichroism spectrum between 185 11m al1d
340 nm. Measured at the maximum at 290.5 nm, L'.E is + 2.2 The particle density takes into account both the true density
to + 2.5. Measured at the maximum at 192.5 11m, L'.f: is - 4.3 al1d the intraparticulate porosity (sealed and/or experimentally
to - 5. non-accessible open pores). Thus, particle density depends on
(15)-(+)- or antipodal (lR)-( - )-ammonium lO-camphorsulfo- the value of the volume determined, which in turn depends
nate R can also be used. on the method of measurement. The particle density can be
determined using one of the 2 following methods.
The gas pycnometric density is determined by measuring
0112010:20242 the volume occupied by a known mass of powder, which is
equivalent to the volume of gas displaced by the powder using
2.2.42. DENSITY OF SOLIDS a gas displacement pycnometer (2.9.23). In gas pycnometric
density measurements, the volume determined excludes the
The density of solids corresponds to their average mass volume occupied by open pores; however, it includes the
per unit volume and typically is expressed in grams per cubic volume occupied by sealed pores or pores inaccessible to
centimetre (g/cm 3) although the International Unit is the the gas. Due to the high diffusivity of helium, which is the
kilogram per cubic metre (1 g/cm 3 == 1000 kg/m 3 ). preferred choice of gas, most open pores are accessible to
Unlike gases and liquids whose density depends only on the gas. Therefore, the gas pyenometric density of a finely
temperature and pressure, the density of a solid also depends milled powder is generally not very different from the true
on its assembly and therefore varies with the crystal structure density. Hence, this density is the best estimate of the true
and degree of crystallinity. density of an amorphous or partially crystalline sample and
When a solid is amorphous or partially amorphous, its is therefore widely applicable for processed pharmaceutical
density may further depend upon the history of preparation, powder samples.
treatment and storage. The mercury porosimeter density is also called granular density.
Therefore, unlike fluids, the densities of 2 chemically With this method the volume determined includes the volume
equivalent solids may be different, and this difference occupied by sealed pores or pores inaccessible to mercury;
reflects a difference in solid -state structure. The density of however, it includes the volume only from open pores
constituent particles is an important physical characteristic smaller than sorne size limito This pore-size limit or minimal
of pharmaceutical powders. access diameter depends 011 the maximal mercury intrusion
The density of a solid particle can assume different values pressure applied during the measurement, and under normal
depending on the method used to measure the volume of operating pressures the mercury do es not penetrate the
the particle. 1t is useful to distinguish 3 levels of expression finest pores accessible to helium. Various granular densities
of density: can be obtained from one sample since, for each applied
- the true density, which only includes the solid fraction of mercury intrusion pressure, a density can be determined that
the material; in case of crystalline material, the true density corresponds to the pore-size limit at that pressure.
is also called crystal density;

68 See the information seetion on general monographs (ca ver pages)


EUROPEAN PHARMACOPOEIA 8.0 2.2,43. Mass spectrometry

BULK AND TAPPED DENSITY - particle-beam interface: the mobile phase, which may flow
The bulk density of a powder ineludes the contribution of at arate of up to 0.6 mLlmin, is nebulised in a desolvation
interparticulate void volume. Hence, the bulk density depends chamber such that only the analytes, in neutral form, reach
on both the density of powder particles and the spatial the ion sOurce of the apparatus; this technique is used
arrangement of partieles in the powder bed. for compounds of relatively low polarity with molecular
masses ofless than 1000 Da,
The bulk density of a powder is often very difficult to measure
with good reproducibility since the slightest disturbance of - moving-belt interface: the mobile phase, which may flow
the bed may result in a new density. Thus, it is essential in at arate of up to 1 mLlmin, is applied to the surface of a
reporting bulk density to specify how the determination was moving belt; after the solvent evaporates, the components
made. to be analysed are successively carried to the ion source
The bulk density and the tapped density are determined as of the apparatus where they are ionised; this techniqlle is
mentioned in chapter 2.9.34. Bulk density and tapped density. rather poorly suited to very polar or heat-Iabile compounds.
Other types of coupling (electrospray, thermospray,
atmospheric-pressure chemical ionisation) are considered to
0112008:20243 be ionisation techniques in their own right and are described
in the section on modes of ionisation.
2,2.43. MAS S SPECTROMETRY Sllpercritical fluid chromatography/mass spectrometry.
The mobile phase, usually consisting of supercritical cm'bon
Mass spectrometry is based on the direct measurement of dioxide enters the gas state after passing a heated restrictor
the ratio of the mass to the number of positive or negative between the column and the ion source.
elementar y charges of ions (miz) in the gas phase obtained
from the substance to be analysed. This ratio is expressed in Capillary electropnoresis/mass spectrometry. The eIuent
atomic mass units (l a.m.u. = one twelfth the mas s of 12C) or is introduced into the ion source, in some cases after adding
in daltons (l Da = the mass of the hydrogen atom). another solvent so that flow rates of the order of a few
The ions, produced in the ion source of the apparatus, are microlitres per minute can be attained. This technique is
accelerated and then separated by the analyser before reaching limited by the smal! quantities of sample introduced and the
the detector. Al! of these operations take place in a chamber need to use volatile buffers.
where a pumping system maintains a vacuum of 10- 3 to
10- 6 Pa. MODES OF IONISATION
The resulting spectrum shows the relative abundance of the Electron impacto The sample, in the gas state, is ionised by
various ionic species present as a function of miz. The signal a beam of electrons whose energy (usually 70 eV) is greater
corresponding to an ion will be represented by several peaks than the ionisation energy of the sample. In addition to the
corresponding to the statistical distribution of the various molecular ion M+, fragments characteristic of the molecular
isotopes of that ion. This pattern is called the isotopic profile structure are observed. This technique is limited mainly by the
and (at least for small molecules) the peak representing need to vaporise the sample. This makes it ul1suited to polar,
the most abundant isotopes for each atom is called the heat-Iabile or high molecular mass compounds. Electron
monoisotopic peak. impact is compatible with the coupling of gas chromatography
Information obtained in mass spectrometry is essentially to mas s spectrometry and sometimes with the use oí liquid
qualitative (determination of the molecular mas s, information chromatography.
on the structure fram the fragments observed) or quantitative Chemical ianisation. This type of ionisation involves a
(using internal or external standards) with limits of detection reagent gas such as methane, ammonia, nitrogen oxide,
ranging fram the picomole to the femtomole. nitrogen dioxide or oxygen. The spectrum is characterised
INTRODUCTION OF THE SAMPLE by ions of the (M + H)' or (M - Ht types, 01' adduct ions
formed from the analyte and the gas used. Fewer fragments
The very first step of an analysis is the introduction of are produced than with electron impact. A variant of this
the sample into the apparatus without overly disturbing technique is used when the substance is heat -labile: the
the vacuum. In a common method, called direct liquid sample, applied to a filament, is very rapidly vaporised by the
introduction, the sample is placed on the end of a cylindrical Joule-Thomson effect (desorption chemical ionisation).
rad (in a quartz crucible, on a filament or on a metal surface).
This rad is introduced into the spectrometer after passing Fast-atom bombardment (FAB) ar fast-ion bombardment
through a vacuum lock where a primary intermediate ionisation (liquid secondary-ian mass spectrometry
vacuum is maintained between atmospheric pressure and the LSIMS). The sample, dissolved in a viscous matrix such as
secondary vacuum of the apparatus. glycerol, is applied to a metal surface and ionised by a beam of
Other introduction systems allow the components of a neutral atoms such as argo n or xenon or high-kinetic-energy
caesium ions. Ions of the (M + H)+ or (M - H)- types or adduct
mixture to be analysed as they are separated by an apprapriate
ions formed from the matrix or the sample are produced.
apparatus connected to the mass spectrometer.
This type of ionisation, well suited to polar and heat-Iabile
Gas chromatography/mass spectrometry. The use of compounds, allows molecular masses of up to 10 000 Da to
suitable columns (capillary or semi-capillary) allows the end be obtained. The technique can be combined with liquid
of the column to be introduced directly into the source of the chromatography by adding 1 per cent to 2 per cent of glycerol
apparatus without using a separator. to the mobile phase; however, the flow rates must be very low
Liquid chromatography/mass spectrometry. This (a few microlitres per minute). These ionisation techniques
combination is particularly useful for the analysis of polar also allow thin-Iayer chromatography plates to be analysed by
compounds, which are insufficiently volatile or too heat-Iabile applying a thin layer of matrix to the surface of these plates.
to be analysed by gas chromatography coupled with mass Field desorption and field ianisation. The sample is
spectrometry. This method is complicated by the difficulty of vaporised near a tungsten filament covered with microneedles
obtaining ions in the gas phase from a liquid phase, which (jield ionisation) or applied to this filament (jield desorption).
requires very special interfaces such as: A voltage of about 10 kV, applied between this filament and a
- direct liquid introduction: the mobile phase is nebulised, counter-electrode, ionises the sample. These two techniques
and the solvent is evaporated in front of the ion source of mainly produce molecular ions M+, and (M + H)' ions and are
the apparatus, used for low polarity and/or heat-labile compounds.

General Notices (1) apply to all monographs and other texts 69


202.43. Mass spectrometry EUROPEAN PHARMACOPOEIA 800

Matrix-assisted laser desorption ionisation (MALDI). The energy fiIter and allows the resolving power of the instrument
sample, in a suitable matrix and deposited ona metal support, to be increased appreciably. The maximum resolving power
is ionised by a pulsed laser beam whose wavelength may range of sueh an instrument (double sector) ranges from 10000 to
from UV to IR (impulses lasting from a picosecond to a few 150000 and in most cases allows the value of miz ratio s to
nanoseconds). This mode of ionisation plays an essential role be calculated accurately enough to determine the elemental
in the analysis of very high molecular mass compounds (more composition of the corresponding ions. For monocharged
than 100 000 Da) but is limited to time-of flight analysers (see ions, the mass range is from 2000 Da to 15 000 Da. Some ion s
below). may decompose spontaneously (metastable transitions) or by
Electrospray. This mode of ionisation is carried out at colliding with a gas (collision-activated dissociation (CAD))
atmospheric pressure. The samples, in solution, are introduced in field-free regions between the ion source and the detector.
into the source through a capillary tube, the end of which has Examination of these decompositions is very useful for the
a potentíal of the order of 5 kV A gas can be used to facilitate determination of the structure as well as the characterisation
nebulisation. Desolvation of the resulting microdroplets of a specific compound in a mixture and involves tandem mass
produces singly or multiply charged ions in the gas phase. spectrometry. There are many such techniques depending on
The t10w rates vary from a few microlitres per minute to the region where these decompositions occur:
1 mLlmin. This technique is suited to polar compounds and - daughter-ion mode (determination ofthe decomposition
to the investigation of biomolecules with molecular mas ses of ions of a given parent ion): B/E = constant, MIKES
up to 100 000 Da. It can be coupled to liquid chromatography (Mass-analysed Ion Kinetic Energy Spectroscopy),
or capillary electrophoresis. - parent-ion mode (determination of al! ions which by
Atmospheric-pressure chemical ionisation (APCl). decomposition give an ion with a specifie miz ratio):
Ionisation is carried out at atmospheric pressure by the B2/E = constant,
actíon of an electro de maintained at a potentíal of several - neutral-Ioss mode (detection of all the ions that lose the
kilovolts and placed in the path of the mobile phase, which is same fragment):
nebulised both by thermal etlects and by the use of a stream
BIE(l - EIEo)1I2 = constant, where Eo is the basic voltage
of nitro gen. The resulting ion s carry a single charge and are of
of the electric sector.
the (M + H)" type in the positive mode and of the (M - Ht
type in the negative mode. The high flow rates that can be Quadrupoles. The analyser consists of four parallel metal
used with this mode of ionisatíon (up to 2 mL/min) make this rods, which are cylindrical or hyperbolic in cross-section.
an ideal technique for coupling to liquid chromatography. They are arranged symmetrically with respect to the trajectory
of the ions; the pairs diagonally opposed about the axis of
Thermospray. The sample, in the mobile phase consisting
symmetry of rods are connected electrically. The potentials to
of water and organic modifiers and containing a volatile
the two pairs of rods are opposed. They are the resultant of a
electrolyte (generally ammonium acetate) is introduced in
constant component and an alternating component. The ions
nebulised form after having passed through a metal capillary
produced at the ion source are transmitted and separated by
tube at controlled temperature. Acceptable flow rates are of
varying the voltages applied to the rods so that the ratio of
the order of 1 mLlmin to 2 mLlmin. The ions of the electrolyte
continuous voltage to alternating voltage remains constant.
ionise the compounds to be analysed. This ionisation process
The quadrupoles usually have a mass range of 1 a.m.u. to
may be replaced or enhaneed by an eleetrical discharge
2000 a.m.u., but some may range up to 4000 a.m.u. Although
of about 800 volts, notably when the solvents are entirely
they have a lower resolving power than magnetic sector
organic. This technique is compatible with the use ofliquid
analysers, they nevertheless allow the monoisotopic profile of
chromatography coupled with mass spectrometry.
single charged ion s to be obtained for the entire mas s range. It
is possible to obtain spectra using three quadrupoles arranged
ANALYSERS
in series, Ql' Q2' Q3 (Q2 serves as a collision cell and is not
Differences in the performance of analysers depend mainly 011 really an analyser; the most commonly used collision gas is
two parameters: argon).
- the range over which miz ratios can be measured, ie, the The most common types of scans are the following:
mass range, - daughter-ion mode: Q¡ selects an miz ion whose fragments
- their resolving power characterised by the ability to separate obtained by collision in Q2 are analysed by Q3'
two ions of equal intensity with miz ratios differing by L'l.M, - parent-ion mode: Q3 filters onlya specific miz ratio, while
and whose overlap is expressed as a given percentage of Q¡ scans a given mass range. Only the ions decomposil1g to
valley definition; for example, a resolving power (MI L'l.M) of give the ion selected by Q3 are detected,
1000 with 10 per cent valley definition allows the separation
of miz ratio s of 1000 and 1001 with the intensity returning - neutral 1055 mode: Q¡ and Q3 scan a certain mass range
to 10 per cent aboye baseline. However, the resolving power but at an offset corresponding to the loss of a fragment
may in some cases (time-of-flight analysers, quadrupoles, characteristic of a product or family of compounds.
ion-trap analysers) be defined as the ratio between the It is also possible to obtain spectra by combining quadrupole
molecular mass and peak width at half height (50 per cent analysers with magnetic or electrostatic sector instruments;
valley definition). su eh instruments are called hybrid mass spectrometers.
Magnetic and electrostatic analysers. The ions produced Ion-trap analyser. The principIe is the same as for a
in the ion source are accelerated by a voltage V, and focused quadrupole, this time with the electric fields in three
towards a magnetic analyser (magnetic field B) or an dimensions. This type of analyser allows product-ion spectra
electrostatic analyser (electrostatic freId E), depending on the over several generations (MSIJ) to be obtained.
configuration of the instrument. They follow a trajectory of Ion-cydotron resonance analysers. Ions produced in a cel!
radius r according to Laplace's law: and subjected to a uniform, intense magnetic field move in
m B 2r2 circular orbits at frequencies which can be directly correlated
z 2V to their miz ratio by applying a Fourier transform algorithm.
This phenomenon is ealled ion-cyclotron resonance.
Two types of scans can be used to collect and measure the Analysers of this type consist of superconducting magnets and
various ions produced by the ion source: a scan of B holding are capable of very high resolving power (up to 1000 000 and
V fixed or a scan of V with constant B. The magnetíc analyser more) as well as MS" spectra. However, very low pressures are
is usually followed by an dectrie sector that acts as a kinetie required (of the order of 10- 7 Palo

70 See the information section on general monographs (cover pages)


EUROPEAN PHARMACOPOEIA 8.0 2.2.44. TOC in water for pharmaceutical use

Time-of-flight analysers. The ions produced at the ion 0112008:20244


source are accelerated at a voltage V of 10 kV to 20 kV. They
pass through the analyser, consisting of a field-free tube, 25 cm
to 1.5 m long, generally called a J7ight tube. The time (t) for 2.2.44, TOTAL ORGANIC CAREON IN
an ion to travel to the detector is proportional to the square WATER FOR PHARIVíACEUTICAL USE
root of the miz ratio. Theoretically the mass range of such an
analyser is infinite. In practice, it is limited by the ionisation Total organic carbon (TOC) determination is an indirect
or desorption method. Time-of-flight analysers are mainly measure of organic substances present in water for
used for high molecular mass compounds (up to several phannaceutical use. TOC determination can also be used
hundred thousand daltons). This technique is very sensitive (a to monitor the performance of various operations in the
few picomoles of product are sufficient). The accuracy of the preparation of medicines.
measurements and the resolving power of such instruments
may be improved considerably by using an electrostatic mirror A variety of acceptable methods is available for determining
(reflectron). TOe. Rather than prescribing a given method to be used,
this general chapter describes the procedures used to qualify
the chosen method and the interpretation of results in limit
SIGNAL ACQUISITION tests. A standard solution is analysed at suitable intervals,
depending on the frequency of measurements; the solution
There are essentially three possible modes. is prepared with a substance that is expected to be easily
Complete spectrum mode. The entire signal obtained over oxidisable (for example, sucrose) at a concentration adjusted to
a chosen mass range is recorded. The spectrum represents give an instrument response corresponding to the Toe limit
the relative intensity of the different ionic species present as to be measured. The suitability of the system is determined by
a function of miz. The results are essentially qualitative. The analysis of a solution prepared with a substance expected to
use of spectral reference libraries for more rapid identification be oxidisable with difficulty (for example, 1,4-benzoqllinone).
is possible. The various types of apparatus used to measure TOC in
Fragmentornetric rnode (Selected-ion monitoring). The water for pharmaceutical use have in common the objective
acquired signal is limited to one (single-ion monitoring (SIM)) of completely oxidising the organic molecules in the sample
or several (multiple-ion monitoring (MIM)) ions characteristic water to produce carbon dioxide followed by measurement of
of the substance to be analysed. The limit of detection the amount of carbol1 dioxide produced, the result being used
can be considerably reduced in this mode. Quantitative or to calculate the carbon concentration in the water.
semiquantitative tests can be carried out using external or The apparatus used must discriminate between organic and
internal standards (far example, deuterated standards). Such
inorganic carbon, the latter being present as carbonate. The
tests cannot be carríed out with time-of-flight analysers.
discrimination may be effected either by measuring the
Fragrnentornetric double mass spectrometry mode inorganic carbon and subtracting it fram the total carbon,
(multiple reaction monitoring (MRM)). The unimolecular or by purging inorganic carbon from the sample before
ar bimolecular decomposition of a chosen precursor ion oxidisation. Purging may also entrain organic molecules,
characteristic of the substance to be analysed is followed but such purgeable organic carbon is present in negligible
specifically. The selectivity and the highly specific nature of quantities in water for pharmaceutical use.
this mode of acquisition provide excellent sensitivity levels
Apparatus. Use a calibrated instrument installed either on-line
and make it the most appropriate for quantitative studies
or off-lineo Verify the system suitability at suitable intervals as
using suitable internal standard s (for example, deuterated
described below. The apparatus must have a limit of detectiol1
standards). This type of analysis can be performed only on
specified by the manufacturer of 0.05 mg or les s of carbon
apparatus fitted with three quadrupoles in series, ion-trap
per litre.
analysers or cyclotron-resonance analysers.
TOe water. Use highly purified water complying with the
following specifications:
CALIBRATION
- conductivity: not greater than 1.0 flS·cm- 1 at 25 oC,
Calibration allows the corresponding miz value to be
- total organic carbon: not greater than 0.1 mg/L.
attributed to the detected signa!. As a general rule, this is done
using a reference substance. This calibration may be external Depending on the type of apparatus used, the content of
(acquisition file separate from the analysis) or internal (the heavy metals and copper may be critica!. The manufacturer's
reference substance(s) are mixed with the substance to be instructions should be followed.
examined and appear on the same acquisition file). The
Glassware preparation. Use glassware that has been
number of ions or points required for reliable calibration
scrupulously cleaned by a method that will remove organic
depends on the type of analyser and on the desired accuracy
matter. Use Toe water for the final rinse of glassware.
of the measurement, for example, in the case of a magnetic
analyser where the miz ratio varies exponentially with the Standard solution. Dissolve sucrose R, dried at 105 oC for
value of the magnetic field, there should be as many points 3 h in TOe water to obtain a solution containing 1.19 mg of
as possible. sucrose per litre (0.50 mg of carbon per litre).
Test solutiol1. Using al! due care to avoid contamination,
SIGNAL DETECTION AND DATA PROCESSING collect water to be tested in an airtight container leaving
minimal head-space. Examine the water with minimum delay
10ns separated by an analyser are converted into electric to reduce contamination from the container and its closure.
signals by a detection system su eh as a photomultiplier or
System suitability solution. Dissolve 1,4-benzoquinone R in
an electron multiplier. These signals are amplified before
TOe water to obtain a solution having a concentration of
being re-converted into digital signals for data processing,
0.75 mg of lA-benzoquinone per litre (0.50 mg of carbon
allowing various functions such as calibration, reconstruction
per litre).
of spectra, automatic quantification, archiving, creation or use
of libraries of mass spectra. The various physical parameters TOe water control. Use Toe water obtained at the same time
required for the functioning of the apparatus as a whole are as that used to prepare the standard solution and the system
controlled by computer. suitability solution.

General Notices (1) apply to all monographs and other texts 71


2.2.45. Supercritical fluid chromatography EUROPEAN PHARMACOPOEIA 8.0

Control solutions. In addition to the TOC water control, The composition, pressure (density), temperature and flow
prepare suitable blank solutions or other solutions needed rate of the prescribed mobile phase may either be constant
for establishing the baseline or for calibration adjustments throughout the whole chromatographic procedure (isocratic,
following the manufacturer's instructions; run the appropriate iso dense, isothermic elution) or may vary according to a
blanks to zero the instrument. defined programme (gradient elution of the modifier, pressure
System suitability. Run the following solutions and record (density), temperature or t10w rate).
the responses: TOC water (r,J; standard solution (rJ; system Detecíors
suitability solution Calculate the percentage response Ultraviolet/visible (UV/Vis) spectrophotometers and flame
efficiency using the expression: ionisation detectors are the most commonly employed
T ss - Tw X 100 detectors. Light scattering detectors, infrared absorption
Ts - Tw spectrophotometers, thermal conductivity detectors or other
special detectors may be used.
The system is suitable if the response efficiency is not less than
85 per cent and not more than 115 per cent of the theoretical METHOD
response. Prepare the test solution(s) and the reference solution(s) as
Procedure. Run the test solution and record the response (reJ. prescribed. The solutions must be free from solid particles.
The test solution complies with the test if ru is not greater than Criteria for assessing the suitability of the system are described
rs - rw' in the chapter on Chromatographic separation techniques
The method can also be applied using on-line instrumentation (2.2.46). The extent to which adjustments of parameters of the
that has been adequately calibrated and shown to have chromatographic system can be made to satisfy the criteria of
acceptable system suitability. The location of instrumentation system suitability are also given in this chapter.
must be chosen to ensure that the responses are representative
of the water used.
0412009:20246
0112008:20245
2.2046. CHROMATOGRAPHIC
2.2.45. SUPERCRITICAL FLUID
CHROMATOGRAPHY SEPARATION TECHNIQUES
Chromatographic separation techniques are multi-stage
Supercritical fluid chromatography (SFC) is a method of
separation methods in which the components of a sample
chromatographic separation in which the mobile phase is a
are distributed between 2 phases, one of which is stationary,
. Huid in a supercritical or a subcritical state. The stationary
while the other is mobile. The stationary phase may be a
phase, contained in a column, consists of either finely divided
solid or a liquid supported on a solid or a gel. The stationary
solid particles, such as a silica or porous graphite, a chemically
phase may be packed in a column, spread as a layer, or
modified stationary phase, as used in liquid chromatography,
distributed as a film, etc. The mobile phase may be gaseous
or, for capillary columns, a cross-linked liquid film evenly
or liquid or supercritical fluid. The separation may be based
coated on the walls of the column.
on adsorption, mass distribution (partition), ion-exchange,
SFC is based 011 mechanisms of adsorption or mass etc., or may be based on differences in the physico-chemical
distribution. properties of the molecules such as size, mass, volume, etc.
APPARATUS This chapter contains definitions and calculations of common
The apparatus usually consists of a cooled pumping system, parameters and generally applicable requirements for system
an injector, a chromatographic column, contained in an oven, suitability. PrincipIes of separation, apparatus and methods
a detector, a pressure regulator and a data acquisition device are given in the following general methods:
(or an integrator or a chart recorder). - paper chromatography (2.2.26);
Pumping system - thin-layer chromatography (2.2.27);
Pumping systems are required to deliver the mobile phase - gas chromatography (2.2.28);
at a constant flow rateo Pressure fluctuatiol1s are to be - liquid chromatography (2.2.29) ;
minimised, e.g. by passing the pressurised solvent through a - size-exclusion chromatography (2.2.30);
pulse-damping device. Tubing and connections are capable of
withstanding the pressures developed by the pumping system. - supercritical fluid chromatography (2.2.45).
Microprocessor controlled systems are capable of accurately DEFINITIONS
delivering a mobile phase in either constant or varying The system suitability and acceptance criteria in monographs
conditions, according to a defined programme. In the case of have been set using parameters as defined below. With some
gradient elution, pumping systems which deliver solvent(s) equipment, certain parameters, such as the signal-to-noise ratio
from several reservoirs are available and solvent mixing can and resolution, can be calculated using software provided by
be achieved on either the low or high-pressure side of the the manufacturero It is the responsibility of the user to ensure
pump(s). that the calculation methods used in the software are equivalent
Injectors to the requirements of the European Pharmacopoeia and to
Injection may be carried out directly at the head of the column make any necessary corrections if this is not the case.
using a valve. Chromatogram
Stationary phases A graphical or other representation of detector response,
Stationary phases are contained in columns which have been effluent COl1centration or other quantity used as a measure
described in the chapters on Liquid chromatography (2.2.29) of effluent concentration, versus time or volume. Idealised
(packed columns) and Gas chromatography (2.2.28) (capillary chromatograms are represented as a sequence of Gaussian
columns). A capillary column has a maximum internal peaks 011 a baseline (Figure 2.2.46.-1).
diameter (0) of 100 [1m. Peak
Mobile phases The portion of a chromatogram recording the detector
Usually the mobile phase is carbon-dioxide which may contain response when a single component (or 2 or more unresolved
a polar modifier such as methanol, 2-propanol or acetonitrileo components) is eluted from the column.

72 See the information section on general monographs (cover pages)


EUROPEAN PHARMACOPOEIA 8.0 2.2.46. Chromatographic separation techniques

Volume (V)
Iio'
Time (1)

Figure 2.2.46.-l.

The peak may be defined by the peak area, or the peak Retention factor (k)
height (h) and the peak width at half-height (w,), or the The retention factor (also known as mass distribution ratio
peak height (h) and the peak width between the points of (D m ) or capacity factor (k')) is defined as:
inflection (w,). In Gaussian peaks (Figure 2.2.46.-1) there is
the following relationship:
k = amount of component in stationary phase = J{ ~
amount of component in mobile phase e VM

distribution constant (also known as equilibrium


distribution coefficient);
Time required for elution of a component (Figure 2.2.46.-1, Vs volume of the stationary phase;
baseline scale being in minutes). VM volume of the mobile phase.
Retention volume (VR )
The retention factor of a component may be determined from
the chromatogram using the following equation:
Volume of the mobile phase required for elution of a
component. It may be calculated from the retention time and k = tR - tM
the flow rate (F) in millilitres per minute using the following tM
equation:
Total mobile phase time (t,)
In size-exclusion chromatography, retention time of a
component whose molecules are smaller than the smallest
gel pores (Figure 2.2.46.-2).
Total mobile phase volume (V,)
Time required for elution of an unretained component In size-exclusion chromatography, retention volume of a
(Figure 2.2.46.-1, baseline scale being in minutes). In component whose molecules are smaller than the smallest
size-exclusion chromatography, the symbol tu (see below) is gel pores. It may be calculated from the total mobile phase
used. time and the flow rate (F) in millilitres per minute using the
following equation :
Hold-up volume (VM )
Vi = tt X F
Volume of the mobile phase required for elution of an
unretained component. It may be calculated from the hold-up Retention time of an unretained compollnd Uo)
time and the flow rate (F) in millilitres per minute using the In size-exclusion chromatography, retention time of a
following equation: component whose molecules are larger than the largest gel
pores (Figure 2.2.46.-2).
Retention volume of an unretained compound (Vo)
In size-exclusion chromatography, retention volume of a
In size-exclusion chromatography, the symbol Vo (see below) component whose molecules are larger than the largest gel
is used. pores. It may be calculated from the retention time of an

General Narices (1) apply ta all managraphs and ather texts 73


2.2.46. Chromatographic separati.on techniques EUROPEAN PHARMACOPOEIA 8.0

~e ~---------------------------
O
5l"M-----------
i!!
LC and GC ü
O
Q)
ID
O
N------

Size-exclusion
chromatography

107

106 <f)
<f)
ro
E
105~:>
()
Q)
o
104 ;;;;

10 3
Time (1)

Volume (V)

Figure 2.2.46.-2.

unretained compound and the flow rate (F) in millilitres per


minute using the following equation:

Vo = to x F _A

Distribution constant (Ko)


In size-exclusion chromatography, the elution characteristics
of a component in a particular column may be given by
-O_B
the distribution constant (also referred to as distribution a
coefficient), which is calculated using the following equation:
b
Ko = tR - to
tt - to
Retardation factor (R p)
_c
The retardation factor (also known as retention factor (R f )),
used in planar chromatography, is the ratio of the distance
from the point of application to the centre of the spot and
the distance travelled by the solvent front from the point of A. mobile phase front B. spot C. line of application
application (Figure 2.2.46.-3).
Figure 2.2.46.-3.
b
RF = - Plate number (N)
a
The column performance (apparent efficiency) may be
b migration distance of the component; calculated from data obtained under either isothermal,
a
isocratic or isodense conditions, depending on the
migration distan ce of the solvent front. technique, as the plate number (also referred to as number of

74 See the information section on general monographs (cover pages)


EUROPEAN PHARMACOPOEIA 8,0 2.2.46. Chromatographic separation techniques

theoretical plates), using the following equation, the values of width of the peak at one-twentieth of the peak
t R and w h being expressed in the same units : height;
d distance between the perpendicular dropped from
N = 5,54 ( tR \ 2
the peak maximum and the leading edge of the
U'h) peak at one-twentieth of the peak height,

retention time of the peak corresponding to the An As value of LO signifies symmetry, When As > LO, the peak
component; is tailing, When < LO, the peak is fronting,
width of the peak at half-height

The plate number varies with the component as well as with


the column, the column temperature, the mobile phase and
the retention time,
DweU volume (D)
The dwell volume (also known as gradient delay volume)
is the volume between the point at which the eluents meet
and the top of the column, 1t can be determined using the
following procedure,
Column: replace the chromatographic column byan
appropriate capillary tubing (e,g, 1 m x 0,12 mm),
Mobile phase:
- mobile phase A: water R;
- mobile phase B: 0,1 per cent V/V solution of acetone R;

Time Mobile phase A Mobile phase B


(min) (per cent V/V) (per cent V/V) Figure 2.2.46,-5
0-20 100 -7 O O -7 100
Resolutlon (RJ
20 - 30 O 100
The resolution between peaks of 2 components
(Figure 2,2.46,-1) may be calculated using the following
Flow rate: set to obtain sufficient back-pressure (e,g, equation:
2 mL/min),
L18 (tm - tRl)
Detection: spectrophotometer at 265 nm, Rs = --'--'-----'-
Whl + Wh2
Determine the time (tos) in minutes when the absorbance has
increased by 50 per cent (Figure 2,2.46,-4), t R2 > tRI
tRI' t R2 retention times of the peaks;
D = tD X F
peak widths at half-height
tos - 0,5t G (in minutes);
In quantitative planar chromatography, using densitometry,
pre-defined gradient time (= 20 min); the migration distances are used instead of retention times
F flow rate (in millilitres per minute), and the resolution between peaks of 2 components may be
calculated using the following equation:
LI8a (RF2 - R Fl )
Rs = -----'-------'-
'Whl + Wh2
100% - - - - - - - - - -,"" - - : : : . . . _ - - - -
,
retardation factors of the peaks;
peak widths at half-height;
a migration distance of the solvent front

Peak- to- vaHey ratio


The peak-to-valley ratio may be employed as a system
suitability criterion in a test for related substances when
baseline separation between 2 peaks is 110t achieved
l
(Figure 2.2.46,-6),
tO.5 Time
H
p/v = ---.E
Hu
Figure 2.2.46,-4
Symmetry factor (AJ height aboye the extrapolated baseline of the minor
The symmetry factor of a peak (Figure 2,2.46,-5) is calculated peak;
using the following equation: height aboye the extrapolated baseline at the lowest
point of the curve separating the minor and major
A _ WO.05
peaks,
s - 2d

General Notices (1) apply to all monographs and other texts 75


2.2.46. Chromatographic separation techniques EUROPEAN PHARMACOPOEIA 8.0

-- Figure 2.2.46.-7.
System repeatability
The repeatability of response is expressed as an estimated
percentage relative standard deviation (s,(%)) of a consecutive
series of measurements for not fewer than 3 injections or
applications of a reference solution, and is calculated using
Figure 2.2.46.-6 the following equation:
Relative retelltion (r)
Sr (%) = 1~0 L (y¡ - y)2
Relative retention is calculated as an estimate using the Y n - 1
following equation:
Yr individual values expressed as peak are a,
peak height, 01' ratio of areas by the internal
standardisation method;
y mean of individual values;
tRi retention time of the peak of interest;
tR;t retention time of the reference peak (usually n number of individual values.
the peak corresponding to the substance to be
examined) ; SYSTEM SUITABILITY
tM hold-up time. The various components of the equipment employed must
be qualified and be capable of achieving the performance
The unadjusted relative retention (rc;) is calculated using the required to conduct the test or assay.
following equatiol1: The system suitability tests represent an integral part of the
method and are used to ensure adequate performance of the
chromatographic system. Apparent efliciency, retention factor
(mass distribution ratio), resolution, relative retention and
symmetry factor are the parameters that are usually employed
Unless otherwise indicated, values for relative retentiol1 stated in assessing the performance of the column. Factors that may
in monographs correspond to unadjusted reJative retention. affect the chromatographic behaviour indude:
In planar chromatography, the retardation factors RPst and RFí - the composition, ionic strength, temperature and apparent
are used instead of tRst and tRi' pH of the mobile phase;
- flow rate, column dimensions, column temperature and
Signal-to-noise ratio (S/N)
pressure;
The short-term noise influences the precision of quantification. - stationary phase characteristics including type of
The signal-to-noise ratio is calculated using the following chromatographic support (particle-based or monolithic),
equation: partide or macropore size, porosity, specilic surface area;
- reversed -phase and other surface-modification of the
S/N = 2H stationary phases, the extent of chemical modification (as
h
expressed by end-capping, carbon loading etc.).
H height of the peak (Figure 2.2.46.-7) corresponding The following requirements and any supplementary
to the component concerned, in the chromatogram requirements given in the individual monograph are to be
obtained with the prescribed reference solution, fulfilled unless otherwise prescribed:
measured from the maximum of the peak to the - in a related substances test or assay, for a peak in the
extrapolated baseline of the signal observed over chromatogram obtained with a reference solution used for
a distance equal to at least 5 times the width at quantilication, the symmetry factor is 0.8 to 1.5, unless
half-height; otherwise prescribed;
h range of the noise in a chromatogram obtained - in an assay of an active substance where the value is 100 per
after injection or application of a blank, observed cent for apure substance, the maximum permitted relative
over a distance equal to at least 5 times the width standard deviation (s"(%)",,,J for the delined limits is
at half-height of the peak in the chromatogram calculated for a series of inj ections of the reference solution
obtained with the prescribed reference solution using the following equation:
and, if possible, situated equally around the place
where this peak would be found. Sr (%)mo" = tKB.¡ri
90%,n-l

76 See the information section on general monographs (cover pages)


EUROPEAN PHARMACOPOEIA 8.0 2.2.46. Chromatographk separation techniques

K constant (0.349), obtained from the expression relative adjustment allows a range of7-13 per cent whereas
K = !J.,Q x in which 0.6 represents a 2 per cent absolute adjustment alJows a range of 8-12 per
V2 V2 cent, the relative value therefore being the larger; for a minor
the required percentage relative standard
deviation after 6 injections for B = l.0; component at 5 per cent of the mobile phase, a 30 per cent
relative adjustment allows a range of 3.5-6,5 per cent whereas
B upper limit given in the definition of the a 2 per cent absolute adjustment allows a range of 3-7 per
individual monograph minus 100 per cent; cent, the absolute value being the larger in this case; no other
n number of replicate injections of the reference component is altered by more than 10 per cent absolute,
solution (3 : : : n S 6); pH of the aqueous component of the mobile phase: ± 0,2 pH,
Student's t at the 90 per cent probability level unless otherwise prescribed, 01' ± 1.0 pH when non-ionisable
(double sided) with n-l degrees offreedom. substances are to be examined.
Unless otherwise prescribed, the maximum permitted Concentratiol1 of salts in the buffer component of a mobile
relative standard deviation do es not exceed the appropriate phase: ± 10 per cent.
value given in Table 2,2.46.-1. This requirement does not Application volume: 10-20 per cent of the prescribed volume
apply to tests for related substances. if using fine particle size plates (2-10 [1m),
Table 2.2.46,-1. - Repeatability requirements Liquid chromatography: isocratic elution
Composition of the mobile phase: the amount of the minor
Number of individual injecHons
solvent component may be adjusted by ± 30 per cent relative
3 4 5 6 or ± 2 per cent absolute, whichever is the larger (see example
above); no other component is altered by more than 10 per
B (per cent) Maximum permitted relative standard deviation
cent absolute,
2,0 0.41 0,59 0.73 0,85 pH of tiJe aqueous component of the mobile phase: ± 0,2 pH,
2,5 0,52 0,74 0,92 1.06 unless otherwise prescribed, or ± LO pH when non-ionisable
substances are to be examined.
3,0 0,62 0,89 1.10 1.27
Concentration of salts in the buffer component of a mobile
~ in a related substances test, the limit of quantification phase: ± 10 per cent.
(corresponding to a signal-to- noise ratio of 10) is equal to Flow rate: ± 50 per cent; a larger adjustment is acceptable
or les s than the disregard limit. when changing the column dimensions (see the formula
Compliance with the system suitability criteria is required below).
throughout the chromatographic procedure, Depending on Column·parameters
various factors, such as the frequency of use of the procedure
Stationary piJase:
and experience with the chromatographic system, the analyst
chooses an appropriate verification scheme to monitor this, ~ no change of the identity of the substituent of the
stationary phase permitted (e,g, no replacement of C18
ADJUSTMENT OF CHROMATOGRAPHIC CONDITIONS by C8);
The extent to which the various parameters of a ~ particle size: maximum reduction of 50 per cent; no
chromatographic test may be adjusted to satisfy the system in crease permitted.
suitability criteria without fundamentally modifying the Column dimensions:
methods are listed below, Adjustment of conditions with
~ length: ± 70 per cent;
gradient elutions is more critical than with isocratic elutions,
since it may lead to shifts in peaks to a different step of the ~ internal diameter: ± 25 per cen!.
gradient, thus leading to the incorrect assignment of peaks, Vvhen column dimensions are changed, the flow rate may
and to the masking of peaks or a shift such that elution be adjusted as necessary using the following equation:
occurs beyond the prescribed elution time, Changes other
than those indicated require revalidation of the method, The 12d~
F2 = FI~-?
chromatographic condiÚons described have been validated lId ~
during the elaboration of the monograph.
The system suitability tests are included to verify that the F¡ flow rate indicated in the monograph, in
separation required for satisfactory performance of the test or millilitres per minute;
assay is achieved. Nonetheless, since the stationary phases are
Fl adjusted flow rate, in millilitres per minute;
described in a general way and there is such a variety available
commercially, with differences in chromatographic behaviour, l¡ length of the column indicated in the
some adjustments of the chromatographic conditions may monograph, in millimetres;
be necessary to achieve the prescribed system suitability 12 length of the column used, in milJimetres;
requirements, With reversed-phase liquid chromatographic
methods in particular, adjustment of the various parameters d¡ internal diameter of the column indicated in the
will not always result in satisfactory chromatography, In that monograph, in millimetres;
case, it may be necessary to replace the column with another d2 internal diameter of the columl1 used, in
of the same type (e,g, octadecylsilyl silica gel), which exhibits millimetres,
the desired chromatographic behaviour, The Knowledge
database 011 the EDQM website usually contains information Temperature: ± 10 oC, where the operating temperature is
011 the coJumn(s) used during monograph elaboration,
specified, unless otherwise prescribed,
For critical parameters the adjustments are defined clearly in Detector wavelength: no adjustment permitted,
the monograph to ensure the system suitability. Injection volume: may be decreased, provided detection and
Thin-Iayer chromatography and papel' chromatography repeatability of the peak( s) to be determined are satisfactory;
no increase permitted,
Composition of the mobile phase: the amount of the minor
solvent component may be adjusted by ± 30 per cent relative Liquid chromatography: gradient elution
or ± 2 per cent absolute, whichever is the larger; for a minor Adjustment of chromatographic conditions for gradient
component at 10 per cent of the mobile phase, a 30 per cent systems requires greater caution than for isocratic systems,

General Notices (1) apply ta all monagraphs and o/her texts 77


2.2.46. Chromatographic separation techniques EUROPEAN PHARMACOPOEIA 8.0

Composition of the mobile phase/gradient elution: minar 12 length of the column used, in millimetres;
adjustments of the composition of the mobile phase and the
gradient are acceptable provided that: d¡ internal diameter of the column indicated in the
monograph, in millimetres;
- the system suitability requirements are fulfilled;
d2 internal diameter of the column used, in
- the principal peak(s) elute(s) within ± 15 per cent ofthe millimetres.
indicated retention time(s); Temperature: ± 5 oC, where the operating temperature is
- the final composition of the mobile phase is not weaker in specified, unless otherwise prescribed.
elution power than the prescribed composition. Detector : 110 adjustment permitted.
Where compliance with the system suitability requirements Injection volume: may be decreased, provided detection and
cannot be achieved, it is often preferable to consider the dwell repeatability of the peak(s) to be determined are satisfactory;
volume or to change the column. no increase permitted,
Dwell volume. The configuration of the equipment employed Gas chromatography
may significantly alter the resolution, retention time and Column parameters
relative retentions described. Should this occur, it may be due
Stationary phase:
to excessive dwell volume. Monographs preferably in dude an
isocratic step before the start of the gradient programme so - particle size: maximllm reduction of 50 per cent; no
that an adaptation can be made to the gradient time points increase permitted (packed columns);
to take account of differences in dwell volume between the - film thickness: - 50 per cent to + 100 per cent (capillary
system used for method development and that actually used. columns).
It is the user's responsibility to adapt the !ength of the isocratic Column dimensions:
step to the analytical equipment used. lf the dwell volume
used during the elaboration of the monograph is given in the - length: ± 70 per cent;
monograph, the time points (t min) stated in the gradient table - internal diameter: ± 50 per cent.
may be replaced by adapted time points (te min), calculated Flow rate: ± 50 per cent.
using the following equation:
Temperature: ± 10 per cent.
Injection volume and split volume: may be adjusted, provided
detection and repeatability are satisfactory,
Supercritical fluid chromatography
D dwell volume, in millilitres;
Composition of the mobile phase: for packed columns, the
Do dwell volume used far development of the method, amount of the minor solvent component may be adjusted by
in millilitres; ± 30 per cent relative or ± 2 per cent absolute, whichever is
F flow rate, in millilitres per minute. the larger; no adjustment is permitted for a capillary column
system.
The isocratic step introduced for this purpose may be omitted Detector wavelength: 110 adjustment permitted.
if validation data for application of the method without this
Column parameters
step is available,
phase:
pH of the aqueous component of the rnobile : no
adjustment - particle size: maximum reduction of 50 per cent; no
increase (packed columns).
Concentration in the buffer component of a mobile Column dimensions:
phase: no adjustment permitted.
- length: ± 70 per cent;
Flow rate: adjustment is acceptable when changing the column - internal diameter:
dimensions (see the formula below),
± 25 per cent (packed columns);
Column parameters
± 50 per cent (capillary columns).
Stationary phase: Flow rate: ± 50 per cent.
- no change of the identity of the substituent of the Temperature: ± 5 oC, where the operating temperature is
stationary phase permitted (e.g. no replacement of C18 specified.
bye8);
Injection volume: may be decreased, provided detection and
- particle size: no adjustment permitted. repeatability are satisfactory; 110 increase permitted.
Column dimensions: QUANTIFICATION
- length: ± 70 per cent; Peaks due to solvents and reagents or arising from the
mobile phase or the sample matrix are disregarded during
- internal diameter: ± 25 per cent.
quantification,
When column dimensions are changed, the flow fate may - Detector sensitivity. The detector sensitivity is the signa!
be adjusted as necessary using the following equation: output per unit concentration or unit mass of a substance
in the mobile phase entering the detector. The relative
12 d ;
F2 = Fl -- 2 detector response factor, commonly referred to as response
lld 1 factor, expresses the sensitivity of a detector for a given
substance relative to a standard substance, The correction
F¡ flow rate indicated in the monograph, in factor is the reciprocal of the response factor.
millilitres per minute; - External standard method. The concentration of the
F2 adjusted flow rate, in millilitres per minute; component(s) to be analysed is determined by comparing
the response(s) (peak(s)) obtained with the test solution
I¡ Jength of the column indicated in the to the response(s) (peak(s)) obtained with a reference
monograph, in millimetres; solution.

78 See the information section on general monographs (cover pages)


EUROPEAN PHARMACOPOEIA 8.0 2.2.47. CapiUaryelectrophoresis

- Internal standard method. Equal amounts of a component The migration velocity of an analyte under an electric field
that will be resolved from the substance to be examined of intensity E, is determined by the electrophoretic mobility
(the internal standard) are introduced into the test of the analyte and the electro-osmotic mobility of the buffer
solution and a reference solution. The internal standard inside the capillary. The electrophoretic mobility of a solute
is chosen such that it does not react with the substance (fAcp) depends 011 the characteristics of the salute (electric
to be examined, is stable and do es not contain impurities charge, molecular size and shape) and those of the buffer in
with the same retention time as that of the substance to which the migration takes place (type and ionic strength of the
be examined. The concentration of the substance to be electrolyte, pH, viscosity and additives). The electrophoretic
examined is determined by comparing the ratio of the peak velocity (v ep ) of a solute, assul11ing a spherical shape, is given
areas OI peak heights due to the substance to be examined by the equation:
and the internal standard in the test solution with the ratio
of the peak areas or peak heights due to the substance to
be examined and the internal standard in the reference
l/ep = J.1.ep X E =
67rr¡r
x (~)
L
(-q )
solution.
q effective charge of the solute,
- Normalisation procedure. The percentage content of a
r¡ viscosity of the electrolyte solution,
component of the substance to be examined is calculated
by determining the area of the corresponding peak as a r Stoke's radius of the solute,
percentage of the total area of al! the peaks, excluding those
V applied voltage,
due to solvents or reagents or arising from the mobile
phase or the sample matrix, and those at or below the L totallength of the capillary.
disregard limit.
When an electric field is applied through the capillary filled
- Calibration procedure. The relationship between the
with buffer, a flow of solvent is generated inside the capillary,
measured or evaluated signal (y) and the quantity called electro-osmotic flow. The velocity of the electro-osmotic
(concentration, mass, etc.) of substance (x) is determined flow depends on the electro-osmotic mobility (fAco) which in
and the calibration function is ealculated. The analytieal turn depends on the charge density on the capillary internal
results are calculated from the measured signal or evaluated wall and the buffer characteristics. The electro-osmotic
signal of the analyte by means of the inverse function. velocity (v ca) is given by the equation:

In tests for related substanees for both the external standard l/eo = J.1.eo X E = (é~) X (~)
method, when a dilution of the test solution is used for
eomparison, and the normalisation proeedure, anyeorreetion E dielectric constant of the buffer,
faetors indicated in the monograph are applied (i.e. when the
response factor is outside the range 0.8-1.2). zeta potential of the capillary surface.

When the related substances test prescribes the total of The velocity of the solute (v) is given by:
impurities or there is a quantitative determination of an
impurity, it is important to choose an appropriate threshold
l/ = l/ep + l/ea
setting and appropriate conditions for the integration of the The electrophoretic mobility of the analyte and the
peak areas. In such tests the disregard limit, Le. the limit at eleetro-osmotic mobility may act in the same direction or in
or below which a peak is disregarded, is generally 0.05 per opposite directions, depending on the charge of the solute.
cent. Thus, the threshold setting of the datacollection system In normal capillary electrophoresis, anions will migrate
corresponds to at least half of the disregard limito Integration in the opposite direction to the electro-osmotic flow and
of the peak are a of any impurity that is not eompletely their velocities will be smaller than the electro-osl11otic
separated frol11 the principal peak is preferably performed by velocity. Cations will migrate in the same direction as the
valley-to-valleyextrapolation (tangential skil11). electro-osmotic flow and their velocities will be greater than
the electro-osmotic velocity. Under conditions in which
there is a fast electro-osmotic velocity with respect to the
electrophoretic velocity of the solutes, both eations and anions
can be separated in the same runo
The time (t) taken by the solute to migrate the distance (1)
frol11 the injection end of the capillary to the detection point
(capillary effective length) is given by the expression:
01/2010:20247 1 1x L
t= ~---
corrected 7.1 l/ep + l/eo (J.1.ep + J.1.eo) X V
In general, uncoated fused-silica capillaries aboye pH 3 have
negative charge due to ionised silanol groups in the inner
2.2.47. CAPILLARY wall. Consequently, the electro-osmotic flow is from anode to
ELECTROPH ORESIS(5) cathode. The electro-osmotic flow must remain constant from
run to run if good reproducibility is to be obtained in the
migration velocity of the solutes. For some applications, it may
be necessary to reduce or suppress the electro-osmotic flow by
GENERAL PRINCIPLES modifying the inner wall of the capillary or by changing the
concentration, composition and/or pH of the buffer solution.
Capillary electrophoresis is a physical method of analysis After the introduction of the sample into the capillary,
based on the migration, inside a capillary, of charged analytes each analyte ion of the sample migrates within the
dissolved in an electrolyte solution, under the influence of a background eleetrolyte as an independent zone, according
direct -current electric field. to its electrophoretic mobility. Zone dispersion, that is

(5) This chapter has undergone pharmacopoeia! barmonisation. See chapter 5.8. Pharrnacopocial harmonisation.

General Notices (1) apply to all monographs and other texts 79


2.2.47. CapiHaryelectrophoresis EUROPEAN PHARMACOPOEIA 8.0

the spreading of each solute band, results from different and degassed to avoid bubble formation that could interfere
phenomena. Under ideal conditions the sole contribution with the detection system or interrupt the electrical contact
to the solute-zone broadening is molecular diffusion of the in the capillary during the separation runo A rigorous rinsing
solute along the capillary (longitudinal diffusion). In this ideal procedure should be developed for each analytical method to
case the efficiency of the zone, expressed as the number of achieve reproducible migration times of the solutes.
theoretical plates (N), is given by:
CAPILLARY ZONE ELECTROPHORESIS
N = (Pep + Pea) X V X 1 PRINCIPLE
2 X D X L In capillary zone electrophoresis, analytes are separated in a
capillary containing only buffer without any anticonvective
D molecular diffusion coefficient of the solute in the medium. With this technique, separation takes place because
buffer. the different components of the sample migrate as discrete
In practice, other phenomena such as heat dissipation, sample bands with different velocities. The velocity of each band
adsorption onto the capillary wall, mismatched conductivity depends on the electrophoretic mobility of the solute and the
between sample and buffer, length of the injection plug, electro-osmotic flow in the capillary (see General PrincipIes).
detector ceU size and unlevelled buffer reservoirs can also Coated capillaries can be used to in crease the separation
significantly contribute to band dispersion. capacity of those substances adsorbing on fused-silica surfaces.
Separation between 2 bands (expressed as the resolution, R) Using this mode of capillary electrophoresis, the
can be obtained by modifying the electrophoretic mobility analysis of both small (Mr < 2000) and large molecules
of the analytes, the electro-osmotic mobility induced in the (2000 < M r < 100000) can be accomplished. Due to the
capillary and by increasing the efficiency for the band of each high efficiency achieved in capillary zone electrophoresis,
analyte, according to the equation: separation of molecules having only minute differences in
their charge-to-mass ratio can be effected. This separation
VN (Pepb ~ Pepa)
Rs = ----;"--"----'---':--'-- mode also allows the separation of chiral compounds by
4 Cflep + Pea) addition of chiral selectors to the separation buffer.
OPTIMISATION
pepa and pepb electrophoretic mobilities of the Optimisation of the separation is a complex process where
2 analytes separated, several separation parameters can playa major role. The main
mean electrophoretic mobility of the factors to be considered in the development of separations are
instrumental and electrolytic solution parameters.
2 analytes 7lep = ~ (Pepb + Pepa).
Instrumental parameters
APPARATUS Voltage. A Joule heating plot is useful in optimising the applied
An apparatus for capillary electrophoresis is composed of: voltage and capillary temperature. Separation time is inversely
proportional to applied voltage. However, an increase in the
- a high-voltage, controllable direct-current power supply;
voltage used can cause excessive heat production, giving rise
- 2 buffer reservoirs, held at the same level, containing the to temperature and, as a result thereof, viscosity gradients
prescribed ano die and cathodic solutions; in the buffer inside the capillary. This effect causes band
- 2 electrode assemblies (the cathode and the anode), broadening and decreases resolution.
immersed in the buffer reservoirs and connected to the Polarity. Electrode polarity can be normal (anode at the inlet
power suppIy; and cathode at the outlet) and the electro-osmotic flow will
- a separation capillary (usually made offused-silica) which, move toward the cathode. If the electrode polarity is reversed,
when used with sorne specific types of detectors, has an the electro-osmotic flow is away fr0111 the outlet and only
optical viewing window aligned with the detector. The charged analytes with electrophoretic mobilities greater than
ends of the capillary are placed in the buffer reservoirs. the electro-osmotic flow will pass to the outlet.
The capillary is filled with the solution prescribed in the Temperature. The main effect of temperature is observed on
monograph; buffer viscosity and electrical conductivity, and therefore on
- a suitable injection system; migration velocity. In sorne cases, an increase in capillary
- a detector able to monitor the amount of substances of temperature can cause a con forma tion al change in proteins,
interest passing through a segment of the separation modifying their migration time and the efficiency of the
capillary at a given time; it is usually based on absorption separation.
spectrophotometry (UV and visible) or fluorimetry, but Capillary. The dimensions of the capillary (length and
conductimetric, amperometric or mass spectrometric internal diameter) contribute to analysis time, efficiency of
detection can be useful for specific applications; indirect separations and load capacity. Increasing both effective length
detection is an alternative method used to detect and totallength can decrease the electric fields (working
non-UV-absorbing and non-fluorescent compounds; at constant voltage) which increases migration time. For a
- a thermostatic system able to maintain a constant given buffer and electric field, heat dissipation, and hence
sample band-broadening, depend on the internal diameter
temperature inside the capillary is recommended to obtain
of the capillary. The latter also affects the detection limit,
a good separation reproducibility;
depending on the sample volume injected and the detection
- a recorder and a suitable integrator or a computer. system employed.
The definition of the injection process and its automation Since the adsorption of the sample components on the capillary
are critical for precise quantitative analysis. Modes of walllimits efficiency, methods to avoid these interactions
injection in dude gravity, pressure or vacuum injection should be considered in the development of a separation
and electrokinetic injection. The amount of each sample method. In the specific case of proteins, several strategies
component introduced electrokinetically depends on its have been devised to avoid adsorption on the capillary wall.
electrophoretic mobility, leading to possible discrimination Sorne of these strategies (use of extreme pH and adsorption of
using this injection mode. positively charged buffer additives) only require modification
Use the capillary, the buffer solutions, the preconditioning of the buffer composition to prevent protein adsorption. In
method, the sample solution and the migration conditions other strategies, the internal wall of the capillary is coated
prescribed in the monograph of the considered substance. The with a polymer, covalently bonded to the silica, that prevents
employed electrolytic solution is filtered to remove partides interaction between the proteins and the negatively charged

80 See the information section on general monographs (cover pages)


EUROPEAN PHARMACOPOEIA 8.0 2.2.47. Capillaryelectrophoresis

silica surface. For this purpose, ready-to-use capillaries dodecyl sulfate and the samples are denatured by heating in
with coatings consisting of neutral-hydrophilic, cationic and a mixture of sodium dodecyl sulfate and 2-mercaptoethanol
anionic polymers are available. or dithiothreitol before injection. When non-reducing
Electrolytic solution parameters conditions are used (for example, analysis of an intact
antibody), 2-mercaptoethanol and dithiothreitol are not used.
Buffer type and concentration. Suitable buffers for capillary Separation in cross-linked gels can be optimised by modifying
electrophoresis have an appropriate buffer capacity in the the separation buffer (as indicated in the capillary zone
pH range of choice and low mobility to minimise current electrophoresis section) and controlling the gel porosity during
generation. the gel preparation. For cross-linked polyacrylamide gels, the
Matching buffer-ion mobility to solute mobility, whenever porosity can be modified by changing the concentration of
possible, is important for minimising band distortion. The acrylamide and/or the proportion of cross-linker. As a rule,
type of samplesolvent used is also important to achieve a decrease in the porosity of the gelleads to a decrease in the
on-column sample focusing, which increases separation mobility of the solutes. Due to the rigidity of these gels, only
efficiency and improves detection. electrokinetic injection can be used.
An increase in buffer concentration (for a given pH) decreases Dynamically coated gels are hydrophilic polymers, such as
electro-osmotic flow and solute velocity. linear polyacrylamide, cellulose derivatives, dextran, etc.,
Buffer pH. The pH of the buffer can affect separation by which can be dissolved in aqueous separation buffers giving
modifying the charge of the analyte or additives, and by rise to a separation medium that also acts as a molecular sieve.
changing the electro-osmotic flow. In protein and peptide These separation media are easier to prepare than cross-linked
separation, changing the pH of the buffer from aboye to polymers. They can be prepared in a vial and filled by pressure
below the isoelectric point (pI) changes the net charge of the in a wall-coated capillary (with no electro-osmotic flow).
solute from negative to positive. An increase in the buffer pH Replacing the gel before every injection generally improves
generally increases the electro-osmotic flow. the separation reproducibility. The porosity of the gels can be
increased by using polymers of higher molecular mass (at a
Organic solvents. Organic modifiers (methanol, acetonitrile, given polymer concentration) or by decreasing the polymer
etc.) may be added to the aqueous buffer to increase the concentration (for a given polymer molecular mass). A
solubility of the solute or other additives and/or to affect the reduction in the gel porosity leads to a de crease in the mobility
degree of ionisation of the sample components. The addition of the solute for the same buffer. Since the dissolution of these
of these organic modifiers to the buffer generally causes a polymers in the buffer gives low viscosity solutions, both
decrease in the electro-osmotic flow. hydrodynamic and electrokinetic injection techniques can be
Additives for chiral separations. For the separation of optical used.
isomers, a chiral selector is added to the separation buffer.
The most commonly used chiral selectors are cyelodextrins, CAPILLARY ISOELECTRIC FOCUSING
but crown ethers, polysaccharides and proteins may also be PRINCIPLE
used. Since chiral recognition is governed by the different
interactions between the chiral selector and each of the In isoelectric focusing, the molecules migrate under the
enantiomers, the resolution achieved for the chiral compounds influence of the electric field, so long as they are charged, in
depends largely on the type of chiral selector used. In this a pH gradient generated by ampholytes having pI values in
regard, for the development of a given separation it may be a wide range (poly-aminocarboxylic acids), dissolved in the
useful to test cyelodextrins having a different cavity size (a-, separation buffer.
~-, or y-cyelodextrin) or modified cyclodextrins with neutral The three basic steps of isoelectric focusing are loading,
(methyl, ethyl, hydroxyalkyl, etc.) or ionisable (aminomethyl, focusing and mobilisation.
carboxymethyl, sulfobutyl ether, etc.) groups. When using Loading step. Two methods may be employed:
modified cyelodextrins, batch-to-batch variations in the
degree of substitution of the cyelodextrins must be taken - loading in one step: the sample is mixed with ampholytes
into account since it will influence the selectivity. Other and introduced into the capillary either by pressure or
factors controlling the resolution in chiral separations are vacuum;
concentration of chiral selector, composition and pH of the - sequentialloading: a leading buffer, then the ampholytes,
buffer and temperature. The use of organic additives, such as then the sample mixed with ampholytes, again ampholytes
methanol or urea can also modify the resolution achieved. alone and finally the terminating buffer are introduced
into the capillary. The volume of the sample must be small
CAPILLARY GEL ELECTROPHORESIS enough not to modify the pH gradient.
PRINCIPLE Focusing step. When the voltage is applied, ampholytes
In capillary gel electrophoresis, separation takes place inside migrate toward the cathode or the anode, according to their
a capillary filled with a gel that acts as a molecular sieve. net charge, thus creating a pH gradient from ano de (lower pH)
Molecules with similar charge-to-mass ratios are separated to cathode (higher pH). During this step the components to be
according to molecular size since smaller molecules move separated migrate until they reach a pH corresponding to their
more freely through the network of the gel and therefore isoelectric point (pI) and the current drops to very low values.
migrate faster than larger molecules. Different biological Mobilisation step. If mobilisation is required for detection,
macromolecules (for example, proteins and DNA fragments), use one of the following methods.
which often have similar charge-to-mass ratios, can thus be
separated according to their molecular mass by capillary gel - in the first method, mobilisation is accomplished during
electrophoresis. the focusing step under the effect of the electro-osmotic
flow; the electro-osmotic flow must be small enough to
CHARACTERISTICS OF GELS allow the focusing of the components;
2 types of gels are used in capillary electrophoresis:
permanently coated gels and dynamically coated gels. - in the second method, mobilisation is accomplished by
Permanently coated gels, such as cross-linked polyacrylamide, applying positive pressure after the focusing step;
are prepared inside the capillary by polymerisation of the - in the third method, mobilisation is achieved after the
monomers. They are usually bonded to the fused-silica wall focusing step by adding salts to the cathode reservoir or
and cannot be removed without destroying the capillary. the anode reservoir (depending on the direction chosen
If the gels are used for protein analysis under reducing for mobilisation) in order to alter the pH in the capillary
conditions, the separation buffer usually contains sodium when the voltage is applied. As the pH is changed, the

General Notices (1) apply to al! monographs and other texts 81


2.2.47. Capillary dectrophoresis EUROPEAN PHARMACOPOEIA 8.0

proteins and ampholytes are mobilised in the direction of analyte migration velocity will depend only on the partition
the reservoir which contains the added salts and pass the coefficient between the micelle and the aqueous buffer. In the
detector. electropherogram, the peaks corresponding to each uncharged
The separation achieved, expressed as L'lpI, depends on the solute are always between that of the eleetro-osmotic flow
pH gradient (dpH/dx), the number of ampholytes having marker and that of the micelle (the time elapsed between these
different pI values, the molecular diffusion coefficient (D), two peaks is called the separation window). For electrically
the intensity of the electric field (E) and the variation of charged solutes, the migration velocity depends on both the
the electrophoretic mobility of the analyte with the pH partition coefficient of the solute between the micelle and the
(-d¡L/ dpH): aqueous butfer, and on the electrophoretic mobility of the
solute in the absenee of micelle.
D (dpH/dx) Since the mechanism in MEKC of neutral and weakly ionised
6pI =3x solutes is essentially chromatographic, migration of the solute
E (-d¡L/dpH)
and resolution can be rationalised in terms of the retention
OPTIMISATION factor of the solute (k'), also referred to as mass distribution
The main parameters to be considered in the development ratio (D rn ), whieh is the ratio of the number of moles of solute
of separations are: in the micelle to those in the mobile phase. For a neutral
compound, k' is given by:
Voltage. Capillary isoeJectric focusing utilises very high
electric fields, 300 V/cm to 1000 VI cm in the focusing step.
CapiUary. The electro-osmotic flow must be reduced or
suppressed depending on the mobilisation strategy (see
aboye). Coated capillaries tend to reduce the electro-osmotic
flow. tR migration time of the solute,
Solutions. The ano de buffer reservoir is filled with a solution t o analysis time of an unretained solute (determined
with a pH lower than the pI of the most acidic ampholyte by injecting an electro-osmotic flow marker which
and the cathode reservoir is filled with a solution with a pH does not enter the micelle, for instance methanol),
higher than the pI of the most basic ampholyte. Phosphoric t me micelle migration time (measured by injecting a
acid for the anode and sodium hydroxide for the cathode are micelle marker, such as Sudan III, whieh migrates
frequently used. while continuously associated in the micelle),
Addition of a polymer, such as methylcellulose, in the K partition coefficient of the solute,
ampholyte solution tends to suppress convective forces (if
any) and electro-osmotic flow by increasing the viscosity. Vs volume of the micellar phase,
Commercial ampholytes are available covering many pH VM volume of the mobile phase.
ranges and may be mixed if necessary to obtain an expanded
pH range. Broad pH ranges are used to estimate the isoelectric Likewise, the resolution between 2 closely-migrating solutes
point whereas narrower ranges are employed to improve (RJ is given by:
aceuracy. Calibration ean be done by correlating migration
time with isoeleetric point for a series of protein markers.
VN a -1 k' C:~J
1-
During the focusing step precipitation of proteins at their R., = 4 x -a- X k~ ~ 1 x
1 + k~ x (~)
isoelectric point can be prevented, if necessary, using buffer
additives such as glycerol, surfaetants, urea or zwitterionic t mc
buffers. However, depending on the concentration, urea
denatures proteins. N number of theoretical plates for one of
the solutes,
MICELLAR ELECTROKINETIC CHROMATOGRAPHY selectivity,
(MEKC)
retention factors for both solutes,
PRINCIPLE
respectively (k'll > k'J.
In micellar electrokinetic chromatography, separation takes
place in an electrolyte solution which contains a surfactant Similar, but 110t identical, equations give k' and R, values for
at a eoncentration aboye the critical micellar concentration eleetrically charged solutes.
(eme). The solute molecules are distributed between the OPTIMISATION
aqueous buffer and the pseudo-stationary phase composed of The main parameters to be considered in the development
micelles, aecording to the partition coefficient of the solute. of separations by MEKC are instrumental and electrolytic
The technique can therefore be considered as a hybrid of solution parameters.
electrophoresis and chromatography. It is a teehnique that
can be used for the separation of both neutral and charged Instrumental parameters
solutes, maintaining the efficiency, speed and instrumental Voltage. Separation time is inversely proportional to applied
suitability of capillary electrophoresis. One of the most widely voltage. However, an in crease in voltage can cause excessive
used surfactants in MEKC is the anionic surfactant sodium heat production that gives rise to temperature gradients and
dodecyl sulfate, although other surfaetants, for example viscosity gradients of the buffer in the cross-section of the
cationic surfactants such as cetyltrimethylammonium salts, capillary. This effect can be significant with high conductivity
are also used. buffers such as those containing micelles. Poor heat dissipation
The separation mechanism is as follows. At neutral and causes band broadening and decreases resolution.
alkaline pH, a strong electro-osmotic flow is generated Temperature. Variations in capillary temperature affeet the
and moves the separation buffer ions in the direction of partition coefficient of the solute between the buffer and the
the cathode. If sodium dodecyl sulfate is employed as the micelles, the critical micellar concentration and the viscosity
surfactant, the electrophoretic migration of the anionic micelle of the buffer. These parameters eontribute to the migration
is in the opposite direction, towards the anode. As a result, the time of the solutes. The use of a good cooling system improves
overall micelle migration velocity is slowed down compared to the reproducibility of the migration time for the solutes.
the bulk flow of the electrolytic solution. In the case of neutral Capillary. As in capillary zone electrophoresis, the dimensions
solutes, sinee the analyte can partition between the micelle and of the capillary (length and internal diameter) contribute to
the aqueous buffer, and has no electrophoretic mobility, the analysis time and efficiency of separations. Increasing both

82 See the information seetion on general monographs (ca ver pages)


EUROPEAN PHARMACOPOEIA 8.0 2.2.47. Capillaryelectrophoresis

effective length and totallength can decrease the electric CALCULATIONS


fields (working at constant voltage), increase migration time From the values obtained, calculate the content of the
and improve the separation efficiency, The internal d¡ameter component or components being examined. When prescribed,
controls heat dissipation (for a given buffer and electric field) the percentage content of one or more components of the
and consequently the sample band broadening, sample to be examined is calculated by determining the
Electrolytic solution parameters corrected area(s) ofthe peak(s) as a percentage of the total
of the corrected areas of al! peaks, excluding those due to
Surfactant type and concentration. The type of surfactant, solvents or any added reagents (normalisation p1'ocedure).
in the same way as the stationary phase in chromatography, The use of an automatic integration system (integrator or data
affects the resolution since it modifies separation selectivity. acquisition and processing system) is recommended.
AIso, the log k' of a neutral compound increases linearly with
the concentration of surfactant in the mobile phase. Since SYSTEM SUITABILITY
resolution in MEKC reaches a maximum when k' approaches In order to check the behaviour of the capillary electrophoresis
the value of Jtme/ta, modifying the concentration of system, system suitability parameters are used. The choice
surfactant in the mobile phase changes the resolution of these parameters depends on the mode of capillary
obtained. electrophoresis used. They are: retention factor (k') (only for
Buffer pH. Although pH do es not modify the partition micellar electrokinetic chromatography), apparent number of
coefficient of non-ionised solutes, it can modify the theoretical plates (N), symmetry factor (A,) and resolution
electro-osmotic flow in uncoated capillaries, A decrease in the (RJ In previous sections, the theoretical expressions for N and
buffer pH decreases the electro-osmotic flow and therefore R, have been described, but more practical equations that allow
increases the resolution of the neutral solutes in MEKC, these parameters to be calculated from the electropherograms
resulting in a longer analysis time. are given below.
Organic solvents. To improve MEKC separation of APPARENT NUMBER OF THEORETICAL PLATES
hydrophobic compounds, organic modifiers (methanol, The apparent number of theoretical plates (N) may be
propano!, acetonitrile, etc.) can be added to the electrolytic calculated using the expression:
solution, The addition of these modifiers usually decreases
migration time and the selectivity of the separation. Since N = 5.54 X
t
( Wh R)2
the addition of organic modifiers affects the critical micellar
concentration, a given surfactant concentration can be used
only within a certain percentage of organic modifier before the tR migration time or distan ce along the baseline from
micellisation is inhibited or adversely affected, resulting in the the point of injection to the perpendicular dropped
absence of micelles and, therefore, in the absence of partition. from the maximum of the peak corresponding to
The dissociation of micelles in the presence of a high content the component,
of organic solvent do es not always mean that the separation w¡, width of the peak at half-height.
will no longer be possible; in some cases the hydrophobic
interaction between the ionic surfactant monomer and the RESOLUTION
neutral solutes forms solvophobic complexes that can be The resolution (R,) between peaks of similar height of
separated electrophoretically. 2 components may be calculated using the expression:
Additives for chiral separations, For the separation of
enantiomers using MEKC, a chiral selector is induded in the
micellar system, either covalently bound to the surfactant or
added to the micellar separation electrolyte. Micelles that
have a moiety with chiral discrimination properties indude
salts of N-dodecanoyl-L-amino acids, bile salts, etc. Chiral migration times 01' distan ces along the
resolution can also be achieved using chiral discriminators, baseline from the point of injection to
such as cyclodextrins, added to the electrolytic solutions the perpendiculars dropped from the
which contain micellised achiral surfactants. maxima of two adjacent peaks,
w "' and W ll2 peak widths at half-height.
Other additives. Several strategies can be carried out to modify
selectivity, by adding chemicals to the buffer. The addition of When appropriate, the resolution may be calculated by
several types of cydodextrins to the buffer can also be used to measuring the height of the valley (H,) between 2 partly
reduce the interaction ofhydrophobic solutes with the micelle, resolved peaks in a standard preparation and the height of the
thus increasing the selectivity for this type of compound. smaller peak (Hp) and calculating the peak-to-valley ratio:
The addition of substances able to modify solute-micelle p Hp
interactions by adsorption on the latter, is used to improve the v Hv
selectivity of the separations in MEKC. These additives may
be a second surfactant (ionic or non-ionic) which gives rise SYMMETRY FACTOR
to mixed micelles or metallic cations which dissolve in the The symmetry factor (A) of a peak may be calculated using
micelle and for111 co-ordination complexes with the solutes. the expression:
A _ Wa.05
QUANTIFICATION , - 2d
Peak areas must be divided by the corresponding migration
time to give the corrected area in order to: Wo.os width of the peak at one-twentieth of the peak
height,
- compensate for the shift in migration time from run to run, d distance between the perpendicular dropped from
thus reducing the variation of the response, the peak maximum and the leading edge of the
- compensate for the different responses of sample peak at one-twentieth of the peak height.
constituents with different migration times.
Tests for area repeatability (standard deviation of afeas
Where an internal standard is used, verify that no peak of the or of the area/migration-time ratio) and for migration
substance to be examined is masked by that of the internal time repeatability (standard deviation of migration time)
standard. are introduced as suitability parameters, Migration time

General Notices (1) apply ta all monographs and ather texts 83


2.2A8. Raman spectrometry EUROPEAN PHARMACOPOEIA 8.0

repeatability provides a test for the suitability of the capillary or:


washing procedures. An alternative practice to avoid the lack - an interferometer with a detector that records the intensity
of repeatability of the migration time is to use migration time of the scattered light over time, and a data-handling device
relative to an internal standard. that converts the data to the frequency or wavenumber
A test fOl' the verification of the signal-to- noise ratio for a domain by a Fourier-transform calculation.
standard preparation (or the determination of the limit of
PREPARATION OF THE SAMPLE
quantification) may also be useful for the determinatiol1 of
related substances. Raman spectra can be obtained from solids, liquids and
gases either directly, or in glass containers or tubes, generally
SIGNAL-TO-NOISE RATIO
without prior sample preparation or dilution.
The detectiol1 limit and quantification limit correspond
to signal-to-noise ratio s of 3 and 10 respectively. The A major limitation of Raman spectrometry is that impurities
signal-to- noise ratio (SI N) is calculated using the expression: may cause fluorescence that interferes with the detectiol1 of
the much weaker Raman signal. Fluorescence may be avoided
S 2H by choosing a laser source with a longer wavelength, for
N h example in the near infrared, as the exciting lineo The intensity
of certain Raman lines may be enhanced in a number of
H height of the peak corresponding to the component ways, for instan ce in Resonance Raman (RR) and by Surface
concerned, in the electropherogram obtained with Enhanced Raman Spectrometry (SERS).
the prescribed reference solution, measured from Due to the narrow focus of the irradiating laser beam, the
the maximum of the peak to the extrapolated spectrum is typically obtained from only a few microlitres of
baseline of the signal observed over a distance sample. Hence, sample inhomogeneities must be considered,
equal to twenty times the width at half-height, unless the sample volume is increased, for example by rotation
h range of the background in an electropherogram of the sample.
obtained after injection of a blank, observed over
a distance equal to twenty times the width at the IDENTIFICATION AND QUANTITATION USING
half-height of the peak in the electropherogram REFERENCE SUBSTANCES
obtained with the prescribed reference solution Prepare the substance to be examined and the reference
and, if possible, situated equally around the place substance by the same procedure and record the spectra under
where this peak would be found. the same operational conditions. The maxima in the spectrum
obtained with the substance to be examined correspond
in position and relative intensity to those in the spectrum
obtainecl with the reference substance (CRS).
0112008:20248 When the spectra recorded in the solid state show differences
in the positiol1s of the maxima, treat the substance to be
2,2048, RAMAN SPECTROMETRY examined and the reference substance in the same manner
so that they crystallise or are produced in the same form,
Raman spectrometry (inelastic light scattering) is a or proceed as described in the monograph, then record the
light-scattering process in which the specimen under spectra.
examination is il'radiated with intense monochromatic light While Beer-Lambert's law is not valid for Raman spectrometry,
(u5ually laser light) and the light scattered from the specimen Raman il1tensity is directly proportional to the concentration
i5 analysed for frequency shifts. of the scattel'ing species. As for other spectroscopic techniques,
Raman spectrometry is complementary to infrared quantitatiol1 can be performed using known amounts or
spectrometry in the sense that the two techniques both probe concentrations of refel'ence substances. Owing to the small
the molecular vibrations in a material. However, Raman and spatial resolution of the technique, care must be taken to
infrared spectrometry have different relative sensitivities ensure representative samples of standards and unknowns, fol'
for different functional groups. Raman spectrometry is example by making sure that they are in the same physical
particularly sensitive to non-polar bonds (e.g. C-C single or state or by using an internal standard for liquid samples.
multiple bonds) and less sensitive to polar bonds. Hence,
IDENTIFICATION AND QUANTITATION USING
water, which has a strong infrared absorption spectrum, i5 a
SPECTRAL LIBRARIES AND STATISTICAL METHODS
weak Raman scatterer and is thus well suited as a solvent for
FOR CLASSIFICATION AND CALIBRATION
Raman spectrometry.
Control of instrument performance. Use the apparatus
Apparatus: Spectrometers fol' recording Raman spectra according to the manufacturer's instructions and carry out
typically con5i5t of the following components: the prescribed calibrations and system performance tests
- a monochromatic light source, typically a laser, with a at regular intervals, depending on the use of the apparatus
wavelength in the ultraviolet, visible or near-infrared and the substances to be examined. When using Raman
regíon, spectrometry for quantitative determinations, or when setting
- suitable optics (lens, minors or optical-fibre assembly) up spectral reference libraries for (chemometric) classification
which directs the irradiating light to and collects the or calibration, particular care should be taken to ensure
scattered light from the sample, that corrections are made or measures are taken to control
the variability in wavenumber and response-intensity of the
- an optical device (monochromator or filter) that transmits
instrumentation.
the frequency-shifted Raman scattering and prevents the
intense incident frequency (Rayleigh scattering) from Verification of the wavenumber seale. Verify the wavenumber
reaching the detector, scale of the Raman shift (normally expressed in reciprocal
centimetres) using a suitable standard which has characteristic
- a dispersing device (grating or prism monochromator) maxima at the wavenumbers under investigation, for example,
combined with wavelength-selecting slits and a detector an organic substance, an Ne lamp or Ar+ plasma lines from
(usuallya photomultiplier tube), an argon -ion laser.
or: The calibration measurement should be matched to the sample
- a dispersing device (grating or prism) combined with a type, Le. a solid calibration sample should be used for solid
multichannel detector (usually a charge-coupled device samples and a liquid calibration sample for liquid samples.
(CCD)), Choose a suitable substance (e.g. indene, cyclohexane or

84 See the information section on general monographs (caver pages)


EUROPEAN PHARMACOPOEIA 8.0 2.2.54. Isoelectric focusing

naphthalene) for which accurate wavenumber shifts have Method. Prepare and examine the sample in the same
been established (see Table 2.2.48.-1). The indene sample manner as for the establishment of the database. A suitable
can favourably be placed in an NMR tube, evacuated and mathematical transformation of the Raman spectrum may be
sealed under inert gas, and stored cool in the dark to avoid calculated to facilitate spectrum comparison or quantitative
degradation of the sample. prediction.
Table 2.2.48.-1. - Wavenumber shifts (and acceptable Comparison of the spectra or transforms of the spectra
tolerances) of cyclohexane, indene and naphthalene. or quantitative prediction of properties or amounts in
the material in question may involve the use of a suitable
cyelohexane A indene B naphthalene A chemometric or statistical classification or calib1'ation
3056.4 (± l.5)
technique.

2938.3 (± l.5)
0112008:20249
2923.8 (± l.5)

2852.9 (± l.5)
2.2.49. FALLING HALL VISCOMETER
1609.7 (± l.0) 1576.6 (± l.0)
METHOD
1444.4 (± l.0) 1552.6 (± l.0)
The determination of dynamic viscosity of Newtonian liquids
1464.5 (± l.0)
using a suitable falling ball viscometer is performed at
1266.4 (± l.0) 1205.2 (± l.0) 1382.2 (± l.0) 20 ± 0.1 oC, unless otherwise prescribed in the monograph.
1157.6 (± l.0) 1147.2 (± l.0)
The time required for a test ball to fall in the liquid to be
examined from one ring mark to the other is determined. If
1028.3 (± l.0) 1018.6 (± 1.0) 102l.6 (± l.0) no stricter limit is defined for the equipment used the result
801.3 (± 1.0) 730.5 (± l.0) 763.8 (± l.0)'
is valid only if 2 consecutive measures do not differ by more
than 1.5 per cent.
533.9 (± l.0) 513.8 (± l.0) Apparatus. The falling ball viscometer consists of: a glass
A Standard guide for Raman shift standards for spectrometer calibration
tube enclosed in a mantle, which allow precise control of
(American Society for Testing and Materials ASTM E 1840). temperature; six balls made of glass, nickel-iron or steel with
B D. A. Carter, W. R. Thompson, C. E. Taylor and J. E. Pemberton, different densities and diameters. The tube is fixed in such
Applied Spectroscopy, 1995,49 (11),1561-1576. a way that the axis is inclined by 10 ± 1° with regard to the
Verificatíon of the response-intensity seale. The absolute and vertical. The tube has 2 ring marks which define the distance
relative intensities of the Raman bands are affected by several the ball has to roll. Commercially available apparatus is
factor s including: supplied with tables giving the constants, the density of the
balls and the suitability of the different balls for the expected
- the state of polarisation of the irradiating light, range of viscosity.
- the state of polarisation of the collection optics, Method. Fill the clean, dry tube of the viscometer, previously
- the intensity of the irradiating light, brought to 20 ± 0.1 oC, with the liquid to be examined,
- differences in instrument response, avoiding bubbles. Add the ball suitable for the range of
viscosity of the liquid so as to obtain a falling time not less than
- differences in focus and geometry at sample, 30 S. Close the tube and maintain the solution at 20 ± 0.1 oC
- differences in packing density for solid samples. for at least 15 mino Let the ball run through the liquid between
Appropriate acceptance criteria will vary with the application the 2 ring marks once without measurement. Let it run again
but a day-to-day variation of ± 10 per cent in relative band and measure with a stop-watch, to the nearest one-fifth of a
intensities is achievable in most cases. second, the time required for the ball to rol! from the upper to
the lower ring mark. Repeat the test run at least 3 times.
Establishment of a spectral reference líbrary. Record the spectra
of a suitable number of materials which have been fully tested Calculate the dynamic viscosity '1 in millipascal seconds using
(e.g. as prescribed in a monograph) and which exhibit the the formula:
variation (manufacturer, batch, crystal modification, particle
1) = k (PI - P2) X t
size, etc.) typical of the material to be analysed. The set of
spectra represents the information that defines the similarity
border or quantitative limits, which may be used, e.g. to k constant, expressed in millimeter squared per
second squared,
identify the substance or control the amount formed in a
manufacturing process. The number of substances in the p¡ density of the ball used, expressed in grams per
database depends on the specific application. The collection of cubic centimetre,
spectra in the database may be represented in different ways P2 density of the liquid to be examined, expressed
defined by the mathematical technique used for classification in grams per cubic centimetre, obtained by
or quantitation. multiplying its relative density d~~ by 0.9982,
The selectivity of the database which makes it possible
to identify positively a given material and distinguish it falling time of the ball, in seconds.
adequately from other materials in the database is to be
established during the validation procedure. This selectivity 0112010:20254
must be challenged on a regular basis to ensure ongoing
validity of the database; this is especially necessary after any 2.2.54. ISOELECTRIC FOCUSING(6)
major change in a substance (e.g. change in supplier or in the
manufacturing process of the material) or in the set-up of the GENERAL PRINCIPLES
Raman instrument (e.g. verification of the wavenumber and Isoelectric focusing (IEF) is a method of electrophoresis
response repeatability of the spectrometer). that separates proteins according to their isoelectric point.
This database is then valid for use only with the originating Separation is carried out in a slab of polyacrylamide or
instrument, al' with a similar instrument, provided the agarose gel that contains a mixture of amphoteric electrolytes
transferred database has been demonstrated to remain valido (ampholytes). When subjected to an electric field, the

(6) This chapter has undergone pharmacopoeial harmonisation. See chapter 5.8. Pharmacopoeial hannonísalion.

General Notices (1) apply to all monographs and other texts 85


2.2.54. Isoelecíric focusing EUROPEAN PHARMACOPOEIA 8.0

ampholytes migrate in the gel to create a pH gradient. In sorne when the density is measured using a densitometer or similar
cases gels containing an immobilised pH gradient, prepared instrumentation to determine the relative concentration of
by incorporating weak acids and bases to specifrc regions of protein in the bands subject to validation.
the gel network during the preparation of the gel, are used.
When the applied proteins reach the gel fraction that has a pH APPARATUS
that is the same as their isoelectric point (pI), their charge is An apparatus for IEF consists of:
neutralised and migration ceases. Gradients can be made over - a controllable generator for constant potential, current
various ranges of pH, according to the mixture of ampholytes and power; potentials of 2500 V have been used and
chosen. are considered optimal under a given set of operating
conditions; a supply of up to 30 W of constant power is
THEORETICAL ASPECTS recommended;
When a protein is at the position of its isoelectric point, it - a rigid plastic IEF chamber that contains a cooled plate, of
has no net charge and cannot be moved in a gel matrix by suitable material, to support the gel;
the electric freld. 1t may, however, move from that position - a plastic cover with platinum electro des that are connected
by diffusion. The pH gradient forces a protein to remain to the gel by means of paper wicks of suitable width, length
in its isoelectric point position, thus concentrating it; this and thickness, impregnated with solutions of anodic and
concentrating effect is called "focusing". Increasing the cathodic electrolytes.
applied voltage or reducing the sample load result in improved
separation of bands. The applied voltage is limited by the ISOELECTRIC FOCUSING IN POLYACRYLAMIDE GELS:
heat generated, which must be dissipated. The use of thin gels DETAILED PROCEDURE
and an effrcient cooling plate controlled by a thermostatic The following method is a detailed description of an IEF
circulator prevents the burning of the gel whilst allowing sharp procedure in thick polyacrylamide slab gels, which is used unless
focusing. The separation is estimated by determining the otherwise stated in the monograph.
minimum pI difference (.6.pI), which is necessary to separate
2 neighbouring bands: PREPARATION OF THE GELS
Mould. The mouId (see Figure 2.2.54.-1) is composed of
D (dpH/dx) a glass plate (A) on which a polyester film (B) is placed to
ll..pI = 3 x
E (-d¡.¡/dpH) facilitate handling of the gel, one or more spacers (C), a second
glass pi ate (D) and clamps to hold the structure together.
D diffusion coefficient of the protein,
dpH pH gradient,
dx
E intensity of the electric freld, in volts per
centimetre,
d¡.¡ variation of the solute mobility with the pH in
dpH the regio n close to the pI.

Since D and - d¡.¡ for a given protein cannot be altered, the


dpH
separation can be improved by using a narrower pH range and
by increasing the intensity of the electric fieId.
Resolution between protein bands on an IEF gel prepared
with carrier ampholytes can be quite good. Improvements
in resolution may be achieved by using immobilised pH
gradients where the buffering species, which are analogous to Figure 2.2.54.-1 - Mould
carrier ampholytes, are copolymerised within the gel matrix. 7.5 per cent polyacrylamide gel. Dissolve 29.1 g of
Proteins exhibiting pIs differing by as little as 0,02 pH units acrylamide R and 0.9 g of methylenebisacrylamide R in 100 mL
may be resolved using a gel prepared with carrier ampholytes of water R. To 2.5 volumes of this solution, add the mixture
while immobilised pH gradients can resolve proteins differing of ampholytes specified in the monograph and dilute to
by approximately 0.001 pH units. 10 volumes with water R. Mix carefully and degas the solution.
PRACTICAL ASPECTS Preparation of the mouId. Place the polyester film on the
lower glass plate, apply the spacer, place the second glass
Special attention must be paid to sample characteristics and/or plate and fit the clamps. Before use, place the solution
preparation. Having salt in the sample can be problematic on a magnetic stirrer and add 0.25 volumes of a 100 giL
and it is best to prepare the sample, if possible, in deionised solution of ammonium persulfate R and 0,25 volumes of
water or 2 per cent ampholytes, using dialysis or gel filtration tetramethylethylenediamine R. Immediately fill the space
if necessary. between the glass plates of the mould with the solution.
The time required for completion offocusing in thin-layer METHOD
polyacrylamide gels is determined by placing a coloured Dismantle the mould and, making use of the polyester film,
protein (e.g. haemoglobin) at different positions on the gel transfer the gel onto the cooled support, wetted with a few
surface and by applying the electric field: the steady state is millilitres of a suitable liquid, taking care to avoid forming air
reached when all applications give an identical band pattern. bubbIes. Prepare the test solutions and reference solutions as
In sorne protocols the completion of the focusing is indicated specified in the monograph. Place strip s of paper for sample
by the time elapsed after the sample application. application, about 10 mm x 5 mm in size, on the gel and
The IEF gel can be used as an identity test when the migration impregnate each with the prescribed amount of the test and
pattern on the gel is compared to a suitable standard reference solutions. AIso apply the prescribed quantity of
preparation and IEF calibration proteins, the IEF gel can a solution of proteins with known isoelectric points as pH
be used as a limit test when the density of a band on IEF is markers to calibrate the gel. In sorne protocols the gel has
compared subjectively with the density of bands appearing in pre-cast slots where a solution of the sample is applied instead
a standard preparation, or it can be used as a quantitative test of using impregnated paper strips. Cut 2 strips of paper to the

86 See the information section on general monographs (caver pages)


EUROPEAN PHARMACOPOEIA 8.0 2.2.55. Peptide mapping

length of the gel and impregnate them with the electrolyte - the use of gel additives such as non-ionic detergents (e.g.
solutions: acid for the anode and alkaline for the cathode. The octylglucoside) or zwitterionic detergents (e.g., CHAPS or
compositions of the anode and cathode solutions are given CHAPSO), and the addition of ampholyte to the sample, to
in the monograph. Apply these paper wicks to each side of prevent proteins from aggregating or precipitating.
the gel several millimetres from the edge. Fit the cover so
that the electro des are in contact with the wicks (respecting POINTS TO CONSIDER
the anodic and cathodic poles). Proceed with the isoelectric Samples can be applied to any are a on the gel, but to protect
focusing by applying the electrical parameters described in the proteins from extreme pH environments samples should
the monograph. Switch off the current when the migration of not be applied close to either electro de. During method
the mixture of standard proteins has stabilised. Using forceps, development the analyst can try applying the protein in
remove the sample application strips and the 2 electro de 3 positions on the gel (i.e. middle and both ends); the pattern
wicks. Immerse the gel in fixing solution for isoelectric focusing of a protein applied at opposite ends of the gel may not be
in polyaClylamide gel R. Incubate with gentle shaking at identical.
room temperature for 30 mino Drain off the solution and add A phenomenon known as cathodic drift, where the pH
200 mL of destaining solution R. Incubate with shaking for 1 h. gradient decays over time, may occur if a gel is focused too
Drain the gel, add coomassie staining solution R. Incubate for long. Although not well understood, electroendoosmosis
30 mino Destain the gel by passive diffusion with destaining and absorption of carbon dioxide may be factors that lead to
solution R until the bands are well visualised against a clear cathodic drift. Cathodic drift is observed as focused protein
background. Locate the position and intensity of the bands in migrating off the cathode end of the gel. Immobilised pH
the electropherogram as prescribed in the monograph. gradients may be used to address this problem.
VARIATIONS TO THE DETAILED PROCEDURE (SUBJECT Efficient cooling (approximately 4 OC) of the bed that the gel
TO VALIDATION) lies on during focusing is important. High field strengths used
during isoelectric focusing can lead to overheating and affect
Where reference to the general method on isoelectric focusing the quality of the focused gel.
is made, variations in methodology or procedure may be
made subj ect to validation. These include:
0112010:20255
- the use of commercially available pre-cast gels and of
commercial staining and destaining kits,
- the use of immobilised pH gradients,
2.2.55. PEPTIDE MAPPING(7)
- the use of rod gels, Peptide mapping is an identity test for proteins, especially
- the use of gel cassettes of different dimensions, including those obtained by rDNA technology. It involves the chemical
ultra-thin (0.2 mm) gels, or enzymatic treatment of a protein resulting in the formation
of peptide fragments followed by separation and identification
- variations in the sample application procedure, including of these fragments in a reproducible manner. 1t is a powerful
different sample volumes or the use of sample application test that is capable of identifying almost any single amino acid
masks or wicks other than paper, changes resulting from events such as errors in the reading of
- the use of alternate running conditions, including variations complementary DNA (cDNA) sequences or point mutations.
in the electric field depending on gel dimensions and Peptide mapping is a comparative procedure because the
equipment, and the use of fixed migration times rather information obtained, compared to a reference substance
than subj ective interpretation of band stability, similarly treated, confirms the primary structure of the
- the inclusion of a pre-focusing step, protein, is capable of detecting whether alterations in structure
- the use of automated instrumentation, have occurred, and demonstrates pro ces s consistency and
gene tic stability. Each protein presents unique characteristics
- the use of agarose gels.
which must be well understood so that the scientific and
VALIDATION OF ISO-ELECTRIC FOCUSING analytical approaches permit validated development of a
PROCEDURES peptide map that provides sufficient specificity.
Where alternative methods to the detailed procedure are This chapter provides detailed assistance in the application of
employed they must be validated. The following criteria may peptide mapping and its validation to characterise the desired
be used to validate the separation: protein, to evaluate the stability of the expression construct of
cells used for recombinant DNA products and to evaluate the
- formation of a stable pH gradient of desired characteristics,
consistency of the overall process, to assess product stability
assessed for example using coloured pH markers of known
as well as to ensure the identity of the protein, or to detect the
isoelectric points,
presence of protein variant.
- comparison with the electropherogram provided with the
PepUde mapping is not a general method, but il1volves
chemical reference substance for the preparation to be
developing specific maps for each unique protein. Although
examined,
the technology is evolving rapidly, there are certain methods
- any other validation criteria as prescribed in the that are generally accepted. Variatiol1s of these methods will
monograph. be indicated, when appropriate, in specific monographs.
SPECIFIED VARIATIONS TO THE GENERAL METHOD A peptide map may be viewed as a fingerprint of a protein
and is the end product of several chemical processes that
Variations to the general method required for the analysis of provide a comprehensive understanding of the protein being
specific substances may be specified in detail in monographs. analysed. 4 principal steps are necessary for the development
These include: of the procedure: isolation and purification of the protein, if
- the addition of urea in the gel (3 M concentration is often the protein is part of a formulation; selective cleavage of the
satisfactory to keep protein in solution but up to 8 M can be peptide bonds; chromatographic separation of the peptides;
used) : some proteins precipitate at their isoelectric point; and analysis and identification of the peptides. A test sample
in this case, urea is included in the gel formulation to keep is digested and assayed in parallel with a reference substance.
the protein in solution; if urea is used, only Íresh solutions Complete cleavage of peptide bonds is more likely to occur
should be used to prevent carbamylation of the protein; when enzymes such as endoproteases (e.g., trypsin) are used,
- the use of alternative staining methods; instead of chemical cleavage reagents. A map must contain

(7) This chapter ha~ undergone pharmacopoeial harmonisation. See chaptcr 5.8. Pharmacopoeíal hUrllIOllisatiol1.

General Notices (1) apply to all monographs and other texts 87


2.2.55. Peptide mapping EUROPEAN PHARMACOPOEIA 8.0

enough peptides to be meaningful. On the other hand, if there To allow the enzyme to have fuU access to deavage sites and
are too many fragments, the map might lose its specificity permit sorne unfolding of the protein, it is often necessary to
because many proteins will then have the same profiles. reduce and alkylate the disulfide bonds prior to digestion.
Digestion with trypsin ean introduce ambiguities in the
peptide map due to side reactions oecurring during the
ISOLATION AND PURIFICATION digestion reaction, such as non-specific cleavage, deamidatioll,
disulfide isomerisation, oxidation of methionine residues,
Isolation al1d purification are necessary for analysis of bulk or formation of pyroglutamic groups created from the
drugs or dosage forms containing interfering excipients and deamidation of glutamine at the N-terminal side of a peptide.
carrier proteins and, when required, will be specified in the Furthermore, peaks may be produced by autohydrolysis of
monograph. Quantitative recovery of protein from the dosage trypsin. Their intensities depend on the ratio of trypsin to
form must be validated. protein. To avoid autohydrolysis, solutions of proteases may be
prepared at a pH that is not optimal (e.g. at pH 5 for trypsin),
which would mean that the enzyme would not become active
SELECTIVE CLEAVAGE OF PEPTIDE BONDS until diluted with the digest buffer.
The selection of the approach used for the cleavage of peptide Establishment of optimal digestion conditions. Faetors
bonds will depend on the protein under test. This selection that affeet the completeness and effeetiveness of digestion of
process involves determination of the type of cleavage to be proteins are those that could affect any chemical or enzymatic
employed, enzymatic or chemical, and the type of cleavage reactions.
agent within the chosen eategory. Several deavage agents and pH of the reaction milieu. The pH of the digestion mixture
their specificity are shown in Table 2.2.55.-1. This list is not is empiricaHy determined to ensure the optimisation of the
aH-inclusive and will be expanded as other cleavage agents performance of the given cleavage agent. For example, when
are identified. using cyanogen bromide as a cleavage agent, a highly aeidie
environment (e.g. pH 2, formie acid) is necessary; however,
Pretreatment of sample. Depending on the size or the when using trypsin as a deavage agent, a slightly alkaline
configuration of the protein, different approaches in the environment (pH 8) is optimal. As a general rule, the pH of
pretreatment of samples can be used. If trypsin is used as the reaction milieu must not alter the chemical integrity of the
a cleavage agent for proteins with a molecular mass greater protein during the digestion and must not change during the
than 100000 Da, Iysine residues must be protected by course of the fragmentation reaction.
citraconylation or maleylation; otherwise, too many peptides
Temperature. A temperature between 25 oC and 37 oC is
will be generated.
adequate for most digestions. The temperature used is
Pretreatment of the deavage agent. Pretreatment of cleavage intended to minimise chemical side reactions. The type
agents, espeeially enzymatic agents, might be neeessary of protein under test will dietate the temperature of the
for purification purposes to ensure reproducibility of the reaction milieu, because sorne proteins are more susceptible
map. For example, trypsin used as a cleavage agent will to denaturation as the temperature of the reaction increases.
have to be treated with tosyl-L-phenylalanine chloromethyl For example, digestion of reeombinant bovine somatropin
ketone to inactivate chymotrypsin. Other methods, such is conducted at 4 oC, because at higher temperatures it will
as purification of trypsin by high performance liquid precipitate during digestion.
chromatography (HPLC) or immobilisation of enzyme on a Time. lf sufficient sample is available, a time course study is
gel support, have been successfully used when only a small considered in order to determine the optimum time to obtain
amount of protein is available. a reproducible map and avoid incomplete digestion. Time of
Pretreatment of the protein. Under certain conditions, it digestion varies from 2 h to 30 h. The reaetion is stopped by
might be necessary to concentrate the sample or to separate the addition of an acid which does not interfere in the map
the protein from excipients and stabilisers used in formulation or by freezing.
of the product, if these interfere with the mapping proeedure. Amount of cleavage agent used. Although excessive amounts
Physical procedures used for pretreatment can in dude of cleavage agent are used to accomplish a reasonably rapid
ultrafiltration, column chromatography and lyophilization. digestion time (i.e. 6-20 hours), the amount of cleavage agent
Other pretreatments, such as the addition of chaotropic agents is minimised to avoid its contribution to the chromatographic
(e.g. urea) can be used to unfold the protein prior to mapping. map pattern. A protein to protease ratio between 20: 1 and
Table 2.2.55.-1. - Examples of cleavage agents
Type Agent Spedficity

Enzymatic Trypsin (EC 3.4.21.4) e-terminal side of Arg and Lys

Chymotrypsin (EC 3.4.21.1) e-terminal side ofhydrophobic residues (e.g. Leu, Met,
Ala, aromatics)
Pepsiu (EC 3.4.23.1 and 2) Non-specific digest

Lysyl endopeptidase (Lys-C endopeptidase) (EC 3.4.21.50) e·terminal side of Lys

Glutamyl endopeptidase (from S. aureus strain V8) e-terminal side of Glu and Asp
(EC 3.4.21.19)
Peptidyl-Asp metallo-endopeptidase (endoprotei- N-terminal side of Asp
nas e Asp-N)
Clostripain (EC 3.4.22.8) e-terminal side of Arg

Chemical Cyanogen bromide e-terminal side of Met

2-Nitro-5-thio-cyanobenzoic acid N-terminal side of eys

0- Iodosobenzoic acid e·terminal side of Trp and Tyr

Dilute acid Asp and Pro

BNPS-skatole Trp

88 See the information section on general monographs (cover pages)


EUROPEAN PHARMACOPOEIA 8.0 2.2.55. Peptide mapping

200:1 is generaHy used. It is recommended that the cleavage Isocratic elution. Isocratic HPLC systems using a single
agent is added in 2 01' more stages to optimise cleavage. mobile phase are used on the basis of their cOllvenience of
Nonetheless, the nnal reaction volume remains small enough use and improved detector responses. Optimal composition
to facilitate the next step in peptide mapping, the separation of a mobile phase to obtain clear resolution of each peak is
step. To sort out digestion artifacts that might interfere with sometimes difncnlt to establish. Mobile phases for which
the subsequent analysis, a blank determination is performed, slight changes in component ratios or in pH signincantly
using a digestion control with aH the reagents, except the test affect retention times of peaks in peptide maps must not be
protein. used in isocratic HPLC systems.
CHROMATOGRAPHIC SEPARATION Other parameters. Temperature control of the colul11n is
usually necessary to achieve good reproducibility. The flow
Many techniques are used to separate peptides for mapping.
rates for the mobile phases range from 0.1-2.0 mLlmin, and
The selection of a technique depends on the protein being
the detection of peptides is perforl11ed with a UV detector
mapped. Techniques that have been successfully used for at 200-230 nm. Other methods of detection have been used
separation of peptides are shown in Table 2.2.55-2. In this
(e.g. post-column derivatisation), but they are not as robust
section, a most widely used reversed-phase HPLC method
or versatile as UV detection.
is described as one of the procedures of chromatographic
separation. Validation. This section provides an experimental means
The purity of solvents and mobile phases is a critical factor in for measuring the overall performance of the test method.
HPLC separation. HPLC-grade solvents and water that are The acceptance criteria for system suitability depend on
commercially available, are recommended for reversed-phase the identincation of critical test parameters that affect data
HPLC. Dissolved gases present a problem in gradient systems interpretation and acceptance. These critical parameters
where the solubility of the gas in a solvent may be les s in are also criteria that monitor peptide digestion and peptide
a mixture than in a single solvento Vacuum degassing and analysis. An indicator that the desired digestion endpoint
agitation by sonication are often used as useful degassing has been achieved is shown by comparison with a reference
procedures. When solid particles in the solvents are drawn standard, which is treated in the same manner as the
into the HPLC system, they can damage the sealing of pump test protein. The use of a reference substance in parallel
valves or clog the top of the chromatographic column. Both with the test protein is critical in the development and
pre- and post-pump nltration is also recommended. establishment of system suitability limits. In additian, a
chromatogram is included with the reference substance
Table 2.2.55-2. - Techniques used for the separation of peptides for additional comparison purposes. Other indicators may
Reversed-phase high performance liquid chromatography (HPLC) include visual inspectian of protein ar peptide solubility, the
absence of intact protein, 01' measurement of responses of a
Ion-exchange chromatography (lEC)
digestion-dependent peptide. The critical system suitability
Hydrophobic interaction chromatography (HIC) parameters for peptide analysis will depend 011 the particular
mode of peptide separation and detection and on the data
Polyacrylamide gel electrophoresis (PAGE), non-denaturating
analysis requirements.
Sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS-PAGE)
When peptide mapping is used as an identification test, the
Capillary electrophoresis (CE) system suitability requirements for the identined peptides
Paper chromatography-high voltage (PCHV) cover selectivity and precision. In this case, as well as when
identincation of variant protein is done, the identincation
High voltage-paper electrophoresis (HVPE) of the primary structure of the peptide fragments in the
peptide map provides both a verincation of the known
Chromatographic column. The selection of a primary structure and the identification of protein variants by
chromatographic column is empirically determined for each comparison with the peptide map of the reference substance
protein. Columns with 10 nm or 30 nm pore size with silica for the specined protein. The use of a digested reference
support can give optimal separation. For smaller peptides, substance for a given protein in the determination of peptide
octylsilyl silica gel for chromatography R (3-10 flm) and resolution is the method of choice. For an analysis of a variant
octadecylsilyl silica gel for chromatography R (3-10 flm) protein, a characterised mixture of a variant and a reference
column packings are more efncient than butylsilyl silica gel for substance can be used, especially if the variant peptide is
chromatography R (5-10 flm). located in a less-resolved regia n oí the map. The index of
Solvento The most commonly used solvent is water with pattern consistency can be sil11ply the number of major
acetonitrile as the organic modiner to which not more than peptides detected. Peptide pattern consistency can be best
0.1 per cent trifluoroacetic acid is added. If necessary, add defined by the resolution of peptide peaks. Chromatographic
propyl alcohol or isopropyl alcohol to solubilise the digest parameters, such as peak-to-peak resolution, maximum peak
components, provided that the addition does not unduly width, peak area, peak tailing factors, and calumn efficiency,
increase the viscosity of the components. may be used to denne peptide resolution. Depending on the
Mobile phase. Buffered mobile phases containing phosphate protein under test and the method of separation used, single
are used to provide some flexibility in the selection of pH peptide or multiple peptide resolution requirements may be
conditions, since shifts of pH in the 3.0-5.0 range enhance the necessary.
separation of peptides containing acidic residues (e.g. glutamic The replicate analysis of the digest af the reference substance
and aspartic acids). Sodium or potassium phosphates, for the protein under test yields measures of precision and
ammonium acetate, phosphoric acid at a pH between 2 quantitative recovery. Recovery of the identined peptides is
and 7 (or higher for polymer-based supports) have also been generally ascertained by the use of internal or external peptide
used with acetonitrile gradients. Acetonitrile containing standards. The precision is expressed as the relative standard
trifluoroacetic acid is used quite often. deviation (RSD). Differences in the recovery and precision
Gradient. Gradients can be linear, nonlinear, or include of the identined peptides are to be expected; therefore, the
step functions. A shallow gradient is recommended in order system suitability limits will have to be established far both the
to separate complex mixtures. Gradients are optimised to recovery and the precision of the identined peptides. These
provide clear resolution of 1 or 2 peales that will become limits are unique for a given protein and will be specined in
"marker" peaks for the test. the individual monograph.

General Notices (1) apply to all monographs and other texts 89


2.2.56. Amino acid analysis EUROPEAN PHARMACOPOEIA 8.0

Visual comparison of the relative retentions, the peak is no 105s of resolution due to scale-up. E1uates corresponding
responses (the peak are a or the peak height), the number of to specific peptide peaks are collected, vacuum-concentrated,
peaks, and the overall elution pattern is completed initially. It and chromatographed again, if necessary. Amino acid
is then complemented and supported by mathematical analysis analysis of fragments may be limited by the peptide size. If
of the peak response ratios and by the chromatographic profile the N-terminus is blocked, it may need to be c1eared before
of a 1:1 (V/V) mixture of sample and reference substance sequencing. C-terminal sequencing of proteins in combination
digest. If all peaks in the sample digest and in the reference with carboxypeptidase and matrix-assisted laser desorption
substance digest have the same relative retentions and peak ionisation coupled to time-of-flight analyser (MALDI-TOF)
response ratios, then the identity of the sample under test is can also be used for characterisation purposes.
confirmed. The use of MS for characterisation of peptide fragments
If peaks that initially eluted with significantly different is by direct infusion of isolated peptides or by the use of
relative retentions are then observed as single peaks in the on-Iine LC-MS for structure analysis. In general, it includes
1: 1 mixture, the initial difference would be an indication of e1ectrospray and MALDI-TOF-MS, as well as fast-atom
system variability. However, if separate peaks are observed in bOl11bárdment (FAB). Tandem MS has also been used to
the 1: 1 mixture, this would be evidence of the nonequivalence sequence a modified protein and to determine the type of
of the peptides in each peak. If a peak in the 1: 1 mixture amino acid modification that has occurred. The comparison
is significantly broader than the corresponding peak in the of mass spectra of the digests before and after reduction
sample and reference substance digest, it may indicate the provides a method to assign the disulfide bonds to the various
presence of different peptides. The use of computer-aided sulfydryl-containing peptides.
pattern recognition software for the analysis of peptide If regions of the primary structure are not clearly demonstrated
mapping data has been proposed and applied, but issues by the peptide map, it might be necessary to develop a
related to the validation of the computer software preclude its secondary peptide map. The goal of a validated method of
use in a compendial test in the near future. Other automated characterisation of a protein through peptide mapping is to
approaches have been used that employ mathematical reconcile and account for at least 95 per cent of the theoretical
formulas, models, and pattern recognition. Such approaches composition of the protein structure.
are, for example, the automated identification of compounds
by IR spectroscopy and the application of diode-array UV
spectral analysis for identification of peptides. These methods 01/2010:20256
have limitations due to inadequate resolutions, co-elution of
fragments, or absolute peak response differences between 2.2.56. AMINO ACID ANALYSIS(8)
reference substance and sample digest fragments.
The numerical comparison of the peak retention times and Amino acid analysis refers to the methodology used to
peak afeas or peak heights can be done for a selected group determine the amino acid composition or content of proteins,
of relevant peaks that have been correctly identified in the peptides, and other pharmaceutical preparations. Proteins and
peptide maps. Peak areas can be calculated using 1 peak peptides are macromolecules consisting of covalently bonded
showing relatively small variation as an internal reference, amino acid residues organised as a linear polymer. The
keeping in mind that peak area integration is sensitive to sequence of the amino acids in a protein or peptide determines
baseline variation and likely to introduce error in the analysis. the properties of the molecule. Proteins are considered large
Alternatively, the percentage oí each peptide peak height molecules that commonly exist as folded structures with a
relative to the sum of all peak heights can be caiculated for specific conformation, while peptides are smaller and may
the sample under test. The percentage is then compared to consist of only a few amino acids. Amino acid analysis can
that of the corresponding peak of the reference substance. be used to quantify proteins and peptides, to determine
The possibility of auto-hydrolysis of trypsin is monitored the identity of proteins or peptides based 011 their amino
by producing a blank peptide map, that is, the peptide map acid composition, to support protein and peptide structure
obtained when a blank solution is treated with trypsin. analysis, to evaluate fragmentation strategies for peptide
The minimum requirement for the qualification of peptide mapping, and to detect atypical amino acids that might be
mapping is an approved test procedure that includes system present in a protein or peptide. It is necessary to hydrolyse
a protein/peptide to its individual amino acid constituents
suitability as a test control. In general, early in the regulatory
process, qualification of peptide mapping for a protein is before amino acid analysis. Following protein/peptide
hydrolysis, the amino acid analysis pro ce dure can be the same
sufficient. As the regulatory approval process for the protein
as that practiced for free amino acids in other pharmaceutical
progresses, additional qualifications of the test can include
a partial validation of the analytical procedure to provide preparations. The amino acid constituents of the test sample
assurance that the l11ethod will perform as intended in the are typically derivatised for analysis.
developl11ent of a peptide map for the specified protein. APPARATUS
ANALYSIS AND IDENTIFICATION OF PEPTIDES Methods used for amino acid analysis are usually based on
a chromatographic separation of the amino acids present in
This section gives guidance on the use of peptide mapping
the test sample. Current techniques take advantage of the
during development in support of regulatory applications.
automated chromatographic instrumentation designed for
The use of a peptide map as a qualitative tool do es not require analytical methodologies. An amil10 acid analysis instrument
the complete characterisation of the individual peptide will typically be a low-pressure or high-pressure Iiquid
peaks. However, validation of peptide l11apping in support chromatograph capable of generating mobile phase gradients
of regulatory applications requires rigorous characterisation that separate the amino acid analytes on a chromatographic
of each of the individual peaks in the peptide map. Methods column. The instrument must have post -column derivatisation
to characterise peaks range from N-terminal sequencing of capability, unless the sample is analysed using precolumn
each peak followed by amino acid analysis to the use of mass derivatisation. The detector is usually an ultraviolet/visible
spectroscopy (MS). or fluorescence detector depending on the derivatisation
For characterisation purposes, when N-terminal sequencing method used. A recording device (e.g., integrator) is used for
and amino acids analysis are used, the analytical separatiol1 is transforming the analogue signal from the detector and for
scaled up. Since scale-up might affect the resolution of peptide quantitation. It is preferred that instrumentation be dedicated
peaks, it is neces5ary, using empirical data, to assure that there particularly for amino acid analysis.

(8) This chapter has undergone pharmacopoeial harmonisation. See chapter 5.8. Pharmacopoeial hnrmonisatioll.

90 See the information section on general rnonographs (cover pages)


EUROPEAN PHARMACOPOEIA 8.0 2.256. Amino acid analy§is

GENERAL PRECAUTIONS REPEATABILITY


Background contamination is always a concern for the analyst COl1sistent high quality amino acid analysis results from an
in performing amino acid analysis. High purity reagents are analyticallaboratory require attention to the repeatability of
necessary (e.g., low purity hydrochloric acid can contribute the assay. During analysis of the chromatographic separation
to glycine contamination). Analytical reagents are changed of the amino acids or their derivatives, numerous peaks can
routinely every few weeks using only high-pressure liquid be observed 011 the chromatogram that correspond to the
chromatography (HPLC) grade solvents. Potential microbial amino acids. The large number of peaks makes it necessary
contamination and foreign material that might be present to have an amino acid analysis system that can repeatedly
in the solvents are reduced by filtering solvents before use, identify the peaks based on retention time and integrate the
keeping solvent reservoirs covered, and not plaeing amino peak are as for quantitation. A typical repeatability evaluation
acid analysis instrumentation in direct sunlight. involves preparing a standard amino aeid solution and
analysing many replicates (e.g., 6 analyses or more) ofthe
Laboratory practices can determine the quality of the amino same standard solution. The relative standard deviation (RSD)
acid analysis. Place the instrumentation in a low traffic area of is determined for the retention time and integrated peak
the laboratory. Keep the laboratory dean. Clean and calibrate area of each amino aeid. An evaluation of the repeatability
pipets according to a maintenance schedule. Keep pipet tips is expanded to indude multiple assays conducted over
in a covered box; the analysts may not han dIe pipet tips with several days by different analysts. Multiple assays indude the
their hands. The analysts may wear powder-free latex or preparation of standard dilutions from starting materials
equivalent gloves. Limit the number of times a test sample vial to determine the variation due to sample handling. The
is opened and dosed because dust can contribute to elevated amino acid composition of a standard protein (e.g., bovine
levels of glycine, serine, and alanine. serum albumin) is often ana1ysed as part of the repeatability
A well-maintained instrument is necessary for acceptable evaluation. By evaluating the replicate variation (i.e., RSD),
amino acid analysis results. lf the instrument is used on a the laboratory can establish analyticallimits to ensure that the
routine basis, it is to be checked daily for leaks, detector analyses from tlle laboratory are under control. It is desirable
and lamp stability, and the ability of the column to maintain to establish the lowest practical variation limits to ensure
resolution of the individual amino acids. Clean or replace al! the best results. Areas to focus on to lower the variability of
instrument filters and other maintenance items on a routine the amino acid analysis indude sample preparation, high
schedule. background spectral interference due to quality of reagents
and/or laboratory practices, instrument performance and
REFERENCE MATERIAL maintenance, data analysis and interpretation, and analyst
performance and habits. Al! parameters involved are fully
Acceptable amino acid standards are commercially available investigated in the scope of the validation work.
for amino acid analysis and typically consist of an aqueous
mixture of amino acids. When determining amino acid SAMPLE PREPARATION
composition, protein or peptide standards are analysed with Accurate results from amino acid analysis require purified
the test material as a control to demonstrate the integrity of protein and peptide samples. Buffer components (e.g., salts,
the entire procedure. Highly purified bovine serum albumin urea, detergents) can interfere with the amino acid analysis
has been used as a pro te in standard for this purpose. and are removed from the sample before analysis. Methods
that utilise post -column derivatisation of the amino acids
CALIBRATION OF INSTRUMENTATION are generally not affected by buffer components to the
extent seen with pre-column derivatisation methods. It is
Calibration of amino aeid analysis instrumentation typically
desirable to limit the number of sample manipulations to
involves analysing the amino acid standard, which consists
reduce potential background contamination, to improve
of a mixture of amino acids at a number of concentrations,
analyte recovery, and to reduce ¡abour. Common techniques
to determine the response factor and range of analysis for
used to remove buffer components from protein samples
each amino acid. The concentration of each amino acid in the
indude the following methods: (1) injecting the protein
standard is known. In the calibration procedure, the analyst
sample onto a reversed-phase HPLC system, removing the
dilutes the amino aeid standard to several different analyte
protein with a volatile solvent containing a sufficient organic
levels within the expected linear range of the amino acid
component, and drying the sample in a vacuum cel1trifuge;
analysis technique. Then, replicates at each of the different
(2) dialysis against a volatile buffer or water; (3) centrifugal
analyte levels can be analysed. Peak are as obtained for each
ultrafiltration for buffer replacement with a volatile buffer or
amino aeid are plotted versus the known concentration for
water; (4) preeipitating the pro te in from the buffer using an
each of the amino acids in the standard dilution. These
organic solvent (e.g., acetone); (5) gel filtration.
resuIts will allow the analyst to determine the range of amino
acid concentrations where the peak are a of a given amino INTERNAL STANDARDS
acid is an approximately linear function of the amino acid It is recommended that an internal standard be used to
concentration. It is important that the analyst prepare the monitor physical and chemicallosses and variations during
samples for amino aeid analysis so that they are within the amino acid analysis. An accurately known amount of
analyticallimits (e.g., linear working range) of the technique internal standard can be added to a protein solution prior to
employed in order to obtain accurate and repeatable results.
hydrolysis. The recovery of the internal standard gives the
4 to 6 amino aeid standard levels are analysed to determine general recovery of the amino acids of the protein solution.
a response factor for each amino acid. The response factor Free amino acids, however, do not behave in the same way as
is calculated as the average peak are a or peak height per protein-bound amino acids during hydrolysis, whose rates of
nanomole of amino acid present in the standard. A calibration releas e or destruction are variable. Therefore, the use of an
file consisting of the response factor for each amino acid is internal standard to correct for los ses during hydrolysis may
prepared and used to calculate the concentration of each give unreliable results. It will be necessary to take this point
amino aeid present in the test sample. This calculation into consideration when interpreting the results. Internal
involves dividing the peak are a corresponding to a given standards can also be added to the mixture of amino acids
amino acid by the response factor for that amino acid to after hydrolysis to correct for differences in sample application
give the nanomoles of the amino acid. For routine analysis, and changes in reagent stability and flow rates. Ideally, an
a single-point calibration may be sufficient; however, the internal standard is an unnaturally occurring primary amino
calibration file is updated frequently and tested by the analysis acid that is commereially available and inexpensive. It should
of analytical contr01s to ensure its integrity. aIso be stabIe during hydrolysis, its response factor should

General Natices (1) apply ta all managraphs and ather texts 91


2.2.56. Amino add analysis EUROPEAN PHARMACOPOEIA 8.0

be linear with concentration, and it needs to elute with a (e.g., incomplete hydrolysis or destruction oflabile amino
unique retention time without overlapping other amino acids. acids). Complete proteolysis, using a mixture of proteases,
Commonly used amino acid standards include norleucine, has been used but can be complicated, requires the proper
nitrotyrosine, and a-aminobutyric acid. contro1s, and is typically more applicable to peptides than
proteins.
PROTEIN HYDROLYSIS
During initial analyses of an unknown protein, experiments
Hydrolysis of protein and peptide samples is necessary for with various hydrolysis time and temperature conditions are
amino acid analysis of these molecules. The glassware used conducted to determine the optimal conditions.
for hydrolysis must be very clean to avoid erroneous results.
Glove powders and fingerprints on hydrolysis tubes may cause METHOD 1
contamination. To clean glass hydrolysis tubes, boil tubes for Acid hydrolysis using hydrochloric acid containing phenol
1 h in 1 M hydrochloric acid or soak tubes in concentrated is the most common procedure used for protein/peptide
nitric acid or in a mixture of equal volumes of concentrated hydrolysis preceding amino acid analysis. The addition of
hydrochloric acid and nitric acid. Clean hydrolysis tubes phenol to the reaction prevents the halogenation of tyrosine.
are rinsed with high-purity water followed by a rinse with Hydrolysis solution. 6 M hydrochloric acid containing
HPLC grade methanol, dried overnight in an oven, and stored 0.1 per cent to 1.0 per cent of phenol.
covered until use. Alternatively, pyrolysis of clean glassware at
500 oC for 4 h may also be used to eliminate contamination Procedure
from hydrolysis tubes. Adequate disposable laboratory Liquid phase hydrolysis. Place the protein or peptide sample in
material can also be used. a hydrolysis tube, and dry (the sample is dried so that water
Acid hydrolysis is the most common method for hydrolysing a in the sample willnot dilute the acid used fol' the hydrolysis).
protein sample before amino acid analysis. The acid hydrolysis Add 200 I1L of hydrolysis solution per 500 ¡.tg oflyophilised
technique can contribute to the variation of the analysis due protein. Freeze the sample tube in a dry ice-acetone bath,
to complete or partia! destruction of severa! amino acids: and flame seal in vacuo. Samples are typically hydrolysed at
tryptophan is destroyed; serine and threonine are partially 110 oC for 24 h in vacuo 01' in an inert atmosphere to prevent
destroyed; methionine might undergo oxidation; and cysteine oxidation. Longer hydrolysis times (e.g., 48 h and 72 h)
is typically recovered as cystine (but cystine recovery is are investigated if there is a concern that the protein is not
usually poor because of partial destruction or reduction to completely hydrolysed.
cysteine). Application of adequate vacuum (less than 200 11m Vapour phase hydrolysis. This is one of the most common acid
of mercury or 26.7 Pa) or introduction of an inert gas (argon) hydrolysis procedures, and it is preferred fol' microanalysis
in the headspace of the reaction vessel can reduce the level of when only small amounts of the sample are available.
oxidative destruction. In peptide bonds involving isoleucine Contamination of the sample from the acid reagent is also
and valine the amido bonds ofIle-He, Val-Val, Ile-Val, and minimised by using vapour phase hydrolysis. Place vials
Val-Ile are partially cleaved; and asparagine and glutamine containing the dried samples in a vessel that contains an
are deamidated, resulting in aspartic acid and glutamic appropriate amount of hydrolysis solution. The hydrolysis
acid, respectively. The loss of tryptophan, asparagine, and solution do es not come in contact with the test sample. Apply
glutamine during an acid hydrolysis limits quantitation to an inert atmosphere or vacuum (less than 200 ~lIn of mercury
17 amino acids. Some of the hydrolysis techniques described or 26.7 Pa) to the headspace of the vessel, and heat to about
are used to address these concerns. Some oí the hydrolysis 110 oC for a 24 h hydrolysis time. Acid vapour hydrolyses the
techniques described (i.e., Methods 4-11) may cause dl'ied sample. Any condensation of the acid in the sample
modifications to other amino acids. Therefore, the benefits of vials is to be minimised. After hydrolysis, dry the test sample
using a given hydrolysis technique are weighed against the in vacuo to remove any residual acid.
concerns with the technique and are tested adequately before METHOD2
employing a method other than acid hydrolysis.
Tryptophan oxidation during hydrolysis is decreased by using
A time-course study (i.e., amino acid anaiysis at acid mercaptoethanesulfonic acid as the reducing acid.
hydrolysis times of 24 h, 48 h and 72 h) is often employed
to analyse the starting concentration of amino acids that Hydrolysis solution. 2.5 M mercaptoethanesulfonic acid
are partially destroyed or slow to cleave. By plotting the solution.
observed concentration of labile amino acids (e.g., serine and Vapour phase hydrolysis. Dry about 1 flg to 100 ~lg of the
threonine) versus hydrolysis time, the 1ine can be extrapolated protein/peptide under test in a hydrolysis tube. Place the
to the origin to determine the starting concentration of these hydrolysis tube in a larger tube with about 200 I1L oí the
amino acids. Time-course hydrolysis studies are also used hydrolysis solution. Seal the larger tube in vacuo (about 50 flm
with amino acids that are slow to cleave (e.g., isoleucine and of mercury or 6.7 Pa) to vaporise the hydrolysis solution.
valine). During the hydrolysis time course, the analyst will Heat the hydrolysis tube to 170-185 oC for about 12.5 mino
observe a plateau in these residues. The level of this plateau After hydrolysis, dry the hydrolysis tube in vacuo Íor 15 min
is taken as the residue concentration. If the hydrolysis time is to remove the residual acid.
too long, the residue concentration of the sample will begin to METHOD3
decrease, indicating destruction by the hydrolysis conditions.
Tryptophan oxidation during hydrolysis is prevented by using
An acceptab1e alternative to the time-course study is to subject thioglycollic acid (TGA) as the reducing acid.
an amino acid calibration standard to the same hydrolysis
conditions as the test sample. The amino acid in free form Hydrolysis solution. 7 M hydrochloric acid containing 1 per
may not completely represent the rate of destruction of labile cent of phenol, 10 per cent of trifluoroacetic acid and 20 per
amino acids within a peptide or protein during the hydrolysis. cent of thioglycollic acid.
This is especially true for peptide bonds that are slow to cleave Vapour phase hydrolysis. Dry about 10 flg to 50 flg of the
(e.g., Ile-Val bonds). However, this technique will allow the protein/peptide under test in a sample tube. Place the sample
analyst to account Íor some residue destruction. Microwave tube in a larger tube with about 200 I1L of the hydrolysis
acid hydrolysis has been used and is rapid but requires solution. Seal the larger tube in vacuo (about 50 11m of
special equipment as well as specia1 precautions. The optimal mercury or 6.7 Pa) to vaporise the TGA. Heat the sample
conditions for microwave hydrolysis must be investigated tube to 166 oC for about 15-30 mino After hydrolysis, dry the
for each individual protein/peptide samp1e. The microwave sample tube in vacuo fol' 5 min to remove the residual acid.
hydro1ysis technique typically requires only a few minutes, but Recovel'Y of tryptophan by this method may be dependent on
even a deviation of one minute may give inadequate results the amount of sample present.

92 See the information section on general monographs (caver pages)


EUROPEAN PHARMACOPOEIA 8.0 2.2.56. Amino add analysis

METHOD4 the pyridylethylation reaction can cause modifications to the


Cysteine/cystine and methionine oxidation is performed with a-amino terminal group and the E-amino group of lysine in
performic acid before the protein hydrolysis. the protein.
Oxidation solution. Use performic acid freshly prepared by METHOD8
mixing 1 volume of hydrogen peroxide solution (30 per cent) Cysteine/cystine reduction and alkylation is accomplished by
and 9 volumes of anhydrous formic acid and incubating at a liquid phase pyridylethylation reaction.
room temperature for 1 h. Stock solutions. Prepare and filter 3 solutions: 1 M
Procedure. Dissolve the protein/peptide sample in 20 flL Tris-hydrochloride pH 8.5 containing 4 mM disodium edetate
of anhydrous formic acid and heat at 50 oC for 5 min; then (stock solution A), 8 M guanidine hydrochloride (stock
add 100 flL of the oxidation solution. Allow the oxidation solution B), and 10 per cent of 2-mercaptoethanol (stock
to proceed forlO-30 mino In this reaction, cysteine is solution C).
converted to cysteic acid and methionine is converted to Reducing solution. Prepare a mixture of 1 volume of stock
methionine-sulfone. Remove the excess reagent from the solution A and 3 volumes of stock solution B to obtain a
sample in a vacuum centrifuge. The oxidised protein can buffered solution of 6 M guanidine hydrochloride in 0.25 M
then be acid hydrolysed using Method 1 or Method 2. This tris-hydrochloride.
technique may cause modifications to tyrosine residues in the
presence of halides. Procedure. Dissolve about 10 flg ofthe test sample in 50 flL of
the reducing solution, and add about 2.5 flL of stock solution C.
METHOD5
Store under nitro gen or argo n for 2 h at room temperature in
Cysteine/cystine oxidation is accomplished during the liquid the dark. To achieve the pyridylethylation reaction, add about
phase hydrolysis with sodium azide. 2 flL of 4-vinylpyridine to the protein solution, and incubate
Hydrolysis solution. To 6 M hydrochloric acid containing for an additional 2 h at room temperature in the darle Desalt
0.2 per cent of phenol, add sodium azide to obtain a the protein/peptide by collecting the protein/peptide fraction
final concentration of 2 giL. The added phenol prevents from a reversed-phase HPLC separation. The collected sample
halogenation of tyrosine. can be dried in a vacuum centrifuge before acid hydrolysis.
Liquid phase hydrolysis. Conduct the protein/peptide METHOD9
hydrolysis at about 110 oC for 24 h. During the hydrolysis, Cysteine/cystine reduction and alkylation is accomplished by
the cysteine/cystine present in the sample is converted to a liquid phase carboxymethylation reaction.
cysteic acid by the sodium azide present in the hydrolysis
Stock solutions. Prepare as directed for Method 8.
solution. This technique allows better tyrosine recovery
than Method 4, but it is not quantitative for methionine. Carboxymethylation solution. Prepare a 100 giL solution
Methionine is converted to a mixture of the parent methionine of iodoacetamide in alcohol.
and its 2 oxidative products, methionine-sulfoxide and Buffer solution. Use the reducing solution, prepared as
methionine-sulfone. described for Method 8.
METHOD6 Procedure. Dissolve the test sample in 50 flL of the buffer
Cysteine/cystine oxidation is accomplished with dimethyl solution, and add about 2.5 11L of stock solution C. 5tore
sulfoxide (DMSO). under nitro gen or argon for 2 h at room temperature in the
Hydrolysis solution. To 6 M hydrochloric acid containing dark. Add the carboxymethylation solution in a ratio 1.5 fold
0.1 per cent to 1.0 per cent of phenol, add dimethyl sulfoxide per total theoretical content of thioIs, and incubate for an
to obtain a final concentration of 2 per cent V/V. additional 30 min at room temperature in the dark. If the thiol
content of the protein is unknown, then add 5 flL of 100 mM
Vapour rhase hydrolysis. Conduct the protein/peptide
iodoacetamide for every 20 nmol of protein present. The
hydrolysis at about 110 oC for 24 h. During the hydrolysis,
reaction is stopped by adding excess of 2-mercaptoethanol.
the cysteine/cystine present in the sample is converted to
Desalt the protein/peptide by collecting the protein/peptide
cysteic acid by the DMSO present in the hydrolysis solution.
fracHon from a reversed-phase HPLC separation. The
As an approach to limit variability and compensate for partial
collected sample can be dried in a vacuum centrifuge befo re
destruction, it is recommended to evaluate the cysteic acid
acid hydrolysis. The S-carboxyamidomethyl-cysteine formed
recovery from oxidative hydrolysis of standard proteins
will be converted to S-carboxymethyl-cysteine during acid
containing 1-8 mol of cysteine. The response factors from
hydroJysis.
protein/peptide hydrolysates are typically about 30 per cent
lower than those for non-hydrolysed cysteic acid standards. METHOD 10
Because histidine, methionine, tyrosine, and tryptophan Cysteinel cystine is reacted with dithiodiglycolic acid or
are also modified, a complete compositional analysis is not dithiodipropionic acid to produce a mixed disulfide. The
obtained with this technique. choice of dithiodiglycolic acid or dithiodipropionic acid
METHOD7 depends 011 the required resolution of the amino acid analysis
method.
Cysteine/cystine reduction and alkylation is accomplished by
a vapour phase pyridylethylation reaction. Redudng solution. A 10 giL solution of dithiodiglycolic acid
(or dithiodipropionic acid) in 0.2 M sodium hydroxide.
Reducing solution. Transfer 83.3 flL of pyridine, 16.7 flL of
4-vinylpyridine, 16.7 flL of tributylphosphine, and 83.3 flL of Procedure. Transfer about 20 flg of the test sample to a
water to a suitable container and mix. hydrolysis tube, and add 5 flL of the reducing solution.
Add 10 flL of isopropyl alcohol, and then remove al! of the
Procedure. Add the protein/peptide (between 1 and 100 flg)
sample liquid by vacuum centrifugation. The sample is then
to a hydrolysis tube, and place in a larger tube. Transfer
hydrolysed using Method l. This method has the advantage
the reducing solution to the large tube, seal in vacuo (about
that other amino acid residues are not derivatised by side
50 flm of mercury or 6.7 Pa), and heat at about 100 oC for
reactions, and that the sample does not need to be desalted
5 mino Then remove the inner hydrolysis tube, and dry it in
a vacuum desiccator for 15 min to remove residual reagents. prior to hydrolysis.
The pyridylethylated sample can then be acid hydrolysed METHOD 11
using previously described procedures. The pyridylethylation Asparagine and glutamine are converted to aspartic acid and
reaction is performed simultaneously with a protein standard glutamic acid, respectively, during acid hydrolysis. Asparagine
sample containing 1-8 mol of cysteine to evaluate the and aspartic acid residues are added and represented by Asx,
pyridylethyl-cysteine recovery. Longer incubation times for while glutamine and glutamic acid residues are added and

General Notices (1) apply to all monographs and other texts 93


2.2.56. Amino acid analysis EUROPEAN PHARMACOPOEIA 8.0

represented by G/x. Proteins/peptides can be reacted with analogue signal is visualised by means of a data acquisition
bis(l,l-trifluoroacetoxy)iodobenzene (BTI) to convert the system, and the peak are as are integrated for quantification
asparagine and glutamine residues to diaminopropionic acid purposes.
and diaminobutyric acid residues, respectively, upon acid METHOD 1 - POST-COLUMN NINHYDRIN
hydrolysis. These conversions allow the analyst to determine DERIVA TISA TION
the asparagine and glutamine content of a protein/peptide in
Ion -exchange chromatography with post -column ninhydrin
the presence of aspartic acid and glutamic acid residues.
derivatisation is one of the most common methods employed
Reducing solutions. Prepare and filter 3 solutions: a solution for quantitative amino acid analysis. As a rule, a lithium-based
of 10 mM trifluoroacetic acid (Solution A), a solution of cation-exchange system is employed for the analysis of
5 M guanidine hydrochloride and 10 mM trifluoroacetic the more complex physiological samples, and the faster
acid (Solution B), and a freshly prepared solution of sodium-based cation-exchange system is used for the
dimethylformamide containing 36 mg of BTI per millilitre more simplistic amino acid mixtures obtained with protein
(Solution C). hydrolysates (typically containing 17 amino acid components).
Procedure. In a clean hydrolysis tube, transfer about 200 ¡.tg Separation of the amino acids on an ion -exchange eolumn is
of the test sample, and add 2 mL of Solution A or Solution B accomplished through a combination of changes in pH and
and 2 mL of Solution C. Sea! the hydrolysis tube in vacuo. cation strength. A temperature gradient is often employed
Heat the sample at 60 oC for 4 h in the dark. The sample to enhance separation.
is then dialysed with water to remove the excess reagents. When the amino acid reacts with ninhydrin, the reaetant has
Extraet the dialysed sample 3 times with equa! volumes of a characteristic purple or yellow colour. Amino acids, except
butyl aeetate, and then lyophilise. The protein can then imino acid, give a purple colour, and show an absorption
be acid hydrolysed using previously described procedures. maximum at 570 nm. The imino acids su eh as proline give a
The a,~-diaminopropionic and a,y-diaminobutyric acid yellow colour, and show an absorption maximum at 440 nm.
residues do not typically resolve from the lysine residues upon The post -column reaction between ninhydrin and amino acids
ion-exchange chromatography based on amino acid analysis. eluted from the column is monitored at 440 nm and 570 nm,
Therefore, when using ion-exchange as the mode of amino and the chromatogram obtained is used for the determination
acid separation, the asparagine and glutamine contents are the of amino acid composition.
quantitative difference in the aspartic acid and glutamic acid The detection limit is considered to be 10 pmol for most of the
content assayed with underivatised and BTI -derivatised acid amino acid derivatives, but 50 pmol for the proline derivative.
hydrolysis. The threonine, methionine, cysteine, tyrosine, and Response linearity is obtained in the range of 20-500 pmol
histidine assayed content can be altered by BTI derivatisation; with correlation coefficients exceeding 0.999. To obtain good
a hydrolysis without BTI will have to be performed if the composition data, samples larger than 1 ¡.tg before hydrolysis
analyst is interested in the eomposition of these other amino are best suited for this amino acid analysis of protein/peptide.
aeid residues of the protein/peptide.
METHOD 2 - POST-COLUMN OPA DERIVATISATION
o-Phthalaldehyde (OPA) reacts with primary amines in the
METHODOLOGIES OF AMINO ACm ANALYSIS:
presence of thiol compound, to form highly fluorescent
GENERAL PRINCIPLES
isoindole products. This reaction is used for the post-column
Many amino acid analysis techniques exist, and the choice derivatisation in analysis of amino acids by ion-exchange
of any one technique often depends on the sensitivity chromatography. The rule of the separation is the same
required from the assay. In general, about one-half of as Method l.
the amino acid analysis techniques employed rely on Although OPA do es not react with secondary amines (imino
the separation of the free amino acids by ion-exchange acids such as proline) to form fluorescent substances, the
chromatography followed by post-column derivatisation oxidation with sodium hypochlorite or chloramine T allows
(e.g., with ninhydrin or o-phthalaldehyde). Post-column secondary amines to react with OPA. The procedure employs a
derivatisation techniques can be used with samples that strongly acidic cation-exchange column for separation of free
contain small amounts of buffer components, (such as salts amino acids followed by post-column oxidation with sodium
and urea) and generally require between 5 ¡.tg and 10 ¡.tg hypochlorite or chloramine T and post-column derivatisation
of protein sample per analysis. The remaining amino acid using OPA and a thiol compound such as N-acetyl-L-cysteine
techniques typically involve pre-column derivatisation or 2-mercaptoethanol. The derivatisation of primary amino
of the free amino acids (e.g., phenyl isothiocyanate; acids is not noticeably affected by the continuous supply of
6-aminoquinolyl-N-hydroxysuccinimidyl carbamate or sodium hypochlorite or chloramine T.
0- phthalaldehyde; (dimethylamino )azobenzenesulfonyl
chloride; 9-fluorenylmethyl chloroformate; and Separation of the amino acids on an ion -exchange column
7 -fluoro-4-nitrobenzo-2-oxa-1,3-diazole) followed by is accomplished through a combination of changes in pH
reversed-phase HPLC. Pre-column derivatisation techniques and cation strength. After post -column derivatisation of
are very sensitive and usually require between 0.5 ¡.tg and 1.0 ¡.tg eluted amino acids with OPA, the reactant pass es through
of protein sample per analysis but may be influenced by buffer the fluorometric detector. Fluorescence intensity of
salts in the samples. Pre-column derivatisation techniques OPA-derivatised amino acids are monitored with an excitation
may also result in multiple derivatives of a given amino acid, wavelength of 348 nm and an emission wavelength of 450 nm.
which complicates the result interpretation. Post-column The detection limit is considered to be a few ten s of pico mole
derivatisation techniques are generally influenced less level for most ofthe OPA-derivatised amino acids. Response
by performance variation of the assay than pre-column linearity is obtained in the range of a few picomole level to a
derivatisation techniques. few tens of nanomole leve!. To obtain good compositional
data, samples larger than 500 ng of protein/peptide before
The following methods may be used for quantitative
hydrolysis are recommended.
amino acid analysis. Instruments and reagents for these
procedures are available eommercially. Furthermore, many METHOD 3 - PRE-COLUMN PITC DERIVA TISATION
modifications of these methodologies exist with different Phenylisothiocyanate (PITC) reacts with amino acids to form
reagent preparations, reaction procedures, chromatographic phenylthiocarbamyl (PTC) derivatives which can be detected
systems, etc. Specific parameters may vary according to the with high sensitivity at 254 nm. Therefore, pre-column
exact equipment and procedure used. Many laboratories will derivatisation of amino acids with PITC followed by a
use more than one amino acid analysis technique to exploit reversed-phase HPLC separation with UV detection is used
the advantages offered by each. In each of these methods, the to analyse the amino acid composition.

94 See the information section on general monographs (caver pages)


EUROPEAN PHARMACOPOEIA 8.0 2.2.56. Amino add analysis

After the reagent is removed under vacuum, the derivatised Pre-column derivatisation of amino acids with OPA is
amino acids can be stored dry and frozen for several weeks followed by a reversed-phase HPLC separation. Because
with no significant degradation. If the solution for injectiol1 of the instability of the OPA-amino acid derivative, HPLC
is kept cold, no noticeable loss in chromatographic response separation and analysis are performed immediately following
occurs after 3 days. derivatisation. The liquid chromatograph is equipped with a
Separation of the PTC-amino acids on a reversed-phase HPLC fluorometric detector for the detection of derivatised amino
acids. Fluorescence intensity ofOPA-derivatised amino acids
with an octadecylsilyl (ODS) column is accomplished through
is monitol'ed with an excitation wavelength of 348 nm and an
a combination of changes in concentrations of acetonitrile
emission wavelength of 450 nm.
and buffer ionic strength. PTC-amino acids eluted from the
column are monitored at 254 nm. Detection limits as low as 50 fmol via fluorescence have been
reported, although the practicallimit of analysis remains at
The detection limit is considered to be 1 pmol fol' most of the
1 pmo!.
PTC-amino acids. Response linearity is obtained in the range
of 20-500 pmol with correlation coefficients exceeding 0.999. METHOD 6 - PRE-COLUMN DABS-Cl DERIVATISATION
To obtain good compositional data, samples larger than 500 ng Pre-column derivatisation of amino acids with
of protein/peptide before hydrolysis are recommended. (dimethylamino )azobenzenesulfonyl chloride (DABS-CI)
METHOD 4 - PRE-COLUMN AQC DERIVITISATION followed by reversed-phase HPLC separation with visible light
detection is used.
Pre-column derivatisation of amino acids with
6-amino quinolyl-N -hydroxysuccinimidyl carbamate DABS-CI is a chromophoric reagent employed for the
(AQC) followed by reversed-phase HPLC separation with labelling of amino acids. Amino acids labelled with DABS-CI
fluorometric detection is used. (DABS-amino acids) are highly stable and show an absorption
maximum at 436 nm.
AQC reacts with amino acids to form stable, fluorescent
unsymmetric urea derivatives (AQC-amino acids) which DABS-amino acids, al! naturally occurring amino acid
are l'eadily amenable to analysis by reversed-phase HPLC. derivatives, can be separated on an ODS column of a
Therefore, pre-column derivatisation of amino acids with reversed-phase HPLC by employing gradient systems
AQC followed by reversed-phase HPLC separation with consisting of acetonitrile and aqueous buffer mixture.
fluorimetl'ic detection is used to analyse the amino acid Separated DABS-amino acids eluted from the column are
composition. detected at 436 nm in the visible region.
Separation of the AQC-amino acids on a reversed-phase HPLC This method can analyse the imino acids such as proline
with an ODS column is accomplished through a combination together with the amino acids at the same degree of
of changes in concentrations of acetonitrile and buffer ionic sensitivity, DABS-CI derivatisation method permits the
strengh. Selective fluorescence detection of the derivatives simultaneous quantification of tryptophan residues by
with an excitation wavelength at 250 nm and an emission previous hydrolysis of the protein/peptide with sulfonic acids
wavelength at 395 nm allows for the direct injection of the such as mercaptoethanesulfonic acid, p-toluenesulfonic acid
reaction mixture with no significant interfel'ence from the or methanesulfonic acid described in Method 2 under Protein
only major fluorescent reagent by-product, 6-aminoquinoline. hydrolysis. The other acid-labile residues, asparagine and
Excess reagent is rapidly hydrolysed (t ll2 <15 s) to yield glutamine, can also be analysed by previous conversion inta
6-aminoquinoline, N-hydroxysuccinimide and carbon dioxide, diaminopropionic acid and diaminobutyric acid, respectively,
and after 1 min no further derivatisation can take place. by treatment of protein/peptide with BTI described in
Method 11 under Protein hydrolysis.
Peak areas for AQC-amino acids are essentially unchanged fol'
at least 1 week at room temperature. Therefore AQC-amino The non-proteinogenic amino acid norleucine cannot be used
acids have more than sufficient stability to allow for overnight as an internal standard in this method as this compound is
automated chromatographic analysis. eluted in a chromatographic region crowded with peaks of
primary amino acids. Nitrotyrosine can be used as an internal
The detection limit is considered to range from about standard because it is eluted in a clean region.
40 fmol to 320 fmol for each amino acid, except for cystein.
The detection limit for cystein is approximately 800 fmo!. The detection limit of DABS-amino acid is about 1 pmo!.
Response linearity is obtained in the range of 2.5-200 flM with As little as 2-5 pmol of an individual DABS-amino acid can
correlation coefficients exceeding 0.999. Good compositional be quantitatively analysed with l'eliability, and only 10-30 ng
data can be obtained from the analysis of derivatised protein of the dabsylated protein hydrolysate is required for each
hydrolysates derived from as little as 30 ng of protein/peptide. analysis.
METHOD 5 - PRE-COLUMN OPA DERIVA TISATION METHOD 7 - PRE-COLUMN FMOC-Cl DERIVA TISA TION
Pre-column derivatisation of amino acids with Pre-column derivatisation of amino acids with
o-phthalaldehyde (OPA) followed by reversed-phase HPLC 9-fluorenylmethyl chloroformate (FMOC-Cl) followed by
separation with fluorometric detection is used. This technique reversed-phase HPLC separation with fluorometric detection
does not detect amino acids that exist as secondary amines is used.
(e.g., proline). FMOC-Cl reacts with both primary and secondary amino
OPA in conjunction with a thiol reagent reacts with primary acids to form highly fluorescent products. The reaction
amine groups to form highly f1uorescent isoindole products. proceeds under mild conditions in aqueous solution and is
2-Mercaptoethanol or 3-mercaptopropionic acid can be used completed in 30 s. The derivatives are stable, only the histidine
as the thio!. OPA itself does not fluoresce and consequently derivative showing any breakdown. Although FMOC-CI
produces no interfering peaks. In addition, its solubility and is fluorescent itself, the reagent excess and fluorescent
stability in aqueous solution, along with the rapid kinetics for side-products can be eliminated without IOS5 of FMOC-amino
the reaction, make it amenable to automated derivatisation acids.
and analysis using an autosampler to mix the sample with the FMOC-amino acids are separated by a reversed-phase HPLC
reagent. However, lack of reactivity with secondary amino using an ODS column. The separation is carried out by
acids has been a predominant drawback. This method does gradient elution varied linearly from a mixture of 10 volumes
not detect amino acids that exist as secondary amines (e.g., of acetonitrile, 40 volumes of methanol and 50 volumes of
proline). To compensate for this drawback, this technique may acetic acid buffer to a mixture of 50 volumes of acetonitrile
be combined with another technique described in Method 7 and 50 volumes of acetic acid buffer and 20 amino acid
or Method 8. derivatives are separated in 20 mino Each derivative eluted

General Notíces (1) apply to all monographs and other texts 95


2.2.56. Amina acid analysis EUROPEAN PHARMACOPOEIA 8.0

from the column is monitored by a fluorometric detector in which m is the recovered quantity, in nanomoles, of the
set at an excitation wavelength of 260 nm and an emission amino acid under test; and M, is the average molecular mass
wavelength of 313 nm. for that amino acid, corrected for the mass of the water
The detection limit is in the low femtomole range. A linearity molecule that was eliminated during peptide bond formation.
range ofO.1-50 flM is obtained for most ofthe amino acids. The sum of the masses of the recovered amino acids will give
an estimate of the total mass of the protein analysed after
METHOD 8 - PRE-COLUMN NBD-F DERIVA TISA TION
appropriate correction for partially and completely destroyed
Pre-column derivatisation of amino acids with amino acids. If the molecular mass of the unknown protein is
7 -fluoro-4-nitrobenzo-2-oxa-l,3-diazole (NBD-F) followed by available (i.e., by SDS-PAGE analysis or mass spectroscopy),
reversed-phase HPLC separation with fluorometric detection the amino acid composition of the unknown protein can be
is used. predicted. Calculate the number of residues of each amino
NBD-F reacts with both primary and secondary amino acid using the formula:
acids to form highly fluorescent products. Amino acids are
m
derivatised with NBD-F by heating to 60 oC for 5 mino
NBD-amino acid derivatives are separated on an ODS
column of a reversed-phase HPLC by employing a gradient
elution system consisting of acetonitrile and aqueous buffer
mixture, and 17 amino acid derivatives are separated in in which m is the recovered quantity, in nano111oles, of the
35 mino E-Aminocaproic acid can be used as an internal amino acid under test; M is the total mass, in micrograms, of
standard, because it is eluted in a clean chromatographic the protein; and is the molecular mass of the unknown
region. Each derivative eluted from the column is monitored protein.
by a fluorometric detector set at an excitation wavelength of
480 nm and an emission wavelength of 530 nm. Known pmtein samples. This data analysis technique can be
The sensitivity of this method is almost the same as for used to investigate the amino acid composition and protein
the pre-column OPA derivatisation method (Method 5), concentration of a protein sample of known molecular mass
excluding proline to which OPA is not reactive, and might be and amino acid composition using the amino acid analysis
advantageous for NBD-F against OPA. The detectionlimit data. When the compositiol1 of the protein being analysed
for each amino acid is about 10 fmol. Profile analysis can be is known, one can exploit the fact that some amino acids
achieved with about 1.5 mg of protein hydrolysates in the are recovered well, while other amino acid recoveries may
pre-column reaction mixture. be compromised because of complete 01' partial destruction
(e.g., tryptophan, cysteine, threonine, serine, methionine),
DATA CALCULATION AND ANALYSIS incomplete bond cleavage (i.e., for isoleucine and valine) and
When determining the amino acid content of a protein/peptide free amino acid contamination (i.e., by glycine and serine).
hydrolysate, it should be noted that the acid hydrolysis step
destroys tryptophan and cysteine. Serine and threonine are Because those amino acids that are recovered best represent
partially destroyed by acid hydrolysis, while isoleucine and the protein, these amino acids are chosen to quantify the
valine residues may be only partially cleaved. Methionine can amount of protein. Well-recovered amino acids are, typically,
undergo oxidation during acid hydrolysis, and some amino aspartate-asparagine, glutamate-glutamine, alanine, leucine,
acids (e.g., glycine and serine) are common contaminants. phenylalanine, lysine, and arginine. This list can be modified
Application of adequate vacuum (iess than 200 flm of based on experience with one's own analysis system. Divide the
mercury or 26.7 Pa) or introductiol1 of inert gas (argon) in quantity, in nanomoles, of each of the well-recovered amino
the headspace of the reaction vessel during vapour phase acids by the expected number of residues for that amino acid
hydrolysis can reduce the level of oxidative destruction. to obtain the protein content based 011 each well-recovered
Therefore, the quantitative results obtained for cysteine, amino acid. Average the protein content results calculated.
tryptophan, threonine, isoleucine, valine, methionine, glycine, The protein content determined for each of the well-recovered
and serine from a protein/peptide hydrolysate may be variable amino acids should be evenly distributed about the mean.
and may warrant further investigation and consideration. Discard protein content values for those amino acids that
have an unacceptable deviation from the mean. Typically
Amino Add Mole Percent. This is the number of specific greater than 5 per cent variation fro111 the mean is considered
amino acid residues per 100 residues in a protein. This unacceptable. Recalculate the mean protein content from the
result may be useful for evaluating amino acid analysis data remaining values to obtain the protein content of the sample.
when the molecular mass of the protein under investigation Divide the content of each amino acid by the calculated mean
is unknown. This information can be used to corroborate protein content to determine the amino acid composition of
the identity of a protein/peptide and has other applications. the sample by analysis.
Carefully identify and integrate the peaks obtained as directed
for each procedure. Calculate the mole percent for each amino Calculate the relative compositional error, in percentage, using
acid present in the test sample using the formula: the formula:
lOOr u
100m
r
ms
in which r u is the peak response, in nanomoles, of the amino
acid under test; and r is the sum of peak responses, in
in which m is the experimentally determined quantity, in
nanomoles, for al! amino acids present in the test sample.
nanomoles peY amino acid residue, of the amino acid under
Comparison of the mole percent of the amino acids under test
test; and m 5 is the known residue value for that amino acid.
to data from known proteins can help establish or corroborate
The average relative compositional error is the average of
the identity of the sample protein.
the absolute values of the relative compositional errors of
Unknown Protein Samples. This data analysis technique can the individual amino acids, typically excluding tryptophan
be used to estimate the protein concentration of an unknown and cysteine from this calculation. The average relative
protein sample using the amino acid analysis data. Calculate compositional error can provide important information on
the mass, in micrograms, of each recovered amino acid using the stability of analysis run over time. The agreement in the
the formula: amino acid composition between the protein sample and the
mNlr lmown composition can be used to corroborate the identity
1000 and purity oí the protein in the sample.

96 See the informa/ion section on monographs (ca ver pages)


EUROPEAN PHARMACOPOEIA 8.0 2.2.57. Inductively wupled plasma-atomic emission spectrometry

0112008:20257 - wavelength dispersive devices consisting of diffraction


gratings, prisms, filters or interferometers;
2.2.57. INDUCTIVELY COUPLED - detectors converting radiant energy into electrical energy;
- data-acquisition unit.
PLASMA-ATOMIC EMISSION
SPECTROMETRY INTERFERENCE
Interference is anything that causes the signal from an analyte
GENERAL PRINCIPLE in a sample to be different from the signal for the same
Inductively coupled plasma-atomic emission spectrometry concentration of that analyte in a calibration solution. The
(ICP-AES) is an atomic emission spectrometry method that well-known chemical interference that is encountered in flame
uses an inductively coupled plasma (ICP) as the excitation atomic absorption spectrometry is usually weak in ICP-AES.
source. In rare cases where interference occurs, it may be necessary to
An ICP is a highly ionised inert gas (usually argon) increase the RF power or to reduce the inner support-gas flow
with equal numbers of electrons and ions sustained by a to eliminate it. The interference in ICP-AES can be of spectral
radio-frequency (RF) field. The high temperature reached origin or even the result of high concentrations of certain
in the plasma successively desolvates, vaporises, excites elements or matrix compounds. Physical interference (due to
- atomic emission spectrometry (AES) detection - and differences in viscosity and surface tension of the sample and
ionises - mass spectrometry (MS) detection - atoms from calibration standards) can be minimised by dilution of the
the sample. Detection limits are, generally, in the lower sample, matrix matching, use of internal standards or through
nanogram (ICP-MS) to microgram (ICP-AES) per litre range. application of the method of standard additionso
Another type of interference occasionally encountered in
The plasma is formed by a tangential stream of support gas
ICP-AES is the so-called 'easily ionised elements (ElEs) effect'.
through a 'torch', i.e. a system consisting of 3 concentric
The EIEs are those elements that are ionised much more
quartz tubes. A metal coil (the load coil) surrounds the top
end of the torch and is connected to a radio-frequency (RF) easily, for example alkaline metals and alkaline earths. In
generator. Power (usually 700-1500 W) is applied through the samples that contain high concentrations of EIEs (more than
0.1 per cent), suppression or enhancement of emission signals
coil and an oscillating magnetic field corresponding to the
is likely to occur.
frequency of the generator (in most cases 27 MHz, 40 MHz)
is formed. The plasma forms when the support gas is made Spectral interference. This may be due to other lines or shifts
conductive by exposing it to an electric discharge, which in background intensity. These lines may correspond to argo n
produces seed electrons and ions. Inside the induced magnetic (observed aboye 300 nm), OH bands due to the decomposition
field, the charged particles (electrons and ions) are forced of water (at about 300 nm), NO bands due to the interaction of
to flow in a closed annular path. As they meet resistance the plasma with the ambient air (between 200 nm and 300 nm),
to their flow, heating takes place producing additional and other elements in the sample, especially those present at
ionisation. The process occurs almost instantaneously, and high concentrations. The interference falls into 4 different
the plasma expands to its fuI! strength and dimensions. The categories: simple background shift, sloping background shift,
radio-frequency oscillation of the power applied through the direct spectral overlap, and complex background shift.
coi! causes radio-frequency electric and magnetic fields to Absorption interference. This arises when part of the
be set up in the are a at the top of the torcho When a spark emission from an analyte is absorbed before it reaches
(produced by a Tesla tube or sorne other seeding device) the detector. This effect is observed particularly when the
is applied to the support gas flowing through the torch, concentration of a strongly emitting element is so high that
sorne e1ectrons are stripped from the support gas atoms. the atoms or ions of that element that are in the lower energy
These electrons are then caught up in the magnetic field and state of transition absorb significant amounts of the radiation
acceleratedo Adding energy to the electrons by the use of a coi! emitted by the relevant excited specieso This effect, known
is known as inductive coupling. These high-energy electrons as self-absorption, determines the upper end of the linear
in turn collide with other support -gas atoms, stripping off still working range for a given emission lineo
more electrons. The collisional ionisation of the support gas
continues in a chain reaction, breaking down the gas into a Multicomponent spectral fitting. Multiple emission-line
physical plasma consisting of support-gas atoms, electrons determinations are commonly used to overcome problems
and support-gas ions. The plasma is then sustained within the with spectral interferences. A better, more accurate method
torch and load coi! as radio-frequency energy is continually for performing spectral interference corrections is to
transferred to it through the inductive coupling processo use the information obtained with advanced detector
systems through multicomponent spectral fitting. This
The ICP appears as an intense, very bright, plume-shaped quantifies not only the interference, but also the background
plasma. At the base the plasma is toroidal, and this is referred contribution from the matrix, thereby creating a correction
to as the induction region (IR), i.e. the regio n in which the formula. Multicomponent spectral fitting utilises a multiple
inductive energy transfer from the load coil to the plasma linear-squares model based on the analysis of pure analyte,
takes place. The sample is introduced through the induction the matrix and the blank, creating an interference-corrected
regio n into the centre of the plasma. mathematical model. This permits the determination of the
analyte emission in a complex matrix with improved detection
APPARATUS
limits and accuracy.
The apparatus consists essentially of the following elements :
PROCEDURE
- sample-introduction system consisting of a peristaltic
pump delivering the solution at constant flow rate into a SAMPLE PREPARATION AND SAMPLE INTRODUCTION
nebuliser; The basic goal for the sample preparation is to ensure that the
radio-frequency (RF) generator; analyte concentration falls within the working range of the
instrument through dilution or preconcentratiol1, and that the
- plasma torch; sample-containing solution can be nebulised in a reproducible
- transfer optics focussing the image of the plasma at the manner.
entrance slit of the spectrometer; radial viewing is better Several sample-introduction systems tolerate high acid
for difficult matrices (alkalis, organics), whereas axial concentrations, but the use of sulfuric and phosphoric
viewing gives more intensity and better detection limits in acids can contribute to background emission observed in
simple matrices; the ICP spectra. Therefore, nitric and hydrochloric acids

General Notices (1) apply to all monographs aná other texts 97


2.2.58. Inductively coupled plasma-mass spectrometry EUROPEAN PHARMACOPOEIA 8.0

are preferable. The availability of hydrofluoric acid-resistant linear and quadratic weighting functions are applied to the
(for example perfluoroalkoxy polymer) sample-introduction data to find the most appropriate weighting function to be
systems and torches also allows the use ofhydrofluoric acid. In employed.
selecting a sample-introduction method, the requirements for If the means compared to the calibration curve show a
sensitivity, stability, speed, sample size, corrosion resistance deviation from linearity, two-dimensionallinear regression
and resistance to clogging have to be considered. The use of is used.
a cross-flow nebuliser combined with a spray chamber and
ACCURACY
torch is suitable for most requirements. The peristaltic pumps
used for ICP-AES usually deliver the standard and sample Verify the accuracy preferably by using a certified reference
solutions at arate of 1 mL/ min or less. material (CRM). Where this is 110t possible, perform a test
for recovery.
In the case of organic solvents being used, the introduction of
oxygen must be considered to avoid organic layers. Recovery. For assay determinations a recovery of 90 per
cent to 11 O per cent is to be obtained. The test is not valid
CHOICE OF OPERA TING CONDITIONS if recovery, for example for trace-element determination,
The standard operating conditions prescribed by the is outside of the range 80 per cent to 120 per cent of the
manufacturer are to be followed. Usual!y, different sets of theoretical value. Recovery may be determined on a suitable
operating conditions are used for aqueous solutions and for reference solution (matrix solution) spiked with a known
organic solvents. Suitable operating parameters are to be quantity of analyte (concentration raIlge that is relevant to
properly chosen: the samples to be determined).
- wavelength selection; REPEATABILITY
- support-gas flow rates (outer, intermediate and inner tubes The repeatability is 110t greater than 3 per cent for an assay
of the torch); and not greater than 5 per cent for an impurity test.
- RF power; LIMIT OF QUANTIFICATION
- viewing position (radial or axial); Verify that the limit of quantification (for example, determined
using the 10 (J approach) is below the value to be measured.
- pump speed;
- conditions for the detector (gain/voltage for photomultiplier
tube detectors, others for array detectors) ; 0112008:20258
- integration time (time set to measure the emission intensity
at each wavelength). 2.2.58. INDUCTIVELY COUPLED
CONTROL OF INSTRUMENT PERFORMANCE
PLASMA-MASS SPECTROMETRY
System suitability
The following tests may be carried out with a multi-element
1nductively coupled plasma-mass speetrometry (ICP-MS) is a
control solution to ensure the adequate performance of the
mass spectrometry method that uses an inductively coupled
ICP-AES system:
plasma (ICP) as the ionisation source. The basic principIes of
- energy transfer (generator, torch, plasma); measurement of ICP formation are described in chapter 2.2.57 on inductively
the ratio Mg JI (280.270 nm)/Mg I (285.213 nm) may be coupled plasma-atomic emission speetrometry (ICP-AES).
used;
ICP-MS utilises the ability of the ICP to generate charged
- sample transfer, by checking nebuliser efficiency and ions from the element species within a sample. These ions
stability; are then directed into a mass spectrometer, which separates
- resolution (optical system), by measuring peak widths at them according to their mass-to-charge ratio (miz). Most
halfheight, for example As (189.042 nm), Mn (257.610 nm), mass spectrometers have a quadrupole system or a magnetic
Cu (324.754 11m) or Ba (455.403 nm); sector. 10ns are transported from the plasma through 2 eones
(sampler and skimmer eones, forming the interface region)
- analytical performance, by calculating detection limits of
to the ion optics. The ion optics consist of an electrostatic
selected elements over the wavelength range.
lens, which takes ions from an area at atmospheric pressure
VALIDATION OF THE METHOD to the mass filter at a vacuum of 10- 8 Pa or less, maintained
with a turbomolecular pump. After their filtration, ions of the
Satisfactory performance of methods prescribed in selected massl charge ratio are direeted to a detector (channel
monographs is verified at suitable time intervals. electromultiplier, Faraday cup, dynodes), where ion eurrents
LINEARITY are converted into electrical signals. The element is quantified
Prepare and analyse not fewer than 4 reference solutions over according to the number of ions arriving and generating
the calibration range plus a blank. Perform not fewer than electrical pulses per unit time.
5 replicates. The sample-introduction system and data-handling techniques
The calibration curve is calculated by least -square regression of an ICP-AES system are also used in ICP-MS.
from al! measured data of the calibration test. The regression
APPARATUS
curve, the means, the measured data and the confidence
interval of the calibration curve are plotted. The operating The apparatus consists essentially of the following elements:
method is valid when: - sample-introduction system, consisting of a peristaltie
- the correlation coefficient is at least 0.99; pump delivering the solution at constant flow rate into a
nebuliser;
- the residuals of each calibration level are randomly
- radio-frequency (RF) generator;
distributed around the calibration curve.
- plasma torch;
Calculate the mean and relative standard deviation for the
lowest and for the highest calibration leve!. - interface region including eones to transport ions to the
ion optics;
When the ratio of the estimated standard deviations of the
- mass spectrometer;
lowest and the highest calibration level is less than 0.5 or
greater than 2.0, a more precise estimation of the calibration - detector;
curve may be obtained using weighted linear regression. Both - data-acquisition unit.

98 See the information section 011 general mOl1ographs (cover pages)


EUROPEAN PHARMACOPOEIA 8.0 2.258. Inductively coupled plasma-mass spectrometry

INTERFERENCE CONTROL OF INSTRUMENT PERFORMANCE


Mass interference is the major problem, for example by System suitability
isobaric species that significantly overlap the mass signal of the - Tuning of the instrument allows to monitor and adjust
ions of interest, especially in the central part of the mass range the measurement before running samples. ICP-MS mass
(for example 40-80 a.m.u.). The combination of atomic ions accuracy is checked with a tuning solution containing
leads to polyatomic or molecular interferences (i.e. 4°Ar160 several isotopes covering the whole range of masses, for
with 56Fe or 4°Ar"oAr with SOSe). Matrix interference may also exan1ple 9Be, 59CO, 89y, 1I5In, HOCe and 209Bi.
occur with some analytes. Some samples have an impact on
- Sensitivity and short- and long-term stability are recorded.
droplet formation or 011 the ionisation temperature in the
The instrument parameters (plasma condition, ion lenses
plasma. These phenomena may lead to the suppression of
and quadrupole parameter) are to be optimised to obtain
analyte signals. Physical interference is to be circumvented by
the highest possible number of counts.
using the method of internal standardisation or by standard
addition. The element used as internal standard depends on - Tuning for resolution and mass axis is to be done with a
the element to be measured: 59CO and 115In, for example, can solution of Li, Y and TI to ensure an acceptable response
be used as internal standards. over a wide range of masses.
The prime characteristic of an ICP-MS instrument is its - Evaluation of the efficiency of the plasma to decompose
resolution, i.e. the efficiency of separation of 2 close masses. oxides has to be performed in order to minimise these
Quadrupole instruments are, from this point of view, inferior interferences, The ratio Ce/CeO and/or Ba/BaO is a good
indicator, and a levelless than about 3 per cent is required.
to magnetic-sector spectrometers.
- Reduction of the formation of double-charged ions is made
PROCEDURE with Ba and Ce. The ratio of the signal for double-charged
ions to the assigned element should be less than 2 per cent.
SAMPLE PREPARATIONS AND SAMPLE INTRODUCTION
- Long-term stability is checked by running a standard
The sample preparation usually involves a step of digestion of
first and at the end of the sample sequence, controlling
the matrix by a suitable method, for example in a microwave
whether salt deposits on the cones have reduced the signal
oven. Furthermore, it is important to ensure that the
throughout tlle runo
analyte concentration falls within the working range of the
instrument through dilution or preconcentration, and that the VALIDATION OF THE METHOD
sample-containing solution can be nebulised in a reproducible
Satisfactory performance of methods prescribed in
manner,
monographs is verified at suitable time intervals.
Several sample-introduction systems tolerate high acid LINEARITY
concentrations, but the use of sulfuric and phosphoric acids
Prepare and analyse not fewer than 4 reference solutions over
can contribute to background emission. Therefore, nitric
the calibration range plus a blank. Perform not fewer than
and hydrochloric acids are preferable. The availability of
5 replicates.
hydrofluoric acid-resistant (for example perfluoroalkoxy
polymer) sample-introduction systems and torches also allows The calibration curve is calculated by least-square regressiol1
the use ofhydrofluoric acid. In selecting a sample-introduction from all measured data of the calibration test. The regression
method, the requirements for sensitivity, stability, speed, curve, the means, the measured data and the confidence
sample size, corrosion resistance and resistan ce to clogging interval of the calibration curve are plotted. The operating
have to be considered. The use of a cross-flow nebuliser method is valid when:
combined with a spray chamber and torch is suitable for - the correlatiol1 coefficient is at least 0.99;
most requirements. The peristaltic pumps usually deliver the - the residuals of each calibration leve! are randomly
standard and sample solutions at arate of 20-1000 flLlmin. distributed around the calibration curve.
In tlle case of organic solvents being used, the introduction of Calculate the mean and relative standard deviation for the
oxygen must be considered to avoid organic layers, lowest and for the highest calibration leve!.
CHOICE OF OPERATING CONDITIONS Wllen the ratio of the estimated standard deviations of the
The standard operating conditions prescribed by the lowest and the highest calibration level is less than 0.5 or
manufacturer are to be followed. Usually, different sets of greater than 2.0, a more precise estimation of the calibration
operating conditions are used for aqueous solutions and for curve may be obtained using weighted linear regression. Both
organic solvents. Suitable operating parameters are to be linear and quadratic weighting functions are applied to the
properly chosen: data to find the m05t appropriate weighting function to be
employed.
- se!ection of eones (material of sampler and skimmer);
lf the means compared to the calibration curve show a
- support-gas flow rates (outer, intermediate and inner tubes deviation from linearity, two-dimensionallinear regressiol1
of the toreh); is used.
- RF power; ACCURACY
- pump speed; Verify tlle accuracy preferably by using a certified reference
- selection of one or more isotopes of the element to be material (CRM). Where this is not possible, perform a test
measured (mass). for recovery.
Recovery. For assay determinations a recovery of 90 per
ISOTOPE SELECTION cent to 110 per cent is to be obtained. The test is not valid
if recovery, for example for trace-element determination, is
Isotope selection is made using several criteria. The m05t
outside the rauge 80 per cent to 120 per cent of the theoretical
abundant isotope for a given element is selected to obtain
value. Recovery may be determined on a suitable reference
maximum sensitivity. Furthermore, an isotope with the least
solution (matrix solution) spiked with a known quantity of
interference from other species in the sample matrix and
analyte (concentration range that is relevant to the samples
from the support gas should be selected. Information about
to be determined).
isobaric interferenees and interferences from polyatomic ions
of various types, for example hydrides, oxides, chlorides, etc., REPEATABILITY
is usually available in the software of ICP-MS instrument The repeatability is not greater than 3 per cent for an assay
manufacturers. and 110t greater than 5 per cent for an impurity test.

General No/ices (1) apply to all monographs and other texts 99


2.2.59. Glycan analysis of glycoproteins EUROPEAN PHARMACOPOEIA 8.0

LIMIT OF QUANTIFICATION - in the type of glycosylation (N- or O-linked);


Verify that the limit of quantification (for example, determined - in the oligosaccharide structures (extensions, branching
using the 10 (J approach) is below the value to be measured. and linkage).
This heterogeneity in glycosylation results in a set of gIycoforms
for one specific glycoprotein. These variations arise because,
unlike transcription and translation, glycosylation is a
0112011:20259 non -template post -translational modification process. The
glycosylation pattern at a given site depends on many
factors including the cell-specific and/or growth-dependent
2.2.59. GLYCAN ANALYSIS OF availability of glycosyltransferases and exo-glycosidases found
GLYCOPROTEINS in the Golgi apparatus and endoplasmic reticulum. Protein
glycosylation is also influenced by the protein structure, the
1. INTRODUCTION production process, the host-vector expression system and the
Glycan analysis is a test to analyse glycan moieties of ceU culture conditions.
glycoproteins. It may involve:
- whole glycoprotein analysis; 2. GLYCAN ANALYSIS PROCEDURES
- separation and detection of protein glycoforms; Heterogeneity in glycosylation can be assessed by 4 distinct
- analysis of glycopeptides obtained after enzymatic and complementary approaches:
treatment of the glycoprotein; - analysis of the intact glycoprotein;
- analysis of released glycans obtained after chemical or
enzymatic treatment of the glycoprotein. - analysis of glycopeptides;
Monosaccharide analysis may complement information - analysis of released glycans;
obtained by glycan analysis.
- monosaccharide analysis.
Glycosylation can playa predominant role in determining the
function, pharmacokinetics, pharmacodynamics, stability, The present section provides methods and general
and immunogenicity of biotherapeutics. Glycosylation, requirements used for glycan analysis of glycoproteins
unlike transcription, is a non-template-driven enzymatic containing N- and O-linked glycans.
modification process that results in glycan heterogeneity. The Glycan analysis is usually a multistep process. There are
manufacturing procedure also has an influence on glycan numerous methodologies for glycan analysis. This variety
heterogeneity. Glycoprotein glycan analysis may therefore be is a consequence of the diversity and complexity of glycan
an important test to identify variations in the glycosylation structures, of the available technologies and detection systems,
pattern of the glycoprotein and/or monitor the consistency of and of the wide range of approaches depending on the level of
the glycosylation pattern during production. information required.
Glycan analysis can be a comparative pro ce dure, because
Figure 2.2.59.-1 provides an overview of glycan analysis
the information obtained, compared to a similarly treated
analytical procedures that can be employed to apply the chosen
reference substance, confirms product consistency.
approach(es). Many variations of the same techniques and
This chapter provides approaches used for glycoprotein glycan conditions are available depending on the glycan structures
analysis and requirements for the application of methods and and origino
validation of methods.
Isolation and purification. Isolation and purification may be
Glycan analysis is not a single general method, but involves necessary for analysis ofbulk drug substances or dosage forms
the application of specific procedures and the development of containing interfering excipients, and, when required, will be
specific glycan maps for each unique glycoprotein. Specific described in the specific monograph.
procedures are therefore indicated in relevant specific
monographs. 2-1. ANALYSIS OF INTACT GLYCOPROTEIN
1-1. PROTEIN GLYCOSYLATION
Analysis of the intact glycoprotein provides informatíon on
the overall pattern of glycosylatiol1 of the glycoprotein.
There are 3 main types of enzymatic glycosylation found in
proteins: This approach provides limited information when the
- N-glycosylation, which involves the addition of molecule is large and contains multiple glycosylation sites.
oligosaccharides to the nitro gen on the terminal amide Methods such as capillary electrophoresis (CE) (2.2.47) and
group of asparagine; mass spectrometry (MS) (2.2.43) can be used. Size-based
O-glycosylation, which involves the addition of techniques, such as size-exclusion chromatography (2.2.30)
oligosaccharides to the hydroxyl groups of serine, and sodium dodecyl sulfate polyacrylamide gel electrophoresis
threonine, and/or hydroxyproline; (SDS-PAGE) (2.2.31), may provide information on the
C-glycosylation, which involves the addition of an glycosylation status of a protein. If the degree of sialylation
a-mannopyranose to the C2-carbon of the indole ring of significantly contributes to the biological activity of the
tryptophan. glycoprotein, ion-exchange chromatography (2.2.46),
isoelectric focusing (IEF) (2.2.54) or CE (2.2.47) may be
Non-enzymatic additions, also known as glycation, can occur
performed to monitor sialylation. The technique must
when proteins are incubated with reducing sugars.
be chosen according to its suitability to provide a reliable
This chapter describes analytical methods for the N- and correlation between the degree of sialylation and the
O-linked glycosylations, which are the most commonly found bioactivity of the product.
in glycoprotein medicinal products.
2-2. ANALYSIS OF GLYCOPEPTIDES
1-2. HETEROGENEITY OF THE PROTEIN Analysis of glycopeptides provides information on site-specific
GLYCOSYLATION gIycosylation properties, on the degree of occupancy, and on
Different levels of glycan heterogeneity can appear during the the oligosaccharide structures. It involves proteolytic digestion
production of glycoproteins. This heterogeneity may result of the glycoproteín. Approaches to site-specific cleavage of the
from variations: protein backbone are given in general chapter 2.2.55. Peptide
- in the degree of occupancy (full, partia!, unoccupied); mapping.

100 See the information section on general monographs (caver pages)


EUROPEAN PHARMACOPOEIA 8.0 2.2.59. Glycan analysi.s of glycoproteins

ANALYSIS OF INTACT GL YCOPROTEIN

MS
IEF
CE
--1l> ion-exchange chromatography
Size-exclusion chromatography
SDS-PAGE

ANAL YSIS OF GL YCOPEPTIDES


Diree! analysis
I GLYCOPROTEIN I

Separation prior lo direet analysisl MS


Proteolysis
LC/CE 1

PRETREATMENT J i
Deglyeosylation
(isolation & purifieation) --1l> 1 1
'-----
·1 of the digest
if neeessary
:
...
ANAL YSIS OF RELEASED GL YCANS Analysis of unlabelled glyeans
HPAEC-PAD
DeSiaIYlation~[ PGC-MS
MS

Release of glyeans
--1l> 0- and/or N-linked glycans:
enzymes or chemicals H
Analysis of labelled glyeans
Exo-glyeosidases
treatment

Labelling
of glycans l i MS

~I Separation
LC/CE
'CE
MS
Le
ANAL YSIS OF MONOSACCHARIDES

HPAEC-PAD
Acid PGC-MS
~
hydrolysis H Fluorophore
labelling
1_
----->- CE
LC

HColorimetrie methods[

CE: Capillaryelectrophoresis LC: Liquid chromatography


HPAEC-PAD: High-pH anion-exchange chromatography with pulsed MS: Mass spectrometry
amperometric detection PGC: Porous graphite chromatography
IEF: Isoelectric focusing SDS-PAGE: Sodium dodecyl sulfate polyacrylamide gel
electrophoresis
Figure 2.2.59.-1. - Overview ofglyean analysis proeedures

After proteolysis of the glycoprotein, the following approaches Deglycosylation of the glycopeptides. Identification of the
can be chosen. different glycosylation sites of a glycoprotein is made
Direet analysis by MS (2.2.43). Care should be taken that the possible by comparing peptide maps obtained by proteolytic
glycopeptide signal is not suppressed due to the presence of digestion of the intact glycoprotein to those obtained
other peptides, where glycopeptides represent a minor portion when the glycoprotein is deglycosylated previously or
of the total peptide mixture and where signal intensities are following proteolytic digestion. The peptide mass gives
lower than those of non-glycosylated peptides. information about the glycosylation sites and by calculating
the mass difference between the intact glycopeptide
Separation prior to analysis by MS. This additional step and the deglycosylated glycopeptide, it is possible to
overcomes the problems raised aboye. Enrichment or obtain information about the attached glycans concerning
fractionation techniques can be used either in parallel with or composition and heterogeneity. Approaches to deglycosylation
sequentially to direct analysis. Separation techniques such of the protein backbone are given in section 2-3-1. A separation
as liquid chromatography (LC) (2.2.29) and CE (2.2.47) are step can be performed after or before deglycosylation.
suitable. These techniques may be interfaced with MS to allow
online MS measurements.

General Notices (1) apply to all monographs and other texts 101
2.2.59. Glycan analysis of glycoproteins EUROPEAN PHARMACOPOEIA 8.0

2-3. ANALYSIS OF RELEA SED CLYCANS Analysis of glycans provides information on the various
Analysis of released glycans provides a convenient way to populations of glycans present 011 the protein (high-mannose,
obtain information on the various populations of glycans hybrid, complex). Information on the relative amounts
present on the protein (bi-, tri-, and tetra-antennary of branched structures might be obtained by analysis of
pro file ). The degree of sialylation can also be addressed desialylated glycans.
at this stage. Depending on the chosen method, prior A separation step may be required. 1t implies the use of Le
derivatisation/labelling may be needed to allow the detection (2.2.29) and CE (2.2.47) as intermediate techniques. Le
of the glycans. (2.2.29) can be used preparatively with individual fractions
Analysis of released glycans gene rally involves the release and being collected (usually labelling is required) or can be directly
purification of glycans from the reaction mixture, followed by coupled to MS (2.2.43).
the labelling/derivatisation ofthe glycans, where needed; the 2-3-2-1. Analysis of unlabelled glycans
glycans are then profiled (fractionation or separation).
Native glycans can be analysed by high-pH anion-exchange
2-3-l. Release of glycans chromatography with pulsed amperometric detection
The selection of the approach used for the releas e of glycans (HPAEC-PAD), porous graphite chromatography (PGC) and
will depend on the glycoprotein under test. The cleavage MS (2.2.43).
agent to be employed is chosen according to the type of HPAEC-PAD has high sensitivity and can also separate some
cleavage needed and level of information required. Enzymatic linkage isomers. Response factors of the different signals
or chemical cleavage may be used. TabIe 2.2.59.-1 gives a are not equal for the different oligosaccharide structures.
non-exhaustive list of enzymatic cleavage agents and their Absolute quantification of the glycan is not possible unless an
specificity. oligosaccharide reference library is available. Quantification
Digestion efficiency is generally dependent on the accessibility can be obtained by comparison with a well-characterised
of the glycans on the protein and hence the protein can be reference standard of the substance being tested, or by relating
denatured to maximise glycosylation site exposure, unless it is the peak area of each glycan to the total peak area of al!
desirable to distinguish between surface and buried glycans. glycans in the map.
Chemical cleavage agents might also be used, using for PGC can also be used to separate native glycans because of its
example hydrazine or alkaline borohydride for ~-elimination. higher selectivity compared to the conventional non-polar
columns. A PGC-electrospray-ionisation-MS approach can be
Table 2.2.59.-l. - Examples of enzymatic cleavage agents applied for direct glycan analysis.
Agents Spedl1dty 2-3-2-2. Analysis of labelled glycans
N-Iinked glycans release Labelling of glycans
Hydrolysis of an Nl-(acetyl-~-D- The type of derivatisation carried out will depend on the
glucosaminyl)asparagine residue method used to detect glycans: UV or fluorescent.
in which the glucosamine residue Derivatisation with fluorescent labels is the most commonly
Peptide- N"- (N-acety!-~-
may be further glycosylated,
glucosaminyl)asparagine amidas e used technique for labelling glycans at their reducing end by
to y;eld a (substitnted)
(EC 3.5.1.52)
N-acetyl- ~- D-glncosaminylamine reductive amination. One label can be attached to every single
and a peptide containillg an mono- and oligo-saccharide, allowing the determination of
aSlnrtate resioue
molar quantities. Table 2.2.59.-2 gives a non-exhaustive list
Release of N-glycan chain but no of commonly used fluorescent labels and suitable analytical
- Peptide N-glycosidase F (PNGase F) re!ease of N-glyean chain eontaining
(al-3)-linked eore fucose
techniques.

- Peptide N-glycosidase A (PNGase A)


Release of N-glyean chain eontaining Table 2.2.59.-2. - Examples ojfluorescent labels and suitable
(al-3)-linked core fucose tcchniques
Endohydrolysis of lhe
N,N'-diacetylchitobiosyl unit Name Acronym Analytical techniques
Mannosyl-glycoprotein
in high-mannose
endo- ~- N- acetylglucosaminidase 2-Aminobenzoic acid 2-AA LC (2.2.29), MS (2.2.43)
(EC 3.2.1.96) glycopeptides/ glycoproteins
containillg lhe 2-Aminobenzamide 2-AB LC (2.2.29), MS (2.2.43)
- [Mall( GlcNAc) 2J Asn structnre
- Endo-~-N-acetylglucosaminidase F Re!ease of high -mannose, hybrid 2-Aminopyridine 2-AP LC (2.2.29), MS (2.2.43)
(endo F) and eomplex oligosaecharides
2-Amino-9(lOH)- AMAC Gel electrophoresis (2.2.23)
- Endo-~-N-acetylglucosal11inidase H Release ofhigh-l11annose, hybrid acridinone
(endo H) oligosaccharides
Trisodium 8-aminopyrene-
APTS CE (2.2.47)
O-Iinked glyeans release 1,3,6-trisulfonic acid
Glycopeptide Hydrolysis of terminal Permethylation of glycans may also be used when MS (2.2.43)
a-N-acetylgalactosaminidase (EC D- galactosyl-N-acetyl-a- D- is used alone for detection. It is based on the methylation of
3.2.1.97)* galactosaminidic residnes
the oligosaccharides.
* This enzyl11e has lil11ited usage because of its high substrate specificity.
Analysis of labelled glycans
2-3-2. Anaiysis of glycans Labelled glycans can be analysed by analytical techniques su eh
Released glycans can be analysed or profiled by as Le (2.2.29), CE (2.2.47) and MS (2.2.43).
chromatographic, electrophoretic and mass spectrometric According to the separation properties of the glycans, glycans
techniques, and in general by a combination of these can be profiled and quantified by several Le (2.2.29) systems
techniques. The choice of the method can be grouped using an appropriate label: reversed-phase (separation by
according to the nature of the glycans and level of information hydrophobicity), normal-phase (separation by size), and
required. anion-exchange (separation by charge) Le.

102 See the information sec/ion on general monographs (cover pages)


EUROPEAN PHARMACOPOEIA 8.0 2.2.59. Glycan analysis of glycoproteins

2-4. MONOSACCHARIDE ANALYSIS identificatiol1 of individual structures can require multi -step
Monosaccharide analysis provides information on the approaches using enzymatic and chemical reactions,
monosaccharide composition of a glycoprotein. Analysis of separation techniques and online or offline detection methods,
monosaccharides can be performed using either colorimetric and will most commonly use the charge-to-mass ratio of a
or separation methods. molecular ion, determined using a suitable mass spectrometric
method as the final basis for structure assignment.
2-4-1. Colorimetric methods 3-1-2. Comparative confirmati.on ofidentity
The colorimetric methods, which are based on chemical During routine application of the analytical method, the
staining, provide information on the quantity of specifl.c classes identity of the analytical target may be conflrmed by
of sugars such as sialic acids, neutral sugars and hexosamines. comparison with process or system suitability reference
standards. These may be generated from known,
2-4-2. Separation methods well-characterised glycoproteins, which may be of the same
The separation methods generate quantitative informatiol1 general class as the product being tested (e.g. fetuin for
on the overall monosaccharide composition. The methods complex N-linked glycoproteins), or may be derived from a
require acid hydrolysis pre-treatment of the oligosaccharide well-characterised batch of the product being tested, which
chains of the intact glycoprotein or released glycans, prior has been established as a reference standard. The following
to anaIysis. To reIease sialic acids, mild acid hydrolysis considerations apply to comparative assignment of structural
or enzymatic treatment is employed. The hydrolysis identity:
step is a signifl.cant so urce of variability and may require - in the case of a validated high reproducibility of the
product -specific validation. retention times, the absolute retention times can be used
Methods for separation and quantification of monosaccharides for correet assignment;
indude: - alternatively, a glycan marker can be injected at the
- the use ofHPAEC-PAD and PGC-MS, which allow beginning and at the end of the testing sequence and
the determination of molar quantities of native checked for any drifts in the retention times; based on these
monosaccharides (sialic acids, neutral sugars and alcohol reference chromatograms the glycans of the test samples
sugars) ; can be assigned;
fluorophore labelling of monosaccharides followed by - in cases where no standard is available to assign a11 glycan
separation methods such as reversed-phase or ion-exchange peaks in the test sample, absolute or normalised retention
chromatography, or CE. times can be used to monitor and ¡abe! unidentified glycan
peaks.
3. EVALUATION AND ANALYSIS OF DATA 3-2. CONFIRMATION OF COMPLIANCE OF THE
Data obtained from analytical methods for the analysis of SUBSTANCE BEING TESTED WITH QUALITATIVE
glycans can be analysed and evaluated for 3 different purposes: REQUIREMENTS
- confirmation oí identity of individual structures or families At this level of evaluation, the analytical results obtained
of structures; with the product being tested are evaluated to demonstrate
compliance with specifications. Typically this is achieved by
- confl.rmation of compliance of the substance being tested
comparison with data obtained in parallel using a reference
with qualitative requirements;
standard of the substance being tested. In the data
- conÍirmation of compliance of the substance being tested it is necessary:
with quantitative requirements. - io establish that the analytical result obtained using the
Specific considerations with respect to reference standards reference standard is broadly comparable to the expected
and method development of each leve! of analysis are set out result, to verify the suitability of the system; for example,
in sections 4 and 5 respectively. in a glycan mapping procedure, this would be achieved
by comparison of the map obtained with the reference
3-1. CONFIRMATION OF IDENTITY OF INDIVIDUAL substance with a provided speeimen map obtained during
STRUCTURES OR FAMILIES OF STRUCTURES establishment of the reference substance, and by ensuring
The analytical target for a glycan ana!ysis method may be an compliance with al! stated system suitability criteria;
individual monosaccharide (e.g. sialic aeid, fucose), a defined - to demonstrate similarity of the maps obtained with
oligosaccharide structure (e.g. tetra -sialylated, tetra -antennary the reference substance and the test substance, using
glycan) or a family of structures sharing a common analytical any specifl.c compliance criteria given in the speeific
feature (e.g. tetra -sialylated glycans, tri -antennary glycans, monograph.
glycoprotein isoforms with the same charge). Confl.rmation of 3-3. CONFIRMATION OF COMPLIANCE OF THE
the identity of the analytical target is an essential step in the SUBSTANCE BEING TESTED WITH QUANTITATIVE
analysis and evaluation of data, and can be achieved absolutely, REQ UIREMENTS
by verification of molecular structure, or comparatively, by
3-3-1. Quantitative measurement of analyte levels and
comparison with al1 appropriate reference standard.
express ion of results
In some cases, e.g. measurement of sialic acid or other
3 -1-1. Absolute confirmation ofidentity monosaccharides, data can be expressed in order to obtain a
Absolute confirmation of the identity of glycan structures is molar ratio of sialic acid to glycoprotein. Data is calcu!ated
typically achieved during product development, and should by re fe rene e to a reference standard for sialic acid and to a
not necessarily be the target of routine analysis. Identity validated method of protein determination. Either the internal
of the analytical target will be assigned by reference to a or external standard method may be used (see general chapter
known molecular property of the molecule. Such absolute 2.2.46. Chramatagraphic separatian techniques).

General Natices (1) apply to all monographs and other texts 103
2.2.59. Glycan analysis oí glycoproteins EUROPEAN PHARMACOPOEIA 8.0

3-3-2. Quantitative expressions of separation pro file Release and Ísolation of oligosaccharides. The approach
chosen for the releas e of glycans will depend on the protein
Pro files or distribution patterns may be expressed numerically under test and will be based 011 the types of glycosylation, i.e.
in a number of ways, including the normalisation procedure; N- or O-linked glycosylation. Non-compendial approaches
the percentage content of each analytical target, e.g. glycan available for the release of glycans must be optimised in
entity, is calculated by determining the response of the glycan order to ascertain a quantitative profiling of al! glycan
entity as a percentage of the total response of aH the entities, entities. Factors that impact cleavage efficiency, such as
excluding those due to solvents or any added reagents, and enzyme-to-protein concentration ratio, temperature, reaction
those below the disregard limit. In addition, numerical time course, and denaturation of protein prior to digestion,
expressions such as the Z number, which are method- and must be optimised.
product-specific and defined in specific monographs, can be
used. It is noteworthy that the enzymatic/chemical reaction must
not alter the glycan composition, e.g. not destroy sialic acid
residues. Where there is more than one glycosylation site,
4. REFERENCE STANDARD S the enzymatic treatment should proportionally release all
oligosaccharide moieties attached to the protein, independent
Reference standards for glycan analysis serve 2 functions: of their structure and their individual position in the protein.
the verification of the suitability of the system and the Reproducible recovery of all glycan entities from the reaction
confirmation that the article under test complies with specified mixture must be confirmed.
requirements.
Derivatisation oC released glycans. Derivatisation is
The reference standards used for system suitability may be : usually carried out according to non-compendial protocols.
Therefore, the reproducible derivatisation of all glycan entities
- a reference substance for the substance being tested; must be verified. This may be achieved through optimisation
- glycan moities liberated from a fully characterised reference of the reaction conditions such as amount of the derivatisation
reagent, reaction temperature and time. The derivatisation
standard of the substance being tested;
reaction must not change the glycan composition, e.g. not
- well-characterised glycan moities liberated from destroy sialic acid residues.
glycoproteins (e.g. fetuin, IgG); Separation, identification and system suitability. The
methods employed for glycan analysis must be capable of
- glycan markers characterised for identity and purity.
detecting and separating different glycan moieties to ascertain
The reference standard used for compliance of the glycoprotein a reliable identification and quantification.
under test is a preparation of the substances being tested. 1t
is noted that glycan analysis procedures described in specific The acceptance criteria for system suitability, which also cover
monographs prescribe the use of a reference standard for glycan cleavage, recovery and analysis, depend on the critical
the substance being tested and for which the glycan analysis test parameters that affect the outcome of the resulto
procedure has been validated.
A comparison between the glycan map of the substance
under test and that of a reference substance, being treated
5. POINTS TO CONSIDER IN METHOD DEVELOPMENT in the same conditions, is an indicator to evaluate the
performance of the analytical procedure. In order to
This section provides means for measuring the overall further confirm the obtained results, the analyses may be
performance of the method during development. The extent repeated with an orthogonal method. The use of a reference
of method development and analytical validation is selected on standard (e.g. reference substance of the product being
the basis of their suitability for a specific producto Depending examined, system suitability glycan marker) is essential in the
on the chosen approach, several steps are necessary for glycan establishment of system suitability parameters and validation
analysis, for example: of the analytical procedure.
- isolation and purification (or desalting) of the glycoprotein; Reproducibility of quantitative expression (e.g. Z number
estimation) of glycan profiles must be verified.
- enzymatic (or chemical) treatment of the glycoprotein to
selectively releas e either N- or O-linked glycans from the Determinatiol1 of site occupancy based 011 relative
protein backbone; quantities of glycosylated and non-glycosylated peptides.
Where site occupancy is estimated by comparison of
- isolation and purification of the released glycans; glycosylated and non-glycosylated peptides from an
enzymatically digested glycoprotein, reproducible cleavage of
- verification of released sialic acid and monosaccharide
both forms of the peptide must be demol1strated.
residues;

- chromophore labelling of the released glycans;

- separation of the glycans, native or fluorescence labelled; 6. GLYCAN ANALYSIS DECISION-MAKING FRAMEWORK

- glycan identification and quantification (e.g. determination This decision-making framework is given for information
of the Z number) ; and does not constitute a mandatory part of the European
Pharmacopoeia.
- determination of site occupancy based on relative quantities
of glycosylated and non-glycosylated peptides. The choice of procedures used to analyse glycans is established
Protein isoJation and purification. Isolation and purification according to the leve! of information required to ensure
of the glycoprotein from its matrix may be necessary to the quality of the glycoprotein and is set up during the
remove all interfering substances (e.g. excipients, salts) and, development phase of the product.
when required, will be specified in the specific monograph.
This must be performed in a reproducible manner in order to Figure 2.2.59.-2 provides guidance in the choice of methods to
guarantee a quantitative recovery of the protein. be used when glycan analysis is required.

104 See the information section on general monographs (cave/' pages)


EUROPEAN PHARMACOPOEIA 8.0 2.2.60. Melting point - instrumental method

Glycan analysis necessary for the glycoprotein

Design glycan analysis methods

Small protein, limited glycosylation >---i~ Analysis of inlact glycoprotein (e.g.


and/or simple slructures? mass spectrometric analysis)
Ves L-----------r-----------~
No

Large molecule, complex branched glycans and/or


multiple glycosylation sites

Knowledge of site-specific Ves


glycosylation required?
>--+1 Site-specific glycosylation analysis

No

Negative charge content determination No Detailed glycan


Sialic acid and/or monosaccharides analysis required?
determination
Ves STOP

Design methods for analysis of cleaved glycans (e.g. antennary


profile, identificalion of individual slructure)

Figure 2.259.-2. - Guidance on methods to be used when glyean analysis is required

04/2008:20260 The temperature at which the sensor signal first leaves its
initial value is defined as the beginning of melting, and the
temperature al which the sensor signal reaches its final value
202.60. MELTING POINT - is defined as the end of melting, or the melting point.
INSTRUMENTAL METHOD Use glass capillary tubes that are open at one end, abont
100 mm long, with an external diameter of 1.3-1.5 mm and
This chapter describes the measurement of melting point
an internal diameter of 0.8- 1.3 mm. The wall thickness of the
by the capillary method using an instrumental method of
tube is 0.1-0.3 mm.
determination.
Sorne apparatuses allow for the determination of the melting
APPARATUS point 011 more than 1 capillary tube.
There are 2 modes of automatic observation arrangements:
METHOD
- mode A: by light transmission thraugh the capillary tube
loaded with the sample; Introduce into the capillary tube a sufficient amount of the
substance to be examined, previously treated as described in
- mode B: by light being reflected fram the sample in the the monograph, to form in each tube a compact column about
capillary tube. 4 mm high, and allow the tubes to stand for the appropriate
In both modes, the capillary tube sits in a hollow of a time at the prescribed temperature.
metal block, which is heated electrically and controlled by a Proceed as follows or according to the manufacturer's
temperature sensor placed in another hollow of the metal instructions. Heat the heating block until the temperature is
block The heating block is capable of being maintained about 5 oC below the expected melting point
accuratelyat a pre-defined temperature (± 0.1 OC) by the
heating element, and ofbeing heated at a slow and steady rate Place the capillary tube in the heating block with the dosed
of 1 °C/min, after an initial isothermal periodo end downwards. Start the temperature programme. When
the substance starts melting, it changes its appearance in the
In mode A, a beam oflight shines through a horizontal hollow capillary tube. As a result, the temperature of the heating
and crosses the capillary tube. A sensor detects the beam at block is recorded automatically following the signal changes
the end of the cylindrical hole after the capillary tube. from the photosensor due to light transmission (mode A,
In mode B, a beam of light illuminates the capillary tube from Figure 2.2.60.-1), or following image processing (mode B,
the front and the sensor records the image. Figure 2.2.60.-2).
Sorne apparatuses allow for the visual determination of the Carry out the test on 2 other samples and calculate the mean
melting point value of the 3 results.

General Notices (1) apply ta all monographs and other texts 105
2.2.61. Characterisation of crystaHine solids calorimetry EUROPEAN PHARMACOPOEIA 8.0

A
CALIBRATION

The temperature scale of the apparatus is checked periodically


by measuring the melting point of certified reference materials.
Use capillary tubes having the same dimensions as those used
for the determination of the melting point (see Apparatus).

Prepare 3 capillary tubes for each of at least 2 certified


reference materials. Carry out the test and calculate the mean
value of the 3 results for each material.

SYSTEM SUITABILITY

In addition to the calibratio11, carry out a verification, before


the measurements, using a suitable certified reference material
whose melting point is close to that expected for the substance
to be examined.

Prepare 3 capillary tubes. Carry out the test and calculate the B
mean value of the 3 results.
D e
The mean value is within the tolerance given on the certificate
supplied with the certified reference material.

G
E
A

A. glass capillary tube E. heating block


E B. sample F. light source

C. imagesensor G. transparent plate


D. temperature sensor

Figure 2.2.60.~2. - Mode B: reflexion

01/2013:20261

F
2.2.61. CHARACTERISATION
OF CRYSTALLINE SOLIDS BY
E) B
MICROCALORIMETRY AND
SOLUTION CALORIMETRY
e
For the purpose of this chapter, crystalline material, partially
crystalline material and amorphous material are considered as
o solids.
INTRODUCTION ~ THE CONCEPT OF CRYSTALLINITY
The perfectly ordered crystallattice with every molecule
in its expected lattice position is an ideal that is seldom, if
ever, achieved. The other extreme is the amorphous sta te,
in which a solid contains the maximum possible density of
imperfections (defects ofvarious dimensionalities), such that
E
a1l10ng~range order is 10st while only the short~range order,
imposed by its nearest neighbours, remains. Real crystals lie
somewhere between these 2 extremes. A crystal's position on
A. glass capillary tube D. temperature sensor a scale bounded by these 2 extremes is termed crystallinity.
Al! real crystals, even in the pure state, possess sorne
B. sample E. heating block lattice imperfections or defects, which increase both the
C. photosensor F. light sourec energy (enthalpy under conditions oí c011stant atmospheric
pressure) and the disorder (expressed as the entropy) of
the crystallattice. A crystal with a relatively low density
Figure 2.2.60.~ 1. - Mode A: transmission of imperfections is said to be highly crystalline and to

106 See the information section on general monographs (cover pages)


EUROPEAN PHARMACOPOEIA 8.0 2.2.61. Characterisation of crystalline solids by calorimetry

possess a high crystallinity. By contrast, a particle with a APPARATUS


relatively high density of imperfections is said to be partially Microcalorimeters are typically designed as twin systems with
amorphous and to possess a low crystallinity. In ideal terms, a a measuring vessel and a reference vessel. Vessels are typically
totally amorphous particle corresponds to zero crystallinity. made of glass or stainless steel. For certain applications
Amorphous particles may contain somewhat ordered domains specially designed vessels which allow the addition of a gas, a
that can act as nuclei for crystallisation; such so-called liquid or a solid material may be used.
amorphous particles are said to possess a low-Ievel but finite
CALIBRATION
crystallinity.
The microcalorimeter is calibrated for heat flow (energy per
The ability to detect and to quantify the amount of amorphous time unit) using either calibrated external or internal electrical
material within a highly crystalline substance is of great heat sources or a suitable standard reaction.
importance during the development and subsequent
manufacture of a pharmaceutical preparation. SENSITIVITY
The sensitivity of the microcalorimetric method can be
In reality, a powder probably contains particles with different
assessed based on an appropriate standard sample analysed
degrees of crystallinity, just as it may contain particles with
according to the corresponding method in conjunction with
varying sizes and shapes. The lower the crystallinity of a
the determination of the instrument baseline noise.
solid, the greater its enthalpy and entropy. The increase in
enthalpy is never totally compensated for by the increase in PROCEDURE
entropy; therefore, the Gibbs free energy, which reflects the Weigh in a suitable vessel an appropriate quantity of the
balance between them, actually increases. Hence, the lower substance to be examined. Close the vessel carefully to avoid
the crystallinity of a material (powder), and consequently any evaporation of solvents and place the vessel in the sample
the greater its amorphous character, the greater its apparent holder. If appropriate, allow the vessel to equilibrate at the
intrinsic solubility and dissolution rate, but the lower its temperature of the measurement before placing it in the
thermodynamic stability. Because of the great relevan ce of measuring position.
these properties, crystallinity is also an important property
and requires measurement by a suitable method. Begin the analysis and record the heat flow, with the time on
the abscissa and the heat flow 011 the ordinate (specify the
In the following chapter, the crystallinity or the content of direction of exothermic or endothermic heat flow).
amorphous parts of a powder are measured by calorimetric
methods such as microcalorimetry or solution calorimetry, DETECTION AND QUANTIFICATION OF AMORPHOUS
although other methods could be used (e.g. see general CONTENT IN POWDERS
chapter 2.9.33. Characterisation of crystalline and partially The amorphous state is metastable with respect to the
crystalline solids by X-ray powder diffraction (XRPD)). crystalline state; recrystallisation may therefore occur.
The measurement of the heat of recrystallisation enables
Many substances are capable of crystallising in more than the amorphous content to be determined by the are a of
one type of crystallattice, which is known as polymorphism. the recrystallisation peak. By relating the output from the
If water or a solvent is incorporated in the crystallattice microcalorimeter for a sample to that obtained from an
the crystals are termed hydrates or solvates. Because of the amorphous standard, it is possible to quantify the amorphous
different crystal packing, and/or molecular conformation content of the sample. The range of amorphous content
and lattice energy, they usually exhibit different physical covered by this method depends on the individual substance
properties. For simplicity, calorimetry measurements for to be tested; in favourable cases limits of detection be!ow 1 per
degree of crystallinity determination discussed hefe as sume cent can be reached.
only one solid crystalline form present in the material of
interest. The theory and experimental technique can be easily Recrystallisation can be initiated by subjecting the sample to
expanded to polymorphic systems with proper consideration higher relative humidity or an atmosphere containing organic
of the enthalpy differences among the polymorphso vapour. The sample is typically placed in an ampoule which
also contains a small test-tube containing an aqueous saturated
METHOD 1 - MICROCALORIMETRY (DETERMINATION salt solution, an organic solvent, or a solvent mixture.
OF AMORPHOUS CONTENT)
The heat of recrystallisation is typically measured using a fixed
Most chemical, physical and biological pro ces ses are associated sample mass placed in a glass or steel vessel. The test -tube
with the exchange of heat. Microcalorimetry is a highly containing a saturated salt solution or an organic solvent is
sensitive technique to monitor and quantify both exothermic chosen to be large enough to allow a full saturation of the
(heat producing) and endothermic (heat absorbing) changes atmosphere aboye the sample. The mass of the sample and the
associated with those processes. The technique allows the nature of the vapour atmosphere aboye the sample are chosen
determination of the rate and extent of chemical reactions, so that recrystallisation occurs in such a way that a distinct
changes of phase or changes of structure. peak is observed, cleariy separated from initial thermal events
Thermal events producing only a fraction of a microwatt caused by introduction of the sample.
can be observed using microcalorimetry. This means that The conditions under which the transition of the amorphous
temperature differences less than 10-6 K must be detectable. phase to a thermodynamically more stable crystalline
Microcalorimetry typically uses the heat flow (heat leakage) state occurs will have a significant impact on the time of
principIe, where the heat produced (or absorbed) in a recrystallisatiol1. In particular, physica! mixtures of purely
thermally defined vessel flows away (or into) in an effort amorphous and crystalline material will behave differently
to re-establish thermal equilibrium with its surroundings. from a partially crystalline material. These effects should be
Exceptional thermal stability with its surrounding has to be considered when developing a method.
achieved either by a heat sink or an electronically regulated
surrounding. A typical response for the recrystallisation of a mainly
amorphous material is shown in Figure 2.2.61.-1. The first
Heat energy from an active sample in the reaction vessel is part of the curve represents several concurrent processes
channelled typically through Pe!tier elements; they act as taking place simultaneously, such as the absorption of water
thermoelectric generators using the Seebeck effect. The heat vapour into the amorphous parts of the powder and the
energy is converted into a voltage signal proportional to the generation of water vapour fram the test -tube. After this
heat flow. initial response there is a large exothermic response caused by
Results are typically presented as a measure of the thermal the recrystallisation of the amorphous material. Also included,
energy produced per unit of time (Watt) as a function of time. but not seen, are the expulsion of excess water from the

General Notices (1) apply to all monographs and other texts 107
2.2.61. Characterisation of crystaHine solids by calorimetry EUROPEAN PHARMACOPOEIA 8.0

recrystallised parts and its condensation. Thus, the are a under The enthalpy of solutiol1 is measured either by an isoperibol
this exothermic recrystallisation response is proportional to (constant perimeter, i.e. jacket) solution calorimeter or by
the heat of recrystallisation. an isothermal (constant temperature) solution calorimeter.
Typically, at least 3 measurements are made with each sample.
2.5 The mean of these values is then calculated. The exact
requirements will depend upon the equipment capability and
degree of accuracy needed.
ISOPERIBOL SOLUTION CALORIMETRY
2
In the isoperibol solution calorimeter, the heat change
during the solution process causes a corresponding change in
temperature of the solvent-solute system (i.e. solution). This
~ 1.5 temperature change is measured by a temperature sensor,
2;
which is wired to an electrical circuit that records an electrical
signal corresponding to the temperature change. Typically,
~
o
this temperature change in an electronic form is measured at
o.. precisely defined time intervals to produce temperature-time
data that are collected, analysed by a computer, and then
plotted. A blank run without addition of the solid solute to
0.5 the solvent normally shows no discernible change in the slope
of the temperature-time plol.
For isoperibol solution calorimeters, response is fairly
rapid, but corrections must be made for any heat losses to
or heat gains from the bath. Therefore, isoperibol solutio11
o 2 3 calorimeters are more advantageous than isothermal solution
Time (Hours) calorimeters when the solution process is relatively fast. For
alJ measurements of enthalpy of solution using isoperibol
solution calorimeters, the choice of solvent is critica!. The
Figure 2.2.61.-1. - Typical microcalorimetric output of power
nature and mass of the solvent and the mass of sample allow
(in 1" W) as a function of time (in hours): amorphous collapse
the total heat change, corresponding to total dissolution of the
peak (I) and crystallisation peak (JI) for mainly amorphous
solid, to proceed to completion within 5 min under vigorous
lactose at 25 oC and 75 per cent relative humidíty
stirring at a constant rotational speed within the range of
METHOD 2 - SOLUTION CALORIMETRY 400-600 r Imin.
(DETERMINATION OF CRYSTALLINITY) The effective heat capacity of the calorimeter ceU and its
Solution calorimetry provides a means of determining contents is determined for every calorimeter runo This
enthalpy of solution (i.e. heat of solution under constant determination is accomplished by electrical heating of the
atmospheric pressure) of a substance. Enthalpy of solution contents of the calorimeter cell. The effective heat capacity is
is defined as the enthalpy of the substance dissolved in the determined according to 1 of 2 protocols: either by making
solution to a defined concentration minus the enthalpy of the 1 determination after ampoule breakage or by making
original substance. The solvent for the dissolution process 1 determination before and a r d determination after ampoule
must be such that the mass of solid dissolves within a time breaJzage and then averaging the 2 results. The accuracy
frame that matches the response time of the calorimeter, as and reliability of the electrical heating are established by
discussed below. The enthalpy of solution is proportional the accuracy and reliability of the aforementioned chemical
to the amount of solid being dissolved. This amount may calibrations.
be defined as 1 mol for molar enthalpy or as 1 g for specific ISOTHERMAL SOLUTION CALORIMETRY
enthalpy. If the substance possesses adequate purity (as
In the isothermal (constant temperature) solution calorimeter,
determined by the degree of accuracy required) and if its
the heat change during the solution process is compensated
molecular mass is known, the molar enthalpy is preferred,
for by an equal but opposite energy change, such that the
otherwise the specific enthalpy must be used. The enthalpy of
temperature ofthe solvent-solute system (i.e. solution)
solution is weakly dependent on both the temperature, which
remains essentially constant. This equal but opposite energy
is usually 25.0 oC, and the final concentration of the dissolved
change is measured and, when its sign is reversed, provides the
solute.
enthalpy of solution. For isothermal calorimeters, response is
1t is usually preferred to express the crystallinity, Pe' of a relatively slow, but the compensation process eliminates the
substance on a percentage scale. This procedure requires effects of heat losses lo or heat gains from the b8oth. Therefore,
2 reference standards, namely a highly crystalline sample isothermal sollltion calorimeters are more advantageous than
assuming 100 per cent crystallinity and having a measured isoperibol solution calorimeters when the solution process
enthalpy of solution of fl.Hg, and an amorphous sample is relatively slow.
assuming O per cent crystallinity and having a measured
enthalpy of solution of fl.H~. From these values and from the SOLUTION CALORIMETER CALIBRATION
measured enthalpy of solution, fl.H;, of the solid under study, To ensure the accuracy of the calorimeter, chemical calibr8otions
the percentage crystallinity of the solid, Pe' may be calculated must be performed on a regular basis. For an endothermic
as follows: solution process, the calibration of the calorimeter is checked
by measuring the heat absorbed during the dissolution of
Pe (%) = 100 (fl.H; - fl.H~) / (fl.H~ - fl.H~) potassium chloride in distilled water at 298.15 K (25.0 OC).
The established enthalpy change in this endothermic process
Clearly, crystallinity expressed 011 a percentage scale depends is 235.5 J/g (17.56 kJlmol). For an exothermic solution
on 3 measured values and the enthalpies of solution may process, the calorimeter is checked by measuring the heat
be replaced by other corresponding physical quantities evolved during the dissolution of 5 g per litre of tromethamine
that depend 011 crystallinity. The value of the percentage [tris(hydroxymethyl)aminomethane, THAM] in a 0.1 mol/L
crystallinity of a sample, however, depends 110t only on the aqueolls hydrochloric acid solution at 298.15 K (25.0 oC). The
nature and method of preparation of the 2 re fe rene e standards, established heat for the aforementioned process is ~ 246.0 J/g
but also 011 the choice of the physical quantity that is measured. (~ 29.80 kJlmol).

108 See the information section on general monographs (cover pages)


EUROPEAN PHARMACOPOEIA 8.0 2.2.65. Voltametric titration

SAMPLE HANDLING Sample size. Usually a few milligrams are used. lf sample sizes
The ehemical and physieal stability of solids may deerease with are variable, the effects of this variation on the appearance
deereasing crystallinity. In particular, solids oflow erystallinity, of the spectrum are validated.
especially amorphous solids, tend to 50rb water vapour from Sample preparation. The test and reference samples must
the atmosphere, leading to crystallisation and a corresponding be comparable in terms of concentration, pH and buffer
gain in crystallinity. For these reasons, anhydrous samples composition. Typically, samples in solution are lyophilised,
whose crystallinity is to be determined must be stored at zero and the dried samples dissolved in deuterated water or a
humidity or below critical humidity levels in sealed chambers buffer in deuterated water. It may be worthwhile to lyophilise
containing a desiccant, preferably containing an indieator of a solution in deuterated water one or more times Cdeuterium
effectiveness. If crystallinity-humidity studies are to be carried exchange') as this reduces the intensity of strong solvent
out, the sample is sto red in a sealed chamber eontaining a signals; volatile process impurities such as ethanol will also
saturated salt solution to provide a defined relative humidity. be lost. Use ofbuffer for the final sample preparation can
reduce aggregation and improve 5pectral reproducibility
by reducing batch-to-batch pH variation. Sorne probes are
07/2011:20264
intolerant to high salt concentrations, but ionic strengths up
to 200 mM sodium chloride are normally tolerated. High salt
concentrations tend to in crease 90° pulse length.
2.2.64. PEPTIDE IDENTIFICATION BY
VERIFICATION OF IDENTITY
NUCLEAR MAGNETIC RESONANCE
Determination of key spectral facfors. Use of a qualitative
SPECTROMETRY approach do es 110t entail stringent requirements 011 spectral
This general ehapter is to be used in conjunction with general parameters (for example, fast pulse repetition rates can
chapter 2.2.33. Nuclear magnetic resonance spectrometry in the be used, as ful! relaxation is not required). The use of
context of peptide identification. The approach to be followed short pulse widths (for example, a 30° pulse) and fast
is qualitative and consists of comparing the nuclear magnetic repetition rates will have no significantly deleterious effect
resonance (NMR) spectrum of a test sample with that of a on spectra, and will allow fas ter acquisition of acceptable
reference sample acquired under identical conditions. signal-to-noise ratios. Variation in the pulse width and
acquisitiol1 time within wide limits will not affect the ability
This general chapter mainly applies to the use of proton NMR to compare spectra. The number of scans collected must give
eH NMR) spectrometry, to confirm the identity of small appropriate signal-to- noise ratios for low intensity resonances
peptide products (up to approximately 15 amino acids). It and therefore a minimum signal-to-noise ratio of 50:1 is
is also applicable when using 13C NMR spectrometry with recommended.
sorne modifications. The scope is restricted to the use of
one-dimensional NMR spectrometry. Identi.fication of characteristic resonances. It is possible
to compare either the complete spectrum or a portion of
GENERAL PRINCIPLES it. Comparison of spectra of relevant samples will highlight
Equipment. Unless otherwise specified, an apparatus with a regions of the spectrum that are distinctive, and comparison
can be constrained to these regions. It is important to define
field strength giving an operating frequency for proton NMR
of at least 300 MHz. resonances from impurities, such as residual solvents, which
may be essentially irrelevant to product quality and which
Spectral acquisition conditions and their optimisation. may vary in intensity between batch es.
After introduction into the magnet, the sample is allowed
Spectral compari,son. See the provisions of general
to come to thermal equilibrium, especially if analysis is
chapter 2.2.33.
carried out at a temperature significantly different from room
temperature: monitoring the lock signal is often a valuable
visual guide to the progress of this process. 01/2013:20265
The spectral width must encompass the complete spectrum
of the peptide, with an empty spectral regio n at each side. 2.2.65. VOLTAMETRIC TITRATION
Typically, a spectral width of 12 ppm or 16 ppm is appropriate.
In voltametric titration the end-point of the titratiol1 is
The following parameters may be optimised to improve
determined by following the variation of the voltage measured
resolution of characteristic peaks: temperature and/or pH
between 2 electro des (either 1 indicator electro de and
primarily, buffer and peptide concentrations. Control of
1 reference electro de or 2 indicator electro des) immersed in
sample temperature is recommended but is not mandatory;
the solution to be examined and maintained at a constant
if not used, the effect of small temperature changes on the
current as a function of the quantity of titrant added.
appearance of the spectrum is validated.
Apparatus. The apparatus comprises an adjustable current
The number of data points collected is such as to define peaks source and a voltmeter; the detection system gene rally
adequately. consists of an indicator electrode (for example, a platinum
Solvent suppression is not recommended but, if used, the electro de, a rotating -dise electrode or a carbon electro de )
inten5ities of peaks close to the solvent resonance may be and a 2nd electro de (for example, a platinum electrode, a
affected and this has to be validated when comparing spectra. rotating-disc electro de or a carbon electrode).
Chemical shift referendng. For samples in aqueous solution, Method. Set the current to the indicator electro de as
sodium 2,2-dimethyl-2-silapentane-5-sulfonate (DSS), sodium prescribed in the monograph and plot a graph of the initial
3-(trimethylsilyl)propionate (TSP) or a deuterated analogue voltage and the values obtained during the titration as
(TSP-d 4 ) are appropriate, and the chemical shift of the methyl functions of the quantity of titrant added. Add the titrant
signals is often set to O ppm. Either the reference material in not fewer than 3 successive quantities equal to a total of
is added at low amounts (l 0-1 00 ppm has been found to be about 80 per cent of the theoretical volume corresponding
appropriate) to the deuterated water used to dissolve the final to the presumed equivalence point. The 3 values must faH
sample, or an easily recognised internal resonance that is on a straight lineo Continue adding the titrant beyond the
consistently present (such as acetate anion) can be used as presumed equivalence point in not fewer than 3 successive
a secondary reference. In this case, a validation spectrum quantities. The values obtained must fal! on another straight
obtained under the same spectral conditions is used to define lineo The point of intersection of the 2 lines represents the
the chemical shift of the secondary standard. end-point of the titration.

General Notices (1) apply to all monographs and other texts 109
2.2.66. Detection and measurement of radioactivity EUROPEAN PHARMACOPOEIA 8.0

Using titration systems for voltametric titration with content must be made with reference to a specified time zone.
2 indicator electrodes, the whole titration curve is recorded The radioactivity at other times may be calculated Írom the
and used to determine the end-point exponential decay equation or from tables.
In general, a correct measurement of radioactivity requires
01/2014:20266 that consideration is given to sorne or all of the following:
Dead-time lo§ses. Due to the finite resolving time (dead
2.2.66. DETECTION AND time) of the detector and its associated electronic equipment,
MEASUREMENT OF RADIOACTIVITY it may be necessary to correct for losses by coincidence. The
resolving time of a counter is the minimum time interval
INTRODUCTION required by the counter to resolve 2 single pulses. Incident
Within the context of the European Pharmacopoeia, the term radiation events at shorter intervals may not be detected or
'radioactivity' is used both to describe the phenomenon of may be detected as a single event with the summed energy.
radio active decay and to express the physical quantity of this These losses are sometimes referred to as 'dead-time losses'.
phenomenon. In the monographs on radiopharmaceutical For a counting system with a fixed dead time T following each
preparations, the detection and measurement of radioactivity count, the tfue count rate, per second, is ca!culated using the
are performed for different purposes: verification of the following expression:
characters, identification, determination of radionuclidic
and radiochemical purity, as well as determination of the
radioactivity in a substance (assay).
Under these assumptions, the measurement can be qualitative,
quantitative or both, depending whether it is directed to the NI the observed count rate, per second;
identification of the radionuclide or the determination of its T the dead time, in seconds.
activity (rate of decay) or both of them.
Radioactive sources can produce various types of emissions, With some equipment this correction is made automatically.
such as alpha particles, electrons, positrons, gamma- and Corrections for los ses by coincidence must be made before
X-rays, according to the radionuclidic composition. the correcHon for background radiation.
Each radionuclide yields characteristic emissions, with Correction fal" decay during measurement. lf the time
specific energies and relative intensities. Such radiations period of an individual measurement, t"" is not negligibly
can be detected as a result of their ionising properties in an short compared with the half-life of the radionuclide, Tu2 '
ionisation chamber but without further characterisation; the decay during this measurement time must be taken into
when they are detected and analysed using a spectrometer, an account. For example, there is a 5 per cent cumulative 10ss of
energy spectrum is obtained. A detailed spectrum analysis is counts due to decay during a counting period that is 15 per
typically used to identify radionuclides present in a sample. cent of the half-life of the radionuclide.
Spectrometry can also be used for quantitative determination
of the radioactivity in sources made of a single radionuclide After having corrected the instrument reading (count rate,
or radionuclide mixtures or of the individual radionuclides ionisation current, etc.) for background signals and, if
present. necessary, for losses due to electronic effects, the instrument
reading corrected to the beginning of the individual
A measurement oí radioactivity is generally performed by
measurement is calculated using the following expression:
counting the number of detected decay events (emissions).
Therefore, the geometry of the sample during the measurement R (At rn )
of radioactivity and the acquisition time strongly influence 1- (c At =)
the result. In general, the measurement geometry must
correspond to a calibrated geometry and the acquisition time
must be long enough to reach sufficient counting statistics. R instrument reading before decay correction, but
already corrected for background signal, etc.;
A measurement of radioactivity can be done in a
stand-alone mode (e.g. using an ionisation chamber or A radionuclide decay constant (1n 2/T,/J;
a spectrometer) or in combination with a separation e base of naturallogarithm;
technique (e.g. radiochromatography) to account for relative
contributions from different radio active chemical species that tln measurement duration.
may be present in a mixture.
Statistics of radioactivity measurement. The results of
MEASUREMENT OF RADIOACTIVITY determinations of radioactivity show variations that derive
A direct determination of the radioactivity of a given sample, mainly from the random nature of nuclear transformations.
in becquerel (Bq), may be carried out if the decay scheme of Counting for any finite time can yield only an estímate of
the radionuclide is known, but in practice many corrections the true rate of nuclear transformations. A sufficient number
are required to obtain accurate results. For this reason, it of counts must be registe red in order to compensate for
is possible to carry out the measurement with the aid of a variations in the number of transÍormations per time. In the
primary standard source or by using measuring instruments case of measurement of radioactivity, the standard deviation
such as an ionisation chamber or a spectrometer calibrated of the recorded counts is the square root of the counts, so at
using suitable standards for the particular radionuclides. least 10000 counts are necessary to obtain a relative standard
deviation of not more than 1 per cent.
A spectrometer is used when measuring the radioactivity of
radionuclides in a mixture, each radionuclide being identified Linearity. The linearity of an instrument is the range of
by its emissions and their characteristic energies. radioactivity for a particular radionuclide over which its
Al! measurements of radioactivity must be corrected for efficiency remains constant.
dead-time losses and by subtracting the background signal The linear range of a radioactivity measurement assembly can
due to radiation in the environment and to spurious signals be determined by repeatedly counting a radioactive sample in
generated in the equipment itself. a fixed geometry as it decays from an activity level that is aboye
The radioactivity of a preparation is stated at a given date. the linear range. After correction for the background signal,
If the half-Me of the radionuclide is less than 70 days, the the naturallogarithm of the count rate data is plotted against
time is also indicated. This statement of the radio active the elapsed time after the first measurement (Figure 2.2.66.-1).

110 See the information section 011 general monographs pages)


EUROPEAN PHARMACOPOEIA 8.0 2.2.66. Detection and measurement of radioactivity

16,- of GBq. It usually comprises a sealed well-type ionisation

-3 ~ -~ -~ ·~- :-0.-1 5-X +-14-.1-8 6- - - - -j


chamber and built-in electronics to convert the detector signal
14
to a measure of radioactivity.
12 The chamber is filled with a gas across which an electrical field
________ is applied. When the gas is ionised by the radiatian emitted by
10 the source, the resulting ionisation current is measured and
related to the radioactivity present in the ionisation chamber.
8
"'a.u The ionisation current is influenced by the applied voltage,
E
6 ~~~~
the energy and the intensity of the radiation and the nature
and pressure of the gas. The instrument settings (calibration
4 factor) may be adjusted to keep a direct relationship between
the ionisation produced by the radiation of a specific
radionuclide and the radioactivity value obtained for each
measurement geometry.
O+-----.~----~------r_----~----~----~
o 20 40 60 80 100 120 As an ionisation chamber measures only the current resulting
Elapsed Time (h) from the overall ionisation produced within the chamber,
it cannot discriminate between the emissions of different
radionuclides.
Figure 2.2.66.-1. ~ Plot showing the measured and extrapolated
count rate (naturallogarithm of counts per second (cps)) from For an accurate measurement of the radioactivity of a specific
a technetium-99m so urce as a function of time, starting with a radionuclide, the measurement must be corrected for the
level of radioactivity above the linear range of the measuring contributions to the ionisation current caused by radionuclidic
equipment impurities present in the preparation.

Linear regression analysis of the central, linear portion of the The activity levels to be measured are limited by saturation
data set yields a slope which is the decay constant Á, which has considerations, the range of the amplifier and the design of the
a characteristic value for each radionuclide: chamber itself. The linearity range of the ionisation chamber
is established as described aboye under Linearity.
lncps=-At+c The ionisation chamber must be shielded to minimise
background signals to an acceptable leve!.
e represents the naturallogarithm of the count rate at t = O of
Method. The sample is positioned inside the well of the
a perfectly linear instrument.
ionisation chamber at a given position, using a holder. After
The resulting regression equation is used to calculate the putting the sample in the ionisation chamber, the activity
theoretical count rate at each time that the actual data were reading is made once the response is stable. Measuring the
recorded. Where the deviation of the measured count rate sample under exactly the same geometrical conditions as
from the theoretical count rate is unacceptably high, the linear the calibration source will yield the most accurate results. If
range of the measuring equipment has been exceeded. necessary, dilute the preparation to be measured to the same
volume as that of the calibration source.
Alternatively, a series of dilutions can be made of a radioactive
solution of known radioactivity concentration. Equal volumes Calibration. The ionisation chamber is calibrated taking into
of each of the dilutions are then counted using standardised account the shape, dimensions, material of the container,
geometry and counter settings. The ratio of the count rate voJume and composition of the solution, the position within
for each sample (after correction for background signals the chamber and the radionuclide being measured. Limits
and decay) to the calculated radioactivity of the respective for uncertainty in calibration can be found in national and
sample in Bq is the counting efficiency. The range over which international regulations.
this ratio is constant is the useable range of the measuring Calibrate the ionisation chamber at least once a year, by
equipment for the radionuclide con cerned. using sources of radionuclides traceable to national or
The limit of detection and the limit of quantification for international standards in the appropriate containers (vial,
equipment and procedures used for radioactivity measurement syringe) with regard to geometry. Establish and implement
must be established before their routine use. subsidiary correction factors to take account of the differing
configurations of the radionuclides to be measured. Perform a
Limit of detection. The limit of detection (LOD) of an linearity check of the instrument's response over the complete
individual procedure is the lowest amount of radioactivity in a range of energies and activities for which the equipment is
sample that can be detected but not necessarily quantified as used.
an exact value. In practical terms this requires an estimate of
the background signal and its standard deviation. The LOD For each setting and before each use (minimum once on
is usually considered to be 3 times the standard deviation of each day of use) perform a constancy check of the ionisation
the background signa!. chamber using standard sources of radionuclides with long
half-lives to verify its calibrated state. A check with a reference
Limit of quantification. The limit of quantification (LOQ) of source, such as caesium-137, must be performed on each
an individual procedure is the lowest amount of radioactivity day of use to verify that the ionisation chamber is still in its
in a sample that can be quantitatively determined with suitable calibrated state.
precisionand accuracy. The LOQ is used particularly for the
determination of impurities and/or degradation products. In MEASUREMENT OF RADIOACTIVITY USING
practical tenns the LOQ is usually considered to be 10 times SOLID-STATE DETECTORS
the standard deviation of the background signa!. Solid-state detectors indude scintillating plastic fluors and
crystals, and semiconductors. Further to their application in
MEASUREMENT OF RADIOACTIVITY USING IONISATION
spectrometry (see section Spectrometry), solid -state detectors
CHAMBERS
can be used for the measurement of radioactivity. In particular,
Apparatus. Ionisation chambers (including dose calibrators) due to their high sensitivity, plastic and crystal scintillation
are the most common equipment for the measurement of detectors are used in counting low levels of radioactivity.
radioactivity in the practice of radiopharmacy. It generally Dead-time losses must be carefully considered with these types
can measure activities from a few ten s of kBq to hundreds of detectors. Semiconductor detector s are used when a higher

General Notices (1) apply to all monographs and other texts 111
2.2.66. Detection and measurement of radioactivity EUROPEAN PHARMACOPOEIA 8.0

energy discrimination is required, for example in mixtures Method.


of radionuclides or when there are potential radionuclidic Half-life. The preparation to be examined is used as such or
impurities with emissions of similar energy. diluted or dried in a capsule after appropriate dilution. The
Apparatus. The equipment consists of a shielded detector radioactive sample is prepared in a manner that will avoid loss
comprising a plasHc or crystal scintillator coupled to a of material during handling. If it is a liquid (solution), it is
photomultiplier, or a semiconductor, which are connected to contained in a closed flask or a sealed tube. If it is a residue
an amplifier and counting electronics. The system may have from drying in a capsule, it is protected by a cover consisting of
an adjustable energy window, used for selecting a counting a sheet of adhesive cellulose acetate or of sorne other material.
regio n of the radionuclide energy spectrum that may be The radioactivity of the sample must be high enough to allow
adjusted by the operator. measurements over a period corresponding to 3 estimated
Instruments have different properties of energy resolution and half-lives but must be, for each measurement, within the
detection efficiency depending on the type of detector and linearity range of the equipment. Correction for dead-time
its volume and geometry. Lower efficiency requires a longer los ses is applied if necessary.
counting time. The same source is measured in the same geometrical
conditions and at intervals usually corresponding to at least
Samples to be measured may be placed in front of the detector
half of the estimated half-life. Each value is tabulated against
or into the well of a well-type detector. Measuring chambers
the time interval from the initial measurement. To avoid
may be enclosed in the detector shielding and single samples
inflllence of decay during measurement, the counting time is
may be introduced using lids or other positioning systems to
the same for al! measurements.
ensure correct measurement geometry.
A graph can be drawn with time as the abscissa and the
A scintillation detector can be used for dynamic radioactivity logarithm of the relative instrument reading (e.g. count rate)
measurement when, for example, the eluate of a liquid as the ordinate. The half-life is calculated from the slope of
chromatograph is directed over or through a detector, see the best linear fit of the measured values against the time
section on Detection and measurement of radioactivity in corresponding to each measurement.
combination with a separation technique.
Approximate half-life. Por this purpose, not fewer than
Method. Ensure that the sample radioactivity gives a counting 3 measurements are made over a period of not less than 114 of
rate in the linearity range of the equipment. The measurement the estimated half-life.
is started after any shielding is in place or the well cover is
replaced and the counting time is selected to reach sufficient The sample to be examined and the instrument to be used
counts for a statistically significant value. comply with the indications given aboye. The data are
processed in the same way as aboye.
Calibration. The detector has to be calibrated by measuring
its efficiency using a source of the radionuclide in question SPECTROMETRY
traceable to national or international standards. Calibration Radionuclides can be identified by their emission spectrum.
in terms of efficiency uses sources such as caesium-137, Each type of emission (i.e. alpha particles, beta particles and
cobalt-60, barium-133 and others covering the desired energy electrons, gamma- and X-rays) requires specific equipment
range. to acquire an emission spectrum. Spectrometers must be
MEASUREMENT OF RADIOACTIVITY USING LIQUID calibrated in order to work properly and the following sections
SCINTILLA TION DETECTORS describe the different equipment and detail the general
Liquid scintillation counting is commonly used for procedures for a reliable measurement.
beta-particle emitting samples, but is also used for GAMMA-RAY SPECTROMETRY
alpha-particle emitting samples. Por the principIes of the
detection of radioactivity using liquid scintillation detectors General principIes. In gamma-ray spectrometry using a
scintillation detector, absorption of gamma- and X-rays results
see under Beta-particle spectrometry below.
in production of light, which is converted into an electrical
Calibration. In order to take into account the loss of
pulse by a photomultiplier. In gamma-ray spectrometry using
counting efficiency due to quenching, the liquid scintillation
a semiconductor detector, absorption of gamma- and X-rays
counter may make use of an external source, typically
results in the immediate production of an electrical pulse.
barium-133 or europium-152, which is brought close to the
sample vial to releas e Compton electrons. The shape of the In both cases the pulse amplitude is proportional to the energy
resulting spectrum is analysed automatically to compute of the absorbed radiation. The most common detectors
a quench-indicating parameter. This parameter can then for gamma- and X-ray spectrometry are thallium-activated
be related to the counting efficiency measuring sources of sodium iodide (NaI(Tl)) scintillation counters and high-purity
known activity at a determined level of quenching agent. The germanium (HPGe) semiconductor detectors.
obtained quench curve allows the determination of the activity A gamma-ray spectrum can be produced by collecting and
of an unknown sample knowing the count rate and the value analysing a sufficient number of pulses.
of the quenching parameter.
Apparatus. A gamma-ray spectrometer usually comprises a
DETERMINATION OF HALF-LIFE shielded measuring chamber where the sample is positioned,
The half-life is a characteristic of the radionuclide that may a detector, an electronic chail1 and a multichannel analyser.
be used for its identification. The half-life is calculated by The shielding of the chamber must be able to reduce the
measuring the variation of radioactivity of a sample to be background signal to a leve! that allows the registration of a
tested as a function of time. Perform the measurements in the correct gamma-ray spectrum.
linearity range of a calibrated instrument.
The measurement chamber has a movable cover or a drawer
Apparatus. Half-life can be measured by using any type of to allow the positioning of the sample. A sample holder
quantitative radioactivity detector provided it is used within may be present to ensure reproducible geometry between
a linearity range throughout the range of activities that are measurements.
present during the measurement and the geometry is not
The duration of measurement is related to the radioactivity
changed during the measurement.
of the target radionuclide and a long period of acquisition
Por preparations containing a radionuclide with a short may be required to achieve the necessary counting statistics.
half-life and when stated in a monograph, determination of Dead-time losses must be carefully cOl1sidered with this type
the approximate half-life contributes to the identification. of detector.

112 See the information section on general monographs (ca ver pages)
EUROPEAN PHARMACOPOEIA 8.0 2.2.66. Detection and measurement of radioactivity

The sensitivity of a NaI(Tl) detector is higher than that of a by using an attenuating material. Introduce the preparation to
germanium detector of the same size. In general, peaks in an be examined in a container into the instrument chamber and
energy spectrum are identified with an uncertainty depending record the spectrum after closing the shielding.
upon the fuI! width of the peak at its half-maximum height Ensure that the container used for quantitative measurements
(FWHM). The energy resolution of a solid-state scintillation is of the same shape, dimensions, volume and material as that
detector is much poorer than that of a semiconductor detector of the calibration standard.
and hence peaks obtained with a semiconductor detector Ensure that the composition of the solution and the position
are much narrower than those obtained with a scintillation of the container in the measuring chamber is the same for
detector. Figure 2.2.66.-2 shows a comparison of the spectra the container for the quantitative measurement as for the
obtained from the same source with the 2 types of detector. calibration standard.
Radionuclide identification. Calibrate the spectrometer in
The different performances of NaI(Tl) and HPGe detectors relation to energy. Determination of the correspondence of the
may limit their use in some spectrometric analyses. energy of the peaks detected from the sample to the energies
prescribed by a monograph is a valid identification test.
For the identification of the radionuclide(s) in a preparation Radionuclidic purity. Calibrate the spectrometer in relation to
and determination of radionuclidic purity, a risk assessment efficiency and energy. Determine the LOQ and resolution of
on the process of radionuclide production must assess the the equipment and ensure that they are in line with the Iimits
potential presence of other radionuclides with photon energies of the radionuclides to be determined. Record the spectrum
in the same range (± 10 per cent) as that of the radionuclide( s) of the preparation.
present in the radiopharmaceutical. Identify the radionuclides present in the preparation to be
examined and determine their radioactivity with the aid of
In case radionuclidic impurities can be present that emit chapter 5.7. Table of physical characteristics of radionuclides.
gamma- or X-rays with an energy in the same range as that of Because the leve! of radionuclidic impurities, expressed as a
the photons emitted by the radionuclide in the preparation, a percentage of the total radioactivity, may increase or deerease
measured peak energy within a maximum interval of ± 2 ke V with time, the measured activity of each impurity must be
or ± 2 per cent (whichever is the larger) with respect to the recalculated to the activity during the period of validity of the
nominal peak energy (see 5.7. Table of physical characteristics preparation. The activities of aH radionuclidic impurities need
of radio n uclides) is sufficient for peak identification. to be summed (taking into aecount the limit of quantification)
and related to the total radioactivity of the preparation.
The sample is placed close to the detector Ol" within a
In the case where such impurities are not expected to be
present, a maximum interval of ± 10 keV or ± 6 per cent well-type detector. AH the events within a pre-set energy
(whichever is the larger) with respect to the nominal peak range are collected and displayed 011 a ratemeter as counts
per second or accumulated over a pre-set period of time. If
energy is acceptable for peak identification.
there is sufficient difference in photon energies emitted by
Method. Ensure that the counting rate of the sample falls the radionuclide(s), a sodium iodide detector can be suitable,
within the linearity range of the equipment. For liquid given its high sensitivity. However, if there is a need to
samples this may be achieved by appropriate dilution; for discriminate emissions of similar energy, a HPGe detector or
solid samples, by increasing the source-to-detector distance or another semiconductor detector is needed.

'100000

10000

'1000
i}<)
e:l
o
(j

'0
~

'D
.Q 100
E
:l
e

10

Erter~ri (f::e \-/)

Figure 2.2.66.-2. - Comparative pulse-height spectra recorded using a thallium-activated sodium iodide scintillator (upper curve)
and a high-purity germanium semiconductor detector (lower curve). The so urce was gamma- and X-ray radiation from the
decay of iodine-131.

General Notices (1) apply lo all monographs and other texts 113
2.2.66. Detection and measurement of radioactivity EUROPEAN PHARMACOPOE1A 8.0

Calibration. Calibration in relation to energy is done by ALPHA -PARTICLE SPECTROMETRY


using the peaks of known sources traceable to national OI For the identification and assay of alpha-particle emitters,
international standards, such as cobalt-57, caesium-137, spectrometry using liquid scintillation is mostly used. The
cobalt-60 and others covering the desired energy range. A principIe is explained in the previous section on beta-particle
calibration in relation to efficiency can be simultaneously spectrometry.
obtained, so that not only the energy spectrum but also the For the identification and determination of radionuclidic
activity of the sample and the radionuclide impurities can purity of alpha-particle emitters, spectrometry using a
be further determined. The calibration of efficiency can be silicon-diode semiconductor detector can be used. Using
performed with a traceable radionuclide source with energy this detector, the absorption of alpha particles resuIts in the
peaks covering the desired range or with the aid of a mixed, immediate production of an electrical pulse. The movement
traceable radionuclide standard with gamma-ray energies of electron-hole pairs created by the interaction of radiation
covering the desired range. induces an electrical charge, which is amplified and measured.
To obtain the efficiency curve, the detector response as a
function of the energy has to be measured using each separate The sample preparation is of crucial importance. After a
sample/detector geometry. For this reason, it is possible chemical separation of the radionuclide of interest, the sample
to carry out the measurement with the aid of a primary is electro-deposited on a stainless steel disk in the form of
standard source. Primary standards may not be available a very thin layer of material to minimise self-absorption.
for radionuclides with a short half-life, e.g. some positron The yield of the whole procedure can be determined
emitters. When measuring, the sample will mostly have to experimental!y by adding a known amount of a tracer, which
be in a container and set at a defined position in relation to will take into account the chemical separation efficiency, the
the detector. The sample/detector geometry is then defined electro-deposition efficiency and the counting efficiency.
by the position of the sample relative to the detector and the For both types of detectors, the pulse amplitude is related
characteristics of the container and sample, e.g. shape, volume to the energy of the absorbed radiation. An alpha-particle
and density. Figure 2.2.66.-3 shows a typical HPGe detector spectrum can be produced by collecting a sufficient number
efficiency curve obtained for a cylindrical container pIaced of pulses.
on top of the detector.
Radiol1uc/ide identification. Determination of the

::::1- -" ---:---~ ---~ ----:~-- ~ --~T-T~l~~~


correspondence of the energy of the peaks detected from the
sample to the energies pl'escribed by a monograph is a valid
o 025 -- -j - - -1- - - - 1- - - - .: - -1- - - - ,. - - - - - ~ ~
-:- - - - - -
identification test.

~ 0020 ---~- __ :____ L ____ : _ _ _ _ : _ _ _ _ ~ _.- - --;----~- --J Calibration. An alpha-particle spectrometer has to be

! --~----:-- -~---¡----j----~---~----j--- ~-- ~


calibrated in relation to energy and efficiency. This is done
0015

w 0010

0005
1 +---- -:- ---------
f --- ~
- - -

-:
I
- - -: - - -
I
~ ~ -j- ---: --
: - -
I
- - - -:- - - - ~ - - - ~ - - - -:- - -
I I I
T - - -
I
J by using the peaks from known sources covering the desired
energy range, such as americium-241 and plutonium-242. Not
al! alpha particles emitted by the source will produce a count
in the system. The probability that an emitted alpha particle
0000 ~ __ ~_~~ ______ ~~~~_,-~'---~'-
will interact with the detector material and produce a count is
o 200 400 600 800 1000 1200 1400 1600 1800 2000 the efficiency of the detector, which depends 011 the geometry.
Energy (keV)

Figure 2.2.66.-3. - Typical HPGe efficiency curve measured DETECTION AND MEASUREMENT OF RADIOACTIVITY
using a dedicated container set on top of the detector IN COMBINATION WITH A SEPARATION TECHNIQUE
BETA-PARTICLE SPECTROMETRY A radioactive preparation may contain the radionuclide
In the case of a beta-particle emitter, a beta-particle in different chemical forms other than the intended one.
spectrometer is necessary to determine the energy distribution Therefore it is necessary to separate the different substances
of the emitted beta particles. 1t is analogous to a gamma-ray containing the radionuclide and determine the percentage of
spectrometer, but frequently uses liquid scintillators to radioactivity due to the radionuclide concerned associated
convert the energy of the beta particles into detectable light, with the stated chemical form and the contribution to the
which can then be analysed. Beta-particle spectrometry is total radioactivity due to the radionuclide concerned coming
mostly achieved by dissolving or suspending the sample in from other substances. For this purpose, instruments for
a liquid scintillation cocktail in transparent or translucent the detection and measurement of l'adioactivity are used in
(glass or pIastic) containers and subsequent counting of the combination with a physico-chemical separation technique.
electrical pulses generated by a photomultiplier from the In principIe, any method of separation may be used.
emitted light. The pulse amplitude is related to the energy Monographs for radiopharmaceutical preparations may
of the absorbed radiation. A beta-particle spectrum can be include the combined use of radioactivity measurement with
produced by collecting a sufficient number of pulses. The paper chromatography (2.2.26), thin-Iayer chromatography
liquid scintillation cocktail is chosen in such a way that (2.2.27), gas chromatography (2.2.28), liquid chromatography
counting errors due to quenching, chemoluminescence, (2.2.29), size-exclusion chromatography (2.2.30) or
phosphorescence, etc., are minimised. Coincidence counting electrophoresis (2.2.31).
with 2 or more photomultipliers is also used to minimise
counts from background radiation, electronics, etc. In al! cases the radioactivity of each analyte is measured after
To differentiate between alpha- and beta-particle emissions, the separation has been achieved using the stated method.
pulse-shape discrimination is commonly used.
Radioactivity measurement may be perfol'med using
Radionuclide identification. Determination of the detectors mounted in series with other detectors in analytical
correspondence of the mean and/or maximum energies in the instruments, such as Iiquid chromatographs, making in-line
energy spectrum from the sample to the energies prescribed detection of analytes, 01' performed off-line, i.e. after the
by a monograph is a valid identification test. analytical separation has been completed, by measuring
Calibration. A common method of energy calibration is to use the radioactivity of eluate fractions obtained after liquid
an unquenched reference sample to determine the maximum chromatographic separation of equal volume or as the
energy of the beta particles emitted by the radionuclide of distribution of radioactivity 011 paper chromatography or
interest. thin-Iayer chromatography supports.

114 See the information section 011 monographs (cover pages)


EUROPEAN PHARMACOPOEIA 8.0 2.2.66. Deíection and measurement of radioactivity

IN-LINE DETECTION AND MEASUREMENT OF OFF-LINE DETECTION AND MEASUREMENT OF


RADIOACTIVITY IN COMBINATION WITH LIQUID RADIOACTIVITY
CHROMATOGRAPHY Liquid chromatography (2.2.29). Provided the retention
Apparatus. Liquid chromatography (see 2.2.29) may be times of the various radiochemical species are reproducibly
used to separate the principal radio active substance of consistent, an alternative method of radioactivity
a radiopharmaceutical preparation from radiochemical quantification is to collect the liquid chromatography effluent
impurities or degradation products. In-line detection is in a series of timed samples (fractions) for off-line analysis
usually obtained by using a scintillation detector connected to for radioactivity content. The radioactivity in the fractions
a ratemeter and recording device. The scintillating material corresponding to the peaks can be expressed as a percentage
of the detector is selected on the basis of the emission to be of the total of the radioactivity in aH fractions, taleing into
detected, e.g. plastic scintillator for beta-particle emissions or account the limit of quantification.
scintillation crystals for gamma- and X-ray radiations. The Method. The sample is applied on the column in the
addition of a liquid scintillation cocktail before the eluate prescribed volume and conditions. Fractions are coHected at
reaches the in-line radioactivity detector may also be used in the end of the chromatographic lineo
the case ofbeta-particle-emitting radionuclides. The volume between the detector used to identify the retention
The simultaneous use of a radioactivity detector and time of the peaks and the collection point is measured and a
other detectors (UV, refractive index, conductimetric, etc.) delay factor is calculated on the basis of the effluent flow rate
and applied to each peak to estimate the time of elution of the
connected in series may be used to identify the substance,
e.g. in relation to the retention time of a known standard, peak at the point of collection. The fractions are collected on
to determine the amount of the substance using a suitable the basis of a fixed time interval or at the time of appearance
estimated from the delay time so that any relevant peak is
reference standard and to measure the radioactivity associated
with such a substance. When different detectors are coupled collected in one or more fractions.
in series, correct the experimentally obtained retention times The radioactivity of each fraction is counted using a calibrated
for the delay in time between the detectors. instrument such as adose calibrator or a scintillation detector,
taking into account the limit of quantification and the linearity.
In liquid chromatography some radiochemical impurities, An elution profile is obtained tabulating the counts per
such as colloidal impurities, may be retained on the column. In fraction against the elution time or volume. The activity of
such cases a separate method is required for the determination fractions belonging to the same peak may be summed and the
of the content of the retained radiochemical impurities . relative percentage calculated to define radiochemical purity.
and the calculation formula for the expression of the total
Thin-layer chromatography (2.2.27) and paper
radiochemical purity takes into account the relative amount of
chromatography (2.2.26). Provided a thin-Iayer
the retained radiochemical impurities.
chromatography or a paper chromatography analytical
One possibility to evaluate such retention problems during method has been validated for the separation of components of
method validation is to evaluate the radioactivity recovery a radio active preparation, the number and relative intensities
from the column by measuring the total radioactivity of the separated spots can be detected and measured using
recovered from the chromatographic equipment with and a radioactivity detector that can relate the radioactivity to a
without the column. specific position on the chromatographic support.
Method. The sample is diluted if necessary and then applied The positions of the spots (peales) may permit chemical
to the column in the prescribed volume and conditions. In identificatiol1 by comparison with solutions of the same
this respect it is important to demonstrate the LOD and LOQ, chemical substances (non-radioactive), using a suitable
and the linearity of the detector throughout the range of detection method.
activities to be measured. Apparatus.
Scanning device. The apparatus gene rally comprises
Flow-through detector. A portion of the tubing where the
a radioactivity detector, such as a position-sensitive
eluate containing the radio active species is flowing is placed
proportional counter or a collimated scintillation detector
in front of or within the detector. Counting efficiency may
be increased using a longer portion of the tube (e.g. making placed at a fixed distance from a scanning platform where the
chromatographic support to be scanned is positioned.
multiple turns in front of or within the detector); however,
this will reduce the ability of the system to separate 2 closely The radioactivity of the sample applied to the chromatography
eluting peaks of radioactivity. support must result in a counting rate in the linearity range
of the equipment and the sample may be diluted if necessary.
When the radiochemical purity test prescribes determination The are a to be scanned is positioned at the reference position
of the total radiochemical impurities or there is a quantitative so that the desired lane is aligned with the detector scanning
determination of an individual impurity, it is important to trip. Adjust the scanning time to allow enough counting time
choose an appropriate threshold setting and appropriate during the runo
conditions for integration of the peak areas. In such tests The detector or the platform may be moved in-plane, along
the disregard limit, i.e. the limit at or below which a peak is the x-axis or the y-axis, so that the entire surface can be
disregarded, is dependent on the method and is related to scanned during a single runo
the limit of detection and limit of quantification. Thus, the
The detector is connected to a suitable counting device, so
threshold setting of the data collection system corresponds to
that the radioactivity revealed can be measured quantitatively
at least half of the disregard limit.
and the count rate related spatially to the surface scanned.
Record the signal of the detectors as a function of time. The radioactivity is automatically reported against the
development distan ce and the profile describes peaks having
Identification of peaks in the radiometric signal
an are a proportional to the number of counts per unit of
(radiochromatogram) is made on the basis of the retention distance.
time of the analytes. The profile from other detectors may be
used for this purpose. Radioactivity counter. In the case where a maximum of only
3 radiochemical components needs to be identified and
Quantification of the different components of chromatogram they are fully separated, the support can be cut into equal
and radiochromatogram profiles is made on the basis of peak strips, each having a size not more than half the length of
areas. Peak areas are usually obtained by direct integration of the support corresponding to the difference between the
the detector signal using commercially available software. retardation factor s of the 2 closest spots. Each single strip

General Notices (1) apply to all monographs and other texts 115
2.2.66. Detection and measurement of radioactivity EUROPEAN PHARMACOPOEIA 8.0

is numbered starting from the origin side and counted Radioactivity may be measured by integration using an
separately. Alternatively, for well-characterised systems the automatic-plotting instrument or a digital counter.
support may be cut into 2 or more unequal portions, folded if
The ratios of the areas under the peaks give the ratíos
necessary to approximately equal geometry before counting.
of the percentages of radioactivity due to the respective
An ionisation chamber or a scintillation counter can be
radiochemical substances.
used for this purpose, provided they are used within the
instrument's linearity range and aboye its LOQ. When the strip s are cut into portions, the ratios of
Autoradiography. This may also be used to acquire an image the quantities of radioactivity measured give the ratio
of the radioactivity distribution on the chromatographic of percentages of radioactivity due to the respective
support. In this case, the response of the system used for radiochemical species.
the acquisition of the image, such as a phosphor imager or a Calibration. It is important to demonstrate the limits of
photographic film, must be shown to be linear with respect detection and quantification, and the linearity of the detector
to the radioactivity in the chromatogram. Otherwise the throughout the range of activities to be measured and in al!
system must be pre-calibrated or exposed at the same time to positions on the support of the chromatographic system.
a series of reference radio active sources, obtained by dilution This may be done by applying samples covering a range of
from a calibrated standard solution, covering the expected activities from 0.1 per cent to 100 per cent of the expected
radioactivity range that may be present on the support. range. Prepare the samples by dilution and apply equal
Method. Deposit the required amount of sample at the origin volumes of each, with drying if necessary. After examining
of the chromatographic support, with drying if necessary the radioactivity pro file using the equipment's standard
to avoid spreading of the spot. Develop the chromatogram settings, the peak areas are integrated for comparison with the
according to the prescribed method. A carrier may be added calculated amount of radioactivity applied to each spot. Verify
when prescribed in a particular monograph. that the response of the detector over the complete length and
In paper and thin-Iayer chromatography, it is preferable not width of the detector path is the same, as the response may
to dilute the preparation to be examined but it is important vary with the detector position.
to avoid depositing such a quantity of radioactivity that The peak-resolving power is influenced by the size of the spot,
counting losses by coincidence (dead-time losses) occur the total radioactivity of the radionuclide and the detector
during measurement of the radioactivity. equipment. It can be checked by applying 5 flL spots separated
After development, the support is dried and the positions by distan ces increasing from 4 mm to 20 mm in 2 mm
of the radio active areas are detected by measurement of increments. The approximate resolution of the detection
radioactivity over the length of the chromatogram, using a system can be determined from the radioactivity pro file as
suitable collimated counter, by autoradiography, or by cutting the distan ce between the 2 spots where the baseline is only
the strip s into portions and counting each portion. just clearly separated.

116 See the information section on general monographs (cover pages)


EUROPEAN PHARMACOPOEIA 8.0

2.3. Identification ...................................................................... 119 2.3.3. Identification of phenothiazines by thin-layer


2.3.1. Identification reactions of ions and functional chromatography...................................................................... 123
groups ...................................................................................... 119 2.3.4. Odour.. ............................................................................. 123
2.3.2. Identification of fatty oils by thin-layer
chromatography...................................................................... 122

General Natices (1) apply ta all managraphs and ather texts 117
EUROPEAN PHARMACOPOEIA 8.0

118 See the information section on general monographs (caver pages)


EUROPEAN PHARMACOPOEIA 8.0 2.3.1. Identification reactions ofions and functional gronps

2.3. IDENTIFICATION AMMONIUM SALTS AND SALTS OF VOLATILE BASES


Dissolve about 20 mg of the substance to be examined in 2 mL
of water R or use 2 mL of the prescribed solution. Add 2 mL of
0112008:20301 dilute sodium hydroxide solution R. On heating, the solution
gives off vapour that can be identified by its odour and by its
alkaline reaction (2.2.4).
2.3.1. IDENTIFICATION REACTIONS
ANTIMONY
OF IONS AND FUNCTIONAL GROUPS
Dissolve with gentle heating about 10 mg of the substance
ACETATES to be examined in a solutiol1 of 0.5 g of sodium potassium
a) Heat the substance to be examined with an equal quantity tartrate R in 10 mL of water R al1d allow to cool: to 2 mL
of oxalic acid R. Acid vapours with the characteristic odour of of this solutiol1, or to 2 mL of the prescribed SOIUtiOI1, add
acetic acid are liberated, showing an acid reaction (2.2.4). sodium sulfide solution R dropwise; al1 orange-red precipitate
is formed which dissolves 011 addition of dilute sodium
b) Dissolve about 30 mg of the substance to be examined in
hydroxide solution R.
3 mL of water R or use 3 mL of the prescribed solution. Add
successively 0.25 mL of lanthanum nitrate solution R, 0.1 mL ARSENIC
of 0.05 M iodine and 0.05 mL of dilute ammonia R2. Heat
Heat 5 mL of the prescribed solution on a water-bath with
carefully to boiling. Within a few minutes a blue precipitate is
an equal volume of hypophosphorous reagent R. A brown
formed or a dark blue colour develops.
precipitate is formed.
ACETYL BARBITURATES, NON-NITRO GEN SUBSTITUTED
In a test-tube about 180 mm long and 18 mm in external Dissolve about 5 mg of the substance to be examined in 3 mL
diameter, place about 15 mg of the substance to be examined, of methanol R, add 0.1 mL of a solution containing 100 giL
or the prescribed quantity, and 0.15 mL of phosphoric acid R. of cobalt nitrate R and 100 giL of calcium chloride R. Mix
Close the tube with a stopper through which passes a small and add, with shaking, 0.1 mL of dilute sodium hydroxide
test-tube about 100 mm long and 10 mm in external diameter solution R. A violet-blue colour and precipitate are formed.
containing water R to act as a condenser. On the outside of
the smaller tube, hang a drop of lanthanum nitrate solution R. BENZOATES
Except for substances hydrolysable only with difficulty, place a) To 1 mL of the prescribed solution add 0.5 mL of ferric
the apparatus in a water-bath for 5 min, then take out the chloride solution R1. A dull-yellow precipitate, soluble in
smaller tube. Remove the drop and mix it with 0.05 mL of ether R, is formed.
0.01 M iodine on a tile. Add at the edge 0.05 mL of dilute
ammonia R2. After 1 min to 2 min, a blue colour develops b) Place 0.2 g of the substance to be examined, treated if
at the junction of the two drops; the colour intensifies and necessary as prescribed, in a test-tube. Moisten with 0.2 mL to
persists for a short time. 0.3 mL of sulfuric acid R. Gently warm the bottom of the tube.
A white sublimate is deposited on the inner wall of the tube.
For substances hydrolysable only with difficulty heat the
mixture slowly to boiling over an open flame and then proceed c) Dissolve 0.5 g of the substance to be examined in 10 mL
as prescribed aboye. of water R or use 10 mL of the prescribed solution. Add
0.5 mL of hydrochloric acid R. The precipitate obtained, after
ALKALOIDS crystallisation from warm water R and drying in vacuo, has a
Dissolve a few milligrams of the substance to be examined, melting point (2.2.14) of 120 oC to 124 oc.
or the prescribed quantity, in 5 mL of water R, add dilute BISMUTH
hydrochloric acid R until an acid reaction occurs (2.2.4), then
a) To 0.5 g of the substance to be examined add 10 mL of dilute
1 mL of potassium iodobismuthate solution R. An orange or
hydrochloric acid R or use 10 mL of the prescribed solution.
orange-red precipitate is formed immediately.
Heat to boiling for 1 mino Cool and filter if necessary. To
ALUMINIUM 1 mL of the solution obtained add 20 mL of water R. A white
or slightly yellow precipitate is formed which on addition of
Dissolve about 15 mg of the substance to be examined in
0.05 mL to 0.1 mL of sodium sulfide solution R turns brown.
2 mL of water R or use 2 mL of the prescribed solution. Add
about 0.5 mL of dilute hydrochloric acid R and about 0.5 mL b) To about 45 mg of the substance to be examined add 10 mL
of thioacetamide reagent R. No precipitate is formed. Add of dilute nitric acid R or use 10 mL of the prescribed solution.
dropwise dilute sodium hydroxide solution R. A gelatinous Boil for 1 mino Allow to cool and filter if necessary. To 5 mL
white precipitate is formed which dissolves on further of the solution obtained add 2 mL of a 100 giL solution of
addition of dilute sodium hydroxide solution R. Gradually thiourea R. A yellowish-orange colour or an orange precipitate
add ammonium chloride solution R. The gelatinous white is formed. Add 4 mL of a 25 giL solution of sodium fluoride R.
precipitate is re-formed. The solution is not decolorised within 30 mino

AMINES, PRIMARY AROMATIC BROMIDES


Acidify the prescribed solution with dilute hydrochloric acid R a) Dissolve in 2 mL of water R a quantity of the substance to
and add 0.2 mL of sodium nitrite solution R. After 1 min to be examined equivalent to about 3 mg of bromide (Br-) or
2 min, add 1 mL of f3-naphthol solution R. An intense orange use 2 mL of the prescribed solution. Acidify with dilute nitric
or red colour and usually a precipitate of the same colour are acid R and add 0.4 mL of si/ver nitrate solution R1. Shake and
produced. allow to stand. A curdled, pale yellow precipitate is formed.
Centrifuge and wash the precipitate with three quantities,
AMMONIUM SALTS each of 1 mL, of water R. Carry out this operation rapidly
To the prescribed solution add 0.2 g of magnesium oxide R. in subdued light disregarding the fact that the supernatant
Pass a current of air through the mixture and direct the gas solution may not become perfectly clear. Suspend the
that escapes just beneath the surface of a mixture of 1 mL of precipitate obtained in 2 mL of water R and add l.5 mL of
0.1 M hydrochloric acid and 0.05 mL of methyl red solution R. ammonia R. The precipitate dissolves with difficulty.
The colour of the indicator changes to yellow. On addition b) Introduce into a small test-tube a quantity of the substance
of 1 mL of a freshly prepared 100 giL solution of sodium to be examined equivalent to about 5 mg of bromide (Br-) or
cobaltinitrite R a yellow precipitate is formed. the prescribed quantity. Add 0.25 mL of water R, about 75 mg

General Notices (1) apply to all monographs and other texts 119
2.3.1. Identification reactions of ions and functional groups EUROPEAN PHARMACOPOEIA 8.0

of lead dioxide R, 0.25 mL of acetic acid R and shake gently. ESTERS


Dry the inside of the upper part of the test-tube with a piece To about 30 mg of the substance to be examined or the
of filter paper and allow to stand for 5 mino Prepare a strip prescribed quantity add 0.5 mL of a 70 giL solution of
of suitable filter paper of appropriate size. Impregnate it by hydroxylamine hydrochloride R in methanol R and 0.5 mL of a
capillarity, by dipping the tip in a drop of decolorised fuchsin 100 giL solution of potassium hydroxide R in ethanol (96 per
solution R and introduce the impregnated part immediately cent) R. Heat to boiling, cool, acidify with dilute hydroehloric
into the tube. Starting from the tip, a violet colour appears acid R and add 0.2 mL of ferric ehloride solution Rl diluted ten
within 10 s that is clearly distinguishable from the red colour times. A bluish-red or red colour is produced.
of fuchsin, which may be visible on a small area at the top of
the impregnated part of the paper strip. IODIDES

CALCIUM a) Dissolve a quantity of the substance to be examined


equivalent to about 4 mg of iodide (I-) in 2 mL of water R or
a) To 0.2 mL of a neutral solution containing a quantity of use 2 mL of the prescribed solution. Acidify with dilute nitrie
the substance to be examined equivalent to about 0.2 mg of aeid R and add 0.4 mL of silver nitrate solution Rl. Shake and
calcium (Ca2 +) per millilitre or to 0.2 mL of the prescribed allow to stand. A curdled, pale-yellow precipitate is formed.
solution add 0.5 mL of a 2 giL solution of glyoxalhydroxyanil R Centrifuge and wash with three quantities, each of 1 mL, of
in ethanol (96 per cent) R, 0.2 mL of dilute sodium hydroxide water R. Carry out this operation rapidly in subdued light
solution R and 0.2 mL of sodium carbonate solution R. Shake disregarding the fact that the supernatant solution may not
with 1 mL to 2 mL of chloroform R and add 1 mL to 2 mL of become perfectly clear. Suspend the precipitate in 2 mL of
water R. The chloroform layer is coloured red. water R and add 1.5 mL of ammonia R. The precipitate does
b) Dissolve about 20 mg of the substance to be examined or not dissolve.
the prescribed quantity in 5 mL of aeetie acid R. Add 0.5 mL b) To 0.2 mL of a solution of the substance to be examined
of potassium ferrocyanide solution R. The solution remains containing about 5 mg of iodide (I-) per millilitre, or to 0.2 mL
clear. Add about 50 mg of ammonium chloride R. A white, of the prescribed solution, add 0.5 mL of dilute sulfuric acid R,
crystalline precipitate is formed. 0.1 mL of potassium dichromate solution R, 2 mL of water R
and 2 mL of ehloroform R. Shake for a few seconds and allow
CARBONATES AND BICARBONATES to stand. The chloroform layer is coloured violet or violet-red.
Introduce into a test-tube 0.1 g ofthe substance to be
examined and suspend in 2 mL of water R or use 2 mL of the IRON
prescribed solution. Add 3 mL of dilute acetic acid R. Close the a) Dissolve a quantity of the substance to be examined
tube immediately using a stopper fitted with a glass tube bent equivalent to about 10 mg of iron (Fe 2+) in 1 mL of water R or
twice at right angles. The solution or the suspension becomes use 1 mL of the prescribed solution. Add 1 mL of potassium
effervescent and gives off a colourless and odourless gas. ferrieyanide solution R. A blue precipitate is formed that does
Heat gently and collect the gas in 5 mL of barium hydroxide not dissolve on addition of 5 mL of dilute hydrochlorie aeid R.
solution R. A white precipitate is formed that dissolves on b) Dissolve a quantity of the substance to be examined
addition of an excess of hydrochlorie acid Rl. equivalent to about 1 mg of iron (Fé+) in 30 mL of water R.
To 3 mL of this solutiol1 or to 3 mL of the prescribed solution,
CHLORIDES add 1 mL of dilute hydrochloric acid R and 1 mL of potassium
a) Dissolve in 2 mL of water R a quantity of the substance to thiocyanate solution R. The solution is coloured red. Take
be examined equivalent to about 2 mg of chloride (Cn or use two portions, each of 1 mL, of the mixture. To one portion
2 mL of the prescribed solution. Acidify with dilute nitric add 5 mL of isoamyl alcohol R or 5 mL of ether R. Shake and
acid R and add 0.4 mL of silver nitrate solution Rl. Shake allow to stand. The organic layer is coloured pink. To the
and allow to stand. A curdled, white precipitate is formed. other portion add 2 mL of mereuric ehloride solution R. The
Centrifuge and wash the precipitate with three quantities, red colour disappears.
each of 1 mL, of water R. Carry out this operation rapidly c) Dissolve a quantity of the substance to be examined
in subdued light, disregarding the fact that the supernatant equivalent to not less than 1 mg of iron (Fe 3+) in 1 mL of
solution may not become perfect1y clear. Suspend the water R or use 1 mL of the prescribed solution. Add 1 mL
precipitate in 2 mL of water R and add 1.5 mL of ammonia R. of potassium ferrocyanide solution R. A blue precipitate is
The precipitate dissolves easily with the possible exception of formed that does not dissolve 011 additiol1 of 5 mL of dilute
a few large particles which dissolve slowly. hydrochloric acid R.
b) Introduce into a test -tube a quantity of the substance to be
examined equivalent to about 15 mg of chloride (CI-) or the LACTATES
prescribed quantity. Add 0.2 g of potassium dichromate R and Dissolve a quantity of the substance to be examined equivalent
1 mL of sulfuric acid R. Place a filter-paper strip impregnated to about 5 mg of lactic acid in 5 mL of water R or use 5 mL of
with 0.1 mL of diphenylcarbazide solution R over the opening the prescribed solution. Add 1 mL of bromine water R and
of the test -tube. The paper turns violet -red. The impregnated 0.5 mL of dilute sulfuric acid R. Heat on a water-bath until
paper must not come into contact with the potassium the colour is discharged, stirring occasionally with a glass
dichromate. rod. Add 4 g of ammonium sulfate R and mix. Add dropwise
and without mixing 0.2 mL of a 100 giL solution of sodium
CITRATES nitroprusside R in dilute sulfuric aeid R. Still without mixing
Dissolve in 5 mL of water R a quantity of the substance to add 1 mL of concentrated ammonia R. Allow to stand for
be examined equivalent to about 50 mg of citric acid or use 30 mino A dark green ring appears at the junction of the two
5 mL of the prescribed solution. Add 0.5 mL of sulfurie acid R liquids.
and 1 mL of potassium permanganate solution R. Warm until
the colour of the permanganate is discharged. Add 0.5 mL LEAD
of a 100 giL solution of sodium nitroprusside R in dilute a) Dissolve 0.1 g of the substance to be examined in 1 mL
sulfurie acid R and 4 g of sulfamie acid R. Make alkaline with of acetie aeid R or use 1 mL of the prescribed solution. Add
concentrated ammonia R, added dropwise until all the sulfamic 2 mL of potassium chromate solution R. A yellow precipitate
acid has dissolved. Addition of an excess of concentrated is formed that dissolves on addition of 2 mL of strong sodium
ammonia R produces a violet colour, turning to violet-blue. hydroxide solution R.

120 See the information section on general monographs (cover pages)


EUROPEAN PHARMACOPOEIA 8.0 2.3.1. Identification reactions ofions and funcHonal groups

b) Dissolve 50 mg of the substance to be examined in 1 mL SILVER


of acetie acid R or use 1 mL of the prescribed solution. Add Dissolve about 10 mg of the substance to be examined in 10 mL
10 mL of water R and 0.2 mL of potassium iodide solution R. of water R or use 10 mL of the prescribed solution. Add 0.3 mL
A yellow precipitate is formed. Heat to boiling for 1 min to of hydrochlorie acid Rl. A curdled, white precipitate is formed
2 mino The precipitate dissolves. Allow to cool. The precipitate that dissolves on addition of 3 mL of dilute ammonia Rl.
is re-formed as glistening, yellow plates.
MAGNESIUM SODIUM
Dissolve about 15 mg of the substance to be examined in a) Dissolve 0.1 g of the substance to be examined in 2 mL of
2 mL of water R or use 2 mL of the prescribed solution. water R or use 2 mL of the prescribed solutioll. Add 2 mL
Add 1 mL of dilute ammonia Rl. A white precipitate is of a 150 giL solution of potassium carbonate R and heat to
formed that dissolves on addition of 1 mL of ammonium boiling. No precipitate is formed. Add 4 mL of potassium
chloride solution R. Add 1 mL of disodium hydrogen phosphate pyroantimonate solution R and heat to boiling. Allow to cool
solution R. A white crystalline precipitate is formed. in iced water and if necessary rub the inside of the test-tube
with a glass rod. A dense white precipitate is formed.
MERCURY
b) Dissolve a quantity of the substance to be examined
a) Place about 0.1 mL of a solution of the substance to be equivalent to about 2 mg of sodium (Na+) in 0.5 mL of
examined on well-scraped copper foil. A dark-grey stain that water R or use 0.5 mL of the prescribed solution. Add 1.5 mL
beco mes shiny on rubbing is formed. Dry the foil and heat in of methoxyphenylacetic reagent R and cool in ice-water for
a test-tube. The spot disappears. 30 mino A voluminous, white, crystalline precipitate is formed.
b) To the prescribed solution add dilute sodium hydroxide Place in water at 20 oC and stir for 5 mino The precipitate
solution R until strongly alkaline (2.2.4). A dense yellow does not disappear. Add 1 mL of dilute ammonia Rl. The
precipitate is formed (mercuric salts). precipitate dissolves completely. Add 1 111L of ammonium
carbonate solution R. No precipitate is formed.
NITRATES
To a mixture of 0.1 mL of nitrobenzene R and 0.2 mL of SULFATES
sulfuric acid R, add a quantity of the powdered substance
a) Dissolve about 45 mg of the substance to be examined in
equivalent to about 1 mg of nitrate (N0 3-) or the prescribed
5 mL of water R or use 5 mL of the prescribed solution. Add
quantity. Allow to stand for 5 mino Cool in iced water and add
1 mL of dilute hydrochloric aeid R and 1 mL of barium ehloride
slowly and with mixing 5 mL of water R, then 5 mL of strong
solution Rl. A white precipitate is formed.
sodium hydroxide solution R. Add 5 mL of acetone R. Shake
and allow to stand. The upper layer is coloured deep violet. b) To the suspension obtained during reaction (a), add 0.1 mL
of 0.05 M iodine. The suspension remains yellow (distinction
PHOSPHATES (ORTHOPHOSPHATES) from sulfites and dithionites), but is decolorised by adding
a) To 5 mL of the prescribed solution, neutralised if necessary, dropwise stannous ehloride solution R (distinction from
add 5 mL of silver nitrate solution Rl. A yellow precipitate is iodates). Boil the mixture. No coloured precipitate is formed
formed whose colour is not changed by boiling and which (distinction from selenates and tungstates),
dissolves on addition of ammonia R.
b) Mix 1 mL of the prescribed solution with 2 mL of TARTRATES
molybdovanadic reagent R. A yellow colour develops. a) Dissolve about 15 mg of the substance to be examined
in 5 mL of water R or use 5 mL of the prescribed solution.
POTASSIUM Add 0.05 mL of a 10 giL solution of ferrous sulfate R and
a) Dissolve 0.1 g of the substance to be examined in 2 mL of 0.05 mL of dilute hydrogen peroxide solution R. A transient
water R or use 2 mL of the prescribed solution. Add 1 mL yellow colour is produced. After the colour has disappeared
of sodium carbonate solution R and heat. No precipitate is add dilute sodium hydroxide solution R dropwise. A violet or
formed. Add to the hot solution 0.05 mL of sodium sulfide purple colour is produced.
solution R. No precipitate is formed. Cool in iced water and
b) To 0.1 mL of a solution of the substance to be examined
add 2 mL of a 150 giL solution of tartaric acid R. Allow to
containing the equivalent of about 15 mg of tartaric acid per
stand. A white crystalline precipitate is formed.
millilitre or to 0.1 mL of the prescribed solution add 0.1 mL of
b) Dissolve about 40 mg of the substance to be examined in a 100 giL solution of potassium bromide R, 0.1 mL of a 20 giL
1 mL of water R or use 1 mL of the prescribed solution. Add solution of resorcinol R and 3 mL of sulfuric aeid R. Heat on a
1 mL of dilute acetic acid R and 1 mL of a freshly prepared water-bath for 5 min to 10 mino A dark-bIue colour develops.
100 giL solution of sodium cobaltinitrite R. A yellow or Allow to cool and pour the solution into water R. The colour
orange-yellow precipitate is formed immediately. changes to red.
SALICYLATES
XANTHINES
a) To 1 mL of the prescribed solution add 0.5 mL of ferric
ehloride solution Rl. A violet colour is produced that persists To a few milligrams of the substance to be examined or the
after the addition of 0.1 mL of acetie acid R. prescribed quantity add 0.1 mL of strong hydrogen peroxide
solution R and 0.3 mL of dilute hydrochloric aeid R. Heat
b) Dissolve 0.5 g of the substance to be examined in 10 mL
of water R or use 10 mL of the prescribed solution. Add to dryness on a water-bath until a yellowish-red residue is
0.5 mL of hydrochlorie acid R. The precipitate obtained, after
obtained. Add 0.1 mL of dilute ammonia R2. The colour of
the residue changes to violet-red.
recrystallisation from hot water R and drying in vacuo, has a
melting point (2.2.14) of 156 oC to 161°C.
ZINC
SILICATES Dissolve 0.1 g of the substance to be examined in 5 111L of
Mix the prescribed quantity of the substance to be examined water R or use 5 mL of the prescribed solution. Add 0.2 mL
in alead or platinum crucible by means of a copper wire with of strong sodium hydroxide solution R. A white precipitate
about 10 111g of sodium fluoride R and a few drops of sulfuric is formed. Add a further 2 mL of strong sodium hydroxide
acid R to give a thin slurry. Cover the crucible with a thin, solution R. The precipitate dissolves. Add 10 mL of ammonium
transparent plate of plastic under which a drop of water R is ehloride solution R. The solution remains clear. Add 0.1 mL
suspended and warm gently. Within a short time a white ring of sodium sulfide solution R. A flocculent white precipitate
is rapidly formed around the drop of water. is formed.

General Notiees (1) apply to all monographs and other texts 121
2.3.2. Identification of fatty oils by TLC EUROPEAN PHARMACOPOEIA 8.0

0112013:20302 Detection: spray with a 100 giL solution of phosphomolybdic


acid R in ethanol (96 per cent) R. Heat the plate at 120 oC for
about 3 min and examine in daylight.
2.3.2. IDENTIFICATION OF The chromatogram obtained typically shows spots comparable
FATTY OILS BY THIN-LAYER to those in Figure 2.3.2.-1.
CHROMATOGRAPHY
METHOD B

METHODA Thin-Iayer chromatography (2.2.27).

Thin-Iayer chromatography (2.2.27). Test solution. Unless otherwise prescribed, dissolve about
20 mg (1 drop) of the fatty oil in 3 mL of methylene chloride R.
Test solution. Unless otherwise prescribed, dissolve about
20 mg (1 drop) of the fatty oil in 3 mL of methylene chloride R. Reference solution. Dissolve about 20 mg (1 drop) of maize
oil R in 3 mL of methylene chloride R.
Reference solution. Dissolve about 20 mg (1 drop) of maize
Plate: a suitable octadecylsilyl silica gel for high performance
oil R in 3 mL of methylene chloride R.
thin-Iayer chromatography as the coating substance.
Plate: a suitable octadecylsilyl silica gel for high performance Mobile phase: methylene chloride R, glacial acetic acid R,
thin-Iayer chromatography as the coating substance. acetone R (20:40:50 V/V/V).
Mobile phase: Application: 1 flL as bands of 8 mm. A suitable automated
apparatus may be used.
- mobile phase A: ether R;
Development: over a path of 7 cm.
- mobile phase B: methylene chloride R, glacial acetic acid R,
acetone R (20:40:50 V/V/V). Drying: in airo

Application: 1 flL. Detection: treat with a 100 giL solution of phosphomolybdic


acid R in ethanol (96 per cent) R. Heat the pI ate at 120 oC for
Development: twice over a path of 0.5 cm with mobile phase A, 3 min and examine in daylight.
then twice over a path of 8 cm with mobile phase B.
The chromatogram obtained typically shows zones comparable
Drying: in airo to those in Figure 2.3.2.-2.

2 3 4 5 7 8 9 10 11 12 13 14 15

1. ara chis oil 4. rapeseed oil 7. linseed oil 10. almond oil 13. evening primrose oil

2. sesame oil 5. soya-bean oil 8. olive oil 11. wheat-germ oíl 14. safflower oil (type 1)

3. maize oil 6. rapeseedoil (erucic acid-free) 9. sunflower oil 12. borage oil 15. safflower oil (type II)

Figure 2.3.2.-1. - Chromatograms for the identification of fatty oils (rnethod A)

122 See the information section on general monographs (caver pages)


EUROPEAN PHARMACOPOEIA 8.0 2.3.4. Odour

1. ara chis oi! 5. soya-bean oil 9. sunflower oi! 13. evening primrose oil

2. sesame oil 6. rapeseed oil (erucic 10. almond oil 14. safflower oil (type I)
acid-free)
3. maize oil 7. linseed oil 11. wheat-germ oil 15. safflower oil (type Ir)

4. rapeseed oil 8. olive oil 12. borage oil 16. hydrogenated ara chis oil

Figure 2.3.2.-2. - Chromatograms for the identification offatty oils (method B)

0112008:20303 Apply separately to the plate 2 flL of each solution and


develop in the dark over a path of 15 cm using a mixture
of 50 mL of light petroleum R and 1 mL of diethylamine R
2.3.3. IDENTIFICATION OF saturated with phenoxyethanol R (i.e. add about 3 mL to
PHENOTHIAZINES BY THIN-LAYER 4 mL of phenoxyethanol R to the aboye mixture of solvents to
give a persistent doudiness on shaking, decant, and use the
CHROMATOGRAPHY supernatant, even if it is doudy). After development place
Examine by thin-layer chromatography (2.2.27) using the plate under ultraviolet light at 365 nm and examine after
kieselguhr G R as the coating substance. Impregnate the a few minutes. The spot in the chromatogram obtained with
plate by placing it in a dosed tank containing the necessary the test solution is similar in position, fluorescence and size
quantity of the impregnation mixture composed of a to the spot in the chromatogram obtained with the reference
solutio~. Spray with a 10 per cent V/V solution of sulfuric
solution containing 10 per cent V/V of phenoxyethanol R
and 50 giL of macrogol 300 R in acetone R so that the plate aCld R 111 alcohol R. The spot in the chromatogram obtained
dlpS about 5 mm beneath the surface of the liquido When the with the test solution is of the same colour as that in the
impregnation mixture has risen at least 17 cm from the lower chromatogram obtained with the reference solution and has
edge of the plate, remove the plate and use immediately for similar stability over a period of at least 20 mino
chromatography. Carry out the chromatography in the same
0112008:20304
direction as the impregnation.
Test solution. Dissolve 20 mg of the substance to be examined 2.3.4.0DOUR
in chloroform R and dilute to 10 mL with the same solvent.
On a watch-glass 6 cm to 8 cm in diameter, spread in a thin
Reference solution. Dissolve 20 mg of the corresponding
layer 0.5 g to 2.0 g of the substance to be examined. After
chemical reference substance (CRS) in chloroform R and dilute
15 min, determine the odour or verify the absence of odour.
to 10 mL with the same solvent.

General Notices (1) apply to all monographs and other texts 123
EUROPEAN PHARMACOPOEIA 8.0

124 See the information section on general monographs (ca ver pages)
EUROPEAN PHARMACOPOEIA 8.0

2.40 Limit tests


2.4. Limit tests ........................................................................... 127 2.4.19. Alkaline impurities in fatty oils .................................. 133
2.4.1. Ammonium .................................................................... 127 2.4.20. Determination of metal catalyst or metal reagent
2.4.2. Arsenic ............................................................................. 127 residues .................................................................................... 133
2.4.3. Calcium ........................................................................... 127 2.4.2l. Foreign oils in fatty oils by thin -layer
2.4.4. Chlorides ......................................................................... 127 chromatography...................................................................... 136
2.4.5. Fluorides .......................................................................... 128 2.4.22. Composition of fatty acids by gas chromatography.. 136
2.4.6. Magnesium ..................................................................... 128 2.4.23. Sterols in fatty oils ........................................................ 139
2.4.7. Magnesium and alkaline-earth metals ........................ 128 2.4.24. Identification and control ofresidual solvents ......... 141
2.4.8. Heavy metals ................................................................... 128 2.4.25. Ethylene oxide and dioxan .......................................... 145
2.4.9. lron .................................................................................. 131 2.4.26. N,N-Dimethylaniline ................................................... 146
2.4.10. Lead in sugars ............................................................... 131 2.4.27. Heavy metals in herbal drugs and fatty oils ............. 147
2.4.11. Phosphates .................................................................... 131 2.4.28. 2-Ethylhexanoie acid ................................................... 148
2.4.12. Potassium ...................................................................... 132 2.4.29. Composition of fatty aeids in oils rieh in omega-3
2.4.13. Sulfates ........................................................................... 132 acids .......................................................................................... 148
2.4.14. Sulfated ash ................................................................... 132 2.4.30. Ethylene glycol and diethylene glycol in ethoxylated
2.4.15. Nickel in polyols ........................................................... 132 substanees ................................................................................ 150
2.4.16. Total ash ........................................................................ 132 2.4.31. Nickel in hydrogenated vegetable oils ....................... 150
2.4.17. Aluminium ................................................................... 132 2.4.32. Total eholesterol in oils rieh in omega-3 acids ......... 151
2.4.18. Free formaldehyde ....................................................... 132

General Notices (1) apply to all monographs and other texts 125
EUROPEAN PHARMACOPOEIA 8.0

126 See the informatíon sectíon on general monographs (caver pages)


EUROPEAN PHARMACOPOEIA 8.0 2.4.4. Chlorides

2.4. LIMIT TESTS After not less than 2 h the stain produced on the mercuric
bromide paper in the test is not more intense than that in the
standard.
0112008:20401
corrected 8.0 METHOD B
Introduce the prescribed quantity of the substance to be
2.4.1. AMMONIUM examined into a test-tube containing 4 mL of hydrochloric
acid R and about 5 mg of potassium iodide R and add 3 mL of
Unless otherwise prescribed, use method A. hypophosphorous reagent R. Heat the mixture on a water-bath
for 15 min, shaking occasionally. Prepare a standard in the
METHODA same manner, using 0.5 mL of arsenic standard solution
Introduce the prescribed solution into a test-tube or dissolve (10 ppm As) R.
the prescribed quantity of the substance to be examined in After heating on the water-bath, any colour in the test solution
14 mL of water R in a test-tube. Make the solution alkaline is not more intense than that in the standard.
if necessary by the addition of dilute sodium hydroxide 5
solution R, dilute to 15 mL with water R and add 0.3 mL of
alkaline potassium tetraiodomercurate solution R. Prepare a
standard by mixing 10 mL of ammonium standard so/ution o
C'?
(1 ppm NH,J R, 5 mL of water R and 0.3 mL of alkaline
potassium tetraiodomercurate solution R. Stopper the
test-tubes.
After 5 min, any yellow colour in the test solution is not more
intense than that in the standard.
METHOD B
In a 25 mL jar fitted with a cap, place the prescribed quantity
of the finely powdered substance to be examined and dissolve
or suspend in 1 mL of water R. Add 0.30 g of heavy magnesium
oxide R. Close immediately after placing a piece of si/ver
manganese paper R 5 mm square, wetted with a few drops
of water R, under the polyethylene cap. Swirl, avoiding
projections of liquid, and allow to stand at 40 oC for 30 mino
If the silver manganese paper shows a grey colour, it is not
more intense than that of a standard prepared at the same
time and in the same manner using the prescribed volume of o
C'?
ammonium standard solution (1 ppm NH,J R, 1 mL of water R
and 0.30 g of heavy magnesium oxide R.

0112008:20402

2.4.2. ARSENIC
Figure 2.4.2.-1. - Apparatus for limit test A for arsenic
METHODA Dimensions in millimetres
The apparatus (see Figure 2.4.2.-1) consists of a 100 mL conical
fiask closed with a ground-glass stopper through which passes 01/2008:20403
a glass tube about 200 mm long and of internal diameter corrected B.O
5 mm. The lower part of the tube is drawn to an internal
diameter of 1.0 mm, and 15 mm from its tip is a lateral orifice 2.4.3. CALCIUM
2 mm to 3 mm in diameter. When the tube is in position in
the stopper, the lateral orifice should be at least 3 mm below All solutions used for this test are prepared with distilled
the lower surface of the stopper. The upper end of the tube water R.
has a perfectly fiat, ground surface at right angles to the axis To 0.2 mL of alcaholic calcium standard solution
of the tube. A second glass tube of the same internal diameter (100 ppm Ca) R add 1 mL of ammonium oxalate solution R.
and 30 mm long, with a similar fiat ground surface, is placed After 1 min add a mixture of 1 mL of dilute acetic acid R and
in contact with the first, and is held in position by two spiral 15 mL of the prescribed solution or of a solution containing
springs. Into the lower tube insert 50 mg to 60 mg of lead the prescribed quantity of the substance to be examined, and
acetate catton R, loosely packed, or a small plug of cotton and shake. Prepare a standard in the same manner using a mixture
a rolled piece of lead acetate paper R weighing 50 mg to 60 mg. of 10 mL of aqueous caleium standard solution (la ppm Ca) R,
Between the fiat surfaces of the tubes place a disc or a small 1 mL of dilute aeetic acid R and 5 mL of distilled water R.
square of mercuric bromide paper R large enough to cover the After 15 min, any opalescence in the test solution is not more
orifice ofthe tube (15 mm x 15 mm). intense than that in the standard.
In the conical fiask dissolve the prescribed quantity of the
substance to be examined in 25 mL of water R, or in the case 01/2008:20404
of a solution adjust the prescribed volume to 25 mL with
water R. Add 15 mL of hydrochloric acid R, 0.1 mL of stannous 2.4.4. CHLORIDES
ehloride solution R and 5 mL of potassium iodide solution R,
allow to stand for 15 min and introduce 5 g of activated zinc R. To 15 mL of the prescribed solution add 1 mL of dilute
Assemble the two parts of the apparatus immediately and nitric acid R and pour the mixture as a single addition into a
immerse the fiask in a bath of water at a temperature such that test-tube containing 1 mL of si/ver nitrate solution R2. Prepare
a uniform evolution of gas is maintained. Prepare a standard a standard in the same manner using 10 mL of ehloride
in the same manner, using 1 mL of arsenic standard solution standard solution (5 ppm Cl) R and 5 mL of water R. Examine
(1 ppm As) R, diluted to 25 mL with water R. the tubes laterally against a black background.

General Notices (1) apply to all monographs and other texts 127
2.4.5. Fluorides EUROPEAN PHARMACOPOE1A 8.0

After standing for 5 min protected from light, any opalescence 0112008:20406
in the test solution is not more intense than that in the
standard. 2.4.6. MAGNESIUM
01/2008:20405 To 10 mL ofthe prescribed solution add 0.1 g of disodium
tetraborate R. Adjust the solution, if necessary, to pH 8.8
to pH 9.2 using dilute hydrochloric acid R or dilute sodium
2.4.5. FLUORIDES hydroxide solution R. Shake with 2 quantities, each of 5 mi,
Introduce into the inner tube of the apparatus (see of a 1 giL solution of hydroxyquinoline R in chloroform R,
Figure 2.4.5.-1) the prescribed quantity of the substance to be for 1 min each time. AlIow to stand. Separate and discard
examined, 0.1 g of acid-washed sand R and 20 mL of a mixture the organic layer. To the aqueous solution add 0.4 mL of
of equal volumes of sulfuric acid R and water R. Heat the butylamine R and 0.1 mL of triethanolamine R. Adjust the
jacket containing tetrachloroethane R maintained at its boiling solution, if necessary, to pH 10.5 to pH 11.5. Add 4 mL
point (146 OC). Heat the steam generator and disti!, collecting of the solution of hydroxyquinoline in chloroform, shake
the distillate in a 100 mL volumetric flask containing 0.3 mL for 1 min, allow to stand and separate. Use the lower layer
of 0.1 M sodium hydroxide and 0.1 mL of phenolphthalein for comparison. Prepare a standard in the same manner
solution R. Maintain a constant volume (20 mL) in the tube using a mixture of 1 mL of magnesium standard solution
during distillation and ensure that the distillate remains (10 ppm Mg) R and 9 mL of water R.
alkaline, adding 0.1 M sodium hydroxide if necessary. Dilute Any colour in the solution obtained from the substance to be
the distillate to 100 mL with water R (test solution). Prepare examined is not more intense than that in the standard.
a standard in the same manner by distillation, using 5 mL
of fluoride standard solution (10 ppm F) R instead of the 0112008:20407
substance to be examined. 1nto two glass-stoppered cylinders
introduce 20 mL of the test solution and 20 mL of the standard
and 5 mL of aminomethylalizarindiacetic acid reagent R.
2.4.7. MAGNESIUM AND
After 20 min, any blue colour in the test solution (originally ALKALINE-EARTH METALS
red) is not more intense than that in the standard. To 200 mL of water R add 0.1 g of hydroxylamine
hydrochloride R, 10 mL of ammonium chloride buffer solution
pH 10.0 R, 1 mL of 0.1 M zinc sulfate and about 15 mg of
mordant black 11 tritura te R. Heat to about 40 oc. Titrate
with 0.01 M sodium edetate until the violet colour changes
to full blue. To the solution add the prescribed quantity of
the substance to be examined dissolved in 100 mL of water R
or use the prescribed solution. lf the colour of the solution
changes to violet, titrate with 0.01 M sodium edetate until the
full blue colour is again obtained.
The volume of 0.01 M sodium edetate used in the second
titration does not exceed the prescribed quantity.

¡<o"'f----180_ 07/2010:20408

2.4.8. HEAVY METALS


The methods described below require the use of thioacetamide
reagent R. As an alternative, sodium sulfide solution R1
(0.1 mL) is usually suitable. Since tests prescribed in
monographs have been developed using thioacetamide
reagent R, if sodium sulfide solution R1 is used instead, it is
necessary to indude also fOI methods A, B and H a monitor
solution, prepared from the quantity of the substance to be
examined prescribed for the test, to which has been added the
volume of lead standard solution prescribed for preparation
of the reference solution. The test is invalid if the monitor
~ solution is not at least as intense as the reference solution.
30 METHODA
o Test solution. 12 mL of the prescribed aqueous solution of
o
<;j'
the substance to be examined.
Reference solution (standard). A mixture of 10 mL of lead
standard solution (1 ppm Pb) R or lead standard solution
(2 ppm Pb) R, as prescribed, and 2 mL of the prescribed
aqueous solution of the substance to be examined.
Blank solution. A mixture of 10 mL of water R and 2 mL of the
prescribed aqueous solution of the substance to be examined.
To each solution, add 2 mL of buffer solution pH 3.5 R. Mix and
add to 1.2 mL of thioacetamide reagent R. Mix immediately.
Examine the solutions after 2 mino
System suitability: the reference solution shows a slight brown
colour compared to the blank solution.
Figure 2.4.5.-1. - Apparatus for limit test for fluorides Result: any brown colour in the test solution is not more
Dimensions in millimetres intense than that in the reference solution.

128 See the information section on general monographs (cover pages)


EUROPEAN PHARMACOPOEIA 8.0 2.4.8. Heavy metals

If the result is difficult to judge, filter the solutions through a - the monitor solution is at least as intense as the reference
suitable membrane filter (nominal pore size 0.45 ¡.tm). Carry solution.
out the filtration slowly and uniformly, applying moderate Result: any brown colour in the test solution is not more
and constant pressure to the piston. Compare the spots on the intense than that in the reference solution.
filters obtained with the different solutions. If the result is difficult to judge, filter the solutions through a
METHOD B suitable membrane filter (nominal pore size 0.45 ¡.tm). Carry
out the filtration slowly and uniformly, applying moderate
Test solution. 12 mL of the prescribed solution of the and constant pressure to the piston. Compare the spots on the
substance to be examined prepared using an organic solvent filters obtained with the different solutiollS.
containing a minimum percentage of water (for example,
dioxan containing 15 per cent of water or acetone containing METHODD
15 per cent ofwater). Test solution. In a silica crucible, mix thoroughly the
Reference solution (standard). A mixture of 10 mL oflead prescribed quantity of the substance to be examined with
standard solution (1 or 2 ppm Pb), as prescribed, and 2 mL of 0.5 g of magnesium oxide Rl. Ignite to dull redness until a
the prescribed solution of the substance to be examined in homogeneous white or greyish-white mass is obtained. If
an organic solvent. Prepare the lead standard solution (1 or after 30 min of ignition the mixture remains coloured, allow
2 ppm Pb) by dilution of lead standard solution (100 ppm Pb) R to cool, mix using a fine glass rod and repeat the ignition. If
with the solvent used for the substance to be examined. necessary repeat the operation. Heat at 800 oC for about 1 h.
Blank solution. A mixture of 10 mL of the solvent used for the
Take up the residue in 2 quantities, each of 5 mL, of a mixture
substance to be examined and 2 mL of the prescribed solution of equal volumes of hydroehloric acid Rl and water R. Add
of the substance to be examined in an organic solvento 0.1 mL of phenolphthalein solution R and then coneentrated
ammonia R until a pink colour is obtained. Cool, add glacial
To each solution, add 2 mL of buffer solution pH 3.5 R. Mix and aeetie aeid R until the solution is decolorised and add 0.5 mL
add to 1.2 mL of thioacetamide reagent R. Mix immediately. in excess. FiIter if necessary and wash the filter. Dilute to
Examine the solutions after 2 mino 20 mL with water R.
System suitability: the reference solution shows a slight brown Referenee solution (standard). Prepare as described for the test
colour compared to the blank solution. solution using the prescribed volume of lead standard solution
Result: any brown colour in the test solution is 110t more (10 ppm Pb) R instead of the substance to be examined and
i11tense than that in the reference solution. drying in an oven at 100-105 oc. To 10 mL of the solution
If the result is difficult to judge, filter the solutions through a obtained add 2 mL of the test solution.
suitable membrane filter (nominal pore size 0.45 ¡.tm). Carry Monitor solution. Prepare as described for the test solution,
out the filtration slowly and uniformly, applying moderate adding to the substance to be examined the volume of lead
and constant pressure to the piston. Compare the spots 011 the standard solution (10 ppm Pb) R prescribed for preparation of
filters obtained with the different solutions. the reference solution and drying in an oven at 100-105 oc. To
10 mL of the solution obtained add 2 mL of the test solution.
METHOD C Blank solution. A mixture of 10 mL of water R and 2 mL of
Test solution. Place the prescribed quantity (not more than 2 g) the test solution.
of the substance to be examined in a silica crucible with 4 mL To 12 mL of each solution, add 2 mL of buffer solution
of a 250 giL solution of magnesium sulfate R in dilute sulfuric pH 3.5 R. Mix and add to 1.2 mL of thioacetamide reagent R.
aeid R. Mix using a fine glass rod. Heat cautiously. If the Mix immediately. Examine the solutions after 2 mino
mixture is liquid, evaporate gentIy to dryness on a water-bath. System suitability:
Progressively heat to ignition and continue heating until an
- the reference solution shows a slight brown colour
almost white or at most greyish residue is obtained. Carry out
the ignition at a temperature not exceeding 800 oc. Allow to compared to the blank solution,
cool. Moisten the residue with a few drops of dilute sulfuric - the monitor solution is at least as intense as the reference
acid R. Evaporate, ignite again and allow to cool. The total solution.
period of ignition must not exceed 2 h. Take up the residue Result: any brown colour in the test solution is not more
in 2 quantities, each of 5 mL, of dilute hydrochloric acid R. intense than that in the reference solution.
Add 0.1 mL of phenolphthalein solution R, then concentrated lf the result is difficult to judge, filter the solutions through a
ammonia R until a pink colour is obtained. Cool, add glacial suitable membrane filter (nominal pore size 0.45 ¡.tm). Carry
acetie acid R until the solution is decolorised and add 0.5 mL out the filtration slowly and uniformly, applying moderate
in excess. Filter if necessary and wash the filter. Dilute to and constant pressure to the piston. Compare the spots on the
20 mL with water R. filters obtained with the different solutions.
Reference solution (standard). Prepare as described for the test
solution, using the prescribed volume of lead standard solution METHOD E
(10 ppm Pb) R instead of the substance to be examined. To Test solution. Dissolve the prescribed quantity of the substance
10 mL of the solution obtained add 2 mL of the test solution. to be examined in 30 mL of water R or the prescribed volume.
Monitor solution. Prepare as described for the test solution, Reference solution (standard). Unless otherwise prescribed,
adding to the substance to be examined the volume of lead dilute the prescribed volume of lead standard solution
standard solution (10 ppm Pb) R prescribed for preparation (1 ppm Pb) R to the same volume as the test solution.
of the reference solution. To 10 mL of the solution obtained Prepare the filtration apparatus by adapting the barrel of a
add 2 mL of the test solution. 50 mL syringe without its piston to a support containing, on
Blank solution. A mixture of 10 mL of water R and 2 mL of the plate, a membrane filter (nominal pore size 3 ¡.tm) and
the test solution. aboye it a prefilter (Figure 2.4.8.-l).
To 12 mL of each solution, add 2 mL of buffer solution Transfer the test solution into the syringe barrel, put the piston
pH 3.5 R. Mix and add to 1.2 mL of thioaeetamide reagent R. in place and then apply an even pressure on it until the whole
Mix immediately. Examine the solutions after 2 mino of the liquid has been filtered. In opening the support and
removing the prefilter, check that the membrane filter remains
System suitability: uncontaminated with impurities. If this is not the case replace
- the reference solution shows a slight brown colour it with another membrane filter and repeat the operation-
compared to the blank solution, under the same conditions.

General Notiees (1) apply to all monographs and other texts 129
2.4.8. Heavy metals EUROPEAN PHARMACOPOEIA 8.0

Membrane
Prefilter filter
\0';·,' ',-" :0';',' '.:' :0'; ·1
1:.:···.·:-.··,:-:-1,-·:-.··,:-:-·1
Membrane Prefilter
filter

00 Prefiltration Filtration of the


00 of the solution solution after
..- Joint
(Method E) addition of the
reagents
(Method E)

13
15.7
17.4
Figure 2.4.8.-1. - Apparatus for the test for heavy metals
Dímensions ín míllimetres
To the prefiltrate or to the prescribed volume of the prefiltrate and rinse into a 50 mL colour comparison tube, ensuring that
add 2 mL of buffer so/ution pH 3.5 R. Mix and add to 1.2 mL the total volume does not exceed 25 mL. Adjust the solution to
of thioacetamíde reagent R. Mix immediately and allow to pH 3.0-4.0, using short range pH indicator paper as external
stand for 10 min and again filter as described aboye, but indicator, with concentrated ammonía Rl (dílute ammonia Rl
inverting the order of the filters, the liquid passing first may be used, if desired, as the specified range is approached),
through the membrane filter before passing through the dilute with water R to 40 mL and mix. Add 2 mL of buffer
prefilter (Figure 2.4.8.-1). The filtration must be carried out solution pH 3.5 R. Mix and add to 1.2 mL of thioaeetamide
slowly and uniformly by applying moderate and constant reagent R. Mix immediately, Dilute to 50 mL with water R
pressure to the piston of the syringe. After complete filtration, and mix.
open the support, remove the membrane filter, and dry using Referenee solution (standard). Prepare at the same time and
filter paper. in the same manner as the test solution, using the prescribed
In parallel, treat the reference solution in the same manner as volume of ¡ead standard solutíon (lO ppm Pb) R.
the test solution. Monitor solution. Prepare as described for the test solution,
Result: the colour of the spot obtained with the test solution adding to the substance to be examined the volume of lead
is not more intense than that obtained with the rderence standard so/utíon (10 ppm Pb) R prescribed for the preparation
solution. of the reference solution.
METHOD F Blank solution. Prepare as described for the test solution,
omitting the substance to be examined.
Test solution. Place the prescribed quantity or volume of the
substance to be examined in a clean, dry, 100 mL long-necked Examine the solutions vertically against a white background
combustion flask (a 300 mL flask may be used if the reaction after 2 mino
foams excessively). Clamp the flask at an angle of 45°. If the System suitabílity:
substance to be examined is a solid, add a sufficient volume - the reference solution shows a brown colour compared to
of a mixture of 8 mL of sulfuríc acíd R and 10 mL of nitríc the blank solution,
acid R to moisten the substance thoroughly; if the substance
- the monitor solution is at least as intense as the reference
to be examined is a liquid, add a few millilitres of a mixture
solution.
of 8 mL of sulfuríc acíd R and 10 mL of nítríc acid R. Warm
gently until the reaction commences, allow the reaction to Result: any brown colour in the test solution is not more
subside and add additional portions of the same acid mixture, intense than that in the reference solution.
heating after each addition, until a total of 18 mL of the acid If the result is difficult to judge, filter the solutions through a
mixture has been added. Increase the amount of heat and boil suitable membrane filter (nominal pore size 0.45 11m). Carry
gently until the solution darkens. Coo!, add 2 mL of nitric out the filtration slowly and uniformly, applying moderate
acid R and heat again until the solution darkens. Continue and constant pressure to the piston. Compare the spots on the
the heating, followed by the addition of nitric acíd R until filters obtained with the different solutions.
no further darkening occurs, then heat strongly until dense,
white fumes are produced. Cool, cautiously add 5 mL of METHOD G
water R, boil gently until dense, white fumes are produced CAUTION: when using high-pressure dígestion vessels the
and continue heating to reduce to 2-3 mL. Cool, cautiously safety precautions and operatíng ínstruetíons given by the
add 5 mL of water R and examine the colour of the solution. manufacturer must be followed. The digestion eye/es have to
If the colour is yellow, cautiously add 1 mL of strong hydrogen be elaborated depending on the type of mícrowave oven to
peroxide solution R and again evaporate until dense, white be used (jor example, energy-controlled microwave ovens,
fumes are produced and reduce to a volume of 2-3 mL. If the temperature-controlled míerowave ovens or high-pressure
solution is still yellow in colour, repeat the addition of 5 mL of ovens). The cye/e must conform to the manufacturers
water R and 1 mL of strong hydrogen peroxide solution R until instructíons. The dígestion cye/e is suitable if a e/ear solutíon is
the solution is colourless. Cool, dilute cautiously with water R obtained.

130 See the information section on general monographs (cover pages)


EUROPEAN PHARMACOPOEIA 8.0 2.4.11. Phosphates

Test solution. Place the prescribed amount of the substance to solutions through a suitable membrane filter (nominal pore
be examined (not more than 0.5 g) in a suitable, clean beaker. size 0.45 11m). Compare the spots on the filters obtained with
Add successively 2.7 mL of sulfuric acid R, 3.3 mL of nitric the different solutions.
acid R and 2.0 mL of strong hydrogen peroxide solutian R System suitability: the spot obtained with the reference
using a magnetic stirrer. Allow the substance to react with a solution shows a brownish-black colour compared to the spot
reagent before adding the next one. Transfer the mixture to obtained with the blank solution.
a dry high-pressure-resistant digestion vessel (fluoropolymer
or quartz glass). Result: the brownish-black colour of the spot obtained with
the test solution is not more intense than that of the spot
Reference solutian (standard). Prepare as described for the test obtained with the reference solution.
solution, using the prescribed volume of lead standard solutian
(10 ppm Pb) R instead of the substance to be examined.
Monitor salution. Prepare as prescribed for the test solution,
adding to the substance to be examined the volume of lead 01/2008:20409
standard salution (10 ppm Pb) R prescribed for the preparation
of the reference solution.
2.4.9. IRON
Blank solutian. Prepare as described for the test solution,
omitting the substance to be examined. Dissolve the prescribed quantity of the substance to be
Close the vessels and place in a laboratory microwave oven. examined in water R and dilute to 10 mL with the same
Digest using a sequence of 2 separate suitable programmes. solvent or use 10 mL of the prescribed solution. Add 2 mL of
Design the programmes in several steps in order to control a 200 giL solution of citric acid R and 0.1 mL of thiaglycollic
the reaction, monitoring pressure, temperature or energy acid R. Mix, make alkaline with ammania R and dilute to
depending on the type of microwave oven available. After the 20 mL with water R. Prepare a standard in the same manner,
first programme allow the digestion vessels to cool before using 10 mL of iran standard solutian (1 ppm Fe) R.
opening. Add to each vessel 2.0 mL of strong hydrogen After 5 min, any pink colour in the test solution is not more
peroxide solutian R and digest using the second programme. intense than that in the standard.
After the second programme allow the digestion vessels to
cool before opening. lf necessary to obtain a clear solution,
repeat the addition of strong hydrogen peroxide solutian R and
the second digestion programme. 01/2008:20410
Coo!, dilute cautiously with water R and rinse into a flask,
ensuring that the total volume do es not exceed 25 mL. 2.4.10. LEAD IN SUGARS
Using short-range pH indicator paper as external indicator,
adjust the solutions to pH 3.0-4.0 with concentrated Determine the lead by atomic absorption spectrometry
ammania Rl (dilute ammonia Rl may be used as the specified (2.2.23, Method II).
range is approached). To avoid heating of the solutions use an Test salutian. Dissolve 20.0 g of the substance to be examined
ice-bath and a magnetic stirrer. Dilute to 40 mL with water R in a mixture of equal volumes of dilute acetic acid R and
and mix. Add 2 mL of buffer salution pH 3.5 R. Mix and add water R and dilute to 100.0 mL with the same mixture of
to 1.2 mL of thiaacetamide reagent R. Mix immediately. Dilute solvents. Add 2.0 mL of a clear 10 giL solution of ammonium
to 50 mL with water R, mix and allow to stand for 2 mino pyrralidinedithiacarbamate R and 10.0 mL of methyl isobutyl
Filter the solutions through a suitable membrane filter ketane R and then shake for 30 s protected from bright light.
(nominal pore size 0.45 11m). Carry out the filtration slowly AlIow the layers to separate and use the methyl isobutyl
and uniformly, applying moderate and constant pressure to ketone layer.
the piston. Compare the spots on the filters obtained with the Reference solutions. Prepare 3 reference solutions in the same
different solutions. manner as the test solution but adding 0.5 mL, 1.0 mL and
System suitability: 1.5 mL respectively of lead standard salution (10 ppm Pb) R in
addition to the 20.0 g of the substance to be examined.
- the spot obtained with the reference solution shows a
brown colour compared to the spot obtained with the blank Set the zero of the instrument using methyl isobutyl ketane R
solution, treated as described for the test solution without the substance
to be examined. Measure the absorbance at 283.3 nm using
- the spot obtained with the monitor solution is at least as
alead hollow-cathode lamp as source of radiation and an
intense as the spot obtained with the reference solution.
air-acetylene flameo
Result: the brown colour of the spot obtained with the test
The substance to be examined contains not more than 0.5 ppm
solution is not more intense than that of the spot obtained
of lead, unless otherwise prescribed.
with the reference solution.

METHOD H
Test solution. Dissolve the prescribed quantity of the substance 0112008:20411
to be examined in 20 mL of the solvent or solvent mixture
prescribed.
Reference salution. Dilute the prescribed volume of lead
2.4.11. PHOSPHATES
standard solutian (10 ppm Pb) R to 20 mL with the solvent or To 100 mL of the solution prepared and, if necessary,
solvent mixture prescribed. neutralised as prescribed add 4 mL of sulfomalybdic
Blank salutian. 20 mL of the solvent or solvent mixture reagent R3. Shake and add 0.1 mL of stannous chlaride
prescribed. salutian Rl. Prepare a standard in the same manner using
To each solution, add 2 mL of buffer salutian pH 3.5 R. Mix. 2 mL of phosphate standard solutian (5 ppm PO,) R and 98 mL
(In same cases precipitation accurs, in which case the specific of water R. After 10 min, compare the colours using 20 mL
managraph would describe re-dissolutian in a defined valume of each solution.
af a given salvent.) Add to 1.2 mL of thioacetamide reagent R. Any colour in the test solution is not more intense than that
Mix immediately and allow to stand for 2 mino Filter the in the standard.

General Natices (1) apply to all manographs and ather texts 131
2.4.12. Potassium EUROPEAN PHARMACOPOEIA 8.0

0112008:20412 Reference solutions. Prepare 3 reference solutions in the same


manner as the test solution but adding 0.5 mL, 1.0 mL and
2.4.12, POTASSIUM 1.5 mL respectively of nickel standard solution (lO ppm Ni) R
in addition to the 20.0 g of the substance to be examined.
To 10 mL of the prescribed solution add 2 mL of a freshly Set the zero of the instrument using methyl isobutyl ketone R
prepared 10 giL solution of sodium tetraphenylborate R. treated as described for preparation of the test solution
Prepare a standard in the same manner using a mixture of omitting the substance to be examined. Measure the
5 mL of potassium standard solution (20 ppm K) R and 5 mL absorbance at 232.0 nm using a nickel hollow-cathode lamp as
of water R. source of radiation and an air-acetylene flameo
After 5 min, any opalescence in the test solution is not more The substance to be examined contains not more than 1 ppm
intense than that in the standard. of nickel, unless otherwise prescribed.
01/2008:20413
corrected 8.0 0112008:20416

2.4,13. SULFATES 2.4.16. TOTAL ASH


All solutions used for this test must be prepared with distilled Heat a silica or p1atinum crucible to redness for 30 min, allow
water R. to coo1 in a desiccator and weigh. Unless otherwise prescribed,
evenly distribute 1.00 g of the substance or the powdered
Add 3 mL of a 250 giL solution of barium chloride R to 4.5 mL herbal drug to be examined in the crucible. Dry at 100 oC to
of sulfate standard solution (10 ppm soJ R1. Shake and allow 105 oC for 1 h and ignite to constant mas s in a muffle furnace
to stand for 1 mino To 2.5 mL ofthis suspension add 15 mL of at 600 oC ± 25 oC, allowing the crucible to cool in a desiccator
the prescribed solution and 0.5 mL of acetic acid R. Prepare a after each ignition. Flames should not be produced at any time
standard in the same manner using 15 mL of sulfate standard during the procedure. lf after prolonged ignition the ash still
solution (10 ppm soJ R instead of the prescribed solution. contains black particles, take up with hot water, filter through
After 5 min, any opalescence in the test solution is not more an ashless filter paper and ignite the residue and the filter
intense than that in the standard. paper. Combine the filtrate with the ash, carefully evaporate
to dryness and ignite to constant mass.
0412010:20414
01/2008:20417
2.4.14. SULFATED ASH(1)
19nite a suitable crucible (for example, silica, platinum, 2.4.17. ALUMINIUM
porcelain or quartz) at 600 ± 50 oC for 30 min, allow to cool
Place the prescribed solution in a separating funnel and
in a desiccator over silica gel or other suitable desiccant and
shake with 2 quantities, each of 20 mL, and then with one
weigh. Place the prescribed amount of the substance to be
10 mL quantity of a 5 giL solution of hydroxyquinoline R in
examined in the crucible and weigh. Moisten the substance to
chloroform R. Dilute the combined chloroform solutions to
be examined with a small amount of sulfuric acid R (usually
50.0 mL with chloroform R (test solution).
1 mL) and heat gently at as low a temperature as practicable
until the sample is thoroughly charred. After cooling, moisten Prepare a standard in the same manner using the prescribed
the residue with a small amount of sulfuric acid R (usually reference solution.
1 mL), heat gently until white fumes are no longer evolved Prepare a blank in the same manner using the prescribed
and ignite at 600 ± 50 oC until the residue is complete1y blank solution.
incinerated. Ensure that flames are not produced at any Measure the intensity of the fluorescence (2.2.21) of the test
time during the procedure. Allow the crucible to co01 in a solution (JI)' of the standard (J2) and of the blank (J3) using
desiccator over silica gel or other suitable desiccant, weigh it an excitant beam at 392 nm and a secondary filter with a
again and calculate the percentage of residue. transmission band centred on 518 nm or a monochromator
lf the amount of the residue so obtained exceeds the prescribed set to transmit at this wave1ength.
limit, repeat the moistening with sulfuric acid R and ignition, The fluorescence (Il- 13) of the test solution is not greater than
as previously, for 30 min periods until 2 consecutive weighings that of the standard (I2- 1J
do not differ by more than 0.5 mg or until the percentage of
residue complies with the prescribed limit. 0112008:20418
The amount of substance used for the test (usually 1-2 g) is
chosen so that at the prescribed limit the mass of the residue 2.4.18. FREE FORMALDEHYDE
(usually about 1 mg) can be measured with sufficient accuracy.
Use method A, unless otherwise prescribed. Method B is
0112008:20415 suitable for vaccines where sodium metabisulfite has been
(orrected 7.0 used to neutralise excess formaldehyde.
METHODA
2.4.15. NICKEL IN POLYOLS For vaccines for human use, prepare a 1 in 10 dilution of the
Determine the nickel by atomic absorption spectrometry vaccine to be examined. For bacterial toxoids for veterinary
(2.2.23, Method II). use, prepare a 1 in 25 dilution of the vaccine to be examined.
Test solutíon. Dissolve 20.0 g of the substance to be examined To 1 mL of the di1ution, add 4 mL of water R and 5 mL of
in a mixture of equal volumes of dilute acetic acid R and acetylacetone reagent R1. Place the tube in a water-bath at
water R and. dilute to 100.0 mL with the same mixture of 40 oC for 40 mino Examine the tubes down their vertical axes.
solvents. Add 2.0 mL of a saturated solution of ammoniurn The solution is not more intensely co1oured than a standard,
pyrrolidinedithiocarbamate R (about 10 giL) and 10.0 mL of prepared at the same time and in the same manner, using
methyl isobutyl ketone R and then shake for 30 s protected 1 mL of a dilution of formaldehyde solution R containing 20 flg
from bright light. Allow the layers to separate and use the offormaldehyde (CH 2 0) per millilitre, instead of the dilution
methyl isobuty1 ketone ¡ayer. of the vaccine to be examined.

(I) This chapter has undergone phannacopoeial harmonisation. See chapter 5.8. Pharmacopoeial harmonisation.

132 See the information section on general monographs (ca ver pages)
EUROPEAN PHARMACOPOEIA 8.0 2.4.20. Determination of metal catalyst or metal reagent residues

METHOD B not possible to describe all suitable sample preparation and


Test solution. Prepare a 1 in 200 dilution of the vaccine to be measurement methods. Therefore, any method that fulfils the
examined with water R. If the vaccine is an emulsion, prepare requirements described in this chapter may be used.
an equivalent dilution using the aqueous phase separated The results of the analysis are acceptable only if the system
by a suitable procedure (see below). If one of the methods suitability has been demonstrated by a suitable test. Before
described below is used for separation of the aqueous phase, a the initial use of a method, the analyst must ensure that the
1 in 20 dilution of the latter is used. method is appropriate for the samples and instruments used.
Reference solutions. Prepare solutions eontaining 0.25 giL, This is accomplished by applying a validation procedure
0.50 giL, 1.00 giL and 2.00 giL of CHp by dilution of to methods 110t described in the specific monograph or
formaldehyde solution R with water R. Prepare a 1 in 200 by a system suitability test for methods described in the
dilution of each solution with water R. monograph. Decision trees for the choice of the sample
To 0.5 mL of the test solution and of each of the reference preparation and the measurement procedures are presented in
solutions in test-tubes, add 5.0 mL of a freshly prepared 0.5 giL Figures 2.4.20.-1 and 2.4.20.-2.
solution of methylbenzothiazolone hydrazone hydrochloride R.
Close the tubes, shake and allow to stand for 60 mino Add 1 mL PROCEDURES
of ferric chloride-sulfamic acid reagent R and allow to stand for
15 mino Measure the absorbance (2.2.25) of the solutions at As a reference procedure is not provided for each metal, matrix
628 nm. Calculate the content of formaldehyde in the vaccine and concentration, the choice of procedure according to
to be examined from the calibration curve established using Figures 2.4.20.-1 and 2.4.20.-2, including sample preparation,
the reference solutions. The test is invalid if the correlation detection technique and instrument parameters, is the
coefficient (r) of the calibration curve is less than 0.97. responsibility of the user.
Emulsions. If the vaccine to be examined is an emulsion, Use the flow chart in Figure 2.4.20.-1 to define the sample
the aqueous phase is separated using a suitable procedure preparation method and the flow chart in Figure 2.4.20.-2 to
and used for preparation of the test solution. The following define the measurement method. The sample preparation
proeedures have been found suitable. method should yield a sufficient quantity of sample to allow
quantification of each metal at the specified limit stated in the
(a) Add 1.0 mL of the vaccine to be examined to 1.0 mL of
specific monograph or the general chapter.
isopropyl myristate R and mix. Add 1.3 mL of 1 M hydrochloric
acid, 2.0 mL of chloroform R and 2.7 mL of a 9 giL solution All suitable sample preparation methods and measurement
of sodium chloride R. Mix thoroughly. Centrifuge at 15 000 g techniques (e.g. 2.2.22. Atomic emission spectrometry (AES),
for 60 mino Transfer the aqueous phase to a 10 mL volumetric 2.2.23. Atomic absorption spectrometry (AAS), 2.2.37. X-ray
flask and dilute to volume with water R. lf this procedure fails fluorescence spectrometry (XRFS), 2.2.57. Inductively
to separate the aqueous phase, add 100 giL of polysorbate 20 R coupled plasma-atomic emission spectrometry (ICP-AES),
to the sodium chloride solution and repeat the procedure but 2.2.58. Inductively coupled plasma-mass spectrometry
centrifuge at 22 500 g. (ICP-MS), 2.4.2. Arsenic, 2.4.8. Heavy metals, 2.4.9. Iron,
2.4.10. Lead in sugars, 2.4.15. Nickel in polyols, 2.4.31. Nickel in
(b) Add 1.0 mL of the vaccine to be examined to 1.0 mL of a
hydrogenated vegetable oils) can be used for the determination
100 giL solution of sodium chloride R and mix. Centrifuge
of metal residues, if the method has been verified before the
at 1000 g for 15 mino Transfer the aqueous phase to a 10 mL
initial use by a system suitability test or a validation procedure
volumetric flask and dilute to volume with water R.
according to this chapter.
(e) Add 1.0 mL of the vaccine to be examined to 2.0 mL
lf no sample preparation and/or measurement method is
of a 100 giL solution of sodium chloride R and 3.0 mL of
described in the specific monograph, a suitable sample
chloroform R and mix. Centrifuge at 1000 g for 5 mino Transfer
preparation and/or measurement method must be developed
the aqueous phase to a 10 mL volumetric flask and dilute to
and validated (see Figures 2.4.20.-1 and 2.4.20.-2).
volume with water R.

0112008:20419 SAMPLE PREPARATION


Sample preparation is critical to the success of elemental
2.4.19. ALKALINE IMPURITIES IN analysis. Many techniques not using direct measurement are
FATTY 0lL5 heavily dependent on sample transporto
If an atomisation system is used, the most conventional means
In a test-tube mix 10 mL ofrecently distilled acetone R and by which samples are introduced into the atomisation system
0.3 mL of water R and add 0.05 mL of a 0.4 giL solution of is by solution nebulisation. In this case, solid samples must
bromophenol blue R in alcohol R. Neutralise the solution if be dissolved in order to be introduced into the atomisation
necessary with 0.01 M hydrochloric acid or 0.01 M sodium system. Samples may be dissolved in any appropriate solvento
hydroxide. Add 10 mL of the oil to be examined, shake and The use of aqueous or dilute nitric acid solutions is strongly
allow to stand. Not more than 0.1 mL of 0.01 M hydrochloric recommended, due to minimal interference with these
acid is required to change the colour of the upper layer to solvents compared to other solvents. Hydrochloric acid,
yellow. hydrofluoric acid, perchloric acid, sulfuric acid and hydrogen
peroxide, at various concentrations, can be used to dissolve
04/2013:20420 the samples. The viscosity of sulfuric acid is greater than
that of the other acids and is to be taken into account as it
2.4.20. DETERMINATION OF METAL can affect the overall fluidity of the solution. The choice
of solvents includes, but is not limited to, the use of dilute
CATALYST OR METAL REAGENT bases, straight or diluted organic solvents, combinations of
RE5IDUES acids or bases, and combinations of organic solvents. Acids,
bases, and hydrogen peroxide of high purity must be used,
INTRODUCTION especially when ICP-MS is employed. For aqueous solutions,
This chapter describes the general approach for the use deionised distilled water R. Diluents must be checked for
determination of metal catalyst or metal reagent residues interference if they are used in an analysis. Because it is not
in substances for pharmaceutical use. As the chemical always possible to obtain organic solvents that are free of
composition of the considered substances and the specification metals, organic solvents of the highest purity possible with
limits for the metal(s) of interest vary considerably, it is regard to metal contaminants must be used. Specifically

General Notices (1) apply to all monographs and other texts 133
2.4.20. Determination of metal catalyst or metal reagent residues EUROPEAN PHARMACOPOEIA 8.0

No Is the procedure described Yes


in the monograph?

Is the sample Is the sample


suitable for suitable for
direct analysis? direct analysis?

No

Is the
compound No
Yes
soluble in
an aqueous
medium?

compound Yes
soluble in
Yes Yes
an organic
medium?

Prepare the
No sample
solutions

Perform
Are the Yes
elements closed-vessel
volatile? digestion

Verify procedure
anci/or instrument
No Perform open- or closed-
vessel digestion ar
incineration

No

Figure 2.4.20.-l. - Metal residues decision tree: sample preparation

for ICP techniques, where samples are introduced into the The suitability of a digestion technique, whether open-
plasma via solution nebulisation, it is important to consider or elosed-vessel, should be supported by spike recovery
the potential matrix effects and interferences that might arise experiments in order to verify that, within an acceptable
from the solvent. The use of an appropriate internal standard tolerance, volatile metals have not been lost during sample
and/or matching the standard matrix with samples should preparation. The digestion cyele is suitable if a clear solution
be applied for ICP-AES and ICP-MS analyses in cases where is obtained.
accuracy and precision are not sufficient. In any case, the It is important to consider the selection of the type, the
selection of an appropriate internal standard should take into material of construction, the pretreatment, and the eleaning
account the metal(s) of interest, ionisation energy, wavelengths of analyticallabware used in elemental analyses. The material
or masses, and the nature of the sample matrix. must be inert and, depending on the specific application,
Where a sample is found not to be soluble in any acceptable resistant to caustics, acids, and/or organic solvents. For sorne
solvent, a variety of digestion or incineration techniques can be analyses, care must be exercised to prevent the adsorption of
employed. These inelude hot-plate digestion, incineration and metal s onto the surface of a vessel, particularly in ultra-trace
microwave-assisted digestions, using open- and elosed-vessel. analyses. Contamination of sample solutions by metals and
The decision regarding the type of digestion technique to be ions present in the container can also lead to inaccurate results.
used depends on the nature of the sample being digested, The use of volumetric glassware that does not comply
as well as on the metal(s) of interest and the concentration with Class A requirements of the appropriate International
range of the metals to be quantified. Open-vessel digestion Standard of the International Organization for Standardization
is not recommended for the analysis of volatile metals. (ISO) is acceptable if the validation or the system suitability

134 See the information section on general monographs (cover pages)


EUROPEAN PHARMACOPOEIA 8.0 2.4.20. Determination of metal catalyst or metal reagent residues

No

Ves
No

Are the validation Perform the Verify the


requirements met? analysis using measurement
monograph procedure system

Ves Is the recovery No


r-----< of standard solution>----------......J
acceptable?

Is the recovery Ves


of standard solution>----------,
acceptable?

Figure 2.4.20.-2. - Metal residues decisian tree: measurement

test of the method using such glassware have experimentally physícochemícal methods, the concentration of a given metal
demonstrated that the method is suitable for the intended in the sample can be calculated from the concentration of the
purpose. metal in the solution using the following expression:
CAUTION: when using high-pressure digestian vessels and
micrawave labaratary equipment, the safety precautians and
e= A x VI X V2
m V3
aperating instructians given by the manufacturer must be
fa llawed. C concentration of metal in the analysed sample, in
MEASUREMENT
micrograms per gram;
A instrument reading of the concentration of the
Method. The choice of the techniques depends mainly on
metal in the sample solution, in micrograms per
the sample matrix and the characteristics and specification
millilitre;
limits of the metal(s) of interest. Analyse according to the
instructions of the manufacturer of the apparatus regarding m mass of the sample in the initial sample solution,
programme and wavelength. in grams;
System suitability. A system suitability test must be carried V¡ volume of the initial sample preparation, in
out on the day of the analysis to ensure that the sample millilitres;
preparation and measurement system are appropriate. V2 total volume of any dilution performed, in
Acceptance criterian far preparatian of sample salutían: a clear millilitres;
solution is obtained. V3 volume of initial sample preparation used in any
dilution performed, in millilitres.
Acceptance criterian far measurement system: the measured
concentration of a standard solution of the metal at a
concentration within the range of the used calibration curve VALIDATION REQUIREMENTS
does not differ from the actual concentration by more than Some validation requirements provided below may differ from
20 per cent. those provided in general chapters of the Ph. Eur. (e.g. 2.2.22
Calculation. The blank value of reagents must be taken into (AES), 2.2.23 (AAS), 2.2.57 (ICP-AES), 2.2.58 (ICP-MS)).
account for the calculation of the content. Upon completion of Before the initial use of the selected procedure, the analyst
the analysis, the concentration of a given metal in the sample must ensure that the sample preparation and measurement
is calculated by the software of the instrument from the method are appropriate for the metal(s), sample matrix and
concentration of the metal in the test solution. lf no calculation instrument used. This is accomplished by following the
software is available or no indication for calculation is given in validation procedure before the initial use and the system
the corresponding general chapter in section 2.2. Physical and suitability test on the day of the analysis.

General Natices (1) apply to all monagraphs and ather texts 135
2.4.21. Foreign oils in fatty oils by thin-layer chromatography EUROPEAN PHARMACOPOEIA 8.0

For metal residues, validation of a limit test must include 0112008:20421


specificity and limit of detection.
The following section defines the characteristics for the 2.4.21. FOREIGN OILS IN FATTY OILS
acceptability of a quantitative procedure. 1t must be BY THIN-LAYER CHROMATOGRAPHY
demonstrated experimentally that such a procedure complies
with the validation requirements, with an appropriate system Examine by thin-layer chromatography (2.2.27) using
suitability test using material spiked with a suitable reference kieselguhr G R as the coating substance. Impregnate a plate by
material. The test materials must be spiked before any sample placing it in a chromatographic tank containing the necessary
preparation steps. For example, if a test material is to be quantity of a mixture of 10 volumes of liquid paraffin R
digested, the material must be spiked at the beginning of the and 90 volumes of light petroleum R so that the plate dips
digestion procedure. about 5 mm beneath the surface of the liquido When the
SPECIFICITY impregnation mixture has risen by at least 12 cm from the
Specificity is the ability to ensure that the analytical procedures lower edge of the plate, remove the plate and allow the solvent
for sample preparation and measurement allow a reliable to evaporate for 5 mino Carry out the chromatography in the
determination of the metal(s) in the presence of components same direction as the impregnation.
(e.g. carrier gas, impurities, matrix) that may be expected to Preparation of the mixture offatty acids. Heat 2 g of the oil
be present. with 30 mL of 0.5 M alcoholic potassium hydroxide under a
Acceptance criteria: the procedure must be able to assess reflux condenser for 45 mino Add 50 mL of water R, allow
unequivocally each metal residue to be determined with to cool, transfer to a separating funnel and extract with
this procedure in the presence of components that may be three quantities, each of 50 mL, of ether R. Discard the ether
expected to be present, including other metal residues, matrix extracts, acidify the aqueous layer with hydrochloric acid R
components, and other sources of interference; specificity is and extract with three quantities, each of 50 mL, of ether R.
demonstrated by complying with the accuracy requirement Combine the ether extracts and wash with three quantities,
for the metal(s) to be determined. each of 10 mL, of water R; discard the washings, dry the ether
RANGE
over anhydrous sodium sulfate R and filter. Evaporate the ether
on a water-bath. Use the residue to prepare the test solution.
Acceptance criterion: range is demonstrated by complying The fatty acids may also be obtained from the soap solution
with the recovery requirement. prepared during the determination of the unsaponifiable
ACCURACY matter.
Verify the accuracy using a certified reference material (CRM) Test solution. Dissolve 40 mg of the mixture of fatty acids
or by performing a test for recovery. obtained from the substance to be examined in 4 mL of
Recovery. Recovery may be determined on a sample of the chloroform R.
substance to be examined, spiked with a known quantity of a Reference solution. Dissolve 40 mg of the mixture of fatty acids
reference standard of the metal (3 concentration levels in the obtained from a mixture of 19 volumes of maize oil R and
range of 50-150 per cent of the intended specification limit, 1 volume of rapeseed oil R in 4 mL of chloroform R.
even if the original concentration of the reference standard is
at the specified value), in triplicate. Apply to the plate 3 ¡.tL of each solution. Develop over a path of
8 cm using a mixture of 10 volumes of water R and 90 volumes
Acceptance criterion: spike recovery is within 70 per cent and of glacial acetic acid R. Dry the plate at 110°C for 10 mino
150 per cent for the mean of 3 replicates at each concentration. Allow to cool and, unless otherwise prescribed, place the plate
in a chromatographic chamber, with a tightly fitting lid, that·
REPEATABILITY
has previously been saturated with iodine vapour by placing
Test samples: either 6 independent samples of the substance iodine R in an evaporating dish at the bottom of the chamber.
to be examined spiked with a suitable reference standard at After some time brown or yellowish-brown spots become
the specified concentration level, or 3 concentration levels visible. Remove the plate and allow to stand for a few minutes.
prepared in triplicate. When the brown background colour has disappeared, spray
Acceptance criterio n : the relative standard deviation is in both with starch solution R. Blue spots appear which may become
cases not more than 20 per cent. brown on drying and again become blue after spraying with
INTERMEDIA TE PRECISION water R. The chromatogram obtained with the test solution
always shows a spot with an Rp of about 0.5 (oleic acid) and
The effect of random events (intra-Iaboratory variations) on
a spot with an Rp of about 0.65 (linoleic acid) corresponding
the analytical precision of the method must be established.
to the spots in the chromatogram obtained with the reference
Acceptable experiments for establishing intermediate
solution. With some oils a spot with an Rp of about 0.75 may
precision include performing the repeatability analysis on
be present (linolenic acid). By comparison with the spot in the
different days, or with different instrumentation, or by
chromatogram obtained with the reference solution, verify the
different analysts. Only 1 of the 3 experiments is required to
absence in the chromatogram obtained with the test solution
demonstrate intermediate precision.
of a spot with an Rp of about 0.25 (erucic acid).
Acceptance criterion: the relative standard deviation is not
more than 25 per cent.
LIMIT OF QUANTIFICATION
Determine the lowest concentration meeting the acceptance
criterion. Use the results from the accuracy study. 0112008:20422
Acceptance criterio n : the limit of quantification is below the correded 6.8
specification limit.
LIMIT OF DETECTION (ONLY APPLICABLE TO LIMIT 2.4.22. COMPOSITION OF FATTY
TESTS) ACIDS BY GAS CHROMATOGRAPHY
Determine the lowest concentration giving a signal clearly
distinct from that obtained with a blank solution. The test for foreign oils is carried out on the methyl esters
Acceptance criterion: the limit of detection is not more than of the fatty acids contained in the oil to be examined by gas
0.5 times the concentration of the specification limito chromatography (2.2.28).

136 See the information section on general monographs (caver pages)


EUROPEAN PHARMACOPOEIA 8.0 2.4.22. Composition of fatty adds by GC

METHODA - signal-to-noise ratio: minimum 5 for the peak due to methyl


myristate in the chromatogram obtained with reference
This method is not applieable to oi/s that eontain glyeerides
solution (b);
offatty aeids with an epoxy-, hydroepoxy-, hydroperoxy-,
eyclopropyl or eyclopropenyl group, or those that contain a
large proportion offatty acids of ehain length less than 8 earbon - number of theoretical plates: minimum 30 000, calculated
atoms or to oils with an aeid value greater than 2.0. for the peak due to methyl stearate in the chromatogram
obtained with reference solution (a).
Test solution. When prescribed in the monograph, dry the oil
to be examined before the methylation step. Weigh 1.0 g of System suitability when using the mixture of calibrating
the oil into a 25 mL round-bottomed flask with a ground-glass substances in Table 2.4.22.-2:
neck fitted with a reflux condenser and a gas port into the flask.
Add 10 mL of anhydrous methanol R and 0.2 mL of a 60 giL - resolution: minimum 4.0 between the peaks due to methyl
solution of potassium hydroxide R in methanol R. Attach the caprylate and methyl caprate in the chromatogram obtained
reflux condenser, pass nitrogen R through the mixture at a with reference solution (a);
rate of about 50 mLlmin, shake and heat to boiling. When
the solution is clear (usually after about 10 min), continue
heating for a further 5 mino Cool the flask under running - signal-to-noise ratio: minimum 5 for the peak due to methyl
water and transfer the contents to a separating funnel. Rinse caproate in the chromatogram obtained with reference
the flask with 5 mL of heptane R and transfer the rinsings solution (b);
to the separating funnel and shake. Add 10 mL of a 200 giL
solution of sodium ehloride R and shake vigorously. Allow to - number of theoretical plates: minimum 15 000, calculated
separate and transfer the organic layer to a vial containing for the peak due to methyl caprate in the chromatogram
anhydrous sodium sulfate R. Allow to stand, then filter. obtained with reference solution (a).

Reference solution (a). Prepare 0.50 g of the mixture of ASSESSMENT OF CHROMATOGRAMS


calibrating substances with the composition described in one Avoid working conditions tending to give masked peaks
of the 2.4.22 tables, as prescribed in the individual monograph (presence of constituents with small differences between
(if the monograph does not mention a specific solution, use retention times, for example linolenic acid and arachidic acid).
the composition described in Table 2.4.22.-1). Dissolve in
Qualitative analysis. Identify the peales in the chromatogram
heptane R and dilute to 50.0 mL with the same solvent.
obtained with reference solution (e) (isothermal operating
Referenee solution (b). Dilute 1.0 mL ofreference solution (a) conditions or linear temperature programming).
to 10.0 mL with heptane R.
When using isothermal operating conditions, the peales may
Referenee solution (e). Prepare 0.50 g of a mixture of fatty acid
also be identified by drawing calibration curves using the
methyl esters that corresponds in composition to the mixture
chromatogram obtained with reference solution (a) and the
of fatty acids indicated in the monograph of the substance
information given in Tables 2.4.22.-1, 2.4.22.-2 or 2.4.22.-3.
to be examined. Dissolve in heptane R and dilute to 50.0 mL
with the same solvent. Commercially available mixtures of
fatty acid methyl esters may also be used. Table 2.4.22.-1. - Mixture of ealibrating substances (jor gas
ehromatography with eapillary column and split inlet system,
Column:
it is recommended that the component with the longest ehain
material: fused silica, glass or quartz; length of the mixture to be examined be added to the ealibration
mixture, when the qualitative analysis is done using ealibratitm
- size: 1= 10-30 m, 0 = 0.2-0.8 mm; curves)
- stationary phase: maerogol 20 000 R (film thickness
0.1-0.5 [lm) or another suitable stationary phase. Mixture of the following substances Composition (per cent m/m)

Carrier gas: helium for ehromatography R or hydrogen for Methyllaurate R


chromatography R. Methyl myristate R 5
Flow rate: 1.3 mLlmin (for a column 0 = 0.32 mm). Methyl palmitate R 10
Split ratio: 1:100 or less, according to the internal diameter of Methyl stearate R 20
the column used (1:50 when 0 = 0.32 mm).
Methyl arachidate R 40
Temperature:
Methyl oleate R 20
- column: in isothermal conditions, 160-200 oC, according to
the length and type of column used (200 oC for a column
30 m long and coated with a layer of macrogol20 000 R); Table 2.4.22.-2. - Mixture of ealibrating substanees (jor gas
if a linear temperature programming is necessary, raise ehromatography with capillary column and split inlet system,
the temperature of the column at arate of 3 °C/min from it is recommended that the component with the longest chain
170 oC to 230 oC, for example; length of the mixture to be examined be added to the calibration
- injeetion port: 250 oC; mixture, when the qualitative analysis is done using ealibration
curves)
- detector: 250 oc.
Mixture of the following substances Composition (per cent m/m)
Detection: flame ionisation.
Methyl caproate R 10
Injection: 1 [lL.
Methyl caprylate R 10
System suitability when using the mixture of calibrating
substances in Table 2.4.22.-1 or Table 2.4.22.-3: Methyl decal10ate R 20

- resolution: minimum 1.8 between the peaks due to methyl Methyllaurate R 20


oleate and methyl stearate in the chromatogram obtained Methyl myristatc R 40
with reference solution (a);

General Notices (1) apply to all monographs and other texts 137
2.4.22. Composition of fatty adds by GC EUROPEAN PHARMACOPOEIA 8.0

Table 2.4.22.-3. - Mixture of calibrating substances (jor gas Fatty add Equivalent chain length
chromatography with capillary column and split inlet system, Alpha-linolenic acid 19.2
it is recommended that the component with the longest chain
length of the mixture to be examined be added to the calibration Arachidic acid 20.0
mixture, when the qualitative analysis is done using calibration Eicosenoic acid (gondoic acid) 20.2
curves)
Arachidonic acid 21.2
Mixture of the following substances Composition (per cent m/m)
Behenic acid 22.0
NIethyl myristate R 5
Erucic acid 22.2
Methyl palmitate R 10
12-0xostearic acid 22.7
Methyl stearate R 15
Ricinoleic acid 23.9
Methyl arachidate R 20
12-Hydroxystearic acid 23.9
Methyl oleate R 20
Lignoceric acid 24.0
Methyl eicosenoate R 10
Nervonic acid 24.2
Methyl behmate R 10

Methyllignocerate R 10
METHOD B
Measure the reduced retention time (t~) of each peak in the This method is not applicable to oils that contain glycerides
chromatogram obtained with reference solution (a). t~ is the offatty acids with an epoxy-, hydroepoxy-, hydroperoxy-,
retention time measured from the solvent peak and not from cyclopropyl or cyclopropenyl group or to oi/s with an acid value
the time of injection. Plot the straight line: greater than 2.0.
Test solution. Introduce 0.100 g ofthe substance to be
lOglO (t~) = f (equivalent chain length) examined into a 10 mL centrifuge tube with a screw cap.
Dissolve with 1 mL of heptane R and 1 mL of dimethyl
The logarithms of t ~ of unsaturated acids are situated on this
carbonate R and mix vigorously under gentle heating
line at points corresponding to non-integer values of carbon (50-60 OC). Add, while still warm, 1 mL of a 12 giL solution
atoms known as 'equivalent chain lengths'; the equivalent
of sodium R in anhydrous methanol R, prepared with the
chain length is the length of the theoretical saturated chain
necessary precautions, and mix vigorously for about 5 mino
that would have the same t ~ as the fatty acid to be identified.
Add 3 mL of distilled water R and mix vigorously for about
For example, linoleic acid has the same t ~ as the theoretical
30 S. Centrifuge for 15 min at 1500 g. Inject 1 I1L of the
saturated fatty acid having 18.8 carbon atoms.
organic phase.
Identify the peaks in the chromatogram obtained with the
Reference solutions and assessment of chromatograms. Where
test solution by means of the straight line and the reduced there is no specific prescription in the individual monograph,
retention times. Equivalent chain lengths are given in
proceed as described under Method A.
Table 2.4.22.-4.
Column:
Quantitative analysis. In general, the normalisation
material: fused silica;
procedure is used in which the sum of the areas of the peaks in -
the chromatogram, except that ofthe solvent, is set at 100 per - size: 1 = 30 m, 0 = 0.25 mm;
cent. The content of a constituent is calculated by determining - stationary phase: macrogol 20 000 R (film thickness
the area of the corresponding peak as a percentage of the sum 0.25 11m).
of the afeas of all the peaks. Disregard any peak with an area Carrier gas: helium for chromatography R.
less than 0.05 per cent of the total area. Flow rate: 0.9 mLlmin.
In certain cases, for example in the presence of fatty acids
with 12 or less carbon atoms, correction factors can be Split ratio: 1:100.
prescribed in the individual monograph to convert peak areas _T_e_m_p_e_r_a_tu_r_e_:_ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ __
in per cent mlm. Time Temperature
(min) (oC)
Table 2.4.22.-4. - Equivalent chain lengths (this value, which is
Column O - 15 100
to be calculated using calibration curves, is given as an example
for a column of macrogol 20 000 R) 15 - 36 100 ~ 225
Fatty acid Equivalent chain length 36 - 61 225
Caproic acid 6.0
Injection port 250
Caprylic acid 8.0
Detector 250
Capric acid 10.0
Detection: flame ionisation.
Lauric acid 12.0
Injection: 1 11L.
Myristic acid 14.0
METHOD C
Palmitic acid 16.0
This method is not applicable to oils that contain glycerides of
Palmitoleic acid 16.3 fatty acids with epoxy-, hydroepoxy-, hydroperoxy-, aldehyde,
ketone, cyclopropyl and cyclopropenyl groups, and conjugated
Margaric acid 17.0
polyunsaturated and acetylenic compounds beca use of partial
Stearic acid 18.0 or complete destruction of these groups.
Oleic acid 18.3
Test solution. Dissolve 0.10 g of the substance to be
examined in 2 mL of a 20 giL solution of sodium hydroxide R
Linoleic acid 18.8 in methanol R in a 25 mL conical flask and boil under
Gamma-linolenic acid 19.0
a reflux condenser for 30 mino Add 2.0 mL of boron
trifluoride-methanol solution R through the condenser and

138 See the information section on general monographs (cover pages)


EUROPEAN PHARMACOPOEIA 8.0 2.4.23. Sterols in fatty oils

boil for 30 mino Add 4 mL of heptane R through the condenser Develop over a path of 17 cm using a mixture of 35 volumes
and boil for 5 mino Cool and add 10.0 mL of saturated sodium of ether R and 65 volumes of hexane R. Dry the plates in a
chloride solution R, shake for about 15 s and add a quantity current of nitrogen R. Spray the plates with a 2 giL solution of
of saturated sodium chloride solution R such that the upper dichlorofluorescein R in anhydrous ethanol R and examine in
phase is brought into the neck of the flask. Collect 2 mL of the ultraviolet light at 254 nm. The chromatogram obtained with
upper phase, wash with 3 quantities, each of 2 mL, of water R the reference solution shows bands due to cholesterol and
and dry over anhydrous sodium sulfate R. betulin. The chromatograms obtained with the test solutions
Reference solutions, chromatographic procedure and assessment show bands with similar Rp values due to sterols. From each of
of chromatograms. Where there is no specific prescription the chromatograms, remove an are a of coating corresponding
in the individual monograph, proceed as described under to the area occupied by the sterol bands and additionally the
Method A. area of the zones 2-3 mm aboye and below the visible zones
corresponding to the reference solution. Place separately
in three 50 mL flasks. To each flask add 15 mL of methylene
chloride R and heat under reflux with stirring, for 15 mino
Filter each solution through a sintered -glass filter (40) (2.1.2)
07/2011:20423 or suitable filter paper and wash each filter with 3 quantities,
each of 15 mL, of methylene chloride R. Place the combined
2.4.23. STEROLS IN FATTY OILS filtrate and washings from each filter separately in 3 flasks,
evaporate under a stream of nitrogen R to 5-10 mL. Transfer
When the monograph do es not specify the method to be used, to a small test tube and evaporate to dryness under a stream
method A is applied. Any change from method A to method B of nitrogen R.
must be validated. Determination of the sterols (GC)
Gas chromatography (2.2.28). Carry out the operations
METHODA protected from humidity and prepare the solutions immediately
Separation of the sterol fraction (TLC) before use.
Prepare the unsaponifiable matter and then isolate the sterol Test solution. To the sterols separated from the substance
fraction of the fatty oil by thin-Iayer chromatography (2.2.27), to be examined by thin-layer chromatography add a freshly
using a TLC silica gel plate R with a 0.2 mm to 0.5 mm layer. prepared mixture of 0.04 mL of chlorotrimethylsilane R, 0.1 mL
Test solution (a). In a 150 mL flask fitted with a reflux of hexamethyldisilazane R and 0.5 mL of anhydrous pyridine R.
condenser, place a volume of a 2 giL solution of betulin R in Allow to stand for at least 5 min and use the liquid phase.
methylene chloride R containing betulin corresponding to Reference solution (a). To 9 parts of the sterols separated
about 10 per cent of the sterol content of the sample used for from rapeseed oil R by thin-Iayer chromatography add 1 part
the determination (e.g. in the case of olive oil add 500 flL, in of eholesterol R. To the mixture add a freshly prepared
the case of other vegetable oils add 1500 flL of the betulin mixture of 0.04 mL of chlorotrimethylsilane R, 0.1 mL of
solution). rf the monograph requires the percentage content hexamethyldisilazane R and 0.5 mL of anhydrous pyridine R.
of the individual sterols in the sterol fraction, the addition Allow to stand for at least 5 min and use the liquid phase.
of betulin may be omitted. Evaporate to dryness under a
Reference solution (b). To the sterols separated from sunflower
current of nitrogen R. Add 5.00 g (m) of the substance to be
examined. Add 50 mL of 2 M alcoholic potassium hydroxide R oil R by thin-layer chromatography add a freshly prepared
mixture of 0.04 mL of chlorotrimethylsilane R, 0.1 mL of
and heat on a water-bath for 1 h, swirling frequently. Cool to
a temperature below 25 oC and transfer the contents of the hexamethyldisilazane R and 0.5 mL of anhydrous pyridine R.
Allow to stand for at least 5 min and use the liquid phase.
flask to a separating funnel with 100 mL of water R. Shake
the ¡iquid carefully with 3 quantities, each of 100 mL, of Column:
peroxide-free ether R. Combine the ether layers in another - material: fused silica;
separating funnel containing 40 mL of water R, shake gently
for a few minutes, allow to separate and reject the aqueous - size: 1 = 20-30 m, 0 = 0.25-0.32 mm;
phase. Wash the ether phase with several quantities, each - stationary phase: poly!methyl(95)phenyl(5)]siloxane R or
of 40 mL, of water R, until the aqueous phase is no longer poly(cyanopropyl)(7)(phenyl)(7)(methyl)(86)siloxane R
alkaline to phenolphthalein. Transfer the ether phase to a (film thickness 0.25 flm).
tared flask, washing the separating funnel with peroxidejree
ether R. Distil off the ether with suitable precautions and add Carrier gas: hydrogen for chromatography R or helium for
6 mL of acetone R to the residue. Carefully remove the solvent chromatography R.
in a current of nitrogen R. Dry to constant mass at 100-105 oc. Linear velocity: 30-50 cm/s (hydrogen) or 20-35 cm/s (helium).
Allow to cool in a desiccator and weigh. Transfer the residue
Split ratio: 1:50 (hydrogen) or 1:100 (helium).
to a small test tube with methylene chloride R. Evaporate
under a stream of nitrogen R to a volume of about 1 mL. Temperature:
Depending on the unsaponifiable content of the oil, adapt the - column: 260 oC;
final concentration of the solution to 25-50 mg/mL.
- injection port: 280 oC;
Test solution (b). Treat 5.00 g of rapeseed oil Ras prescribed
for the substance to be examined, beginning at the words "Add - detector: 290 oc.
50 mL of 2 M alcoho/ic potassium hydroxide R". Detection: flame ionisation.
Test solution (e). Treat 5.00 g of sunflower oil Ras prescribed Injection: 1 11L.
for the substance to be examined, beginning at the words "Add
50 mL of 2 M alcoholie potassium hydroxide R". Identification of peaks: the chromatogram obtained with
reference solution (a) shows 4 principal peaks corresponding
Reference solution. Dissolve 25 mg of eholesterol R and 10 mg to cholesterol, brassicasterol, campesterol and ~-sitosterol and
of betulin R in 1 mL of methylene chloride R. the chromatogram obtained with reference solution (b) shows
Use a separate plate for each test solution. Apply as a band 4 principal peaks corresponding to campesterol, stigmasterol,
of 10 mm, at 20 mm from the base and 10 mm from the left ~-sitosterol and Ll7-stigmastenol. The relative retentions of
edge, 10 flL of the reference solution and as bands of 150 mm, the sterols with reference to ~-sitosterol (main peak) are given
at 20 mm from the base, 0.5 mL oftest solutions (a), (b) or (c). in Table 2.4.23.-1.

General Notices (1) apply to all monographs and other texts 139
2.4.23. Sterols in fatty oils EUROPEAN PHARMACOPOEIA 8.0

Table 2.4.23.-1. - Relative retentions of sterols with reference to Simultaneously prepare under the same conditions the
f3-sítosterol for 2 different columns unsaponifiable matter of sunflower oil R. This will in particular
Poly( cyanopropyl) (7)- Poly[methyl(95)-
serve to locate the sterol fraction to be collected.
(phenyl)(7)- phenyl(5) ]siloxane Separation of the sterol fraction (LC)
(methyl)(86)siloxane
Cholesterol 0.64 0.63 Liquid chromatography (2.2.29).
Test solution. Take up the residue with 3 quantities, each
Brassicasterol 0.70 0.71
of 4 mL, of the solvent used during the preparation of the
24-Methylenecholesterol 0.79 0.80 unsaponifiable matter (generally ether R OI light petroleum R)
and transfer to a 15 mL tube. Evaporate to dryness under
Campesterol 0.82 0.81
a current of nitrogen R. Dissolve the residue in a volume
Campestanol 0.83 0.82 of mobile phase sufficient to obtain a solution with an
approximate concentration of 40 mg/mL. Add a few drops of
Stigmasterol 0.87 0.87
2-propanol Rl to improve the solubility (3 drops are normally
117 -Campesterol 0.93 0.92 sufficient to ensure complete solubilisation). Filter through a
membrane filter (nominal pore size 0.45 [lm).
1l5,23-Stigmastadienol 0.95 0.95
Reference solution. Proceed as described for the test solution
Clerosterol 0.96 0.96
with the unsaponifiable matter obtained with sunflower oil R.
~-Sitosterol Precolumn:
Sitostanol 1.01 1.02 - size: 1= 5 mm, 0 = 4.6 mm;
1l5-Avenasterol 1.03 1.03 - stationary phase: si/ica gel for chromatography R (5 [lm)
with a pore size of 6 nm.
1l5,24-Stigmastadienol 1.09 1.08
Column:
117 -Stigmastenol(I) 1.13 1.12
- size: 1 = 0.25 m, 0 = 4.6 mm;
1l7-Avenasterol !.l8 1.16
- stationary phase: silica gel for chromatography R (5 [lm)
Betulin lA lA with a pore size of 6 nm.
(1) This sterol may also be referred to as 117 -stigmasterol in Iiteratnre. Mobile phase: 2-propanol Rl, hexane R 0:99 V/V).
Flow rate: 1 mL/min.
The peak due to the internal standard (betulin) must be clearly
separated from the peaks due to the sterols to be determined. Detection: spectrophotometer at 210 nm.
For the chromatogram obtained with the test solution, identify Injection: 50 [lL.
the peaks and calculate the percentage content of each sterol Identification of the peaks due to sterols: the sterol fraction
in the sterol fraction of the substance to be examined using elutes at the end of the chromatogram. Locate the fraction
the following expression: to be collected using the chromatogram obtained with the
A reference solution, which shows 2 principal peaks eluting
S x 100 approximately between 23 min and 32 mino Collect the
fraction at the detector outlet in a 15 mL tube with a screw
cap. Evaporate the solvent under a current of nitrogen R.
A are a of the peak due to the component to be
determined; Determination of the sterols (GC)
s sum of the areas of the peaks due to the components Gas chromatography (2.2.28).
indicated in Table 2.4.23.-1; disregard the peak due Test solution. Dissolve the residue of the sterol fraction
to betulin. obtained with the test solution in the previous LC step in
If required in the monograph, calculate the content of each 0.2 mL of anhydrous pyridine R and 0.2 mL of a mixture
sterol in milligrams per 100 grams of the substance to be of 1 volume of chlorotrimethylsilane R and 99 volumes of
examined using the following expression: N,O-bis(trimethylsilyl)trifluoroacetamide R. Stopper the tube
tightly and heat at 80 oC for 20 mino Allow to cool and use
A x mi x 100 the liquid phase.
A' Xm Reference solution. Dissolve the residue of the sterol fraction
obtained with the reference solution in the previous LC step
A are a of the peak due to the component to be in 0.2 mL of anhydrous pyridine R and 0.2 mL of a mixture
determined; of 1 volume of chlorotrimethylsilane R and 99 volumes of
A' area of the peak due to betulin; N,O-bis(trimethylsilyl)trifluoroacetamide R. Stopper the tube
m tightly and heat at 80 oC for 20 mino Allow to cool and use
mass of the sample of the substance to be examined, the liquid phase.
in grams;
A standard of cholesterol (cholesterol R) may also be used,
m mass of betulin R added, in milligrams.
alone or as a mixture with the sterol fraction of sunflower oil.
Proceed with derivatisation as described for the test solution.
METHOD B
Column:
Preparation of the unsaponifiable matter
- material: fused silica;
Prepare the unsaponifiable matter according to the method
- size: 1= 30 m, 0 = 0.25 mm;
stated in the test for unsaponifiable matter of the monograph
on the substance to be examined. Failing this, prepare the - stationary phase: poly[methyl(95)phenyl(5)]siloxane R (film
unsaponifiable matter according to the method described thickness 0.25 [lm).
in chapter 2.5.7. Unsaponifiable matter. After the final Carrier gas: helium for chromatography R.
neutralisation step, evaporate the ethanol, then add 6 mL
of acetone R and evaporate the solvent. Dry the residue at Flow rate: 2.6 mL!min.
100-105 oc. It is not necessary to dry to constant mass. Split ratio: 1:25.

140 See the information section on general monographs (caver pages)


EUROPEAN PHARMACOPOEIA 8.0 2.4.24. Identification and control of residual solvents

Temperature: PROCEDURE
Time Temperature Examine by gas chromatography with static head-space
(min) Ce) injection (2.2.28).
CoJumn 0-38 260 Sample preparation l. This is intended for the control of
38 - 44 2607290 residual solvents in water-soluble substances.
44 - 49 290
Sample solution (1). Dissolve 0.200 g of the substance to be
Injection port 290
examined in water R and dilute to 20.0 mL with the same
Detector 290 solvent.
Sample preparation 2. This is intended for the control of
Deteetion: flame ionisation.
residual solvents in water-insoluble substances.
Injeetion: 1-3 flL (depending on the expected amount of
Sample solution (2). Dissolve 0.200 g of the substance to
sterols in the substance to be examined).
be examined in dimethylformamide R (DMF) and dilute to
Identifieation of peaks: use the chromatogram obtained with 20.0 mL with the same solvent.
the reference solution to identify the peaks due to campesterol,
stigmasterol, ~-sitosterol and L'>7-stigmastenol. Identify the Sample preparation 3. This is intended for the control of
peaks due to the sterols in the chromatogram obtained with N,N-dimethylacetamide and/or N,N-dimethylformamide,
the test solution using the chromatogram obtained with the when it is known or suspected that one or both of these
reference solution and the relative retentions with reference to substances are present in the substance to be examined.
~-sitosterol (main peak) given in Table 2.4.23.-1. Sample solution (3). Dissolve 0.200 g ofthe substance to be
System suitability: reference solution: examined in 1,3-dimethyl-2-imidazolidinone R (DMI) and
dilute to 20.0 mL with the same solvent.
- resolution: minimum 4.0 between the peaks due to
campesterol and stigmasterol. In sorne cases none of the aboye sample preparation
Calculate the percentage content of each sterol in the sterol procedures are appropriate, in which case the diluent to be
used for the preparation of the sample solution and the static
fraction of the substance to be examined using the following
express ion : head-space conditions to be employed must be demonstrated
to be suitable.
A Solvent solution (a). To 1.0 mL of Class 1 residual solvent
S x 100
solution CRS, add 9 mL of dimethyl sulfoxide R and dilute
to 100.0 mL with water R. Dilute 1.0 mL of this solution to
A area of the peak due to the component to be 100 mL with water R. Dilute 1.0 mL ofthis solution to 10.0 mL
determined; with water R.
s sum of the areas of the peaks due to the components The reference solutions correspond to the following limits:
indicated in Table 2.4.23.-1, except betulin.
- benzene: 2 ppm,
- carbon tetrachloride: 4 ppm,
01/2008:20424
corrected 7.2 - 1,2-dichloroethane: 5 ppm,
- 1, l-dichloroethene: 8 ppm,
2.4.24. IDENTIFICATION AND - 1,1,1-trichloroethane: 10 ppm.
CONTROL OF RESIDUAL SOLVENTS Solvent solution (b). Dissolve appropriate quantities of the
Class 2 residual solvents in dimethyl sulfoxide R and dilute to
The test procedures described in this general method may 100.0 mL with water R. Dilute to give a concentration of 1/20
be used: ofthe limits stated in Table 2 (se e 5.4. Residual solvents).
i. for the identification of the majority of Class 1 and Class 2 Solvent solution (e). Dissolve l.00 g of the solvent or solvents
residual solvents in an active substance, excipient or medicinal present in the substance to be examined in dimethyl sulfoxide R
product when the residual solvents are unknown; or water R, if appropriate, and dilute to 100.0 mL with water R.
ii. as a limit test for Class 1 and Class 2 solvents when present Dilute to give a concentration of 1/20 of the limit(s) stated in
in an active substance, excipient or medicinal product; Table 1 or 2 (see 5.4. Residual solvents).
iii. for the quantification of Class 2 solvents when the limits are Blank solution. Prepare as described for solvent solution (c)
greater than 1000 ppm (0.1 per cent) or for the quantification but without the addition of solvent(s) (used to verify the
of Class 3 solvents when required. absence of interfering peaks).
Class 1, Class 2 and Class 3 residual solvents are listed in Test solution. Introduce 5.0 mL of the sample solution and
general chapter 5.4. Residual solvents. 1.0 mL of the blank solution into an injection vial.
Three diluents are described for sample preparation and Reference solution (a) (Class 1). Introduce 1.0 mL of solvent
the conditions to be applied for head-space injection of the solution (a) and 5.0 mL of the appropriate diluent into an
gaseous sample onto the chromatographic system. Two injection vial.
chromatographic systems are prescribed but System A Referenee solution (al) (Class 1). Introduce 5.0 mL of the
is preferred whilst System B is employed normally for sample solution and 1.0 mL of solvent solution (a) into an
confirmation of identity. The choice of sample preparation injection vial.
procedure depends on the solubility of the substance to be
examined and in certain cases the residual solvents to be Referenee solution (b) (Class 2). Introduce 1.0 mL of solvent
controlled. solution (b) and 5.0 mL of the appropriate diluent into an
injection vial.
The following residual solvents are not readily detected by
the head-space injection conditions described: formamide, Referenee solution (e). Introduce 5.0 mL of the sample solution
2-ethoxyethanol, 2-methoxyethanol, ethylene glycol, and 1.0 mL of solvent solution (c) into an injection vial.
N-methylpyrrolidone and sulfolane. Other appropriate Reference solution (d). Introduce 1.0 mL of the blank solution
procedures should be employed for the control of these and 5.0 mL of the appropriate diluent into an injection vial.
residual solvents. Close the vials with a tight rubber membrane stopper coated
When the test procedure is applied quantitatively to control with polytetrafluoroethylene and secure with an aluminium
residual solvents in a substance, then it must be validated. crimped cap. Shake to obtain a homogeneous solution.

General Notiees (1) apply to all monographs and other texts 141
2.4.24. Identification and control of residual solvents EUROPEAN PHARMACOPOEIA 8.0

The following static head-space injection conditions may be determined. The system is suitable if the ehromatogram
used: obtained resembles the ehromatogram shown in
Figure 2.4.24.-2 and the resolution between acetonitrile and
Sample preparation methylene chloride is at least 1.0.
procedure
Operating parameters 2 3 Inject 1 mL of the gaseous phase of the test solution onto
the column described in System A. If in the ehromatogram
Equilibration temperature (oC) 80 105 80 obtained, there is no peak which corresponds to one of
Equilibration time (min) 60 45 45 the residual solvent peaks in the ehromatograms obtained
with referenee solution (a) 01' (b), then the substance to be
Transfer-line temperature (oC) 85 110 105 examined meets the requirements of the test. If any peak in
Carrier gas: Nitrogen for chromatography R or Helium for chromatography R
the ehromatogram obtained with the test solution eorresponds
at an appropriate pressure to any of the residual solvent peaks obtained with referenee
Pressurisation time (5) 30 30 30 solution (a) or (b) then System B is to be employed.

Injection volume (mL) Inject 1 mL of the gaseous phase of reference solution (a)
onto the column described in System B and record the
The chromatographic procedure may be carried out using: chromatogram under sueh conditions that the signal-to-noise
ratio for benzene can be measured. The signal-to-noise ratio
SYSTEM A must be at least 5. A typical ehromatogram is shown in
Figure 2.4.24.-3.
- a fused-silica capillary or wide-bore column 30 m long and
0.32 mm or 0.53 mm in internal diameter coated with Inject 1 mL of the gaseous phase of reference solution (al)
cross-linked 6 per cent polycyanopropylphenylsiloxane and onto the eolumn deseribed in System B. The peaks due to the
94 per cent polydimethylsiloxane (film thickness: 1.8 11m Class I residual solvents are still detectable.
or 311m),
Inject 1 mL of the gaseous phase of referenee solution (b)
- nitrogen for chromatography R or helium for onto the column described in System B and record the
chromatography R as the carrier gas, split ratio 1:5 with a ehromatogram under sueh conditions that the resolution
linear velocity of about 35 cm/s, between acetonitrile and trichloroethene can be determined.
The system is suitable if the ehromatogram obtained
- a flame-ionisation detector (a mass spectrometer may also
resembles the ehromatogram shown in Figure 2.4.24.-4 and
be used or an electron -capture detector for the chlorinated
the resolution between acetonitrile and trichloroethene is at
residual solvents of Class 1),
least 1.0.
maintaining the temperature of the column at 40 oC for
20 min, then raising the temperature at arate of 10 oC Inject 1 mL of the gaseous phase of the test solution onto
per min to 240 oC and maintaining it at 240 oC for 20 min the column described in System B. If in the chromatogram
and maintaining the temperature of the injection port at obtained, there is no peak whieh corresponds to any of the
140 oC and that of the detector at 250 oC, or, where there is residual solvent peaks in the ehromatogram obtained with
interference from the matrix, use: the reference solution (a) or (b), then the substance to be
SYSTEM B examined meets the requirements of the test. If any peak in
the ehromatogram obtained with the test solution eorresponds
- a fused-silica capillary or wide-bore column 30 m long and to any of the residual solvent peaks obtained with reference
0.32 mm or 0.53 mm in internal diameter coated with solution (a) or (b) and confirms the correspondenee obtained
macrogol 20 000 R (film thickness: 0.25 11m), when using System A, then proceed as follows.

- nitrogen for chromatography R or helium for Injeet 1 mL ofthe gaseous phase ofreferenee solution (e) onto
chromatography Ras the carrier gas, split ratio 1:5 with a the eolumn deseribed for System A or System B. If neeessary,
linear velocity of about 35 cm/s. adjust the sensitivity of the system so that the height of the
peak corresponding to the identified residual solvent(s) is at
- a flame-ionisation detector (a mass spectrophotometer least 50 per cent of the full scale of the recorder.
may also be used or an electron -capture detector for the
chlorinated residual solvents of Class 1), Inject 1 mL of the gaseous phase of reference solution (d) onto
maintaining the temperature of the column at 50 oC for the column. No interfering peaks should be observed.
20 min, then raising the temperature at arate of 6 oC per min
to 165 oC and maintaining it at 165 oC for 20 min and Injeet 1 mL of the gaseous phase of the test solution and
maintaining the temperature of the injection port at 140 oC 1 mL of the gaseous phase ofreferenee solution (e) on to the
and that of the detector at 250 oc. column. Repeat these injections twiee more.

Inject 1 mL of the gaseous phase of reference solution (a) The mean area of the peak of the residual solvent(s) in the
onto the column described in System A and record the chromatograms obtained with the test solution is not greater
chromatogram under such conditions that the signal-to-noise than half the mean area of the peak of the corresponding
ratio for 1,1,l-trichloroethane can be measured. The residual solvent(s) in the ehromatograms obtained with
signal-to-noise ratio must be at least 5. A typical referenee solution (c). The test is not valid unless the relative
chromatogram is shown in Figure 2.4.24.-l. standard deviation of the differences in are as between the
analyte peaks obtained from 3 replicate paired injections of
Inject 1 mL of the gaseous phase of reference solution (al) reference solution (e) and the test solution, is at most 15 per
onto the column described in System A. The peaks due to the cent.
Class 1 residual solvents are still detectable.
A flow diagram of the proeedure is shown in Figure 2.4.24.-5.
Inject 1 mL of the gaseous phase of reference solution (b)
onto the column described in System A and record the When a residual solvent (Class 2 or Class 3) is present at a level
chromatogram under such conditions that the resolution of 0.1 per cent or greater then the eontent may be quantitatively
between aeetonitrile and methylene ehloride can be determined by the method of standard additions.

142 See the information section on general monographs (caver pages)


EUROPEAN PHARMACOPOEIA 8.0 2.4.24. Identification and control of residual solvents

2 4/5

+o----~----~2----~3----~4~--~5----~6----~7~----8----~9----~1~O----~1~'1----~1~2----~13----~1~4----m~in

1. 1,1-dichloroethene 2. 1,1,1-trichloroethane 3. carbon tetrachloride 4. benzene 5. 1,2-dichloroethane

Figure 2.4.24.-1. - Typical chromatogram of class 1 solvents using the conditions described for System A and Procedure 1.
Flame-ionisation detector.
3 4 5/6 8 11 14 1617
I
I,
18
I!
10
I
I

15

17

J U 9 _-IV
'--_

o 5 10 15 20 25 min
1. methanol 5. cis-1,2-dichloroethene 9. 1,2-dimethoxyethane 13. pyridine 16. chlorobenzene

2. acetonitrile 6. nitromethane 10. 1,1,2-trichloroethene 14. toluene 17. xylene ortho, meta, para

3. dichloromethane 7. chloroform 11. methylcyclohexane 15. 2-hexanone 18. tetralin

4. hexane 8. cyclohexane 12. l,4-dioxan

Figure 2.4.24.-2. - Chromatogram of Class 2 solvents using the conditions described for System A and Procedure 1.
Flame-ionisation detector.

General Notices (1) apply to all monographs and other texts 143
2.4.24. Identification and control of residual solvents EUROPEAN PHARMACOPOEIA 8.0

2/3

o 3
1. l,l-dichloroethene 2. 1,1,1-trichloroethane 3. carbon tetrachloride 4. benzene 5. 1,2-dichloroethane

Figure 2.4.24.-3. - Chromatogram of Class 1 residual solvents using the conditions described for System B and Procedure 1.
Flame-ionisation detector.
4811 5/10 14 17

3/9
17
16

6 13

O 2 3 4 5 7 8 9 mín

1. rnethanol 5. cis-1,2-dichloroethene 9. 1,2-dirnethoxyethane 13. pyridine 16. chlorobenzene

2. acetonitrile 6. nitrornethane 10. 1,1,2-trichloroethene 14. toluene 17. xylene ortho, meta, para

3. dichlorornethane 7. chloroforrn 11. rnethylcyclohexane 15. 2-hexanone 18. tetralin (tR = 28 rnin)

4. hexane 8. cyclohexane 12. l,4-dioxan

Figure 2.4.24.-4. - Typical chromatogram of class 2 residual solvents using the conditions described for System B and Procedure 1.
Flame-ionisation detector.

144 See the information section on general monographs (cover pages)


EUROPEAN PHARMACOPOEIA 8.0 2.4.25. Ethylene oxide and dioxan

NO PASSES THE TEST


>---~I NO FURTHER ACTION

'.>-N0--flllllli>-ll PASSES THE TEST


NO FURTHER ACTION

PREPARATION OF TEST AND REFERENCE SOLUTIONS

ANHALF THE AREA

Figure 2.4.24.-5. - Diagram relating to the identification of residual solvents and the application of limit tests
01/2014:20425 Head-space gas chromatography (2.2.28).
A. For samples soluble in or miscible with water, the following
2.4.25. ETHYLENE OXIDE AND procedure may be used.
DIOXAN Test solution. Weigh 1.00 g (M r) of the substance to
be examined in a 10 mL vial (other sizes may be used
The test is intended for the determination of residual depending on the operating conditions) and add 1.0 mL of
ethylene oxide and dioxan in samples soluble in water or water R. Close and mix to obtain a homogeneous solution.
dimethylacetamide. For substances that are insoluble or AlIow to stand at 70 oC for 45 mino
insufficiently soluble in these solvents, the preparation of the
sample solution and the head-space conditions to be employed
are given in the individual monograph.

General Notices (1) apply to all monographs and other texts 145
2.4.26. N,N-Dimethylaniline EUROPEAN PHARMACOPOEIA 8.0

Reference solution (a). Weigh l.00 g (M R ) ofthe substance - signal-to-noise ratio: minimum 5 for the peaks due to
to be examined into an identical10 mL vial, add 0.10 mL ethylene oxide and dioxan.
of dioxan solution Rl and 0.50 mL of ethylene oxide Verification of precision
solution R3. Close and mix to obtain a homogeneous
solution. Allow to stand at 70 oC for 45 mino For each pair of injections, calculate for ethylene oxide and
for dioxan the difference in are a between the peaks obtained
Reference solution (b). To 0.50 mL of ethylene oxide with the test solution and reference solution (a). The test is
solution R3 in a 10 mL vial add 0.1 mL of a freshly prepared not valid unless the relative standard deviation of the 3 values
10 mg/L solution of acetaldehyde R and 0.10 mL of dioxan obtained for ethylene oxide is not greater than 15 per cent and
solution Rl. Close and mix to obtain a homogeneous the relative standard deviation of the 3 values obtained for
solution. Allow to stand at 70 oC for 45 mino dioxan is not greater than 15 per cent. rf the weighings used
B. For samples soluble in or miscible with dimethylacetamide, for the test solution and reference solution (a) differ from
the following procedure may be used. 1.00 g by more than 0.5 per cent, the appropriate corrections
Test solution. Weigh l.00 g (M T ) of the substance to must be made.
be examined in a 10 mL vial (other sizes may be used Calculate the content of ethylene oxide or dioxan in parts per
depending on the operating conditions) and add 0.20 mL million from the following expressions:
of water R and l.0 mL of dimethylacetamide R. Close and
mix to obtain a homogeneous solution. Allow to stand at AT x e
90 oC for 45 mino
Reference solution (a). Weigh 1.00 g (MR ) ofthe substance
to be examined into an identical10 mL vial, add 0.10 mL AT area of the peak due to ethylene oxide in the
of dioxan solution R1, 0.10 mL of ethylene oxide solution R2 chromatogram obtained with the test solution;
and 1.0 mL of dimethylacetamide R. Close and mix to AR are a of the peak due to ethylene oxide in
obtain a homogeneous solution. Allow to stand at 90 oC the chromatogram obtained with reference
for 45 mino solution (a);
Reference solution (b). To 0.10 mL of ethylene oxide MT mass of the substance to be examined in the test
solution R2 in a 10 mL vial, add 0.1 mL of a freshly prepared solution, in grams;
10 mg/L solution of acetaldehyde R and 0.10 mL of dioxan
MR mass ofthe substance to be examined in reference
solution R1. Close and mix to obtain a homogeneous
solution (a), in grams;
solution. Allow to stand at 70 oC for 45 mino
Column: C amount of ethylene oxide added to reference
solution (a), in micrograms.
- material: glass or fused silica;
- size: 1 = 30 m, 0 = 0.32 mm; DT x e
- stationary phase: poly(dimethyl)siloxane R (film thickness
1.0 [lm).
Carrier gas: helium for chromatography R or nitrogen for area of the peak due to dioxan in the chromatogram
chromatography R. obtained with the test solution;
Linear velocity: 20 cm/s. DR area of the peak due to dioxan in the chromatogram
obtained with reference solution (a);
Split ratio: 1:20.
Static head-space conditions that may be used: e amount of dioxan added to reference solution (a)
in micrograms.
- equílibration temperature: 70 oC (90 oC for solutions in
dimethylacetamide) ;
- equilibration time: 45 min;
- transfer-line temperature: 75 oC (150 oC for solutions in 0112008:20426
dimethylacetamide) ;
- carríer gas: helium for chromatography R; 2.4.26. N,N-DIMETHYLANILINE
- pressurisation time: 1 min;
METHODA
- injection time: 12 s.
Examine by gas chromatography (2.2.28), using
Temperature: N,N-diethylaniline R as the internal standard.
Time Temperature Internal standard solution. Dissolve 50 mg of
(min) (oC) N,N-diethylaniline R in 4 mL of 0.1 M hydrochloric
Column O- 5 50 acid and dilute to 50 mL with water R. Dilute 1 mL of this
5 - 31 50 ~ 180
solution to 100 mL with water R.
Test solution. Dissolve in a ground-glass-stoppered tube 0.50 g
31 - 32.5 180 ~ 230
of the substance to be examined in 30.0 mL of water R. Add
32.5 - 37.5 230 1.0 mL of the internal standard solution. Adjust the solution
to a temperature of 26-28 oc. Add 1.0 mL of s/rong sodium
Injection port 150
hydroxide solution R and mix until completely dissolved. Add
Detector 250 2.0 mL of trimethylpentane R. Shake for 2 min and allow the
phases to separate. Use the upper layer.
Detection: flame ionisation. Reference solution. Dissolve 50.0 mg of N,N-dimethylaniline R
Injection: a suitable volume, for example 1.0 mL, of the in 4.0 mL of 0.1 M hydrochloric acid and dilute to 50.0 mL
gaseous phase of the test solution and of reference solutions (a) with water R. Dilute 1.0 mL of this solution to 100.0 mL
and (b). Repeat the procedure twice more. with water R. Dilute 1.0 mL of this solution to 30.0 mL with

-
System suitability: reference solution (b):

- resolution: minimum 2.0 between the peaks due to


acetaldehyde and ethylene oxide;
water R. Add 1.0 mL of the internal standard solution and
1.0 mL of strong sodium hydroxide solu/ion R. Add 2.0 mL of
trimethylpentane R. Shake for 2 min and allow the phases to
separate. Use the upper layer.

146 See the information section on general monographs (cover pages)


EUROPEAN PHARMACOPOEIA 8.0 2.4.27. Heavy metals in herbal dmgs and fatty oils

The chromatographic procedure may be carried out using: CA UTION: when using closed high-pressure digestion vessels
and microwave laboratory equipment, be familiar with the
- a fused-silica capillary column 25 m long and
safety and operating instructions given by the manufacturero
0.32 mm in internal diameter coated with cross-linked
polymethylphenylsiloxane R (film thickness 0.52 flm),
- helium for chromatography Ras the carrier gas with a split APPARATUS
ratio 1:20, a column head pressure of 50 kPa and a split The apparatus typically consists of the following:
vent of 20 mLlmin,
- as digestion flasks, polytetrafluoroethylene flasks with a
- a flame-ionisation detector, volume of about 120 mL, fitted with an airtight closure,
- a split-liner consisting of a column about 1 cm a valve to adjust the pressure inside the container and a
long packed with diatomaceous earth for gas polytetrafluoroethylene tube to allow releas e of gas,
chromatography R impregnated with 10 per cent mlm of - a system to make flasks airtight, using the same torsional
poly(dimethyl)siloxane R, force for each of them,
maintaining the temperature of the column at 150 oC for
- a microwave oven, with a magnetron frequency of
5 min, then raising the temperature at arate of 20 oC
2450 MHz, with a selectable output from O to 630 ± 70 W
per min to 275 oC and maintaining it at 275 oC for 3 min and
in 1 per cent increments, a programmable digital computer,
maintaining the temperature of the detector at 300 oC and that
a polytetrafluoroethylene-coated microwave cavity with
of the injection port at 220 oc.
a variable speed exhaust fan, a rotating turntable drive
The retention times are: N,N-dimethylaniline about 3.6 min, system and exhaust tubing to vent fumes,
N,N-diethylaniline about 5.0 mino
- an atomic absorption spectrometer, equipped with
Inject 1 flL of the test solution and 1 flL of the reference hollow-cathode lamps as source of radiation and a
solution. deuterium lamp as background corrector; the system is
fitted with:
METHOD B (a) a graphite furnace as atomisation device for cadmium,
Examined by gas chromatography (2.2.28), using copper, iron, lead, nickel and zinc.
naphthalene R as the internal standard.
(b) an automated continuous-flow hydride vapour
Internal standard solution. Dissolve 50 mg of naphthalene R generation system for arsenic and mercury.
in cyclohexane R and dilute to 50 mL with the same solvent.
Dilute 5 mL of this solution to 100 mL with cyclohexane R.
METHOD
Test solution. To 1.00 g of the substance to be examined
in a ground-glass-stoppered tube add 5 mL of 1 M sodium In case alternative apparatus is used, an adjustment of the
hydroxide and 1.0 mL of the internal standard solution. instrument parameters may be necessary.
Stopper the tube and shake vigorously for 1 mino Centrifuge if Clean all the glassware and laboratory equipment with a
necessary and use the upper layer. 10 giL solution of nitric acid R before use.
Reference solution. To 50.0 mg of N,N-dimethylaniline R add Test solution. In a digestion flask place the prescribed quantity
2 mL of hydrochloric acid R and 20 mL of water R, shake to of the substance to be examined (about 0.50 g of powdered
dissolve and dilute to 50.0 mL with water R. Dilute 5.0 mL drug (1400) (2.9.12) or 0.50 g offatty oil). Add 6 mL of
of this solution to 250.0 mL with water R. To 1.0 mL of the heavy metal-free nitric acid R and 4 mL of heavy metal-free
latter solution in a ground-glass-stoppered tube add 5 mL of hydrochloric acid R. Make the flask airtight.
1 M sodium hydroxide and 1.0 mL of the internal standard
solution. Stopper the tube and shake vigorously for 1 mino Place the digestion flasks in the microwave oven. Carry out
Centrifuge if necessary and use the upper layer. the digestion in 3 steps according to the following programme,
used for 7 flasks each containing the test solution: 80 per cent
The chromatographic procedure may be carried out using:
power for 15 min, 100 per cent power for 5 min, 80 per cent
- a glass column 2 m long and 2 mm in internal diameter power for 20 mino
packed with silanised diatomaceous earth for gas
chromatography R impregnated with 3 per cent mlm of At the end of the cycle allow the flasks to cool in air and to
polymethylphenylsiloxane R, each add 4 mL of heavy metal-free sulfuric acid R. Repeat
the digestion programme. After cooling in air, open each
- nitrogen for chromatography R as the carrier gas at a flow digestion flask and introduce the clear, colourless solution
rate of 30 mLlmin, obtained into a 50 mL volumetric flask. Rinse each digestion
- a flame-ionisation detector, flask with 2 quantities, each of 15 mL, of water R and collect
the rinsings in the volumetric flask. Add 1.0 mL of a 10 giL
maintaining the temperature of the column at 120 oC and that solution of magnesium nitrate R and 1.0 mL of a 100 giL
of the injection port and of the detector at 150 oc. solution of ammonium dihydrogen phosphate R and dilute to
Inject 1 flL of the test solution and 1 flL of the reference 50.0 mL with water R.
solution. Blank solution. Mix 6 mL of heavy metal-free nitric acid R and
4 mL of heavy metal-free hydrochloric acid R in a digestion
flask. Carry out the digestion in the same manner as for the
test solution.
0112008:20427 CADMIUM, COPPER, IRON, LEAD, NICKEL AND ZINC
Measure the content of cadmium, copper, iron, lead, nickel
and zinc by the standard additions method (2.2.23, Method JI),
2.4.27. HEAVY METALS IN HERBAL using reference solutions of each heavy metal and the
instrumental parameters described in Table 2.4.27.-1.
DRUGS AND FATTY OILS
The absorbance value of the blank solution is automatically
Examine by atomic absorption spectrometry (2.2.23). subtracted from the value obtained with the test solution.

General Notices (1) apply to all monographs and other texts 147
2.4.28. 2- Ethylhexanoic add EUROPEAN PHARMACOPOEIA 8.0

Table 2.4.27.-1 Test solution. To 0.300 g of the substance to be examined,


Cd Cn Fe Ni Pb Zn add 4.0 mL of a 33 per cent V/V solution of hydrochloric
HIn
acid R. Shake vigorously for 1 min with 1.0 mL of the internal
Wavelength 228.8 324.8 248.3 232 283.5 213.9
standard solution. Allow the phases to separate (if necessary,
Slit width nm 0.5 0.5 0.2 0.2 0.5 0.5 centrifuge for a better separation). Use the upper layer.
Lamp mA 6 7 5 10 7 Reference solution. Dissolve 75.0 mg of 2-ethylhexanoic acid R
current in the internal standard solution and dilute to 50.0 mL with the
Ignition oC 800 800 800 800 800 800
same solution. To 1.0 mL of the solution add 4.0 mL of a 33 per
temperature cent V/V solution of hydrochloric acid R. Shake vigorously for
1 mino Allow the phases to separate (if necessary, centrifuge
Atomisation oC 1800 2300 2300 2500 2200 2000
temperature
for a better separation). Use the upper layer.
on The chromatographic procedure may be carried out using:
Background off off off off off
corrector - a wide-bore fused-silica column 10 m long and 0.53 mm
Nitrogen L/min 3 3 3
in internal diameter coated with macrogol 20 000
flow 2-nitroterephthalate R (film thickness 1.0 ¡..tm),
helium for chromatography R as the carrier gas at a flow
ARSENIC AND MERCURY
rate of10 mLlmin,
Measure the content of arsenic and mercury in comparison
with the reference solutions of arsenic or mercury at a known - a flame- ionisation detector,
concentration by direct calibration (2.2.23, Method I) using an with the following temperature programme:
automated continuous-flow hydride vapour generation system. Time Temperature Rate Comment
The absorbance value of the blank solution is automatically
subtracted from the value obtained with the test solution.
(min) re) ¡OC/min)
Column 0-2 40 isothermal
Arsenic 2 - 7.3 40 -7 200 30 linear gradient
Sample solution. To 19.0 mL of the test solution or of the blank
solution as prescribed aboye, add 1 mL of a 200 giL solution 7.3 - 10.3 200 isothermal
of potassium iodide R. Allow the test solution to stand at room Injection port 200
temperature for about 50 min or at 70 oC for about 4 mino
Detector 300
Acid reagent. Heavy metal-free hydroehlorie acid R.
Redueing reagent. A 6 giL solution of sodium Inject 1 ¡..tL of the test solution and 1 ¡..tL of the reference
tetrahydroborate R in a 5 giL solution of sodium solution.
hydroxide R.
The test is not valid unless the resolution between the peaks
The instrumental parameters in Table 2.4.27.-2 may be used. due to 2-ethylhexanoic acid (first peak) and the internal
Mercury standard is at least 2.0.
Sample solution. Test solution or blank solution, as prescribed Calculate the percentage content of 2-ethylhexanoic acid from
aboye. the expression:
Acid reagent. A 515 giL solution of heavy metal-free AT X IR x mR x 2
hydrochloric acid R.
AR X IT x mT
Reducing reagent. A 10 giL solution of stannous ehloride R in
heavy metal-free dilute hydroehloric acid R. Ay area of the peak due to 2-ethylhexanoic acid in the
The instrumental parameters in Table 2.4.27.-2 may be used. chromatogram obtained with the test solution,
Table 2.4.27.-2 AR area of the peak dne to 2-ethylhexanoic acid in
As Hg the chromatogram obtained with the reference
solution,
Wavelength nm 193.7 253.7
IT are a of the peak due to the internal standard in the
SUt width nm 0.2 0.5 chromatogram obtained with the test solution,
Lamp current mA 10 4 area of the peak due to the internal standard in
the chromatogram obtained with the reference
Acid reagent flow mL/min 1.0 1.0
rate solution,
Reducing reagent mL/min 1.0 1.0 mass of the substance to be examined in the test
flow rate solution, in grams,
Sample solution flow mL/min 7.0 7.0
mass of 2-ethylhexanoic acid in the reference
rate
solution, in grams.
Absorption cel! Quartz (heated) Quartz
(unheated)
Background off off
corrector
Nitrogen flow rate L/min 0.1 0.1 07/2010:20429

0112008:20428
2.4.29. COMPOSITION OF FATTY
ACIDS IN OILS RICH IN OMEGA-3
2.4.28. 2-ETHYLHEXANOIC ACID ACIDS
Examine by gas chromatography (2.2.28), using The assay may be used for quantitative determination of the
3-cyc/ohexylpropionic aeid R as the internal standard. EPA and DHA content in omega-3-containing products of
Internal standard solution. Dissolve 100 mg of fish oil in different concentrations. The method is applicable
3-cyclohexylpropionic acid R in cyclohexane R and dilute to to triglycerides or ethyl esters and the results are expressed as
100 mL with the same solvento triglycerides or ethyl esters, respective/y.

148 See the information section on general monographs (cover pages)


EUROPEAN PHARMACOPOEIA 8.0 2.4.29. Composition of fatty adds in oils rkh in omega-3 adds

EPAANDDHA Referenee solution (e). Into a 10 mL volumetric flask dissolve


Gas chromatography (2.2.28). Carry out the operations as a sample containing about 55.0 mg of doeosahexaenoie acid
rapidly as possible, avoiding exposure to actinie light, oxidising methyl ester R and about 5.0 mg of tetracos-15-enoie acid
agents, oxidation catalysts (jor example, copper and iron) and methyl ester R in a 50 mg/L solution of butylhydroxytaluene R
airo in trimethylpentane R and dilute to 10.0 mL with the same
The assay is carried out on the methyl or ethyl esters of solution.
(all-Z)-eicosa-5,8,1l,14,17-pentaenoic acid (EPA; 20:5 n-3) Column:
and (all-Z)-docosa-4,7,10,13,16,19-hexaenoic acid (DHA; - material: fused silica;
22:6 n-3) in the substance to be examined. - dimensions: 1 = at least 25 m, 0 = 0.25 mm;
Internal standard. Methyl tricasanoate R. - stationary phase: bonded maerogol20 000 R (film thickness
Test solution (a) 0.2 ¡.tm).
A. Dissolve the mass of sample to be examined according Carrier gas: hydrogen for ehromatography R or helium for
to Table 2.4.29.-1 and about 70.0 mg of the internal ehromatography R.
standard in a 50 mg/L solution of butylhydroxytoluene R in Flow rate: 1 mL/min.
trimethylpentane R and dilute to 10.0 mL with the same
Split ratio: 1:200, alternatively splitless with temperature
solution. Gentle heating (up to 60 OC) may be applied to
control (sample solutions need to be diluted 1/200
dissolve the internal standard.
with a 50 mg/L solution of butylhydroxytoluene R in
Table 2.4.29.-1. trimethylpentane R before injection).
Approximate sum EPA + DHA Mass of sample to be examined Temperature:
(per cent) (g)
Time Temperature
30 - 50 0.4 - 0.5 (min) (oC)
50 - 70 0.3 Column 0-2 170

70 - 90 0.25 2 - 25.7 170 -7 240

25.7 - 28 240
Ethyl esters are now ready for analysis. For triglycerides
continue as described in step B. Injection port 250
B. Introduce 2.0 mL of the solution obtained in step A into Detector 270
a quartz tube and evaporate the solvent with a gentle
current of nitrogen R. Add 1.5 mL of a 20 giL solution of Deteetion: flame ionisation.
sodium hydroxide R in methanol R, cover with nitrogen R, Injeetion: 1 ¡.tL, twice.
cap tightly with a polytetrafluoroethylene-lined cap, mix
and heat on a water-bath for 7 mino Allow to coo!. Add System suitability:
2 mL of baran trichlaride-methanol solutian R, cover with - in the chromatogram obtained with reference solution (b),
nitrogen R, cap tightly, mix and heat on a water-bath for the area per cent composition increases in the following
30 mino Cool to 40-50 oC, add 1 mL of trimethylpentane R, order: methyl palmitate, methyl stearate, methyl arachidate,
cap and shake vigorously for at least 30 S. Immediately methyl behenate; the difference between the percentage
add 5 mL of a saturated sodium ehloride solution R, cover area of methyl palmitate and that of methyl behenate is less
with nitrogen R, cap and shake thoroughly for at least 15 S. than 2.0 area per cent units;
Transfer the upper layer to a separate tube. Shake the - resolution: minimum of 1.2 between the peaks due to
methanollayer once more with 1 mL of trimethylpentane R. docosahexaenoic acid methyl ester and to tetracos-15-enoic
Wash the combined trimethylpentane extracts with 2 acid methyl ester in the chromatogram obtained with
quantities, each of 1 mL, of water R and dry over anhydrous reference solution (c);
sodium sulfate R. Prepare 3 solutions for each sample. - in the chromatogram obtained with test solution (a),
Test solution (b). Dissolve 0.300 g of the sample to be the peaks due to methyl tricosanoate and any
examined in a 50 mg/L solution of butylhydroxytoluene R heneicosapentaenoic acid methyl ester or ethyl ester
in trimethylpentane R and dilute to 10.0 mL with the same (C21:5) present when compared with the chromatogram
solution. Proceed as described for test solution (a). obtained with test solution (b) are clearly separated (if not,
Referenee solution (aJ Dissolve about 70.0 mg of the a correction factor has to be used).
internal standard and 90.0 mg of eicosapentaenoie acid ethyl Calculate the percentage content of EPA and DHA using the
ester CRS in a 50 mg/L solution of butylhydroxytoluene R following express ion and taking into account the assigned
in trimethylpentane R and dilute to 10.0 mL with the same value of the reference substances:
solution. Gentle heating (up to 60 OC) may be applied to
dissolve the internal standard. Ax x A x ,3 x mI x mx,r x _1_ x e x 100
mx.3 Al Ax.r m2
Referenee solution (a 2 ). Dissolve 60.0 mg of doeosahexaenoie
acid ethyl ester CRS and about 70.0 mg of the internal
standard in a 50 mg/L solution of butylhydroxytoluene R mI mass of the internal standard in test solution (a),
in trimethylpentane R and dilute to 10.0 mL with the same in milligrams;
solution. Gentle heating (up to 60 OC) may be applied to
dissolve the internal standard. m2 mass of the sample to be examined in test
solution (a), in milligrams;
For both reference solution (al) and reference solution (a2 )
proceed as described for test solution (a) step A when mX.3 mass of the internal standard in reference
analysing ethyl esters. For analysis of triglycerides, continue solution (al) (EPA determination), or in
with step B in the same manner as for test solution (a) and reference solution (a 2) (DHA determination),
prepare 3 solutions for each sample. in milligrams;
Reference solution (b). lnto a 10 mL volumetric flask dissolve
0.3 g of methyl araehidate R, 0.3 g of methyl behenate R, 0.3 g of mv mass of eicosapentaenaic aeid ethyl es ter CRS
methyl palmitate R and 0.3 g of methyl stearate R in a 50 mg/L in reference solution (al) or docosahexaenaie
solution of butylhydroxytoluene R in trimethylpentane R and acid ethyl ester CRS in reference solution (a 2),
dilute to 10.0 mL with the same solution. in milligrams;

General Notices (1) apply to all monographs and ather texts 149
2.4.30. Ethylene glycol and diethylene glycol in ethoxylated suostances EUROPEAN PHARMACOPOEIA 8.0

are a of the peak due to eicosapentaenoic acid Reference solution (a). Mix 30.0 mg of ethy/ene glyeal R with
ester or docosahexaenoic acid ester in the acetone R and dilute to 100.0 mL with the same solvent. Dilute
chromatogram obtained with test solution (a); 1.0 mL to 10.0 mL with the internal standard solution.
Reference solution (b). Prepare a solution of diethylene glyeal R
are a of the peak due to eicosapentaenoic acid with a concentration corresponding to the prescribed limit
ester in the chromatogram obtained with and using the same solvents as for the preparation of reference
reference solution (aJ or to docosahexaenoic solution (a).
acid ester in the chromatogram obtained with
Column:
reference solution (a 2);
materia/: fused silica,
are a of the peak due to the internal standard - size: 1 = 30 m, 0 = 0.53 mm,
in the chromatogram obtained with test
- stationary phase: macrogol20 000 R (film thickness 1 [lm).
solution (a);
Carrier gas: he/ium for chromatography R.
area of the peak due to the internal standard Flow rate: 10 mLlmin.
in the chromatogram obtained with reference
solution (al) (EPA determination) or with Sp/it ratio: 1:3.
reference solution (a2 ) (DHA determination); Temperature:
Time Temperature
C conversion factor between ethyl ester and
(min) (oC)
triglycerides,
Column 0-40 80 -7 200
C = 1.00 for ethyl esters, 40 - 45 200 -7 230
C = 0.954 for EPA, 45 - 65 230

C = 0.957 for DHA. Injection port 250

Detector 250
TOTAL OMEGA-3 ACIDS
From the assay for EPA and DHA, calculate the Detection: flame ionisation.
percentage content of the total omega-3 acids using the Injection: 2 [lL.
following expression and identifying the peaks from the
Relative retention with reference to 1,2-pentanediol (retention
chromatograms:
time = about 19 min): ethylene glycol = about 0.7; diethylene
EPA+DHA+ An-3 (EPA+DHA) glycol = about l.3.
A EPA + ADRA

EPA percentage content of EPA; 01/2008:20431


DHA percentage content of DHA;
A n _) sum ofthe are as ofthe peaks due to C18:3 n-3,
2.4.31. NICKEL IN HYDROGENATED
C18:4 n-3, C20:4 n-3, C21:5 n-3 and C22:5 n-3 VEGETABLE OILS
esters in the chromatogram obtained with test
Atomic absorption spectrometry (2.2.23, Method I).
solution (b);
CAUTION: when using closed high-pressure digestion vessels
A EPA area of the peak due to EPA ester in the
and microwave laboratory ovens, be familiar with the safety
chromatogram obtained with test solution (b) ;
and operating instructions given by the manufacturero
AmiA area of the peak due to DHA ester in the The reagents magnesium nitrate R and ammonium dihydrogen
chromatogram obtained with test solution (b). phosphate R must be eantrolled for nickel before use. The actual
nickel content is taken into aceaunt in the ca/cu/ation of the
nickel content of the sample.
0112008:20430 Test so/ution. Weigh 0.250 g (m) ofthe substance to be
examined into a suitable high-pressure-resistant digestion
2.4.30. ETHYLENE GLYCOL vessel (fluoropolymer or quartz glass), add 6.0 mL of
nicke/-free nitric acid R and 2.0 mL of strong hydrogen peroxide
AND DIETHYLENE GLYCOL IN solution R. Prepare a blank solution in the same manner.
ETHOXYLATED SUBSTANCES Place the closed vessels in a laboratory microwave oven and
digest with an appropriate programme, e.g. 1000 W for
Ethoxylated substances may contain varied amounts of 40 mino Allow the digestion vessels to cool before opening.
ethylene glycol and diethylene glyeal, as a resu/t of the Add 2.0 mL of strong hydrogen peroxide solution R and repeat
manufacturing process. The following method may be used for the digestion step. Allow the digestion vessels to cool before
the quantitative determination of these substances, in particular opening. Quantitatively transfer to a 25 mL flask, add 0.5 mL
in the case of the following surfactants: macrogolg/ycero/ of a 10 giL solution of magnesium nitrate R and 0.5 mL of a
ricinoleate, macrogolglycerol hydroxystearate, macrogo/lS 100 giL solution of ammonium dihydrogen phosphate R, dilute
hydroxystearate, nonoxinol9 and macrogo/ cetosteary/ ether. to 25.0 mL with water for chromatography R and mix.
Reference solutions. lnto 4 volumetric flasks, introduce
Gas chromatography (2.2.28).
25 IiL, 50 [lL, 75 [lL and 100 [lL of nicke/ standard so/ution
Interna/ standard so/ution. Dissolve 30.0 mg of (5 ppm Ni) R. To each flask, add 0.5 mL of a 10 giL solution
1,2-pentanediol R in acetone R and dilute to 30.0 mL with the of magnesium nitrate R, 0.5 mL of a 100 giL solution
same solvent. Dilute 1.0 mL of this solution to 20.0 mL with of ammonium dihydrogen phosphate R and 6.0 mL of
acetone R. nickel-free nitric acid R and dilute to 25.0 mL with water
Test solution. Dissolve 0.500 g of the substance to be examined for chromatography R. Mix to obtain reference solutions
in the internal standard solution and dilute to 10.0 mL with containing respectively 5 ng/mL, 10 ng/mL, 15 ng/mL and
the same solution. 20 ng/mL (ppb) of nickel.

150 See the information section 011 general monographs (eaver pages)
EUROPEAN PHARMACOPOEIA 8.0 2.4.32. Total cholesterol in oils rich in omega-3 adds

Zero so/ution. In a volumetric flask, introduce 1.0 mL of a carefully a fixed number of times, e.g. 60 times. Discard the
10 giL solution of magnesíum nitrate R, 1.0 mL of a 100 giL aqueous phase, add 5 mL of a 3 per cent solution of potassium
solution of ammonium dihydrogen phosphate R and 12.0 mL hydroxide R to the ether phase and mix carefully 20 times.
of nickel-free nitric acid R. Dilute to 50.0 mL with water for Discard the aqueous phase, add another 5 mL of distilled
chromatography R and mix. water R and mix carefully a further 20 times. Transfer the
Method. Determine the absorbance of each solution ether phase into a small centrifuge tube, avoiding any transfer
at 232.0 nm using a suitable graphite furnace atomic of water. lf an emulsion forms during the process, add a small
absorption spectrometer equipped with a background amount of sodium chloride R to get a separation of the phases.
compensation system, a pyrolytically-coated tube, and a nickel Evaporate to dryness under a gentle stream of nitro gen
hollow-cathode lampo Maintain the drying temperature of with careful heating. Dissolve the residue in 600 ¡.tL of ethyl
the furnace at 120 oC for 35 s after a 5 s ramp, the ashing acetate R.
temperature at 1100 oC for 10 s after a 30 s ramp, the cooling Depending on the expected cholesterol content in the oi!, the
temperature at 800 oC for 5 s after a 5 s decrease, and the solution is further diluted as follows:
atomisation temperature at 2600 oC for 7 S. Use the zero - content less than 3 mgl g: dilute 200 ¡.tL of the solution to
solution to set the instrument to zero. Using the calibration 2.0 mL with ethyl acetate R;
curve, determine the concentrations of the test solution and
the blank solution from the corresponding absorptions. lf - content greater than or equal to 3 mgl g: dilute 20 ¡.tL of the
necessary, dilute with the zero solution to obtain a reading solution to 2.0 mL with ethyl acetate R.
within the ci:tlibrated absorbance range. Reference solution (a). Transfer 1.0 mL of the interna!
Calculate the content of Ni in micrograms per gram (ppm) standard stock/working solution and 1.0 mL of the cholesterol
using the following expression: stock/working solution, depending on the expected cholesterol
content in the oil (see Table 2.4.32.-1), to a 15 mL quartz tube
ex! and continue as described for the test solution, starting with
m x 40 "Evaporate to dryness on a heating block..:'.
Reference solution (b). Mix 1.0 mL of the internal standard
e measured concentration of Ni, in nanograms per stock solution, 1.0 mL of the cholesterol stock solution
millilitre; and 2.0 mL of the a-tocopherol solution in a suitable flask.
f dilution factor of the test solution; Evaporate to dryness under a gentle stream of nitro gen with
careful heating. Dissolve the residue in ethyl aeetate R and
m mass of the substance to be examined, in grams. dilute to 50.0 mL with the same solvent. Dilute 1.0 mL of this
solution to 10.0 mL with ethyl acetate R. The solution may be
stored in a deep-freezer for up to 6 months.
0712010:20432
Table 2.4.32. -l. - Preparatíon af the test and referenee solutíans
Reference Reference
2.4.32. TOTAL CHOLESTEROL IN OILS Test solution
solution (a) solution (b)
RICH IN OMEGA-3 ACIDS les s greater less greater
than than or than than or
This method may be used for the quantitative determinatíon of 3mg/g equal to 3 mg/g equal to
the sum offree and esterified cho/esterol in products offish oils 3~mg/g 3 mg/g
rieh in omega-3 acids (as ethyl esters or triglycerides). Internal
standard + + +
Gas chromatography (2.2.28). stock solution
Internal standard stock so/ution. Dissolve 0.15 g of Internal
(5a)-cholestane R in heptane R and dilute to 50.0 mL with the standard
+ +
working
same solvent. solution
Internal standard working solution. Prepare the solution Cholesterol
immediately before use. Dilute 1.0 mL of the internal standard + +
stock solution
stock solution to 10.0 mL with heptane R. Cholesterol
Cholesterol stock solution. Dissolve 30.0 mg of cho/esterol R in working +
heptane R and dilute to 10.0 mL with the same solvento The solution
solution is dispensed into gas chromatography vials and may a -Tocopherol
+
be sto red in a deep-freezer for up to 6 months. solution
Cholesterol working solution. Prepare the solution immediately Column:
before use. Dilute 1.0 mL of the cholesterol stock solution to - size: 1 = 30 m, el = 0.25 mm (film thickness 0.25 flm);
10.0 mL with heptane R.
- stationary phase: po/y(dimethyl)(diphenyl)siloxane R.
a- Tocopherol solution. Dilute 15.0 mg of a-tacophero/ CRS to
10.0 mL with heptane R. Carrier gas: helium for chromatography R.
Test solution. Weigh 0.100 g ofthe substance to be examined Flow rate: 1.3 mLlmin.
into a 15 mL quartz tube. Add 1.0 mL of the internal standard Temperature:
stock/working solution, depending on the expected cholesterol Time Temperature
content in the oil (see Table 2.4.32.-1). (min) ("e)
Evaporate to dryness on a heating block at 50 oC under a gentle Column o~ 1 170
stream of nitrogen, while mixing. Add 0.5 mL of a 50 per 1 - 38 170 -7 320
cent solution of potassium hydroxide R and 3.0 mL of ethanol
(96 per cent) R. Fill the tube with nitrogen R and cap. Further 38 ~ 40 320
heat on the heating block at 100 oC for 1 h with stirring. Cool
Injection port 320
for about 10 min and add 6 mL of distilled water R. Extract
with 4 quantities, each of 2.5 mL, of ether R, mixing each time Detector 300
for 1 min using a vortex mixer. Transfer the ether phase to
a large centrifuge tube or a separating funnel and wash the Deteetían: flame ionisation.
combined extracts with 5 mL of distilled water R, mixing Injection: 1 ¡.tL.

General Notices (1) apply to all monographs and other texts 151
2.4.32. Total cholesterol in oils rich in omega-3 adds EUROPEAN PHARMACOPOEIA 8.0

System suitability: reference solution (b): mass of (5a)-cholestane in the internal standard
- resolution: minimum 1.2 between the peaks due to stock solution, in grams;
cholesterol and a-tocopherol. F 20 for oils with an expected cholesterol content
Calculate the content of total cholesterol, expressed as greater than or equal to 3 mg/g; 2 for oils
milligrams of cholesterol per gram of oil, using the following with an expected cholesterol content les s than
expression: 3 mg/g;
R response factor.
Calculate the response factor R using the following expression:

area of the peak due to cholesterol in the A3 x m2


chromatogram obtained with the test solution; A4 x m3 x 5
area of the peak due to (5a)-cholestane in the
are a of the peak due to cholesterol in the
chromatogram obtained with the test solution;
chromatogram obtained with reference
mass of the substance to be examined in the solution (a);
test solution, in grams; area of the peak due to (5a)-cholestane in
the chromatogram obtained with reference
solution (a);
m3 mass of cholesterol in the cholesterol stock
solution, in grams.

152 See the information section on general monographs (caver pages)


EUROPEAN PHARMACOPOEIA 8.0

2.5. Assays ................................................................................. 155 2.5.21. Methylpentoses in polysaccharide vaccines .............. 161
2.5.1. Acid value ........................................................................ 155 2.5.22. Uronic acids in polysaccharide vaccines ................... 161
2.5.2. Ester value ....................................................................... 155 2.5.23. Sialic acid in polysaccharide vaccines ....................... 161
2.5.3. Hydroxyl value ............................................................... 155 2.5.24. Carbon dioxide in gases .............................................. 161
2.5.4. Iodine value .................................................................... 155 2.5.25. Carbon monoxide in gases ......................................... 162
2.5.5. Peroxide value ................................................................. 156 2.5.26. Nitrogen monoxide and nitro gen dioxide in gases .. 163
2.5.6. Saponification value ....................................................... 157 2.5.27. Oxygen in gases ............................................................ 163
2.5.7. Unsaponifiable matter.. .................................................. 157 2.5.28. Water in gases ............................................................... 163
2.5.8. Determination of primary aromatic amino- 2.5.29. Sulfur dioxide ............................................................... 164
nitrogen .................................................................................... 157 2.5.30. Oxidising substances ................................................... 164
2.5.9. Determination of nitrogen by sulfuric acid 2.5.31. Ribose in polysaccharide vaccines ............................. 164
digestion .................................................................................. 157 2.5.32. Water: micro determination ....................................... 164
2.5.10. Oxygen-flask method .................................................. 158 2.5.33. Total protein ................................................................. 165
2.5.11. Complexometric titrations .......................................... 158 2.5.34. Acetic acid in synthetic peptides ................................ 168
2.5.12. Water: semi-micro determination ............................. 158 2.5.35. Nitrous oxide in gases ................................................. 168
2.5.13. Aluminium in adsorbed vaccines .............................. 159 2.5.36. Anisidine value ............................................................. 169
2.5.14. Calcium in adsorbed vaccines .................................... 159 2.5.37. Methyl, ethyl and isopropyl methanesulfonate in
2.5.15. Phenol in immunosera and vaccines ......................... 159 methanesulfonic acid ............................................................. 169
2.5.16. Protein in polysaccharide vaccines ............................ 159 2.5.38. Methyl, ethyl and isopropyl methanesulfonate in active
2.5.17. Nucleic acids in polysaccharide vaccines .................. 160 substances ................................................................................ 170
2.5.18. Phosphorus in polysaccharide vaccines .................... 160 2.5.39. Methanesulfonyl chloride in methanesulfonic
2.5.19. O-Acetyl in polysaccharide vaccines ......................... 160 acid ........................................................................................... 171
2.5.20. Hexosamines in polysaccharide vaccines .................. 160

General Notices (1) apply to all monographs and other texts 153
EUROPEAN PHARMACOPOEIA 8.0

154 See the information section on general monographs (caver pages)


EUROPEAN PHARMACOPOEIA 8.0 2.5.4. Iodine value

2.5. ASSAYS appears add sufficient pyridine R to clear it, noting the volume
added. Shake the flask and replace in the water-bath for
0112008:20501 10 mino Withdraw the flask and allow to cool. Rinse the
condenser and the walls of the flask with 5 mL of alcohol R,
previously neutralised to phenolphthalein solution Rl. Titrate
2.5,1. ACID VALUE with 0.5 M alcoholic potassium hydroxide using 0.2 mL of
The acid value lA is the number that expresses, in milligrams phenolphthalein solution Rl as indicator (nI mL of 0.5 M
the quantity of potassium hydroxide required to neutralise the alcoho/ic potassium hydroxide). Carry out a blank test under
free acids present in 1 g of the substance. the same conditions (n 2 mL of 0.5 M alcoholic potassium
Dissolve 10.00 g of the substance to be examined, or the hydroxide).
quantity prescribed, (m g), in 50 mL of a mixture of equal nl) f
f OH = 28.05 (n2 -
+ A
volumes of ethanol (96 per cent) R and light petroleum R3, m
previously neutralised with 0.1 M potassium hydroxide
or 0.1 M sodium hydroxide, unless otherwise specified, METHODB
using 0.5 mL of phenolphthalein solution R1 as indicator. If Introduce the prescribed quantity of the substance to be
necessary, heat to about 90 oC to dissolve the substance to be examined (m g) into a perfectly dry 5 mL conical flask fitted
examined. When the substance to be examined has dissolved, with a ground-glass or suitable plastic stopper and add 2.0 mL
titrate with 0.1 M potassium hydroxide or 0.1 M sodium of propionic anhydride reagent R. Close the flask and shake
hydroxide until the pink colour persists for at least 15 s (n mL gently to dissolve the substance. Allow to stand for 2 h unless
of titrant). When heating has been applied to aid dissolution, otherwise prescribed. Remove the stopper and transfer the
maintain the temperature at about 90 oC during the titration. flask and its contents into a wide-mouthed 500 mL conical
lA = 5.610n flask containing 25.0 mL of a 9 giL solution of aniline R in
m cyclohexane R and 30 mL of glacial acetic acid R. Swirl the
contents of the flask, allow to stand for 5 min, add 0.05 mL
of crystal violet solution R and titrate with 0.1 M perchloric
0112008:20502
acid until an emerald-green colour is obtained (nI mL of
0.1 M perchloric acid). Carry out a blank test under the same
2.5.2. ESTER VALUE conditions (n 2 mL of 0.1 M perchloric acid).
The es ter value lE is the number that expresses in milligrams fOH = 5.610 (nl - n2)
the quantity of potassium hydroxide required to saponify the m
esters present in 1 g of the substance. It is calculated from the
saponification value ls and the acid value lA: To take account of any water present, determine this (y per
cent) by the semi-micro determination of water (2.5.12).
fE = f8 - fA The hydroxyl value is then given by the equation:

fO H = (hydroxyI value as determined) - 31.1y


01/2008:20503

2.5.3. HYDROXYL VALUE 01/2008:20504

The hydroxyl value lOH is the number that expresses in


milligrams the quantity of potassium hydroxide required
2.5.4. IODINE VALUE
to neutralise the acid combined by acylation in 1 g of the The iodine value 1] is the number that expresses in grams the
substance. quantity of halogen, calculated as iodine, that can be fixed in
the prescribed conditions by 100 g of the substance.
METHODA
When the monograph does not specify the method to be used,
Introduce the quantity of the substance to be examined method A is applied. Any change from method A to method B
shown in Table 2.5.3.-1 (m g) into a 150 mL acetylation is validated.
flask fitted with an air condenser, unless another quantity
is prescribed in the monograph. Add the quantity of acetic METHODA
anhydride solution R1 stated in Table 2.5.3.-1 and attach the Unless otherwise prescribed, use the following quantities
air condenser. (Table 2.5.4.-1) for the determination.
Table 2.5.3.-1 Table 2.5.4.-1
Presumed value IOH Quautity of sample Volume of Presumed value Ir Quantity of sample (g)
(g) acetylatiug
reagent (mL) less than 20 1.0
10 - 100 2.0 5.0
20 - 60 0.5 - 0.25
100 - 150 1.5 5.0
60 - 100 0.25 - 0.15
150 - 200 1.0 5.0
more than 100 0.15 - 0.10
200 - 250 0.75 5.0
Introduce the prescribed quantity of the substance to be
250 - 300 0.60 or 1.20 5.0 or 10.0 examined (m g) into a 250 mL flask fitted with a ground-glass
300 - 350 1.0 10.0 stopper and previously dried or rinsed with glacial acetic
acid R, and dissolve it in 15 mL of chloroform R unless
350 - 700 0.75 15.0 otherwise prescribed. Add very slowly 25.0 mL of iodine
700 - 950 0.5 15.0 bromide solution R. Close the flask and keep it in the dark for
30 min unless otherwise prescribed, shaking frequently. Add
Heat the flask in a water-bath for 1 h keeping the level of the 10 mL of a 100 giL solution of potassium iodide R and 100 mL
water about 2.5 cm aboye the level of the liquid in the flask. of water R. Titrate with 0.1 M sodium thiosulfate, shaking
Withdraw the flask and allow to cool. Add 5 mL of water R vigorously until the yellow colour is almost discharged. Add
through the upper end of the condenser. If a cloudiness 5 mL of starch solution R and continue the titration adding

General Notices (1) apply to all monographs and other texts 155
2.5.5. Peroxide value EUROPEAN PHARMACOPOEIA 8.0

the 0.1 M sodium thiosulfate dropwise until the colour is When the monograph does not speeify the method to be used,
discharged (n¡ mL of 0.1 M sodium thiosulfate). Carry out a method A is applied. Any ehange from method A to method B
blank test under the same conditions (n 2 mL of 0.1 M sodium is validated.
thiosulfate).

METHODA
m
Place 5.00 g of the substance to be examined (m g) in a 250 mL
METHOD B conical flask fitted with a ground-glass stopper. Add 30 mL
Unless otherwise prescribed, use the following quantities of a mixture of 2 volumes of chloroform R and 3 volumes of
(Table 2.5.4.-2) for the determination. glacial acetic acid R. Shake to dissolve the substance and add
0.5 mL of saturated potassium iodide solution R. Shake for
Table 2.5.4.-2
exactly 1 min then add 30 mL of water R. Titrate with 0.01 M
Presumed Mass (g) Mass (g) Iodine sodium thiosulfate, adding the titrant slowly with continuous
value 11 (corresponding (corresponding chloride vigorous shaking, until the yellow colour is almost discharged.
to an excess of to an excess of solution (mL)
150 per cent 100 per centICl) Add 5 mL of starch solution R and continue the titration,
ICI) shaking vigorously, until the colour is discharged (n¡ mL of
<3 10 10 25 0.01 M sodium thiosulfate). Carry out a blank test under the
same conditions (n 2 mL of 0.01 M sodium thiosulfate). The
8.4613 10.5760 25
volume of 0.01 M sodium thiosulfate used in the blank titration
5 5.0770 6.3460 25 must not exceed 0.1 mL.
10 2.5384 3.1730 20

20 0.8461 1.5865 20

40 0.6346 0.7935 20

60 0.4321 0.5288 20 METHOD B


80 0.3173 0.3966 20
Carry out the operations avoiding exposure to aetinic light.
100 0.2538 0.3173 20
Place 50 mL of a mixture of 2 volumes of trimethylpentane R
120 0.2115 0.2644 20 and 3 volumes of glacial aeetic aeid R in a conical flask and
140 0.1813 0.2266 20 replace the stopper. Swirl the flask until the substance to be
examined (m g; see Table 2.5.5.-1) has dissolved. Using a
160 0.1587 0.1983 20 suitable volumetric pipette, add 0.5 mL of saturated potassium
180 0.1410 0.1762 20 iodide solution R and replace the stopper. Allow the solution
to stand for 60 ± 1 s, thoroughly shaking the solution
200 0.1269 0.1586 20 continuously, then add 30 mL of water R.
The mass of the sample is such that there will be an excess of
Table 2.5.5.-1
iodine ehloride solution R of 50 per cent to 60 per cent of the
amount added, i.e. 100 per cent to 150 per cent of the amount Expected peroxide Mass of substance
absorbed. to be examined (g)
value Ir
Introduce the prescribed quantity of the substance to be
O lo 12 2.00 lo 5.00
examined (m g) into a 250 mL flask fitted with a ground-glass
stopper and previously rinsed with glacial acetie acid R or 12 lo 20 1.20 lo 2.00
dried, and dissolve it in 15 mL of a mixture of equal volumes
20 lo 30 0.80 lo 1.20
of cyclohexane R and glacial aeetic acid R, unless otherwise
prescribed. If necessary, melt the substance before dissolution 30 lo 50 0.500 lo 0.800
(melting point greater than 50 OC). Add very slowly the
50 lo 90 0.300 lo 0.500
volume of iodine ehloride solution R stated in Table 2.5.4.-2.
Close the flask and keep it in the dark for 30 min, unless
otherwise prescribed, shaking frequently. Add 10 mL of a Titrate the solution with 0.01 M sodium thiosulfate (V¡ mL),
100 giL solution of potassium iodide R and 100 mL of water R. adding it gradually and with constant, vigorous shaking, until
Titrate with 0.1 M sodium thiosulfate, shaking vigorously until the yellow iodine colour has almost disappeared. Add about
the yellow colour is almost discharged. Add 5 mL of stareh 0.5 mL of stareh solution Rl and continue the titration, with
solution R and continue the titration adding the 0.1 M sodium constant shaking especially near the end-point, to liberate all of
thiosulfate dropwise until the colour is discharged (n¡ mL of the iodine from the solvent layer. Add the sodium thiosulfate
0.1 M sodium thiosulfate), Carry out a blank test under the solution dropwise until the bIue colour just disappears.
same conditions (n 2 mL of 0.1 M sodium thiosulfate).
Depending on the volume of 0.01 M sodium thiosulfate used,
1.269 (n2 - nI) it may be necessary to titrate with 0.1 M sodium thiosulfate.
h=---'----'-
m
NOTE: there is a 15 s to 30 s delay in neutralising the starch
indicator for peroxide values of 70 and greater, due to the
tendency of trimethylpentane to float on the surface of the
0112008:20505 aqueous medium and the time necessary to adequately mix the
solvent and the aqueous titrant, thus liberating the Iast traces
2.5.5. PEROXIDE VALUE of iodine. It is recommended to use 0.1 M sodium thiosulfate
for peroxide values greater than 150. A small amount (0.5 per
The peroxide value Ip is the number that expresses in cent to 1.0 per cent m/m) of high HLB emulsifier (for exampIe
milliequivalents of active oxygen the quantity of peroxide polysorbate 60) may be added to the mixture to retard the
contained in 1000 g of the substance, as determined by the phase separation and decrease the time lag in the liberation
methods described below. of iodine.

156 See the information section on general monographs (caver pages)


EUROPEAN PHARMACOPOEIA 8.0 2.5.9. Determination of nitro gen by sulfuric add digestion

Carry out a blank determination (Vo mL). lf the result of several times, each with 40 mL of water R, until the aqueous
the blank determination exceeds 0.1 mL of titration reagent, phase is no longer a1kaline to phenolphthalein. Transfer the
replace the impure reagents and repeat the determination. ether phase to a tared flask, washing the separating funnel
with peroxide-free ether R.
1p = 1000 (VI - Vo) e
Distil off the ether with suitable precautions and add 6 mL of
m
acetone R to the residue. Carefully remove the solvent in a
current of airo Dry to constant mass at 100-105 oc. Allow to
c concentration of the sodium thiosulfate solution
cool in a desiccator and weigh (a g).
in moles, per litre.
. lOOa
Unsapomfiable matter = - - per cent
m
0112008:20506
Dissolve the residue in 20 mL of alcohol R, previously
neutralised to phenolphthalein solution R and titrate with
2.5.6. SAPONIFICATION VALUE 0.1 M ethanolic sodium hydroxide. If the volume of 0.1 M
The saponification value 1s is the number that expresses in ethanolic sodium hydroxide used is greater than 0.2 mL, the
milligrams the quantity of potassium hydroxide required to separation of the layers has been incomplete; the residue
neutralise the free adds and to saponify the esters present in weighed cannot be considered as "unsaponifiable matter". In
1 g of the substance. case of doubt, the test must be repeated.
Unless otherwise prescribed, use the quantities indicated in
Table 2.5.6.-1 for the determination. 0112008:20508
Table 2.5.6.-1
Presumed value 1, Quantity of sample (g)
2.5.8. DETERMINATION OF PRIMARY
<3 20
AROMATIC AMINO-NITROGEN
12 lo 15
Dissolve the prescribed quantity of the substance to be
3 lo 10
examined in 50 mL of dilute hydrochloric acid R or in another
10 lo 40 8 to 12 prescribed solvent and add 3 g of potassium bromide R. Cool
in ice-water and titrate by slowly adding 0.1 M sodium nitrite
40 lo 60 5 lo 8
with constant stirring.
60 lo 100 3 lo 5 Determine the end-point electrometrically or by the use of
100 lo 200 2.5 lo 3 the prescribed indicator.
200 lo 300 1 lo 2
01/2008:20509
300 lo 400 0.5 lo 1

Introduce the prescribed quantity of the substance to be 2.5.9. DETERMINATION OF


examined (m g) into a 250 mL borosilicate glass flask fitted NITRO GEN BY SULFURIC ACID
with a reflux condenser. Add 25.0 mL of 0.5 M alcoholic DIGESTION
potassium hydroxide and a few glass beads. Attach the
condenser and heat under reflux for 30 min, unless otherwise SEMI-MICRO METHOD
prescribed. Add 1 mL of phenolphthalein solution R1 and Place a quantity of the substance to be examined (m g)
titrate immediately (while still hot) with 0.5 M hydrochloric containing about 2 mg of nitro gen in a combustion flask, add
acid (nI mL of 0.5 M hydrochloric acid). Carry out a blank test4 g of a powdered mixture of 100 g of dipotassium sulfate R,
under the same conditions (n 2 mL of 0.5 M hydrochloric acid). 5 g of copper sulfate R and 2.5 g of selenium R, and three glass
I8 = 28.05 (n2 - nI) beads. Wash any adhering particles from the neck into the
m flask with 5 mL of sulfuric acid R, allowing it to run down the
sides of the flask, and mix the contents by rotation. Close
the mouth of the flask loosely, for example by means of a
01/2008:20507 glass bulb with a short stem, to avoid excessive 10ss of sulfuric
add. Heat gradually at first, then increase the temperature
2.5.7. UNSAPONIFIABLE MATTER until there is vigorous boiling with condensation of sulfuric
add in the neck of the flask; precautions should be taken to
The term "unsaponifiable matter" is applied to the substances prevent the upper part of the flask from becoming overheated.
non-volatile at 100-105 oC obtained by extraction with an Continue the heating for 30 min, unless otherwise prescribed.
organic solvent from the substance to be examined after it has Cool, dissolve the solid material by cautiously adding to
been saponified. The result is calculated as per cent mlm. the mixture 25 mL of water R, cool again and place in a
Use ungreased ground-glass glassware. steam-distillation apparatus. Add 30 mL of strong sodium
hydroxide solutíon R and distil immediately by passing steam
Introduce the prescribed quantity of the substance to be
through the mixture. Collect about 40 rnL of distillate in
examined (m g) into a 250 mL flask fitted with a reflux
20.0 mL of 0.01 M hydrochloric acid and enough water R
condenser. Add 50 mL of 2 M alcoho/ic potassium hydroxide R
to cover the tip of the condenser. Towards the end of the
and heat on a water-bath for 1 h, swirling frequently. Cool
distillation, lower the receiver so that the tip of the condenser
to a temperature below 25 oC and transfer the contents of
is aboye the surface of the add. Take precautions to prevent
the flask to a separating funnel with the aid of 100 mL of
any water on the outer surface of the condenser from reaching
water R. Shake the liquid carefully with 3 quantities, each of
the contents of the receiver. Titrate the distillate with 0.01 M
100 mL, of peroxide-free ether R. Combine the ether layers
sodium hydroxide, using methyl red mixed solution R as
in another separating funnel containing 40 mL of water R,
indicator (nI mL of 0.01 M sodium hydroxide).
shake gently for a few minutes, allow to separate and reject
the aqueous phase. Wash the ether phase with 2 quantities, Repeat the test using about 50 mg of glucase R in place of the
each of 40 mL, of water R then wash successively with 40 mL substance to be examined (n 2 rnL of 0.01 M sodium hydroxide).
of a 30 giL solution of potassium hydroxide R and 40 mL of . 0.01401 (n2 - nl)
water R; repeat this procedure 3 times. Wash the ether phase Content of mtrogen = per cent
m

General Notices (1) apply to all monographs and other texts 157
2.5.10. Oxygen-flask method EUROPEAN PHARMACOPOEIA 8.0

0112008:20510 1 mL of 0.1 M sodium edetate is equivalent to 2.431 mg of Mg.


LEAD
2.5.10. OXYGEN-FLASK METHOD Introduce the prescribed solution into a 500 mL conical flask
Unless otherwise prescribed the combustion flask is a conical and dilute to 200 mL with water R. Add about 50 mg of
flask of at least 500 mL capacity of borosilicate glass with xylenol orange tritura te R and hexamethylenetetramine R until
a ground-glass stopper fitted with a suitable carrier for the the solution becomes violet-pink. Titrate with 0.1 M sodium
sample, for example in platinum or platinum-iridium. edetate until the violet-pink colour changes to yellow.
Finely grind the substance to be examined, place the 1 mL of 0.1 M sodium edetate is equivalent to 20.72 mg ofPb.
prescribed quantity in the centre of a piece of filter paper
measuring about 30 mm by 40 mm provided with a small strip ZINC
about 10 mm wide and 30 mm long. If paper impregnated Introduce the prescribed solution into a 500 mL conical
with lithium carbonate is prescribed, moisten the centre of flask and dilute to 200 mL with water R. Add about 50 mg
the papel' with a saturated solution of lithium carbonate R of xylenol orange tritura te R and hexamethylenetetramine R
and dry in an oven before use. Envelop the substance to be until the solution becomes violet-pink. Add 2 g of
examined in the paper and place it in the sample carrier. hexamethylenetetramine R in excess. Titrate with 0.1 M
Introduce into the flask water R or the prescribed solution sodium edetate until the violet-pink colour changes to yellow.
designed to absorb the combustion products, displace the air 1 mL of 0.1 M sodium edetate is equivalent to 6.54 mg of Zn.
with oxygen by means of a tube having its end just aboye the
liquid, moisten the neck of the flask with water R and close 0112013:20512
with its stopper. Ignite the paper strip by suitable means with
the usual precautions. Keep the flask firmly closed during the 2.5.12. WATER: SEMI-MICRO
combustion. Shake the flask vigorously to completely dissolve
the combustion products. Cool and after about 5 min, unless DETERMINATION
otherwise prescribed, carefully unstopper the flask. Wash the The semi-micro determination of water is based upon the
ground parts and the walls of the flask, as well as the sample quantitative reaction of water with sulfur dioxide and iodine
carrier, with water R. Combine the combustion products and in a suitable anhydrous medium in the presence of a base with
the washings and proceed as prescribed in the monograph. sufficient buffering capacity.
Apparatus
0112008:20511 The apparatus consists of a titration vessel with:
corrected 8.0
- 2 identical platinum electro des ;
- tight inlets for introduction of solvent and titrant;
2.5.11. COMPLEXOMETRIC
- an inlet for introduction of air via a desiccant;
TITRATIONS - a sample inlet fitted with a stopper or, for liquids, a septum.
ALUMINIUM Inlet systems for introduction of dry nitro gen or for aspiration
Introduce 20.0 mL of the prescribed solution into a 500 mL of solvents may also be fitted.
conical flask, add 25.0 mL of 0.1 M sodium edetate and The titration is carried out according to the instrument
10 mL of a mixture of equal volumes of a 155 giL solution of supplier's instructions. Care is taken throughout the
ammonium acetate R and dilute acetic acid R. Boil for 2 min, determination to avoid exposure of reagents and solvents to
then cool. Add 50 mL of ethanol R and 3 mL of a freshly atmospheric moisture. The end-point is determined using
prepared 0.25 giL solution of dithizone R in ethanol R. Titrate 2 identical indicator electro des connected to an electrical
the excess of sodium edetate with 0.1 M zinc sulfate until the source that maintains between the electro des either a constant
colour changes from greenish-blue to reddish-violet. current (2.2.65. Voltametric titration) or a constant voltage
1 mL of 0.1 M sodium edetate is equivalent to 2.698 mg of Al. (2.2.19. Amperometrie titration). Where direct titration is
used (method A), addition of titrant causes either a de crease
BISMUTH in voltage where constant current is maintained or an increase
Introduce the prescribed solution into a 500 mL conical flask. in current where constant voltage is maintained, until the
Dilute to 250 mL with water R and then, unless otherwise end-point is reached. Instruments with automatic end-point
prescribed, add dropwise, with shaking, concentrated detection are commonly used.
ammonia R until the mixture becomes cloudy. Add 0.5 mL Standardisation. To the titration vessel, add methanol R,
of nitric acid R. Heat to about 70 oC until the cloudiness dried if necessary, or the solvent recommended by the supplier
disappears completely. Add about 50 mg of xylenol orange of the titrant. Where applicable for the apparatus used,
tritura te R and titrate with 0.1 M sodium edetate until the eliminate residual water from the measurement cell or carry
colour changes from pinkish-violet to yellow. out a pre-titration. Introduce a suitable amount of water in an
1 mL of 0.1 M sodium edetate is equivalent to 20.90 mg of BL appropriate form (water R or a certified reference material)
and carry out the titration, stirring for the necessary time. The
CALCIUM water equivalent is 110t less than 80 per cent of that indicated
Introduce the prescribed solution into a 500 mL conical by the supplier. Standardise the titrant before the first use and
flask, and dilute to 300 mL with water R. Add 6.0 mL of at suitable intervals thereafter.
strong sodium hydroxide solution R and about 200 mg of Unless otherwise prescribed, use Method A.
calconecarboxylie aeid triturate R. Titrate with 0.1 M sodium
edetate until the colour changes from violet to full blue. Method A. Introduce into the titration vessel methanol R, or
the solvent indicated in the monograph or recommended by
1 mL of 0.1 M sodium edetate is equivalent to 4.008 mg of Ca.
the supplier of the titrant. Where applicable for the apparatus
MAGNESIUM used, eliminate residual water from the measurement ceH
Introduce the prescribed solution into a 500 mL conical flask or carry out a pre-titration. Introduce the substance to be
and dilute to 300 mL with water R. Add 10 mL of ammonium examined rapidly and carry out the titration, stirring for the
ehloride buffer solution pH 10.0 R and about 50 mg of mordant necessary extraction time.
blaek 11 triturate R. Heat to about 40 oC then titrate at this Method B. Introduce into the titration vessel methanol R, or
temperature with 0.1 M sodium edetate until the colour the solvent indicated in the monograph or recommended by
changes from violet to ful! bIne. the supplier of the titrant. Where applicable for the apparatus

158 See the information section on general monographs (caver pages)


EUROPEAN PHARMACOPOEIA 8.0 2.5.16. Proíein in polysaccharide vacdnes

used, eliminate residual water from the measurement cell 0.05 mL of methyl orange solution R and neutralise with strong
or carry out a pre-titration. Introduce the substance to be sodium hydroxide solution R (6.5 mL to 7 mL). If a precipitate
examined rapidly and in a suitable state of division. Add an forms dissolve it by adding, dropwise, sufficient dilute sulfuric
accurately measured volume of the titrant, sufficient to give acid R. Transfer the solution to a 250 mL conical flask, rinsing
an excess of about 1 mL or the prescribed volume. Allow to the combustion flask with 25 mL of water R. Add 25.0 mL
stand protected from light for 1 min or the prescribed time, of 0.02 M sodium edetate, 10 mL of acetate buffer solution
with stirring. Titrate the excess of reagent using methanol R pH 4.4 R and a few glass beads and boil gently for 3 mino Add
or the prescribed solvent, containing an accurately known 0.1 mL of pyridylazonaphthol solution R and titrate the hot
quantity of water. solution with 0.02 M copper sulfate until the colour changes
Suitability. The accuracy of the determination with the to purplish-brown. Carry out a blank titration omiUing the
chosen titrant must be verified for each combination of vaccine.
substance, titrant and solvent to be examined. The following 1 mL of 0.02 M sodium edetate is equivalent to 0.5396 mg of Al.
procedure, given as an example, is suitable for samples
containing 2.5-25 mg of water. 0112008:20514
The water content of the substance to be examined is
determined using the reagent/solvent system chosen. 2.5.14. CALCIUM IN ADSORBED
Thereafter, in the same titration vessel, sequential known VACCINES
amounts of water, corresponding to about 50-100 per cent
of the amount found in the substance to be examined, are All solutions used for this test must be prepared using water R.
added in an appropriate form (at least 5 additions) and the Determine the calcium by atomic emission spectrometry
water content is determined after each addition. Calculate the (2.2.22, Method I). Homogenise the preparation to be
percentage recovery (r) after each addition using the following examined. To 1.0 mL add 0.2 mL of dilute hydrochloric acid R
expression: and dilute to 3.0 mL with water R. Measure the absorbance
at 620 nm.
r = 100 W2
Wl
0112008:20515
amount of water added, in milligrams;
2.5.15. PHENOL IN IMMUNOSERA
amount of water found, in milligrams.
AND VACCINES
Calculate the mean percentage recovery (7'). The
reagentl solvent system is considered to be acceptable if 7' is Homogenise the preparation to be examined. Dilute an
between 97.5 per cent and 102.5 per cent. appropriate volume with water R so as to obtain a solution
presumed to contain 15 flg of phenol per millilitre. Prepare
Calculate the regression lineo The x-axis represents the
a series of reference solutions with phenol R containing
cumulative water added whereas the y-axis represents the sum
5 flg, 10 flg, 15 flg, 20 flg and 30 ~lg of phenol per millilitre
of the initial water content determined for the substance (M)
respectively. To 5 mL of the solution to be examined and to
and the cumulative water determined after each addition.
5 mL of each of the reference solutions respectively, add 5 mL
Calculate the slope (b), the intercept with the y-axis (a) and
of buffer solution pH 9.0 R, 5 mL of aminopyrazolone solution R
the intercept of the extrapolated calibration line with the
and 5 mL of potassium ferricyanide solution R. Allow to stand
x-axis (d).
for 10 min and measure the intensity of colour at 546 nm.
Calculate the percentage errors (el and ez) using the following
Plot the calibratiol1 curve and calculate the phenol content of
expressions:
the preparation to be examined.

0112008:20516

2.5.16. PROTEIN IN POLYSACCHARIDE


VACCINES
a the y-axis intercept, in milligrams of water;
Test solution. Use a volumetric flask with a suitable volume for
d the x-axis intercept, in milligrams of water; preparation of a solution containing about 5 mg per millilitre
M water content of the substance, in milligrams of of dry polysaccharide. Transfer the contents of a container
water. quantitatively to the flask and dilute to volume with water R.
Place 1 mL of the solution in a glass tube and add 0.15 mL of a
The reagent/solvent system is considered to be acceptable if: 400 giL solution of trichloroacetic acid R. Shake, allow to stand
- lell and le21 are not greater than 2.5 per cent; for 15 min, centrifuge for 10 min at 5000 r/min and discard
- b is between 0.975 and 1.025. the supernatant. Add 0.4 mL of 0.1 M sodium hydroxide to the
centrifugation residue.
01/2008:20513 Reference solutions. Dissolve 0.100 g of bovine albumin R in
100 mL of 0.1 M sodium hydroxide (stock solution containing
2.5.13. ALUMINIUM IN ADSORBED 1 g of protein per litre). Dilute 1 mL of the stock solution
to 20 mL with 0.1 M sodium hydroxide (working dilution
VACCINES 1: 50 mg of protein per litre). Dilute 1 mL of the stock solution
Homogenise the preparation to be examined and transfer to 4 mL with 0.1 M sodium hydroxide (working dilution
a suitable quantity, presumed to contain 5 mg to 6 mg of 2: 250 mg of protein per litre). Place in 6 glass tubes 0.10 mL,
aluminium, to a 50 mL combustion flask. Add 1 mL of sulfuric 0.20 mL and 0.40 mL of working dilution 1 and 0.15 mL,
acid R, 0.1 mL of nitric acid R and some glass beads. Heat 0.20 mL and 0.25 mL of working dilution 2. Make up the
the solution until thick, white fumes are evolved. If there is volume in each tube to 0.40 mL using 0.1 M sodium hydroxide.
charring at this stage add a few more drops of nitric acid R Prepare a blank using 0.40 mL of 0.1 M sodium hydroxide.
and continue boiling until the colour disappears. Allow to Add 2 mL of cupri-tartaric solution R3 to each tube, shake
cool for a few minutes, carefully add 10 mL of water R and and allow to stand for 10 mino Add to each tube 0.2 mL
boil until a clear solution is obtained. Allow to cool, add of a mixture of equal volumes of phosphomolybdotungstic

General Notices (1) apply to all monographs and other texts 159
2.5.17. Nudeic adds in polysaccharide vaccines EUROPEAN PHARMACOPOEIA 8.0

reagent R and water R, prepared immediately before use. 30 Ilg to 600 Ilg of acetylcholine chloride (O-acetyl). Introduce
Stopper the tubes, mix by inverting and allow to stand in 0.3 mL, 0.5 mL and 1.0 mL in duplicate into 6 tubes (3 reaction
the dark for 30 mino The blue colour is stable for 60 mino If solutions and 3 correction solutions).
necessary, centrifuge to obtain clear solutions. Reference solutíons. Dissolve 0.150 g of acetylcholíne chloride R
Measure the absorbance (2.2.25) of each solution at 760 nm in 10 mL of water R (stock solution containing 15 g of
using the blank as the compensation liquido Draw a calibration acetylcholine chloride per litre). Irnmediately before use,
curve from the absorbances of the 6 reference solutions and dilute 1 mL of the stock solution to 50 mL with water R
the corresponding protein contents and read from the curve (working dilution 1: 300 Ilg of acetylcholine chloride per
the content of protein in the test solution. millilitre). Immediately before use, dilute 1 mL of the stock
solution to 25 mL with water R (working dilution 2: 600 Ilg
0112008:20517 of acetylcholine chloride per millilitre). Introduce 0.1 mL
and 0.4 mL of working dilution 1 in duplicate (reaction and
correction solutions) in 4 tubes and 0.6 mL and 1.0 mL of
2.5.17. NUCLEIC ACIDS IN working dilution 2 in duplicate (reaction and correction
POLYSACCHARIDE VACCINES solutions) in another 4 tubes.
Test solutíon. Use a volumetric flask with a suitable volume for Prepare a blank using 1 mL of water R.
preparation of a solution containing about 5 mg per millilitre Malee up the volume in each tube to 1 mL with water R. Add
of dry polysaccharide. Transfer the contents of a container l.0 mL of 4 M hydrochloric acid to each of the correction
quantitatively to the flask and dilute to volume with water R. tubes and to the blanlc. Add 2.0 mL of alkaline hydroxylamine
Dilute the test solution if necessary to obtain an absorbance solution R to each tube. Allow the reaction to proceed for
value suitable for the instrument used. Measure the absorbance exactly 2 min and add l.0 mL of 4 M hydrochloríc acid to
(2.2.25) at 260 nm using water R as the compensation liquido each of the reaction tubes. Add l.0 mL of a 100 giL solution
of ferric chloríde R in 0.1 M hydrochloric acíd to each tube,
The absorbance of a 1 giL solution of nucleic acid at 260 nm is stopper the tubes and shake vigorously to remove bubbles.
20.
Measure the absorbance (2.2.25) of each solution at 540 11m
using the blank as the compensation liquido For each reaction
0112008:20518 solution, subtract the absorbance of the corresponding
correction solution. Draw a calibration curve from the
2.5.18. PHOSPHORUS IN corrected absorbances for the 4 reference solutions and the
POLYSACCHARIDE VACCINES corresponding content of acetylcholine chloride and read
from the curve the content of acetylcholine chloride in the
Test solution. Use a volumetric flask with a suitable volume for test solution for each volume tested. Calculate the mean of
preparation of a solution containing about 5 mg per millilitre the 3 values.
of dry polysaccharide. Transfer the contents of a container 1 mole of acetylcholine chloride (181.7 g) is equivalent to
quantitatively to the flask and dilute to volume with water R. 1 mole of O-acetyl (43.05 g).
Dilute the solution so that the volume used in the test (1 mL)
contains about 6 Ilg of phosphorus. Transfer l.0 mL of the 01/2008:20520
solution to a 10 mL ignition tube.
Reference solutions. Dissolve 0.2194 g of potassium díhydrogen 2.5.20. HEXOSAMINES IN
phosphate R in 500 mL of water R to give a solution containing
the equivalent of 0.1 mg of phosphorus per millilitre. Dilute POLYSACCHARIDE VACCINES
5.0 mL of the solution to 100.0 mL with water R. Introduce Test solutíon. Use a volumetric flask with a suitable volume for
0.5 mL, l.0 mL and 2.0 mL of the dilute solution into 3 preparation of a solution containing about 5 mg per millilitre
ignition tubes. of dry polysaccharide. Transfer the contents of a container
Prepare a blank solution using 2.0 mL of water R in an ignition quantitatively to the flask and dilute to volume with water R.
tube. Dilute the solution so that the volumes used in the test contain
To aH the tubes add 0.2 mL of sulfuríc acid R and heat in an 125 Ilg to 500 flg of glucosamine (hexosamine). Introduce
oil bath at 120 oC for 1 h and then at 160 oC until white fumes 1.0 mL of the diluted solution into a graduated tube.
appear (about 1 h). Add 0.1 mL of perchloríc acid R and heat at Reference solutions. Dissolve 60 mg of glucosamíne
160 oC until the solution is decolorised (about 90 min). Cool hydrochloride R in 100 mL of water R (stock solution
and add to each tube 4 mL of water R and 4 mL of ammonium containing 0.500 g of glucosamine per litre). Introduce
molybdate reagent R. Heat in a water-bath at 37 oC for 90 min 0.25 mL, 0.50 mL, 0.75 mL, and 1.0 mL of the working dilution
and coo!. Adjust the volume to 10.0 mL with water R. The into 4 graduated tubes.
blue colour is stable for several hours. Prepare a blank using 1 mL of water R.
Measure the absorbance (2.2.25) of each solution at 820 nm Make up the volume in each tube to 1 mL with water R.
using the blank solution as the compensation liquido Draw Add 1 mL of a solution of hydrochloríc acid R (292 giL) to
a calibration curve with the absorbances of the 3 reference each tube. Stopper the tubes and place in a water-bath for
solutions as a function of the quantity of phosphorus in the 1 h. Cool to room temperature. Add to each tube 0.05 mL
solutions and read from the curve the quantity of phosphorus of a 5 giL solution of thymolphthaleín R in alcohol R; add a
in the test solution. solution of sodíum hydroxíde R (200 giL) until a blue colour
is obtained and then 1 M hydrochloric acid until the solution
0112008:20519 is colourless. Dilute the volume in each tube to 10 mL with
water R (neutralised hydrolysates).
2.5.19. Q-ACETYL IN In a second series of 10 mL graduated tubes, place 1 mL of each
POLYSACCHARIDE VACCINES neutralised hydrolysate. Add 1 mL of acetylacetone reagent
(a mixture, prepared immediately before use, of 1 volume
Test solution. Use a volumetric flask with a suitable volume for of acetylacetone R and 50 volumes of a 53 giL solution of
preparation of a solution containing about 5 mg per millilitre anhydrous sodíum carbonate R) to each tube. Stopper the
of dry polysaccharide. Transfer the contents of a container tubes and place in a water-bath at 90 oC for 45 mino Cool to
quantitatively to the flask and dilute to volume with water R. room temperature. Add to each tube 2.5 mL of alcohol R and
Dilute the solution so that the volumes used in the test contain 1.0 mL of dimethylaminobenzaldehyde solution (immediately

160 See the information section on general monographs (cover pages)


EUROPEAN PHARMACOPOEIA 8.0 2.5.24. Carbon dioxide in gases

before use dissolve 0.8 g of dimethylaminobenzaldehyde R in Make up the volume in each tube to 1 mL with water R. Place
15 mL of alcohol R and add 15 mL of hydrochloric acid R) the tubes in iced water and add dropwise and with continuous
and dilute the volume in each tube to 10 mL with alcohol R. stirring to each tube 5.0 mL of borate solution R. Stopper the
Stopper the tubes, mix by inverting and allow to stand in the tubes and place in a water-bath for 15 mino Cool to room
dark for 90 mino Measure the absorbance (2.2.25) of each temperature. Add 0.20 mL of a 1.25 giL solution of carbazole R
solution at 530 nm using the blank as the compensation liquido in ethanol R to each tube. Stopper the tubes and place in a
Draw a calibration curve from the absorbances for the water-bath for 15 mino Cool to room temperature. Measure
4 reference solutions and the corresponding content of the absorbance (2.2.25) of each solution at 530 nm using the
hexosamine and read from the curve the quantity of blank as the compensation liquido
hexosamine in the test solution. Draw a calibration curve from the absorbances for the
5 reference solutions and the corresponding content of
glucuronic acid and read from the curve the quantity of
0112008:20521
glucuronic acid in the test solution for each volume tested.
Calculate the mean of the 3 values.
2.5.21. METHYLPENTOSES IN
POLYSACCHARIDE VACCINES 0112008:20523

Test solution. Use a volumetric flask with a suitable volume 2.5.23. SIALIC ACID IN
for preparation of a solution containing about 5 mg per
millilitre of dry polysaccharide. Transfer the contents of a , POLYSACCHARIDE VACCINES
container quantitatively to the flask and dilute to volume with Test solution. Transfer quantitatively the contents of one or
water R. Dilute the solution so that the volumes used in the several containers to a volumetric flask of a suitable volume
test contain 2 flg to 20 flg of rhamnose (methylpentoses). that will give a solution with a known concentration of about
Introduce 0.25 mL, 0.50 mL and 1.0 mL of the diluted solution 250 flg per millilitre of polysaccharide and dilute to volume
into 3 tubes. with water R. Using a syringe, transfer 4.0 mL of this solution
Reference solutions. Dissolve 0.100 g of rhamnose R in 100 mL to a 10 mL ultrafiltration ceH suitable for the passage of
of water R (stock solution containing 1 g of methylpentose molecules of relative molecular mass less than 50 000. Rinse
per litre). Immediately before use, dilute 1 mL of the stock the syringe twice with water R and transfer the rinsings to the
solution to 50 mL with water R (working dilution: 20 mg of ultrafiltration cell. Carry out the ultrafiltration, with constant
methylpentose per litre). Introduce 0.10 mL, 0.25 mL, 0.50 mL, stirring, under nitrogen R at a pressure of about 150 kPa. Refill
0.75 mL and 1.0 mL of the working dilution into 5 tubes. the cell with water R each time the volume of liquid in it has
Prepare a blank using 1 mL of water R. decreased to 1 mL and continue until 200 mL has been filtered
Make up the volume in each tube to 1 mL with water R. Place and the remaining volume in the ceH is about 2 mL. Using a
the tubes in iced water and add dropwise and with continuous syringe, transfer this residualliquid to a 10 mL volumetric
stirring to each tube 4.5 mL of a cooled mixture of 1 volume flask. Wash the ceH with 3 quantities, each of 2 mL, of water R,
of water R and 6 volumes of sulfuric acid R. Warm the tubes transfer the washings to the flask and dilute to 10.0 mL with
to room temperature and place in a water-bath for a few water R (test solution). In each of2 test-tubes place 2.0 mL of
minutes. Cool to room temperature. Add to each tube 0.10 mL the test solution.
of a 30 giL solution of cysteine hydrochloride R, prepared Reference solutions. Use the reference solutions prescribed in
immediately before use. Shake and allow to stand for 2 h. the monograph.
Measure the absorbance (2.2.25) of each solution at 396 nm Prepare 2 series of 3 test -tubes, place in the tubes of each
and at 430 nm using the blank as compensation liquido For series 0.5 mL, 1.0 mL and 1.5 111L respectively, of the reference
each solution, calculate the difference between the absorbance solution corresponding to the type of vaccine to be examined
measured at 396 nm and that measured at 430 nm. Draw and adjust the volume in each tube to 2.0 mL with water R.
a calibration curve from the absorbance differences for Prepare blank solutions using 2.0 mL of water R in each of
the 5 reference solutions and the corresponding content 2 test -tubes.
of methylpentose and read from the curve the quantity of To all the tubes add 5.0 mL of resorcinol reagent R. Heat at
methylpentose in the test solution for each volume tested. 105 oC for 15 min, cool in cold water and transfer the tubes
Calculate the mean of the 3 values. to a bath of iced water. To each tube add 5 mL of isoamyl
alcohol R and mix thoroughly. Place in the bath of iced
0112008:20522 water for 15 mino Centrifuge the tubes and keep them in
the bath of iced water until the examination by absorption
spectrophotometry. Measure the absorbance (2.2.25) of each
2.5.22. URONIC ACIDS IN supernatant solution at 580 nm and 450 nm using isoamyl
POLYSACCHARIDE VACCINES alcohol R as the compensation liquido For each wavelength,
calculate the absorbance as the mean of the values obtained
Test solution. Use a volumetric flask with a suitable volume for
with 2 identical solutions. Subtract the mean value for the
preparation of a solution containing about 5 mg per millilitre
blank solution from the mean values obtained for the other
of dry polysaccharide. Transfer the contents of a container
solutions.
quantitatively to the flask and dilute to volume with water R.
Dilute the solution so that the volumes used in the test contain Draw a graph showing the difference between the absorbances
4 flg to 40 flg of glucuronic acid (uronic acids). Introduce at 580 nm and 450 nm of the reference solutions as a function
0.25 mL, 0.50 mL and 1.0 mL of the diluted solution into 3 of the content of N-acetylneuraminic acid and read from the
tubes. graph the quantity of N-acetylneuraminic acid (sialic acid) in
the test solution.
Reference solutions. Dissolve 50 mg of sodium glucuronate R
in 100 mL of water R (stock solution containing 0.4 g of
01/2011:20524
glucuronic acid per litre). Immediately before use, dilute 5 mL
of the stock solution to 50 mL with water R (working dilution:
40 mg of glucuronic acid per litre). Introduce 0.10 mL, 2.5.24. CARBON DIOXIDE IN GASES
0.25 mL, 0.50 mL, 0.75 mL, and 1.0 mL ofthe working dilution Gases absorb light at one or more specific wavelengths. This
into 5 tubes. property is widely used to allow highly selective measurement
Prepare a blank using 1 mL of water R. of their concentrations.

General Notices (1) apply to all monographs and other texts 161
2.5.25. Carbon monoxide in gases EUROPEAN PHARMACOPOEIA 8.0

Description and principIe of measurement. The - a U-tube (U2 ) containing peHets of potassium hydroxide R;
concentration of carbon dioxide in other gases can be - a U-tube (U3 ) containing diphosphorus pentoxide R
determined using an infrared anaIyser. dispersed on previously granuIated, fused pum ice ;
The infrared anaIyser generally consists of a light source - a U-tube (U4 ) containing 30 g of recrystallised iodine
emitting broadband infrared radiation, an optical device, pentoxide R in granules, previously dried at 200 oC and kept
a sample cell and a detector. The optical device may be at a temperature of 120 oC (T) during the test; the iodine
positioned either before or after the sampIe ceH and it consists pentoxide is packed in the tube in 1 cm coIumns separated
of one or several optical filters, through which the broadband by 1 cm columns of glass wool to give an effective length of
radiation is passed. The optical device in this case is selected 5cm;
for carbon dioxide. The measurement light beam passes - a reaction tube (P2 ) containing 2.0 mL of potassium iodide
through the sampIe ceH and may also pass through a reference solution R and 0.15 mL of starch solution R.
cell if the anaIyser integrates such a feature (some use an
Method. Flush the apparatus with 5.0 L of argon R and, if
electronic system instead of a reference cell).
necessary, discharge the bIue colour in the iodide solution by
When carbon dioxide is present in the sampIe ceH, absorption adding the smallest necessary quantity of freshly prepared
of energy in the measurement light beam will occur according 0.002 M sodium thiosulfate. Continue flushing until not more
to the Beer-Lambert Iaw and this produces a change in than 0.045 mL of 0.002 M sodium thiosulfate is required after
the detector signal. This measurement signal is compared passage of 5.0 L of argon R. Pass the gas to be examined from
to a reference signal to generate an output reIated to the the cylinder through the apparatus, using the prescribed
concentration of carbon dioxide. The generated signal is volume and the flow rateo FIush the last traces of liberated
linearised in order to obtain the carbon dioxide concentration. iodine into the reaction tube by passing through the apparatus
To prevent the entry of particles into the sensors, which could 1.0 L of argon R. Titrate the liberated iodine with 0.002 M
cause stray-light phenomena, the apparatus is fitted with a sodium thiosulfate. Carry out a bIank test, using the prescribed
suitabIe filter. volume of argon R. The difference between the volumes of
Required technical specifications. When used for a limit 0.002 M sodium thiosulfate used in the titrations is not greater
test, the infrared analyser meets the following technical than the prescribed limito
specifications:
METHOD II
- limit of detection: (generally defined as a signal-to-noise
ratio of 2) maximum 20 per cent of the maximum Gases absorb light at one or more specific wavelengths. This
admissibIe concentration; property is widely used to allow highIy selective measurement
of their concentrations.
- repeatability: maximum relative standard deviation of
10 per cent of the maximum admissibIe concentration, Description and principle of measurement. The
determined on 6 measurements; concentration of carbon monoxide in other gases can be
- linearity: maximum 10 per cent ofthe maximum admissible determined using an infrared analyser.
concentration. The infrared analyser generally consists of a light source
The technical specifications must be met in the presence of emitting broadband infrared radiation, an optical device,
the other gas impurities in the sample. a sampIe cell and a detector. The optical device may be
positioned either before or after the sample ceH; it consists of
one or several optical filters, through which the broadband
01/2011:20525 radiation is passed. The optical device in this case is selected
for carbon monoxide. The measurement light beam pass es
2.5,25. CAREON MONOXIDE IN GASES through the sampIe cell and may also pass through a reference
ceH if the analyser integrates such a feature (some use an
METHOD 1 electronic system instead of a reference ceH).
Apparatus. The apparatus (Figure 2.5.25.-1) consists of the When carbon monoxide is present in the sampIe ceH,
following parts connected in series: absorption of energy in the measurement light beam will
- a U -tube (U¡) containing anhydrous silica gel R impregnated occur according to the Beer-Lambert law and this produces
with chromium trioxide R; a change in the detector signa!. This measurement signal is
- a wash bottle (p¡) containing 100 mL of a 400 giL solution compared to a reference signal to generate al1 output related
of potassium hydroxide R; to the concentration of carbon monoxide. The generated

U1 F1 U2 U3 U4 F2

-1 '_-
- 1
-_: 1-
-_1: : :
-1
_,1:...

Figure 2.5.25.-1. - Apparatus for the determination of carbon monoxide


Dimensions in millimetres

162 See the information sectiol1 011 general mOl1ographs (cover pages)
PHARMACOPOEIA 8.0 2.5.28. Water in gases

signal is linearised in order to obtain the carbon monoxide 01/2009:20527


concentration. To prevent the entry of particles into the
sensors, which could cause stray-light phenomena, the 2.5.27. OXYGEN IN GASES
apparatus is fitted with a suitable filter.
Oxygen in gases is determined using a paramagnetic analysero
Required technical spedfications. Wnen used for a limit test,
the carbon monoxide infrared analyser meets the following The principIe of the method is based on the high paramagnetic
technical specifications: sensitivity of the oxygen molecule. Oxygen exerts a
strong interaction on magnetic fields, which is measured
- limit of detection: (generally defined as a signal-to-noiseelectronically, amplified and converted to a reading of oxygen
ratio of 2) maximum 20 per cent of the maximum concentration. The measurement of oxygen con centra ti on
admissible concentration; is dependent upon the pressure and temperature and, if the
- repeatability: maximum reiative standard deviation of analyser is not automatically compensated for variations in
10 per cent of the maximum admissible concentration, temperature and pressure, it must be calibrated immediately
determined on 6 measurements; prior to use. As the paramagnetic effect of oxygen is linear,
- linearity: me.ximum 10 per cent of the maximum admissible the instrument must have a suitable range with a readability of
concentration. 0.1 per cent or better.
The technical specifications must be met in the presence of Calibration of the instrumento Make the setting in the
the other gas impurities in the sample. following manner:
- set the zero by passing nitrogen Rl through the instrument
until a constant reading is obtained;
- set the scale to 100 per cent by passing oxygen R through
0112008:20526 the instrument at the same flow rate as for nitrogen Rl until
a constant reading is obtained.
Assay. Pass the gas to be examined through the instrument
2.5.26. NITROGEN MONOXIDE AND at a constant flow rate until a constant reading is obtained.
NITRO GEN DIOXIDE IN GASES Record the concentration of oxygen in the gas to be examined.
Nitrogen monoxide and nitro gen dioxide in gases
are determined using a chemiluminescence analyser 01/2008:20528
(Figure 2.5.26.-1).
The apparatus consists of the following: 2.5.28. WATER IN GASES
- a device fOl' filtering, checking and controlling the flow of Water in gases is determined using an electrolytic hygrometer,
the gas to be examined, described below.
- a converter that reduces nitro gen dioxide to nitro gen The measuring ceH consists of a thin film of diphosphorus
monoxide, to determine the combined content of nitro gen pentoxide, between 2 coiled platinum wires which act as
monoxide and nitro gen dioxide. The efficiency of the electro des. The water vapour in the gas to be examined is
converter has to be verified prior to use, absorbed by the diphosphorus pentoxide, which is transformed
to phosphoric acid, an electrical conductor. A continuous
- a controlled-flow-rate ozone generator; the ozone is
voltage applied across the electro des produces electrolysis of
produced by high-voltage electric discharges across
the water and the regeneration of the diphosphorus pentoxide.
two electro des ; the ozone generator is supplied with
The resulting electric current, which is proportional to the
pure oxygen or with dehydrated ambient air and the
water content in the gas to be examined, is measured. This
concentration of ozone obtained must greatly exceed the
system is self-calibrating since it obeys Faraday's law.
maximum content oI' any detectable nitro gen oxides,
Take a sample of the gas to be examined. Allow the gas to
- a chamber in which nitrogen monoxide and ozone can stabilise at room temperature. Purge the cell continuously
react, until a stable reading is obtained. Measure the water content
- a system for detecting light radiation emitted at a in the gas to be examined, making sure that the temperature
wavelength of 1:Z 11m, consisting of a selective optical filter is C011stant throughout the device used to introduce the gas
and a photomultiplier tube. into the apparatus.

Sample flow control

Converter
NO,-> NO /optical filter

Refrigerated chamber

<0(- -Q- Fiíter to eliminate


ozone
111E1
= Photomulti-
plier iube

Ozone generator
system

L
- Controls
- NO - (NO+NO,) cyc!e

NO (NO+N02)

Figure 2.5.26.-1. - Chemiluminescence

General Natices (1) apply ta all monographs and other texts 163
2.5.29. Sulfur dioxide EUROPEAN PHARMACOPOEIA 8.0

0112013:20529 Calculate the content of sulfur dioxide in parts per million


using the following expression:
n
32030 x (V1 - V2 ) x -
2.5.29. SULFUR DIOXIDE m

n molarity of the sodium hydroxide solution used


Introduce 150 mL of water R into the flask (A) (see as titrant.
Figure 2.5.29.-1) and pass carbon dioxide R through the whole
system for 15 min at arate of 100 ± 5 mL/min. To 10 mL of
dilute hydrogen peroxide solution R add 0.15 mL of a 1 giL 01/2008:20530
solution of bromophenol blue R in ethanol (20 per cent V/V) R.
Add 0.1 M sodium hydroxide until a violet-blue colour is 2.5.30. OXIDISING SUBSTANCES
obtained, without exceeding the end-point. Place the solution
Transfer 4.0 g to a glass-stoppered, 125 mL conical flask
in the test-tube (D). Without interrupting the stream of
and add 50.0 mL of water R. Insert the stopper and swirl for
carbon dioxide, remove the funnel (B) and introduce through
5 mino Transfer to a glass-stoppered 50 mL centrifuge tube
the opening into the flask (A) 25.0 g (m g) of the substance to and centrifuge. Transfer 30.0 mL of the clear supernatant to
be examined with the aid of 100 mL of water R. Replace the
a glass-stoppered 125 mL conical flask. Add 1 mL of glacial
funnel. Close the tap of the funnel and add 80 mL of dilute
acetic acid R and 0.5 g to 1.0 g of potassium iodide R. Insert
hydrochloric acid R to the funnel. Open the tap of the funnel
the stopper, swirl, and allow to stand for 25 min to 30 min
to allow the hydrochloric acid solution to flow into the flask,
in the dark. Add 1 mL of starch solution R and titrate with
making sure that no sulfur dioxide escapes into the funnel by
0.002 M sodium thiosulfate until the starch-iodine colour
closing the tap before the last few millilitres ofhydrochloric
disappears. Carry out a blank determination. Not more than
acid solution drain out. Boil for 1 h. Open the tap of the funnel 1.4 mL of 0.002 M sodium thiosulfate is required (0.002 per
and stop the flow of carbon dioxide and also the heating and
cent, calculated as H 2 02)'
the cooling water. Transfer the contents of the test -tube with
the aid of a little water R to a 200 mL wide-necked, conical 1 mL of 0.002 M sodium thiosulfate is equivalent to 34 flg of
flask. Heat on a water-bath for 15 min and allow to coo1. Add oxidising substances, calculated as hydrogen peroxide.
0.1 mL of a 1 giL solution of bromophenol blue R in ethanol
(20 per cent V/V) R and titrate with 0.1 M sodium hydroxide 0112008:20531
until the colour changes from yellow to violet-blue (V¡ mL).
Carry out a blank titration (V2 mL). 2.5.31. RIBOSE IN POLYSACCHARIDE
VACCINES
Test solution. Use a volumetric flask with a suitable vo!ume for
preparation of a solution containing about 5 mg per millilitre
of dry polysaccharide. Transfer the contents of a container
quantitatively to the flask and dilute to volume with water R.
Dilute the solution so that the volumes used in the test contain
2.5 flg to 25 flg of ribose. Introduce 0.20 mL and 0040 mL of
the diluted solution into tubes in triplicate.
Reference solutíons. Dissolve 25 mg of ribose R in water R
and dilute to 100.0 mL with the same solvent (stock solution
containing 0.25 giL of ribose). Immediately before use, dilute
1 mL of the stock solution to 10.0 mL with water R (working
dilution: 25 mg/L ofribose). Introduce 0.10 mL, 0.20 mL,
0040 mL, 0.60 mL, 0.80 mL and 1.0 mL of the working dilution
into 6 tubes.
Prepare a blank using 2 mL of water R.
Make up the volume in each tube to 2 mL with water R.
Shake. Add 2 mL of a 0.5 giL solution offerric chloride R in
hydrochloric acid R to each tube. Shake. Add 0.2 mL of a
100 giL solution of orcinol R in ethanol R. Place the tubes
in a water-bath for 20 mino Coo! in iced water. Measure the
absorbance (2.2.25) of each solution at 670 nm using the blank
as the compensation liquido Draw a calibration curve from
the absorbance readings for the 6 reference solutions and the
corresponding content of ribose and read from the curve the
quantity of ribose in the test solution for each volume tested.
Calculate the mean of the 3 values.

01/2008:20532

2.5.32. WATER: MICRO


DETERMINATION
PRINCIPLE
The coulometric titration of water is based upon the
quantitative reaction of water with sulfur dioxide and iodine in
an anhydrous medium in the presence of a base with sufficient
buffering capacity. In contrast to the volumetric method
Figure 2.5.29.-1.- Apparatus for the determination of sulfur described under (2.5.12), iodine is produced electrochemically
dioxide in the reaction ceH by oxidation of iodide. The iodine

164 See the informatíon section on general monographs (cover pages)


EUROPEAN PHARMACOPOEIA 8.0 2.5.33. Total protein

produced at the anode reacts immediately with the water and 01/2008:20533
the sulfur dioxide contained in the reaction cell. The amount corrected 6.0
of water in the substance is directly proportional to the
quantity of electricity up until the titration end-point. When 2.5.33, TOTAL PROTEIN
all of the water in the ceH has been consumed, the end-point
is reached and thus an excess of iodine appears. 1 mole of Many of the assay methods described in this chapter can be
iodine corresponds to 1 mole of water, a quantity of electricity performed using kits from commercial sources.
of 10.71 C corresponds to 1 mg of water.
METHOD 1
Moisture is eliminated from the system by pre-electrolysis.
Protein in solution absorbs ultraviolet light at a wavelength
Individual determinations can be carried out successively in
of 280 nm, due to the presence of aromatic amino acids,
the same reagent solution, under the following conditions:
mainly tyrosine and tryptophan, in the pro te in structure. This
- each component of the test mixture is compatible with the property can be used for assay purposes. If the buffer used to
other components, dissolve the pro te in has a high absorbance relative to that of
- no other reactions take place, water, an interfering substance is present. This interference
may be obviated by using the buffer as compensation
- the volume and the water capacity of the e1ectrolyte reagent liquid but if the interfering substance produces a high
are sufficient. absorbance, the results may nevertheless be compromised.
Coulometric titration is restricted to the quantitative At low concentratiol1S, protein adsarbed onto the cell may
determination of small amounts of water, a range of 1O ~lg up significantly reduce the content in solution. This can be
to 10 mg of water is recommended. prevented by preparing samples at higher concentration or by
using a non-ionic detergent in the preparation.
Accuracy and precision of the method are predominantly
governed by the extent to which atmospheric moisture is Test solution. Dissolve a suitable quantity of the substance
excluded from the system. Control of the system must be to be examined in the prescribed buffer to obtain a solution
monitored by measuring the amount of baseline drift. having a protein concentration between 0.2 mg/mL and
2 mg/mL.
APPARATUS Reference solution. Prepare a solution of a suitable reference
substance for the protein to be determined, in the same buffer
The apparatus consists of a reaction cel!, electro des and and at the same protein concentration as the test solution.
magnetic stirrer. The reaction cell consists of a large anode
compartment and a smaller cathode compartment. Depending Procedure. Keep the test solution, the reference solution and
on the design of the electrode, both compartments can be the compensation liquid at the same temperature during the
separated by a diaphragm. Each compartment contains performance of this test. Determine the absorbances (2.2.25)
a platinum electrode. Liquid or solubilised samples are of the test solution and the reference solution in quartz ceUs
introduced through a septum, using a syringe. Alternatively, at 280 nm, using the prescribed buffer as the compensation
an evaporation technique may be used in which the sample liquido The response must be linear in the range of protein
is heated in a tube (oven) and the water is evaporated and concentrations to be assayed to obtain accurate results.
carried into the cel! by means of a stream of dry inert gas. The Light scattering. The accuracy of the determinatiol1 of protein
introduction of solid samples into the cell should in general be can be diminished by the scattering oflight by the test sample.
avoided. However, if it has to be done it is effected through a If the proteins in solution exist as particles comparable in size
sealable port; appropriate precautions must be taken to avoid to the wavelength of the measuring light (250 nm to 300 11m),
the introduction of moisture from air, such as working in a scattering of the light beam results in an apparent in crease in
glove box in an atmosphere of dry inert gas. The analytical absorbance of the test sample. To calculate the absorbance at
procedure is controlled by a suitable electronic device, which 280 nm due to light scattering, determine the absorban ces of
also displays the results. the test solutio11 at wavelengths of 320 nm, 325 nm, 330 nm,
335 nm, 340 nm, 345 11m and 350 nm. Plot the logarithm
METHOD of the observed absorbance against the logarithm of the
Fill the compartments of the reaction cell with electrolyte wavelength and determine the standard curve best fitting the
reagent for the micro determination of water R according to plotted points by linear regression. Extrapolate the curve to
the manufacturer's instructions and perform the coulometric determine the logarithm of the absorbance at 280 nm. The
titration to a stable end-point. Introduce the prescribed antilogarithm of this value is the absorbance attributed to
amount of the substance to be examined into the reaction cel!, light scattering. Correct the observed values by subtracting
stir for 30 s, if not otherwise indicated in the monograph, and the absorbance attributed to light scattering from the total
titrate again to a stable end-point. In case an oven is used, absorbance at 280 11m to obtain the absorbance value of the
the prescribed sample amount is introduced into the tube protein in solution. Filtration with a 0.2 flm filter that do es
and heated. After evaporation of the water from the sample 110t adsorb protein or clarification by centrifugation may be
into the titration cel!, the titration is started. Read the value performed to reduce the effect oflight scattering, especially if
from the instrument's output and calculate if necessary the the solution is noticeably turbid.
percentage or amount of water that is present in the substance. Calculations. Use corrected values for the calculations.
When appropriate to the type of sample and the sample Calculate the concentration of protein in the test solution (Cu)
preparation, perform a blank titration. from the following equation:

VERIFICATION OF THE ACCURACY Cu = Cs (Au/As)


Between two successive sample titrations, introduce an where es is the concentration of protein in the reference
accurately weighed amount of water in the same arder of solution and Au and As are the corrected absorbances of the
magnitude as the amount of water in the sample, either as test solution and the reference solution, respectively.
water R or in the form of standard so/ution for the micro
determination of water R, and perfarm the coulometric METHOD 2
titration. The recovery rate is within the range from 97.5 per This method (commonly referred to as the Lowry
cent to 102.5 per cent for an addition of 1000 flg of HP and in assay) is based on the reduction by protein oí the
the range from 90.0 per cent to 110.0 per cent for the addition phosphomolybdotungstic mixed acid chromogen
of 100 flg of Hp. in the phosphomolybdotungstic reagent, which

General Notices (1) apply to all monographs and other texts 165
2.5.33. Total protein EUROPEAN PHARMACOPOEIA 8.0

results in an absorbance maximum at 750 nm. The a vortex mixer and allow to stand at room temperature for
phosphomolybdotungstic reagent reacts primarily with 10 mino Add 0.1 mL of a 720 giL solution of trichloroacetic
tyrosine residues in the protein. Colour development reaches acid R and mix using a vortex mixer. Centrifuge at 3000 g for
a maximum in 20 min to 30 min at room temperature, 30 min, decant the liquid and remove any residualliquid with
after which there is a gradualloss of colour. Because the a pipette. Redissolve the protein pellet in 1 mL of alkaline
method is sensitive to interfering substances, a procedure copper reagent.
for precipitation of the pro te in from the test sample may be
used. Most interfering substances cause a lower colour yield; METHOD 3
however, sorne detergents cause a slight increase in colour. This method (commonly referred to as the Bradford assay)
A high salt concentration may cause a precipitate to formo is based on the absorption shift from 470 nm to 595 nm
Because different protein species may give different colour observed when the acid blue 90 dye binds to protein. The acid
response intensities, the reference substance and test protein blue 90 dye binds most readily to arginine and Iysine residues
must be the same. Where separation of interfering substances in the protein which can lead to variatiol1 in the response of
from the protein in the test sample is necessary, proceed as the assay to different proteins. The protein used as reference
directed below for interfering substances prior to preparation substance must therefore be the same as the protein to be
of the test solution. The effect of interfering substances may determined. There are relatively few interfering substances,
be minimised by dilution, provided the concentration of the but it is preferable to avoid detergents and ampholytes in the
test protein remains sufficient for accurate measurement. test sample. Highly alkaline samples may interfere with the
Use distilled water R to prepare all buffers and reagents used acidic reagent.
for this method. Use distilled water R to prepare al! buffers and reagents used
Test solution. Dissolve a suitable quantity of the substance for this method.
to be examined in the prescribed buffer to obtain a solution Test solution. Dissolve a suitable quantity of the substance
having a concentration within the range of the standard curve. to be examined in the prescribed buffer to obtain a solution
A suitable buffer will produce a solution of pH 10.0 to 10.5. having a concentration within the range of the standard curve.
Reference solutions. Dissolve the reference substance for the Reference solutions. Dissolve the reference substance for the
protein to be determined in the prescribed buffer. Dilute protein to be determined in the prescribed buffer. Dilute
portions of this solution with the same buffer to obtain portions of this solution with the same buffer to obtain
not fewer than five reference solutions having protein not fewer than five reference solutions having protein
concentrations evenly spaced over a suitable range situated concentrations evenly spaced over a suitable range situated
between 5 flg/mL and 100 flg/mL. between 0.1 mg/mL and 1 mg/mL.
Blanlc. Use the buffer used to prepare the test solution and Blank. Use the buffer used to prepare the test solution and
the reference solutions. the reference solutions.
Copper sulfate reagent. Dissolve 100 mg of copper sulfate R Acid blue 90 reagent. Dissolve 0.10 g of acid blue 90 R in
and 0.2 g of sodium tartrate R in distilled water R and dilute 50 mL of alcohol R. Add 100 mL of phosphoric acid R, dilute
to 50 mL with the same solvent. Dissolve 10 g of anhydrous to 1000 mL with distilled water R and mix. Filter the solution
sodium carbonate R in distilled water R and dHute to 50 mL and store in an amber bottle at room temperature. Slow
with the same solvent. Slowly pour the sodium carbonate precipitation of the dye occurs during storage. Filter the
solution into the copper sulfate solution with mixing. Use reagent before using.
within 24 h. Procedure. Add 5 mL of acid bIne 90 reagent lo 0.100 111L of
Alkaline copper reagent. Mix 1 volume of copper sulfate each reference solution, of the test solution and of the blank.
reagent, 2 volumes of a 50 giL solution of sodium dodecyl Mix by inversion. Avoid foaming, which will Jead to poor
sulfate R and 1 volume of a 32 giL solution of sodium reproducibility. Determine the absorbances (2.2.25) of the
hydroxide R. Stofe at room temperature and use within standard solutiol1s and of the test solution at 595 nm, using
2 weeks. the blank as compensation liquido Do 110t use quartz (silica)
Diluted phosphomolybdotungstic reagent. Mix 5 mL of spectrophotometer cells because the dye binds to this material.
phosphomolybdotungstic reagent R with 55 mL of distilled Calculations. The relationship of absorbance to protein
water R. Sto re in an amber bottle, at room temperature. concentration is non-linear; however, ifthe range of
Procedure. To l.0 mL of each reference solution, of the test concentrations used to prepare the standard curve is
solution and of the blank, add 1.0 mL of alkaline copper sufficiently sma11, the latter will approach linearity. Plot
reagent and mix. Allow to stand for 10 mino Add 0.5 mL of the absorbances of the reference solutions against protein
the diluted phosphomolybdotungstic reagent, mix and allow concentrations and use linear regression to establish the
to stand at room temperature for 30 mino Determine the standard curve. From the standard curve and the absorbance
absorbances (2.2.25) of the solutions at 750 nm, using the of the test solution, determine the concentration of protein in
solution from the blank as compensation liquido the test solution.
Cakulations. The relationship of absorbance to protein METHOD 4
concentration is non -linear; however, if the range of This method (eommonly referred to as the bicinchoninic acid
concentrations used to prepare the standard curve is or BCA assay) is based on reduchon of the cuprie (Cu 2+) ion to
sufficiently small, the latter will approach linearity. Plot the cuprous (Cu ll ) ion by protein. The bicinchoninic acid reagent
absorbances of the reference solutions against the pro te in is used to detect the cuprous ion. Few substances interfere
concentrations and use linear regression to establish the with the reaction. When interfering substances are present
standard curve. From the standard curve and the absorbance their effect may be minimised by dilution, provided that
of the test solution, determine the concentration of protein in the concentration of the protein to be determined remains
the test solution. sufficient for accurate measurement. Alternatively, the protein
Interfering suostances. In the following procedure, precipitation procedure given in Method 2 may be used to
deoxycholate-trichloroacetic acid is added to a test sample remove interfering substances. Because different protein
to remove interfering substances by precipitation of proteins species may give different colour response intensities, the
before determination; this technique can also be used to reference protein and protein to be determined must be the
concentrate proteins from a dilute solution. same.
Add 0.1 mL of a 1.5 giL solution of sodium deoxycholate R io Use distilled water R to prepare al! buffers and reagents used
1 mL of a solution of the substance to be examined. Mix using for this method.

166 See the informatian section on general monographs (cover pages)


PHARMACOPOEIA 8.0 2.5.33. Total protein

Test solution. Dissolve a suitable quantity of the substance Biuret reagent. Dissolve 3.46 g of copper sulfate R in 10 l11L of
to be examined in the prescribed buffer to obtain a solution hot distilled water R, and allow to cool (Solution Dissolve
a concentration within the range of the concentrations 34.6 g of sodium citrate R and 20.0 g of anhydrous sodium
of the reference solutions. carbonate R in 80 mL of hot distilled water R, and allow to
solutions. Dissolve the reference substance for the cool (Solution B). Mix solutions A and B and dilute to 200 mL
to be determi11ed in the prescribed buffer. Dilute with distilled water R. Use within 6 l110nths. Do 110t use the
of this soludon with the same buffer to obtain reagent if it develops turbidity or contains any precipitate.
not fewer than five reference solutions having protein Procedure. To one volume of the test solution add an equal
concentrations evenly spaced over a suitable range situated volume of a 60 giL solution of sodiurn hydroxide R and mix.
between 10 ~lg/mL and 1200 flg/mL Immediately add biuret reagent equivalent to 0.4 volumes
Blank. Use the buffer used to prepare the test solution and of the test solution and mix rapidly. Allow to stand at a
the reference solutions. temperature between 15 oC and 25 oC for not less than 15 mino
Within 90 min of addition of the biuret reagent, determine
BCA reagent. Dissolve 10 g of disodium bicinchoninate R, the absorbances (2.2.25) of the reference solutions and of the
20 g of sodium carbonate monohydrate R, 1.6 g of sodium test solution at the maximum at 545 11m, using the blank as
tartrate R, 4 g of sodium hydroxide R, and 9.5 g of sodium compensation liquido Any solution that develops turbidity
hydrogen carbonate R in distilled water R. Adjust, if necessary, or a precipitate is not acceptable for calculation of protein
to pH 11.25 with a solution of sodium hydroxide R 01' a COl1centration.
solution of sodium hydrogen carbonate R. Dilute to 1000 mL
with distilled water R and mix. Cakulations. The relationship of absorbance to protein
concentration is approximately linear within the indicated
Copper-BCA reagent. Mix 1 mL of a 40 giL solution of copper range of protein concentrations for the reference solutions.
sulfate R and 50 mL of BCA reagent. Plot the absorban ces of the reference solutions against protein
Procedure. Mix 0.1 mL of each reference solution, of the concentrations and use linear regression to establish the
test solution and of the blank with 2 mL of the copper-BCA standard curve. Calculate the correlation coefficient for the
reagent. Incubate the solutions at 37 oC for 30 min, note the standard curve. A suitable system is Ol1e that yields a line
time and allow the mixtures to cool to room temperature. having a correlation coefficient not les s than 0.99. From
Within 60 min of the end of incubation, determine the the standard curve and the absorbance of the test solution,
absorbances (2.2.25) of the reference solutions and of the determine the concentration of protein in the test solution.
test solution in quartz cells at 562 nm, using the blank as Interfering substances. To minimise the effect of interfering
compensation liquido After the solutions have cooled to substances, the protein can be precipitated from the test
room temperature, the colour intensity continues to increase sample as follows: add 0.1 volumes of a 500 giL solution of
gradually. trichloroacetic acid R to 1 volume of a solution of the test
Cakulations. The relationship of absorbance to protein sample, withdraw the supernatant and dissolve the
concentration is non -linear; however, if the range of precipitate in a small volume of 0.5 sodium hydroxide. Use
concentrations used to prepare the standard curve is the solution obtained to prepare the test solution.
sufficiently small, the latter will approach linearity. Plot
the absorbances of the reference solutions against protein METHOD 6
concentrations and use linear regression to establish the
standard curve. From the standard curve and [he absorbance This fluorimetric method is based on the derivatisation of
of the test solution, determine the concentration of protein in the protein with o-phthalaldehyde, which reacts with the
the test solution. primary amines of the protein (N-terminal amino acid and
the E-amino group of lysine residues). The sensitivity of the
METHOD 5 assay can be increased by hydrolysing the protein before
adding o-phthalaldehyde. Hydrolysis makes the o.-amino
This method (commonly referred to as the biuret assay) is
group of the constituent amino acids available for reaction
based on the interaction of cupric (Cu 2+) ion with protein in
with the phthalaldehyde reagent. The method requires
alkaline solution and resultant development of absorbance
very small quantities of the protein. Primary amines, such
at 545 nm. This test shows minimal difference between
as tris(hydroxymethyl)aminomethane and amino acid
equivalent IgG and albumin samples. Addition of the sodium
buffers, react with phthalaldehyde and must be avoided
hydroxide and the biuret reagent as a combined reagent,
or removed. Ammonia at high concentrations reacts with
insufficient mixing after the addition of the sodium hydroxide,
phthalaldehyde. The fluorescence obtail1ed when amine reacts
or an extended time between the addition of the sodium
with phthalaldehyde can be unstable. The use of automated
hydroxide solution and the additiol1 of the biuret reagent will
procedures to standardise this procedure may improve the
give IgG sal11ples a higher response than albumin samples.
accuracy and precision of the test.
The trichloroacetic acid l11ethod used to minimise the effects
of interfering substances also can be used to determine the Use distilled water R to prepare all buffers and reagents used
protein content in test sal11ples at concentrations below for this method.
500 flg/mL
Test solution. Dissolve a suitable quantity of lhe substance to
Use distilled water R to prepare all buffers and reagents used be examined in a 9 giL solution of sodium chloride R to obtain
for this method. a solution having a concentration within the range of the
Test solution. Dissolve a suitable quantity of the substance concentrations of the reference solutions. Adjust the solution
to be examined in a 9 giL solution of sodium chloride R to to pH 8 to 10.5 before addition of the phthalaldehyde reagent
obtain a solution having a concentration within the range of Reference solutions. Dissolve the reference substance for
the concentrations of the reference solutions. the protein to be determined in a 9 giL solution of sodium
Reference solutions. Dissolve the reference substance for chloride R. Dilute portions of this solution with a 9 giL solutiol1
the protein to be deterl11ined in a 9 giL solution of sodium of sodium chloride R to obtain 110t fewer than five reference
chloride R. Dilute portions of this solution with a 9 giL solution solutions having protein concentrations evenly spaced over
of sodium chloride R to obtain not fewer than three reference a suitable range situated between 10 Vg/mL and 200 flg/mL
solutions having protein concentrations evenly spaced over a Adjust the solutions to pH 8 to 10.5 before addition of the
suitable range situated between 0.5 mg/mL and 10 mg/mL phthalaldehyde reagent.
Blanlc Use a 9 solution of sodium chloride R. Blank solution. Use a 9 giL solution of sodium chloride R.

General Notices (1) apply to all monographs and other texts 167
2,5.34. Acetic acid in synthetic peptides EUROPEAN PHARMACOPOEIA 8.0

Borate buffer solution. Dissolve 61.83 g of borie acid R in Test solution. Prepare as described in the monograph. The
distilled water R and adjust to pH 10.4 with a solution of concentration of peptide in the solution may be adapted,
potassium hydroxide R. Dilute to 1000 mL with distilled depending on the expected amount of acetic acid in the
water R and mix. sample.
Phthalaldehyde stock solution. Dissolve 1.20 g of Reference so/ution. Prepare a 0.10 giL solution of glacial
phthalaldehyde R in 1.5 mL of methanol R, add 100 mL of acetic acid R in a mixture of 5 volumes of mobile phase B and
borate buffer solution and mix. Add 0.6 mL of a 300 giL 95 volumes of mobile phase A.
solution of macrogol 23 lauryl ether R and mix. Store at room
The chromatographic procedure may be carried out using:
temperature and use within 3 weeks.
Phthalaldehyde reagent. To 5 mL of phthalaldehyde stock - a stainless steel column 0.25 m long and 4.6 mm in
solution add 15 flL of 2-mercaptoethanol R. Prepare at least internal diameter packed with octadecylsi/yl si/ica gel for
30 min before use. Use within 24 h. chromatography R (5 flm),
Procedure. Mix 10 flL of the test solution and of each of the - as mobile phase at a flow rate of 1.2 mL/min:
reference solutions with 0.1 mL of phthalaldehyde reagent and Mobile phase A. Dilute 0.7 mL of phosphorie acid R to
allow to stand at room temperature for 15 mino Add 3 mL of 1000 mL with water R; adjust the pH to 3.0 with strong
0.5 M sodium hydroxide and mix. Determine the fluorescent sodium hydroxide solution R,
intensities (2.2.21) of solutions from the reference solutions
Mobile phase B. Methanol R2,
and from the test solution at an excitation wavelength of
340 nm and an emission wavelength between 440 and 455 nm. Time Mobile phase A Mobile phase B
Measure the fluorescent intensity of a given sample only once, (min) (per cent V/V) (per cent V/V)
since irradiation de creases the fluorescence intensity. O- 5 95 5
Calculations. The relationship of fluorescence to protein 5 - 10 95 -7 50 5 -7 50
concentration is linear. Plot the fluorescent intensities of
the reference solutions against protein concentrations and 10 - 20 50 50
use linear regression to establish the standard curve. From 20 - 22 50 -7 95 50 -7 5
the standard curve and the fluorescent intensity of the test
solution, determine the concentration of protein in the test 22 - 30 95 5
solution.
- as detector a spectrophotometer set at 210 nm.
METHOD 7
lnject 10 flL of the reference solution and 10 flL of the
This method is based on nitro gen analysis as a means of test solution. In the chromatograms obtained, the peak
protein determination. Interference caused by the presence of corresponding to acetic acid has a retention time of 3-4 mino
other nitrogen-containing substances in the test sample can The baseline presents a steep rise after the start of the linear
affect the determination of protein by this method. Nitrogen gradient, which corresponds to the elution of the peptide
analysis techniques destroy the test sample during the analysis from the column. Determine the content of acetic acid in the
but are not limited to protein presentation in an aqueous peptide.
environment.
Pro ce dure A. Proceed as prescribed for the determination of
nitrogen by sulfuric acid digestion (2.5.9) or use commercial
instrumentation for Kjeldahl nitro gen assay.
01/2011 :20535
Procedure B. Commercial instrumentation is available
for nitro gen analysis. Most nitro gen analysis instruments
use pyrolysis (Le. combustion of the sample in oxygen at 2.5.35. NITROUS OXIDE IN GASES
temperatures approaching 1000 oC), which produces nitric
oxide (NO) and other oxides of nitro gen (NO) from the Gases absorb light at one or more specific wavelengths. This
nitrogen present in the substance to be examined. Sorne property is widely used to allow highly selective measurement
instruments convert the nitric oxides to nitro gen gas, which of their concentrations.
is quantified using a thermal-conductivity detector. Other Description and principIe of measurement. The
instruments mix nitric oxide (NO) with ozone (03) to produce concentration of nitrous oxide in other gases can be
excited nitro gen dioxide (N0 2*), which emits light when determined using an infrared analyser.
it decays and can be quantified with a chemiluminescence
The infrared analyser generally consists of a light source
detector. A protein reference material that is relatively pure
emitting broadband infrared radiation, an optical device,
and is similar in composition to the test proteins is used
a sample cell and a detector. The optical device may be
to optimise the injection and pyrolysis parameters and to
positioned either before or after the sample cel! and it consists
evaluate consistency in the analysis.
of one or several optical filters, through which the broadband
Cakulations. The protein concentration is calculated by radiation is passed. The optical device in this case is selected
dividing the nitro gen content of the sample by the known for nitrous oxide. The measurement light beam passes through
nitrogen content of the protein. The known nitro gen the sample cel! and may also pass through a reference cel! if
content of the protein can be determined from the chemical the analyser integrates such a feature (sorne use an electronic
composition of the protein or by comparison with a suitable system instead of a reference ceH).
reference substance.
When nitrous oxide is present in the sample cel!, absorption
of energy in the measurement light beam will occur according
to the Beer-Lambert law and this produces a change in
01/2008:20534 the detector signaL This measurement signal is compared
to a reference signal to generate an output related to the
concentration of nitrous oxide. The generated signal is
2.5.34. ACETIC ACID IN SYNTHETIC linearised in order to obtain the nitrous oxide concentration.
PEPTIDES To prevent the entry of particles into the sensors, which could
cause stray-light phenomena, the apparatus is fitted with a
Examine by liquid chromatography (2.2.29). suitable fiIter.

168 See the information section on general monographs (ca ver pages)
EUROPEAN PHARMACOPOEIA 8.0 2.5.37. MM S, EMS and IMS in methanesulfofik acid

0112008:20536 Reference so/ution Dilute 3.0 mL of reference solution (a)


to 10.0 mL with the internal standard solution.
ANISIDINE VALUE Column:
- material: fused silica;
The anisidine value is defined as 100 times the optical density
- size: 1 = 15 m, (7) = 0.25 mm;
measured in a 1 cm cel! of a solution containing 1 g of the
substance to be examined in 100 mL of a mixture of solvents - stationary phase: )lIIS·llO.>Ca11e R (film thickness
and reagents according to the following method. 1 ¡.tm).
Carrier gas: mntnown11h,! R.
out the operations as as possible, avoiding
exposure to actinic light. Flow rate: lmL/min.
Test solution (a). Dissolve 0.500 g of the substance to be Pu/sed splitless: 250 kPa, 0.25 min.
examined in trimethylpentane R and dilute to 25.0 mL with Temperature:
the same solvent. Thne Temperature
Test solution (b). To 5.0 mL of test solution (a) add 1.0 mL of a (min) (oC)
2.5 giL solution of p-anisidine R in glacial acetic acid R, shake Column o 1 55
and 5tOfe protected from light.
1-9 55 ~ 135
Reference solution. To 5.0 mL of trimethylpentane R add
1.0 mL of a 2.5 giL solution of p-anisidine R in glacial acetic lnjection port 240
acid R, shake and sto re protected from light.
Detector: transfer line 280
Measure the absorbance (2.2.25) of test solution (a) at
the maximum at 350 nm using trimethylpentane R as source 230
the compensation liquido Measure the absorbance of test analyser 150
solution (b) at 350 nm exactly 10 min after its preparation,
usíng the reference solution as the compensation liquido Deteetíon: mass spectrometer as described below; adjust the
Calculate the anisidine value from the expression: detector settings so as to comply with the system suitability
criteria:
25 X (1.2A l - A2 )
- quadrupole mass spectrometer equipped with an electron
ln impact ionisation mode (70 eV);
- mass spectrometer parameters fOI the fragmentometric
absorbance of test solutíon (b) at 350 nm,
mode (single-ion monitoring (SIM)) set as follows:
absorbance of test solution (a) at 350 nm,
Duration of
Substance miz
m mass of the substance to be examined in test monitoring
so!ution (a), in grams. Butyl methanesulfonate (BMS) 56
tR between 7.0 min
and 9.0 min
tR between 2.0 min
Methyl methanesulfonate (MMS) 80
and 3.5 min
04/2011:20537 Ethyl methanesulfonate (EMS) 79
tR between 4.0 111in
and 4.7 111in
tR between 4.7 111in
2.5.37. METHYL, ETHYL AND lsopropyl methanesulfonate (IMS) 123
and 5.5 min
ISOPROPYL METHANESULFONATE IN Injection: 2 ¡.tL.
METHANESULFONIC ACID Relative retentiol1 with reference to the internal
standard (retention time = about 7.6 min):
The following method has been validated for the methyl, ethyl MMS = about 0.3; EMS = about 0.5; IMS = about 0.6.
and isopropyl esters of methanesulfonic acid at concentrations System suitability:
in the range of 0.5 ppm to 100 ppm.
- resolution: minimum 3.0 between the peaks due to EMS
If it is intended to be used to determine levels of and IMS in the chromatogram obtained with reference
methanesulfonic acid esters outside this validated range, for solution (a);
example in early steps of the synthesis prior to their removal,
the concentration of the test solution has to be adjusted - signal-to-noise ratio: minimum 10 for the peaks due to
accordingly. MMS, EMS and IMS in the chromatogram obtained with
reference solution (b) o

Gas chromatography (2.2.28) coupled with mass spectrometry


Calculate the content of MMS, EMS or IMS in parts per
(2.2.43).
million using the following expre5sion:
Internal standard solution. Dilute 7 ¡.tL of butyl
methanesulfonate CRS (BMS) to 10.0 mL with methylene A 2 x h X W l X e x 0.148
chloride R. Dilute 10 ¡.tL of the solution to 100.0 mL with Al x h X W 2
methylene chloride R.
Test solution. Add 0.74 g ofthe substance to be examined to are a of the peak due to MMS, EMS or IMS
lO.O mL of water R and extract with 10.0 mL of the internal in the chromatogram obtained with reference
standard solution. Allow to separate and transfer the organic solution (a);
layer to a vial containing anhydrous sodium sulfate R. Shake area of the peak due to MMS, EMS or IMS in the
and filter. chromatogram obtained with the test solution;
Reference solution (a). Dissolve 50 mg each of methyl C percentage content of MMS, EMS or IMS;
methanesulfonate R (MMS), ethyl methanesulfonate R (EMS)
are a of the peak due to the internal standard
and isopropyl methanesulfonate R (IMS) in the internal
in the chromatogram obtained with reference
standard solution and dilute to 50.0 mL with the same
solution (a);
solution. Dilute 74 ¡.tL of the solution to 10.0 mL with the
internal standard solution. Dilute 100 ¡.tL of this solution to are a of the peak due to the internal standard in the
10.0 mL with the internal standard solution. chromatogram obtained with the test solution;

General Notices (1) apply to all monographs and other texts 169
2.5.38. MM S, EMS and IMS in active substances EUROPEAN PHARMACOPOEIA 8.0

W¡ mass of MMS, EMS or IMS used to prepare Split ratio: 1:20.


reference solution (a), in milligrams; Static head-spaee conditions that may be used:
W2 mass of the substance to be examined in the test equilibration temperature: 60 oC;
solution, in milligrams; - equilibration time: 30 min;
0.148 == dilution factor. - transfer-line temperature: 120 oc.
Temperature:
Time Temperature
0112012:20538 (min) (oC)
Column o- 1 40
2.5.38. METHYL, ETHYL AND 1 - 10 40.." 130
ISOPROPYL METHANESULFONATE IN Injection port 220
ACTIVE SUBSTANCES
Detector transfer line 280
The following general method has been validated for the source 250
determination of methyl, ethyl and isopropyl esters of
methanesulfonic acid (in concentrations between 0.2 ppm and analyser 200
5 ppm) in betahistine mesilate.
lf it is intended to use the method for other active substances, At the end of analysis the temperature of the column is raised
particularly those that contain different concentrations of the to 240 oC and maintained at this temperature for 7 mino
methanesulfonic acid esters, the concentrations of the test Deteetian: mass spectrometer as described below; adjust the
solution and reference solution must be adjusted accordingly detector settings so as to comply with the system suitability
and the method must be suitably validated. criteria; alternatively a suitable electron -capture detector may
be used:
METHOD
- quadrupole mass spectrometer equipped with an electron
Head-space gas chromatography (2.2.28) coupled with mass impact ionisation mode (70 eV);
spectrometry (2.2.43). Prepare the test solution and referenee
- mas s spectrometer parameters for the fragmentometric
solutions immediately before use.
mode (single-ion monitoring (SIM)) set as follows:
Solvent mixture: water R, acetonitrile R (20:80 VIV).
Quantitation ion Qualification ion
Solution A. Dissolve 30 mg of anhydrous sodium thiosulfate R Substance
(miz) (miz)
and 60.0 g of sodium iodide R in water R and dilute to 50.0 mL
with the same solvent. Butyl iodide (Bu!)* 184 127
Internal standard solution. Dilute 10 [1L of butyl Methyl iodide (MeI)* 142 127
methanesulfonate CRS (BMS) to 10.0 mL with the solvent
mixture. Dilute 20 [1L of the solution to 10.0 mL with the Ethyl iodide (Etl)* 156 127
solvent mixture. Dilute 10.0 mL of this solution to 100.0 mL Isopropyl iodide
170 127
with the solvent mixture. (iPrI)*

Test solution. Weigh 25.0 mg of the substance to be examined * formed from BMS, MM S, EMS and IMS in the derivatisation reaction.
into a 20 mL headspace vial, add 0.50 mL of solution A and
Injection: 1 mL of the gas phase of the test solution and
0.50 mL of the internal standard solution, then seal the vial
reference solutions (b) and (c).
immediately with a polytetrafluoroethylene-coated silicon
membrane and an aluminium cap. Following the derivatisation Relative retention with reference to the internal standard
reaetion, a precipita te may be observed, however this does not (BuI) (retention time == about 8.5 min): MeI == about 0.51 ;
affeet the validity of the quantification. EtI == about 0.63; iPrI == about 0.68.
Reference solution (aJ. Dissolve 25.0 mg each of methyl System suitability:
methanesulfonate R (MMS), ethyl methanesulfonate R (EMS) - resolution: minimum 1.5 between the peaks due to EtI
and isopropyl methanesulfonate R (IMS) in toluene R and and iPrI in the chromatogram obtained with reference
dilute to 5.0 mL with the same solvent. Dilute 50 [1L of the solution (c);
solution to 25.0 mL with the internal standard solution. - signal-to-noise ratio: minimum 10 for the peak due to each
Referenee solution (bJ. Dilute 20 [1L ofreference solution (a) alkyl iodide in the chromatogram obtained with reference
to 20.0 mL with the internal standard solution. Introduce solution (b).
0.50 mL of this solution and 0.50 mL of solution A into a
Calculate the content in parts per million of each alkyl
20 mL headspace vial and seal the vial immediately with
methanesulfonate using the following expression:
a polytetrafluoroethylene-coated silicon membrane and an
aluminium cap. A 2 x Ir X W l X e x 0.05
Reference solutian (ej. Dilute 500 I1L of reference solution (a) Al x I 2 X W2
to 20.0 mL with the internal standard solution. Introduce
0.50 mL of this solution and 0.50 mL of solution A into a A¡ area of the peak due to each alkyl iodide in
20 mL headspace vial and seal the vial immediately with the chromatogram obtained with reference
a polytetrafluoroethylene-coated silicon membrane and an solution (c);
aluminium cap. A2 area of the peak due to each alkyl iodide in the
Column: chromatogram obtained with the test solution;
material: fused silica; C percentage content of each ester;
- size: 1 == 30 m, (2) == 0.25 mm; area of the peak due to the internal standard
- stationary phase: polar-deaetivated polyethyleneglycol R in the chromatogram obtained with reference
(film thickness 1 [1m). solution (c);
Carrier gas: helium for chromatography R. area of the peak due to the internal standard in the
Flow rate: 0.5 mL/min. chromatogram obtained with the test solution;

170 See the information sectian on general monagraphs (cover pagesJ


EUROPEAN PHARMACOPOEIA 8.0 2.5.39. Methanesulfonyl chloride in methanesulfonic add

mass of each ester used to prepare reference At the end of analysis the temperature of the column is raised
solution (a), in milligrams; to 270 oC and maintained at this temperature for 8 mino
mass of the substance to be examined in the test Deteetion: mass spectrometer as described below; adjust the
sollltion, in milligrams; detector settings so as to comply with the system suitability
0.05 dillltion factor. criteria:

- quadrupole mass speetrometer equipped with an eleetron


04/2013:20539
impact ionisation mode (70 e V);
2.5.39. METHANESULFONYL - mas s spectrometer parameters for the fragmentometric
CHLORIDE IN METHANESULFONIC mode (single-ion monitoring (SIM)) set as follows:
ACID
Substance miz Duration of monitoring
The following method has been validated for the determination Methanesulfonyl
of methanesulfonyl chloride in methanesulfonic acid at 79 tR between 3.3 min and 6.0 min
chloride
concentrations in the range of 0.05 ppm to 50 ppm. Butyl methane-
56 tR between 6.0 min and 8.0 min
Gas chromatography (2.2.28) coupled with mass spectrometry sulfonate (BMS)
(2.2.43).
Internal standard solution. Dissolve 7 flL of butyl Injection: 5 flL oí the test solution, reference solutions (b)
methanesulfonate CRS (BMS) in methylene ehloride R and and (e), the internal standard solution and methylene
dilute to 10.0 mL with the same solvent. Dilute 5.0 mL of this ehloride R.
solution to 50.0 mL with methylene ehloride R.
Relative retention with reference to the internal standard
Test solutíon. To 5 mL of water R, add 7.4 g of the substance
(BMS) (retention time = about 7.2 min): methanesulfonyl
to be examined and mix slowly. After cooling, add 5.0 mL
chloride = about 0.68.
of methylene ehloride R and 100 flL of the internal standard
solution and shake. Allow to separate and transfer the System suitability:
organic layer to a vial containing 1 g of anhydrous sodium
sulfate R. Repeat the extraction twice with 5.0 mL of methylene - in the chromatogram obtained with the internal standard
ehloride R each time, combine the organic layers and fiIter. solution, there is no peak with the same retention time as
Referenee solution (a). Dissolve 50.0 mg of methanesulfonyl methanesulfonyl chloride;
ehloride R in methylene ehloride R and dilute to 10.0 mL with
the same solvent. Dilute 1.0 mL of the solution to 10.0 mL - resolution: minimum 5.0 between the peaks dne to
with methylene ehloride R. Dilute 300 flL of this solution to methanesulfonyl chloride and BMS in the chrol11atogram
10.0 mL with methylene ehloride R. obtained with reference solution (b) ;
Reference solution (b). Dilute 500 flL ofreference solution (a)
- signal-to-noise ratio: minimum 10 for the peak due to
and 100 flL of the internal standard solution to 15.0 mL with methanesulfonyl chloride in the chromatogram obtained
methylene ehloride R.
with reference solutiol1 (c).
Referenee solution (e). Dilute 25 flL of reference solution (a)
and 100 ~lL of the internal standard solution to 15.0 mL with Calculate the content of methanesulfonyl chloride in parts per
methylene ehloride R. million using the Íollowing expression:
Column:
A 2 x h X W l X e x 1.5
- material: fused silica;
Al x h X W 2
- size: 1 = 15 m, 0 = 0.25 mm;
- stationary phase: poly(dimethyl)siloxane R (film thickness
area of the peak due to methanesulfonyl chloride
1 flm).
in the chromatogram obtained with referenee
Carrier gas: helium for ehromatography R. solution (b);
Flow rate: 1 mLlmin. are a of the peak due to methanesulfonyl chloride in
Pulsed splitless: 60 kPa, 0.1 mino the chromatogram obtained with the test solution;
Temperature: C percentage content oí methanesulfonyl chloride;
Time Temperature
(oC)
are a of the peak due to BMS in the chromatogram
(min)
obtained with reference solution (b);
Column 0-4 40
are a of the peak due to BMS in the chromatogram
4 - 8 40 -7 200 obtained with the test solution;
Injection port 240 l11ass of methanesulfonyl chloride used to prepare
reference solution (a), in milligrams;
Detector: transfer line 280
mass of the sample in the test solution, in
so urce 230
milligral11s ;
analyser 150 1.5 dilution factor.

General Notices (1) apply to all monographs and other texts 171
EUROPEAN PHARMACOPOEIA 8.0

172 See the information section on general monographs (caver pages)


EUROPEAN PHARMACOPOEIA 8.0

Biologi al ests
2.6. Biological tests ............... 175
00 • • • • • • • • • • • • • • • • , . • • • • • • • • • • • • • • • • • • • • • • • • • • • • • • • • 2.6.19. Test for neurovirulence of poliomyelitis vaccine
2.6.1. Sterilityoo 175
. 0 0 • • 00 • • 0 0 . 0 0 • • • • • 00 . . . 0 0 . 0 0 0 0 • • 0 0 . 0 0 0 0 • • • 0 0 0 0 . . . . . . . , . . . . . . . . . . . , . • • • • • , . (oral) ......................................................................................... 202
2.6.2. Mycobacteria,. ...... 178
0 0 . 0 0 0 0 . . . . . . . . . . . . . . . . , . 0 0 0 0 . 0 0 . . . . . . . . . . . . 00 . . . . . . . . . . 00 2.6.20. Anti-A and anti-B haemagglutinins ......... 203 00 . . . 00 . . 00 . . . . . .

2.6.7. Mycoplasmas ............................... 178 00 . . 00 . . 00 . . . . 0 0 . 0 0 0 0 • • 00 . . . . . . 0 0 0 0 2.6.21. Nucleic acid amplification techniques ....................... 204
2.6.8. Pyrogens ......................... 183
00 . . . . . . . . . . . . . . . . . . . . . . . . . 0 0 0 0 0 0 . . . . . . . . . . . . . . . . 2.6.22. Activated coagulation factors .... 209
00 . . 00 . . . 00 . . . . . . . . . . . . . . . . . . . . . .

2.6.9. Abnormal toxicity....... ,. ................ ,. ......................... 184 ,..00. 2.6.24. Avian viral vaccines: tests for extraneous agents in
2.6.10. Histamineoo .. 184
00 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0 0 0 0 . 0 0 . . 00 . . . . . . . . . 0 0 0 0 . . 00 . . 00 . . seed lots ................ 209
00 . . . . . . . . . . . . . . . . . . . . 00 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .

2.6.11. Depressor substances ...... oooo .... oo ...... oooooooooo .. oo .. oooooo.oo.oo. 185 2.6.25. Avian live virus vaccines: tests for extraneous agents in
2.6.12. Microbiological examination of non-sterile products: batches of finished prodllct.. ...... 212
00 . . . 00 . . 00 . . . . . . . . . . . . . . . . . . . . . 00 . . . . . . . . .

microbial enumeration testsoo ... oo .... oooooooooo.oo ........... oo .... oo ....... 185 2.6.26. Test for anti-D antibodies in human
2.6.13. Microbiological examination of non-sterile products: immunoglobulin ........................................................... 215 00 . . . . . . . .

test for specified micro-organisms .............. 189 00 . . . . . . . 00 . . . . . . . . . . . . . . 2.6.27. Microbiological control of cellular products ............ 216
2.6.14. Bacteria! endotoxins .. 194
00 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2.6.30. Monocyte-activatiol1 test... .......................................... 217
2.6.15. Prekallikrein activator.. ................. 198 00 . . . . . . . . . . . . . . . . . . . 0 0 . 0 0 . . . . 2.6.31. Microbiologica! examination of herbal medicinal
2.6.16. Tests for extraneous agents in viral vaccines for human products for oral use and extracts used in their
use ............................................................................................ 198 preparation .............................................................................. 222
2.6.17. Test for anticomplementary activity of 2.6.33. Residual pertussis toxin and irreversibility of pertussis
immunoglobulin ..................................................................... 200 toxoid ....................................................................................... 224
2.6.18. Test fOl' neuroviru!ence of live virus vaccines .......... 202

General Natices (1) apply lo all monographs and ather texts 173
EUROPEAN PHARMACOPOEIA 8.0

174 See the information section on general monographs (cover pages)


EUROPEAN PHARMACOPOEIA 8.0 2.6.1. Stedlity

BIOLOGICAL TESTS the containers in a water-bath or in free-flowing steam until


the pink colour disappears and cooling quiddy, taking care to
prevent the introduction of non-sterile air into the container.
04/2011:20601 Do not use the medium for a longer storage period than has
corrected 7.7 been validated.
Fluid thioglycollate medium is to be incubated at 30-35 oc.
2.6.1. STERILITY(l) For products containing a mercurial preservative that
cannot be tested by the membrane-filtration method, fluid
to substances, preparations or artíe/es
thioglycollate medium incubated at 20-25 oC may be used
which, to the Pharmacopoeia, are required ro be
instead of soya-bean casein digest medium provided that it
sterile. However, a satisfactory result only indicates that no
has been validated as described in growth promotion test.
contaminating micro-organism has been found in the sample
examined in the conditions of the test. Where prescribed or justified and authorised, the following
alternative thioglycollate medium may be used. Prepare a
PRECAUTIONS AGAINST MICROBIAL mixture having the same composition as that of the fluid
CONTAMINATION thioglycollate medium, but omitting the agar and the resazurin
The test for sterility is carried out under aseptic conditions. sodium solution, sterilise as directed aboye. The pH after
In order to achieve such conditiol1s, the test environment sterilisation is 7.1 ± 0.2. Heat in a water-bath prior to use and
has to be adapted to the way in which the sterility test is incubate at 30-35 oC under anaerobic conditions.
performed. The precautions taken to avoid contamination are Soya-bean casein digest medium
such that they do not affect any micro-organisms which are to Pancreatic digest of casein 17.0 g
be revealed in the test. The working conditions in which the
tests are performed are monitored regularly by appropriate Papaie digest of soya-bean meal 3.0 g
sampling of the working are a and by carrying out appropriate Sodiurn chJoride 5.0 g
controls.
Dipotassium hydrogen phosphate 2.5 g
CULTURE MEDIA AND INCUBATION TEMPERATURES
Glucose monohydrate/anhydrous 2.5 g/2.3 g
Media for the test be prepared as described below, or
equivalent media may be used provided that they Water R 1000 mL
comply with the promotion test. pH after sterilisation 7.3 ± 0.2
The following culture media have been found to be suitable for
the test for sterility. Fluid thioglycollate medium is primarily Dissolve the solids in water R, warming slightly to effect
intended for the culture of anaerobic bacteria; however, it will solution. Cool the solution to room temperatureo Add 1 M
also detect aerobic bacteria. Soya-bean casein digest medium sodium hydroxide, if necessary, so that after sterilisation the
is suitable for the culture of both fungi and aerobic bacteria. solution will have a pH of 7.3 ± 0.2. Filter, if necessary, to
clarify, distribute into suitable vessels and sterilise using a
Fluid thioglycollate medium
validated process. Sto re at a temperature between 2 oC and
L-Cystine 0.5 g 25 oC in a sterile well-dosed container, unless it is intended for
Agar 0.75 g immediate use. Do not use the medium for a longer storage
period than has been validated.
Sodiurn chloride 2.5 g
Soya-bean casein digest medium i5 to be incubated at 20-25 oc.
Glucose rnonohydrate/anhydrous 5.5 g/S.O g The media used comply with the tests, carried
Yeast extraet (water-soluble) 5.0 g out before or in parallei with the test on product to be
examined.
Pancreatic digest of casein 15.0 g
Stedlity. Incubate portions of the media for 14 days. No
Sodium thioglyeollate or 0.5 g growth of micro-organisms occurs.
Thioglycollic acid 0.3 mL Growth promotion test of aerobes, anaerobes and fungi.
Test each batch of ready-prepared medium and each batch of
Resazurin sodium solution (1 giL of resazurin l.0 rnL
sodiurn), freshly prepared medium prepared either from dehydrated medium or from
Water R 1000 rnL
ingredients. Suitable strains of micro-organisms are indicated
in Table 2.6.1.-l.
pH after sterilisation 7.1 ± 0.2 Inoculate portions of fluid thioglycollate medium with a
small number (not more than 100 CFU) of the following
Mix the L-cystine, agar, sodium chloride, glucose,
micro-organisms, using a separate portion of medium for
water-soluble yeast extract and pancreatic digest of casein
each of the following species of micro-organism: Clostridium
with the water R and heat until solution is effected. Dissolve
sporogenes, Pseudomonas aeruginosa, Staphylococcus aureus.
the sodium thioglycollate or thioglycollic acid in the solution
Inoculate portions of soya-bean casein digest medium with
and, if necessary, add 1 M sadium hydroxide so that, after
a smallnumber (11ot more than 100 CFU) of the following
sterilisation, the solution will have a pH of 7.1 ± 0.2. If
micro-organisms, using a separate portion of medium for
filtration is necessary, heat the solution again without boiling
each of the following species of micro-organism: Aspergillus
and fiIter while hot through moistened filter paper. Add the
brasiliensis, Bacíllus subtilis, Candida albicans. Incubate for
resazurin sodium so]ution, mix and place the medium in
not more than 3 days in tile case of bacteria and not more
suitable ves seis which provide a ratio of surface to depth of
than 5 days in the case of fungi.
medium such that not more than the upper half of the medium
has undergone a colour change indicative of oxygen uptake at Seed 10t culture maintenance techniques (seed-Iot systems) are
the end of the incubation periodo Sterilise u5ing a validated used so that the viable micro-organisms used for inoculation
process. If the medium is stored, sto re at a temperature are not more than 5 passages removed from the original
between 2 oC and 25 oC in a sterile, airtight container. If master seed -lot.
more than the upper one-third of the medium has acquired The media are suitable if a clearly visible growth of the
a pink colour, the medium may be restored once by heating micro-organisms occurs.

(1) This chapter has undergone pharmacopoeial harmonisation. Sec chaptcr 5.8. Pharmacopocial hannonisatiol1.

General Natices (1) apply to all monographs and other texts 175
2.6.1. SterHity EUROPEAN PHARMACOPOEIA 8.0

Table 2.6.1.-1. - Strains of the test micro-organisms suitable for use in the growth promotion test and the method suitability test
Aerobic bacteria

Staphylococcus aureus ATCC 6538, CIP 4.83, NCTC 10788, NCIMB 9518, NBRC 13276

Bacil/us subtilis ATCC 6633, CIP 52.62, NCIMB 8054, NBRC 3134

Pseudo monas aeruginosa ATCC 9027, NCIMB 8626, CIP 82.118, NBRC 13275
Anaerobic bacterium

Clostridium sporogenes ATCC 19404, CIP 79.3, NCTC 532, ATCC 11437, NBRC 14293

Fungí

Candida albicans ATCC 10231, IP 48.72, NCPF 3179, NBRC 1594

Aspergillus brasiliensis ATCC 16404, lP 1431.83, lMI 149007, NBRC 9455

METHOD SUITABILITY TEST alcoholic solutions and cellulose acetate filters, for example,
Carry out a test as described below under Test for sterility of for strongly alcoholic solutions. Specially adapted filters may
the product to be examined using exactly the same methods be needed for certain products, e.g. for antibiotics.
except for the fo11owing modifications. The technique described below assumes that membranes
Membrane filtration. After transferring the contents of the about 50 mm in diameter will be used. If filters of a different
container or containers to be tested to the membrane add an diameter are used the volumes of the dilutions and the
inoculum of a small number of viable micro-organisms (not washings should be adjusted accordingly. The filtration
more than 100 CFU) to the final portion of sterile diluent apparatus and membrane are sterilised by appropriate means.
used to rinse the filter. The apparatus is designed so 'chat the solution to be examined
can be introduced and filtered under aseptic conditions; it
Direct inoculation. After transferring the content of the permits the aseptic removal of the membrane for transfer to
container or containers to be tested (for catgut and other the medium or it is suitable fo1' carrying out the incubation
surgical sutures for veterinary use: strands) to the culture after adding the medium to the apparatus itself.
medium add an inoculum of a small number of viable
micro-organisms (not more than 100 CFU) to the medium. Aqueous solutions. If appropriate, transfer a small quantity
of a suitable, sterile diluent such as a 1 giL neutral solution
In both cases use the same micro-organisms as those described
of meat or casein peptone pH 7.1 ± 0.2 onto the membrane
aboye under Growth promotion test of aerobes, anaerobes and
in the apparatus and fiIter. The diluent may contain suitable
fungi. Perform a growth promotion test as a positive control.
neutralising substances and/or appropriate inactivating
Incubate a11 the container s containing medium for not more
substances for example in the case of antibiotics.
than 5 days.
If clearly visible growth of micro-organisms is obtained after Transfer the contents of the container or container s to be
the incubation, visually comparable to that in the control tested to the membrane or membranes, if necessary after
vessel without product, either the product possesses no diluting to the volume used in the method suitability test
antimicrobial activity under the conditions of the test or such with the chosen sterile diluent but in any case using 110t les s
activity has been satisfactorily eliminated. The test for sterility than the quantities of the product to be examined prescribed
may then be carried out without further modification. in Table 2.6.1.-2. Filter immediately. If the product has
antimicrobial properties, wash the membrane not less than
If clearly visible growth is not obtained in the presence of the 3 times by filtering through it each time the volume of the
product to be tested, visually comparable to that in the control chosen sterile diluent used in the method suitability test. Do
vessels without product, the product possesses antimicrobial not exceed a washing cycle of 5 times 100 mL per filter, even
activity that has not been satisfactorily eliminated under the if during the method suitability test it has been demonstrated
conditions of the test. Modify the conditions in order to that such a cycle does not ful1y eliminate the antimicrobial
eliminate the antimicrobial activity and repeat the method activity. Transfer the whole membrane to the culture medium
suitability test. or cut it aseptically into 2 equal parts and transfer one half
This method suitability test is performed: to each of 2 suitable media. Use the same volume ofeach
a) when the test for sterility has to be carried out on a new medium as in the method suitability test. Alternatively,
product; transfer the medium onto the membrane in the apparatus.
b) whenever there is a change in the experimental conditions Incubate the media for not less than 14 days.
of the test. Soluble solids. Use for each medium not less than the quantity
The method suitability test may be performed simultaneously prescribed in Table 2.6.1.-2 of the product dissolved in
with the test for sterility of the product to be examined. a suitable solvent such as the solvent provided with the
preparation, water for injections, saline or a 1 giL neutral
TEST FOR STERILITY OF THE PRODUCT TO BE solution of meat or casein peptone and proceed with the test
EXAMINED as described aboye for aqueous solutions using a membrane
The test may be carried out using the technique of membrane appropriate to the chosen solvent.
filtration or by direct inoculation of the culture media Gi/s and oily solutions. Use for each medium not less than
with the product to be examined. Appropriate negative the quantity of the product prescribed in Table 2.6.1.-2. Oils
controls are included. The technique of membrane filtration and oily solutions of sufficiently low viscosity may be filtered
is used whenever the nature of the product permits, that without dilution through a dry membrane. Viscous oils may
is, for filterable aqueous preparations, for alcoholic or oily be diluted as necessary with a suitable sterile diluent such as
preparations and for preparations miscible with or soluble in isopropyl myristate shown not to have antimicrobial activity
aqueous or oily solvents provided these solvents do not have in the conditions of the test. Allow the oil to penetrate the
an antimicrobial effect in the conditions of the test. membrane by its own weight then filter, applying the pressure
Membrane filtration. Use membrane filters having a nominal or suction gradually. Wash the membrane at least 3 times
pore size not greater than 0.45 flm whose effectiveness to by filtering through it each time about 100 mL of a suitable
retain micro-organisms has been established. Cellulose nitrate sterile solution such as 1 giL neutral meat or casein peptone
filters, for example, are used for aqueous, oily and weakly containing a suitable emulsifying agent at a concentration

176 See the information section on general monographs (ca ver pages)
EUROPEAN PHARMACOPOEIA 8.0 2.6.1. Sterility

Table 2.6.1.-2. - Minimum quantity to be used for each medium


Minimum qualltity to be used fOf each medium unless
Quantity per container
otherwise justified and authorised
Liquids

- les s than 1 mL The whole contenls of each container

- 1-40mL Half Ihe contents of each container but nol less Ihan 1 mL

- greater Ihan 40 mL and not greater Ihan 100 mL 20 mL

- greater Ihan lOO mL 10 per cent ofthe conlenls of lhe container bul nol less Ihan 20 mL

Antibiotic liquids 1 mL

Insoluble preparations. creams and ointments to be suspended 01' emulsitied Use the contents of each container to provide not less than 200 mg

Solids

- less than 50 mg The whole contents of each container

- 50 mg or more but less than 300 mg Half the contents of each container but not les s than 50 mg

- 300 mg to 5 g 150 mg

- greater than 5 g 500 mg

Catgut and other surgical sutures for veterinary use 3 sections of a strand (each 30 cm long)

shown to be appropriate in the method suitability test, for aseptic precautions and remove 3 sections of the strand for
example polysorbate 80 at a concentration of 10 giL. Transfer each culture medium. Carry out the test on 3 sections, each
the membrane or membranes to the culture medium or media 30 cm long, cut off from the beginning, the centre and the
or vice versa as described aboye for aqueous solutions, and end of the strand. Use whole strands from freshly opened
incubate at the same temperatures and for the same times. cassette packs. Transfer each section of the strand to the
Ointments and creams. Use for each medium not less than selected medium. Use sufficient medium to cover adequately
the quantities of the product prescribed in Table 2.6.l.-2. the material to be tested (20 mL to 150 mL).
Ointments in a fatty base and emulsions of the water-in-oil
type may be diluted to 1 per cent in isopropyl myristate as OBSERVATION AND INTERPRETATION OF RESULTS
described aboye, by heating, if necessary, to not more than
40 oc. In exceptional cases it may be necessary to heat to not At intervals during the incubation period and at its conclusion,
more than 44 oc. Filter as rapidly as possible and proceed as examine the media for macroscopic evidence of microbial
described aboye for oils and oily solutions. growth. If the material being tested renders the medium
turbid so that the presence or absence of microbial growth
Direct inoculation of the culture medium. Transfer the cannot be readily determined by visual examination, 14 days
quantity of the preparation to be examined prescribed in after the beginning of incubation transfer portions (each not
Table 2.6.l.-2 directly into the culture medium so that the less than 1 mL) of the medium to fresh vessels of the same
volume of the product is not more than 10 per cent of the medium and then incubate the original and transfer vessels
volume of the medium, unless otherwise prescribed. for not les s than 4 days.
rf the product to be examined has antimicrobial activity, carry
out the test after neutralising this with a suitable neutralising If no evidence of microbial growth is found, the product to be
substance or by dilution in a sufficient quantity of culture examined complies with the test for sterility. rf evidence of
medium. When it is necessary to use a large volume of the microbial growth is found the product to be examined does
product it may be preferable to use a concentrated culture not comply with the test fol' sterility, unless it can be clearly
medium prepared in such a way that it takes account of the demonstrated that the test was invalid for causes unrelated
subsequent dilution. Where appropriate, the concentrated to the product to be examined. The test may be considered
medium may be added directly to the product in its container. invalid only if one or more of the following conditions are
fulfilled:
Oily liquids. Use media to which have been added a suitable
emulsifying agent at a concentration shown to be appropriate a) the data of the microbiological monitoring of the sterility
in the method suitability test, for example polysorbate 80 at a testing facility show a fault;
concentration of 10 giL.
b) a review of the testing procedure used during the test in
Ointments and creams. Prepare by diluting to about 1 in 10 question reveals a fault;
by emulsifying with the chosen emulsifying agent in a suitable
sterile diluent such as a 1 giL neutral solution of meat or e) microbial growth is found in the negative controls;
casein peptone. Transfer the diluted product to a medium not d) after determination of the identity of the micro-organisms
containing an emulsifying agent. isolated from the test, the growth of this species or these
Incubate the inoculated media for not less than 14 days. species may be ascribed unequivocally to faults with respect
Observe the cultures several times during the incubation to the material and/or the technique used in conducting the
period. Shake cultures containing oily products gently each sterility test procedure.
day. However when fluid thioglycollate medium is used for
the detection of anaerobic micro-organisms keep shaking If the test is declared to be invalid it is repeated with the same
or mixing to a minimum in order to maintain anaerobic number of units as in the original test.
conditions. lf no evidence of microbial growth is found in the repeat
Catgut and other surgical sutures far veterinary use. Use for test the product examined complies with the test for sterility.
each medium not less than the quantities of the product lf microbial growth is found in the repeat test the product
prescribed in Table 2.6.1.-2. Open the sealed package using examined does not comply with the test for sterility.

General Natices (1) apply ta all monographs and other texts 177
2.6.2. Mycobacteria EUROPEAN PHARMACOPOEIA 8.0

Table 2.6.1.-3. - Minimum number of items to be tested


Minimum number of ítems lo be testea for each medium, unless
Nnmber of ítems in the batch*
otherwise justified and authorised"
Parenteral preparations

- Not more than 100 containers 10 per cent or 4 containers, "\vhichever is the greater

- More than J 00 but not more than 500 containers 10 containers


2 per cent ol' 20 containers (la cantainers far large-volume parenterals)
- More than 500 containers
whichever is les s
Ophthalmic ami other 11On-injectable preparations

- Not more than 200 containers 5 per cent or 2 containers, whichever is the greater

- More than 200 containers JO container s


- If the product is presented in the form of single-dose containers, apply
the scheme shown aboye for preparations for parenteral administration
2 per cent or 5 packages whichever is the greater, up to a maximum total of
Catgut and other surgical sutures jor ve/erinary use
20 packages
Bulk solid produc/s

- Up to 4 containers Each container

- More than 4 container s but not more than 50 containers 20 per cent or 4 containers, whichever is the greater

- More than 50 containers 2 per cent or 10 containers, whichever is the greater

* If the batch size is not known, use ¡he maximum number of items prescribed.
Hlf the contents of OIle container are enough to inocuJate the 2 media, this coJumn gives the number of containers needed for both the media
together.

APPLICATION OF THE TEST TO PARENTERAL lf at the end of the incubation time no growth of mycobacteria
PREPARATIONS, OPHTHALMIC AND OTHER occurs in any of the test media, the preparation complies with
NON-INJECTABLE PREPARATIONS REQUIRED TO the test.
COMPLY WITH THE TEST FOR STERILITY
When using the technique of membrane filtration, use,
whenevel' possible, the whole contents of the container, but 0112008:20607
not less than the quantities indicated in Table 2.6.1.-2, diluting corrected 6.1
where necessary to about 100 mL with a suitable sterile
solution, such as 1 giL neutral meat or casein peptone.
When using the technique of direct inoculation of media,
2.6.7. MYCOPLASMAS
use the quantities shown in Table 2.6.1.-2, unless otherwise V/here the test for mycoplasmas is prescribed for a master
justified and authorised. The tests for bacterial and fungal ceH bank, for a working cen bank, for a virus seed 10t or for
sterility are carried out on the same sample of the product to control cells, both the culture method and the indicator ceH
be examined. When the volume or the quantity in a single culture method are used. Where the test for mycoplasmas is
container is insufficient to carry out the tests, the contents of 2 prescribed for a virus harvest, for a bulk vaccine or for the
01' more containers are used to inoculate the different media. finallot (batch), the culture method is used. The indicator
MINIMUM NUMBER OF ITEMS TO BE TESTED ceH culture method may also be used, where necessary, for
screening of media.
The minimum number of items to be tested in relation to the
size ofthe batch is given in Table 2.6.1.-3. Nudeic acid amplification techniques (NAT) may be used
as an alternative to one or both of the other methods after
Guidelines on the test for sterility are given in general suitable validation.
chapter 5.1.9.
CULTURE METHOD
0112008:20602 CHOICE OF CULTURE MEDIA
The test is carried out using a sufficient number ofboth solid
2.6.2. MYCOBACTERIA and liquid media to ensure growth in the chosen incubation
If the sample to be examined may be contaminated conditions of smallnumbers of mycoplasmas that may be
by micro-organisms other than mycobacteria, treat present in the product to be examined. Liquid media must
it with a suitable decontamination solution, such as contain phenol red. The range of media chosen is shown
acetylcysteine-sodium hydroxide solution or sodium to have satisfactory nutritive properties for at least the
laurilsulfate solution. micro-organisms shown below. The nutritive properties of
each new batch of medium are verified for the appropriate
Inoculate 0.2 mL of the sample in triplicate onto each of micro-organisms in the list. When testing for mycoplasmas in
2 suitable solid media (Li:iwenstein-Jensen medium and the product to be examined, at least 1 of the following species
Middlebrook 7HlO medium are considered suitable). will be induded as a positive control:
lnoculate 0.5 mL in triplicate into a suitable liquid medium.
Incubate all media at 37 oC for 56 days. - Acholeplasma laidlawii (vaccines for human and veterinary
use where an antibiotic has been used during production) ;
Establish the fertility of the media in the presence of the
preparation to be examined by inoculation of a suitable strain - Mycoplasma gallisepticum (where avian material has been
of a Mycobacterium sp. such as BCG and if necessary use a used during production or where the vaccine is intended
suitable neutralising substance. for use in poultry);
If contaminating micro-organisms develop during the first - Mycoplasma hyorhinis (non-avian veterinary vaccines);
8 days of incubation, repeat the test and carry out at the same - Mycoplasma orale (vaccines for human and veterinary use);
time a bacteriological sterility test.

178 See the information section on general monographs (cover pages)


EUROPEAN PHARMACOPOEIA 8.0 2.6.7. Mycoplasmas

- Mycoplasma pneumoniae (vaccines for human use) or other TEST FOR MYCOPLASMAS IN THE PRODUCT TO BE
suitable species ofD-glucose fermenter such as Mycoplasma EXAMINED
Inoculate 10 mL ofthe product to be examined per 100 mL
- Mycoplasma (where avian material has been used of each liquid medium. rf it has been found that a signiflcant
during production or where the vaccine is intended for pH change occurs upon the addition of the product to be
use in poultry). examined, the liquid medium is restored to its original pH
value by the addition of a solution of either sodium hydroxide
The test strains are fleld isolates having undergone a limited or hydrochloric acid. Inoculate 0.2 mL of the product to
number of sub cultures (not more than 15), and are stored be examined 011 each plate of each solid medium. Incubate
frozen or freeze-dried. AÍter cloning, the strains are identifled liquid media for 20-21 days. Incubate solid media for not less
as being of the required species by comparison with type than 14 days, except those corresponding to the 20-21 day
cultures, for example: sub culture, which are incubated for 7 days. At the same time
A.lnid/awii NCTC 10116 CIP 75.27 ATCC 23206 incubate an uninoculated 100 mL portion of each liquid
M. gallisepticum NCTC 10115 CIP 104967 ArCC 19610
medium and agar plates, as a negative control. On days
2-4 after inoculation, subculture each liquid medium by
M. fermentans NCTC 10117 CIP 105680 ATCC 19989 inoculating 0.2 mL on at least 1 plate of each solid medium.
AJ. hyorhillis NCTC10130 CIP 104968 ATCC 17981
Repeat the procedure between the 6th and 8 th days, again
between the 13 th and 15 th days and again between the 19 th
lvI. orale NCTC 10112 CIP 104969 ATCC 23714 and 21 ,t days of the test. Observe the liquid media every
M. pneumol1iae NCTC 10119 CIP 103766 ATCC 15531
2 01' 3 days and if a colour change occurs, sub culture. If a
liquid medium shows bacterial or fungal contamination, the
ivl. synol'iae NCTC 10124 CIP 104970 ATCC 25204 test is invalido The test is valid if at least 1 plate per medium
and per inoculation day can be read. Indude in the test
Acholeplasma laidlawii BRP, Mycoplasma fermentans BRP, positive controls prepared by inoculation of not more than
Mycoplasma hyorhinis BRP, Mycoplasma orale BRP and 100 CFU of at least 1 test micro-organism on agar medium
Mycoplasma syn01~iae BRP are suitable for use as low-passage or into broth medium. Where the test for mycoplasmas is
reference strains. carried out regularly and where possible, it is recommended
to use the test micro-organisms in regular rotation. The test
INCUBATION CONDITIONS
micro-organisms used are those listed under Choice of culture
Incubate liquid media in tightly stoppered containers at media.
35-38 oc. Incubate solid media in microae1'Ophilic conditions
(nitrogen containing S-lO per cent of carbon dioxide and INTERPRETATION OP RESULTS
sufficient humidity to prevent desiccation of the agar surface) At the end o[ the prescribed incubation period, examine al!
at 35-38 oc. inoculated solid media microscopically [or the presence of
mycoplasma colonies. The product complies with the test if
NUTRITIVE PROPERTIES
growth oE typical mycoplasma colonies has not occurred. The
out the test for nutritive properties for each ¡¡ew product do es not comply with the test if growth of typical
medium. Inoculate the chosen media with the mycoplasma colonies has occurred on any of the solid media.
appropriate test mic1'O-organisms; use not more than 100 CFU The test is invalid if 1 or more of the positive cont1'Ols do not
(colony-forming peY 60 mm diameter pi ate containing show of mycoplasmas on at least 1 sub culture plateo
9 mL of solid medium and per 100 mL container of liquid The test is il1valid if 1 or more of the negative contr01s show
medium; use a separate plate and container fol' each species growth of mycoplasmas. If colonies are observed, a
of micro-organismo Incubate the media and make subcultures suitable vaUdated method may used to determine whether
from 0.2 mL ofliquid medium to solid medium at the specified are due to
intervals (see below under Test for mycoplasmas in the
product to be examined). The solid medium complies with the
test if adequate growth is found for each test mic1'O-organism The following section is published for information.
(growth obtained does not differ by a factor greater than 5
from the value calculated with respect to the inoculum). The
liquid medium complíes with the test if growth on agar plates RECOMMENDED MEDIA FOR THE CULTURE METHOD
subcultured from the broth is found for at least 1 sub culture The following media are recommended. Other media may
for each test micro-organismo be used, provided that their ability to sustain the growth of
INHIBITORY SUBSTANCES mycoplasmas has been demonstrated 011 each batch in the
The test for inhibitory substances is carried out once for a presence and absence of the product to be examined.
given product and is repeated whenever there is a change HA YFLICK MEDIA POR THE GENERAL
in production method that may affect the detection of DETECTION OP MYCOPLASMAS)
mycoplasmas.
To demonstrate absence of inhibitory substances, carry out the Liquid medium
test for nutritive properties in the presence and absence of the Beef hearl infusion brotll (1) 90.0 mL
p1'Oduct to be examined. If g1'Owth of a test micro-organism
Horse serum (unheated) 20.0 mL
occurs more than 1 subculture sooner in the absence of
the product to be examined than in its presence, or if plates Yeast extract (250 giL) 10.0 mL
directly inoculated with the product to be examined have
Phenol red (0.6 giL soJution) 5.0 mL
fewer than l/S of the number of colonies of those inoculated
without the product lO be examined, inhibitory substances are Penicillin (20000 IU/mL) 0.25 mL
present and they must be neutralised 01' their effect otherwise
Deoxyribonucleic acid (2 giL solution) 1.2 mL
countered, for example by passage in substrates not containing
inhibitors or dilution in a larger volume of medium before
the test. If dilution is used, larger medium volumes may be Adjust pH 7.8.
used or the inoculum volume may be divided among several Solid medium
100 mL flasks. The effectiveness of the neutralisation or
other process is checked by repeating the test lor inhibitory Prepare as described aboye replacing beef heart infusion broth
substances after neutralisation. by beef heart infusion agar containing 15 gil of agar.

General Notices (1) apply lo all monographs and other texts 179
2.6.7. Mycoplasmas EUROPEAN PHARMACOPOEIA 8.0

FREY MEDIA (RECOMMENDED FOR THE DETECTION (2) Essential vitamins


OF M. SYNOVIAE) Biotin 100 mg
Liquid medium Calcium pantothenate 100mg
Beefheart infusion broth (1) 90.0 mL
Choline chloride 100 mg
Essential vitamins (2) 0.025 mL
Folie acid 100 mg
Glucose monohydrate (500 giL solution) 2.0 mL
i-Inositol 200 mg
Swine serum (inactivated at 56 oC for 30 min) 12.0 mL
Nicotinamide 100 mg
~-Nicotinamide adenine dinucleotide (10 giL solution) 1.0 mL
Pyridoxal hydrochloride 100 mg
Cysteine hydrochloride (lO giL solution) 1.0mL
Riboflavine 10mg
Phenol red (0.6 giL solution) 5.0 mL
Thiamine hydrochloride 100 mg
Penicillin (20 000 !U/mL) 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 (3) Agar, purified
ingredients. Adjust to pH 7.8.
A highly refined agar for use in microbiology and immunology,
Solid medium
prepared by an ion-exchange procedure that results in a
Beefheart infusion broth (1) 90.0 mL
product having superior purity, darity and gel strength. It
Agar, purified (3) 1.4 g contains about:
Water 12.2 per cent
Adjust to pH 7.8, sterilise by autoclaving then add:
Ash 1.5 per cent
Essential vitamins (2) 0.025 mL
Acid-insoluble ash 0.2 per cent
Glucose monohydrate (500 giL solution) 2.0 mL

Swine serum (unheated) 12.0 mL Chlorine o


Phosphate (calculated as p,os) 0.3 per cent
~-Nicotinamide adenine dinucleotide (10 giL solution) 1.0 mL
Totalnitrogen 0.3 per cent
Cysteine hydrochloride (10 giL solution) 1.0 mL
Copper 8 ppm
Phenol red (0.6 giL solution) 5.0 mL
lron 170 ppm
Penicillin (20 000 !U/mL) 0.25 mL
Calcium 0.28 per cent
FRIIS MEDIA (RECOMMENDED FOR THE DETECTION OF
NON-A VIAN MYCOPLASMAS) Magnesium 0.32 per cent

Liquid medium
(4) Hanks' balanced salt solution (modified)
Hanks' balanced salt solution (modified) (4) 800 mL
Sodium chloride 6.4 g
Distilled water 67 mL
Potassium chloride 0.32 g
Brain heart infusion (5) 135 mL
Magnesium sulfate heptahydrate 0.08 g
PPLO Broth (6) 248 mL
Magnesium chloride hexahydrate 0.08 g
Yeast extraet (170 giL) 60 mL
Calcium chloride, anhydrous 0.112 g
Bacitracin 250 mg
Disodium hydrogen phosphate dihydrate 0.0596 g
Meticillin 250 mg
Potassium dihydrogen phosphate, anhydrous 0.048 g
Phenol red (5 giL) 4.5 mL
Distilled water to 800 mL
Horse serum 165 mL

Swine serum 165 mL (5) Brain heart infusion


Calf-brain infusion 200 g
Adjust to pH 7.40-7.45.
Beef-heart infusion 250 g
Solid medium
Hanks' balanced sal! solution (modified) (4) 200 mL Proteos e peptone lOg

DEAE-dextran 200 mg Glucose monohydrate 2g

Agar, purified (3) 15.65 g Sodium chloride 5g

2.5 g
Mix well and sterilise by autoclaving. Cool to 100 oc. Add to Disodium hydrogen phosphate, anhydrous

1740 mL of liquid medium as described aboye. Distilled water to 1000 mL


(1) Beef heart infusion broth
Beef heart (for preparation of the infusion) 500 g (6) PPLO broth
Peptone 10 g Beef-heart infusion 50 g

Sodium chloride 5g Peptone IOg

Distilled water to 1000 mL Sodium chloride 5g

Distilled water to 1000 mL


Sterilise by autoclaving.

180 See the information section on general monographs (caver pages)


EUROPEAN PHARMACOPOEIA 8.0 2.6.7. Mycoplasmas

INDICATOR CELL CULTURE METHOD INTERPRETATION OF RESULTS


Cell cultures are stained with a fluorescent dye that binds The product to be examined complies with the test if
to DNA. Mycoplasmas are detected by their characteristic fluorescence typical of mycoplasmas is not presento The test
particulate or filamentous pattern of fluorescence on the cel! is invalid if the positive controls. do 110t show fluorescence
surface and, if contamination is heavy, in surrounding areas. typical of mycoplasmas. The test is invalid if the negative
Mitochondria in the cytoplasm may be stained but are readily controls show fluorescence typical of mycoplasmas.
distinguished from mycoplasl11as.
NUCLEIC Acm AMPLIFICATION TECHNIQUES (NAT)
If for viral suspensions the interpretation of results is affected
by marked cytopathic effects, the virus may be neutralised NAT (2.6.21) may be used for detection of mycoplasmas
USi11g a specific antiserum that has no inhibitory effects 011 amplification of nucleic acids extracted from a test sample
mycoplasmas or a ceH culture substrate that does not allow with specific primers that reveal the presence of the target
growth of the virus may be used. To del110nstrate the absence nucleic acid. NAT indicate the presence of a particular
of inhibitory effects of serum, carry out the positive control nucleic acid sequence and not necessarily the presence of
tests in the presence and absence of the antiserum. viable mycoplasmas. A number of different techniques are
available. This general chapter does not prescribe a particular
VERIFICATION OF THE SUBSTRATE method for the test. The procedure applied must be validated
Use Vero cells 01' anothe1' ceH culture (for example, the as described, taking accoul1t of the guidelines presented at
production cellline) that is equivalent in effectiveness for the end of this section. Where a commercial kit is used,
detecting mycoplasmas. Test the effectiveness of the cells to be certain elements of the validation may be carried out by the
used by applying the procedure shown below and inoculating manufacturer and information provided to the user but it
not more than 100 CFU or CFU-like micro-organisms of must be remembered that full information on the primers
suitable reference strains of M. hyorhinis and M. orale. The may not be available and that production of the kit may be
following strains have been found to be suitable: ll10dified or discontinued.
M. hyorhinis ATCC 29052 NAT are applied where prescribed in a monograph. They may
also be used instead of the culture method and the indicator
M.orale NCTC 10112 CIP 104969 ATCC 23714
cell culture method after suitable validatiol1.
The cells are suitable if both reference strains are detected. Direct NAT can be applied in the presence of cytotoxic
material and where a rapid method is needed.
The indicator ceUs must be subcultured without an antibiotic
before use in the test. Cell-eulture enrichment followed by NAT: the test sample and
a suitable ceH substrate (as described under the indicator
TEST METHOD
ceu-culture method) are cultured together for a suitable
1. Seed the indicator ceH culture at a suitable density period; the nucleic acids are then ext1'acted from cells and
(for example, 2 x lO" to 2 X 10 5 cells/mL, 4 x 10 3 to supernatant and used far detection by NAT.
2.5 X 104 cells/cm 2 ) that will yield confluence after 3 days of
growth. Inoculate 1 mL of the product to be examined into VALIDATION
the cel! culture vessel and incubate at 35-38 oc. Reference standards are required at various stages during
2. After at least 3 of incubation, when the cells have grown validation and for use as contro15 during routine application
to confluence, make a sub culture on cover slips in suitable of the test. The reference standards may be mycoplasmas or
containe1's or on some other surface example, chambered nucleic acids.
slides) suitable for the test procedure. Seed the cells at low For validatiol1 of the limit of detection, the following species
density so that they reach 50 per cent confluence after 3-5 days represent an optimal selection in terms of the frequency of
of incubation. Complete confluence impairs visualisation of occurrence as contaminants and phylogenetic relationships:
mycoplasmas at1:er staining and must be avoided. - A. laidlawií;
3. Remove the mediul11 and rinse the indicator cells with - M. fermentans;
phosphate buffered saline pH 7.4 R, then add a suitable fixing
- M. hyorhinis (where ceH-culture enrichment is used, a
solution (a freshly prepared mixture of 1 volume of glacial
fastidious strain such as ATCC 29052 is included);
acetie acid R and 3 volumes of methanol R is suitable when
bisbenzimide R is used for staining). - M. ora/e;
4. Remove the fixing solutiol1 and wash the cells with sterile - M. pneumoniae or M. gallisepticum;
water R. Dry the slides completely if they are to be stained - M. synoviae (where there is use of or exposure to avian
more than 1 h later (particular care is needed for staining of material during production);
slides after drying owing to artefacts that may be produced).
arginini;
5. Add a suitable DNA stain and allow to stand for a suitable
- Spiroplasma citri (where there is use of or exposure to
time (bisbenzimide working solution R and a standing time
insect or plant material during production).
of 10 min are suitable).
Demonstration of specificity requires the use of a suitable
6. Remove the stain and rinse the mOll01ayer with water R. range of bacterial species other than mycoplasmas. Bacterial
7. Mount each coverslip, where applicable (a mixture of equal genera with close phylogenetíc relation to mycoplasmas
volumes of glyeerol R and phosphate-eitrate buffer solutian are most appropriate for this validatíon; these include
pH 5.5 R is suitable for mounting). Examine by fluorescence Clostridium, Lactobaeillus and Streptococcus.
(for bisbenzimide stain a 330 nm/380 nm excitation filter and Comparability studies for use of NAT as an alternative method.
an LP 440 nm barrier filter are suitable) at 400 x magnification For each mycoplasma test species:
or greater.
- as an alternative to the culture method: the NAT test system
8. Compare the microscopic appearance of the test cultures must be shown to detect 10 CFU/mL;
with that of the negative and positive controls, exal11ining for
extranuclear fluorescence. Mycoplasmas produce pinpoints or - as an alternative to the indicator ceH culture method: the
filaments over the indicator cell cytoplasm. They may a1so NAT test system must be shown to detect 100 CFU/mL;
produce pinpoints and filaments in the intercellular spaces. or a11 equivalent ¡imit of detection in terms of the number of
Multiple microscopic fields are examined according to the copies of mycoplasma nucleic acid in the test sample (using
protocol established during validation. suitable reference standards of mycoplasma nucleic acid).

General Notices (1) apply to all monographs and other texts 181
2.6.7. Mycoplasmas EUROPEAN PHARMACOPOEIA 8.0

CONTROLS stringency of the test conditions (for both the amplification


Internal controls. Internal control s are necessary for routine and detection steps).
verification of absence of inhibition. The internal control The ability of the NAT to detect a large panel of mycoplasma
may contain the primer binding-site, or some other suitable species will depend on the choice of primers, probes and
sequence may be used. It is preferably added to the test method parameters. This ability should be demonstrated
material before isolating the nudeic acid and therefore using characterised reference paneIs (e.g. reference strains
acts as an overall control (extraction, reverse transcription, provided by the EDQM). Since NAT systems are usuaHy based
amplification, detection). on a mix of primers, the theoretical analysis of primers and
External controls. The external positive control contains a probes by comparison with databases is not recommended,
defined number of target -sequence copies or CFUs from 1 because interpretation of the results may be quite complex and
may not reflect the experimental results.
or more suitable species of mycoplasma chosen from those
used during validation of the test conditions. 1 of the positive Moreover, as it is likely that the primers will detect other
controls is set close to the positive cut-off point to demonstrate bacterial species, the potential cross-detection should be
that the expected sensitivity is achieved. The external negative documented in the validation study. Bacterial genera such
control contains no target sequence but does not necessarily as gram-positive bacteria with close phylogenetic relation
represent the same matrix as the test artide. to mycoplasmas are most appropriate for this validation;
these in dude Clostridium, Lactobacillus and Streptococcus.
INTERPRETATION OP RESULTS
However, this is not an exhaustive list and species to be tested
The primers used may also amplify non-mycoplasmal bacterial will depend on the theoretical ability (based on primers/probes
nucleic acid, leading to false positive results. Procedures sequences) of the NAT system to detect such other species.
are established at the time of validation for dealing with
confirmation of positive results, where necessary. Based on the results from this validation of the specificity,
if a gap in the specificity of the method is identified (such
The following section is published for information. as detection of non-mycoplasmal bacterial nudeic acid),
an appropriate strategy must be proposed in the validation
study to allow interpretation of positive results on a routine
Validation of nudeic acid amplificabon basis. For example, a second test may be performed using an
techniques (NAT) for the detection of alternative method without this specificity gap or using an
mycoplasmas: guidelines official method.
2-2. Detection limito The detection limit of an individual
l. SCOPE analytical procedure is the lowest amount of target nudeic
Nucleic acid amplification techniques (NAT) are either acid in a sample that can be detected but not necessarily
qualitative or quantitative tests for the presence of nucleic acid. quantitated as an exact value.
For the detection of mycoplasma contamination of various
Por establishment of the detection limit, a positive cut-off
samples such as vaccines and ceH substrates, qualitative tests
point should be determined for the nucleic acid amplification
are adequate and may be considered to be limit tests.
analytical procedure. The positive cut -off point (as defined
These guidelines describe methods to validate qualitative
in general chapter 2.6.21) is the mínimum number of target
nucleic acid amplification analytical procedures for assessing
sequence copies per volume of sample that can be detected
mycoplasma contamination. They may also be applicable
in 95 per cent of test runs. This positive cut-off point is
for real-time NAT used as limit tests for the control of
influenced by the distribution of mycoplasmal genomes in the
contaminants.
individual samples being tested and by factors such as enzyme
The 2 characteristics regarded as the most important for efficiency, and can result in different 95 per cent cut-off values
validation of the analytical procedure are the specificity for individual analytical test runs.
and the detection limito In addition, the robustness of the
analytical procedure should be evaluated. To determine the positive cut-off point, a dilution series of
characterised and calibrated (either in CFUs or nudeic acid
For the purpose of this document, an analytical procedure is copies) in-house working strains or EDQM standards should
defined as the complete procedure from extraction of nucleic be tested on different days to examine variation between test
acid to detection of the amplified products. runs.
Where commercial kits are used for part or aH of the analytical Por validation of the limit of detection, the following species
procedure, documented validation points already covered represent an optimal selection in terms of the frequency of
by the kit manufacturer can repIace validation by the user. occurrence as contaminants and phylogenetic relationships:
Nevertheless, the performance of the kit with respect to its
intended use has to be demonstrated by the user (e.g. detection - A. laidlawii;
limit, robustness, cross-detection of other classes ofbacteria). M. fermentans;
NAT may be used as: - M. hyorhinis;
- a complementar y test (for example, for cytotoxic viral - M.orale;
suspensions) or for in-process control purposes; - M. pneumoniae or M. gallisepticum;
- an alternative method to replace an official method M. synoviae (where there is use of or exposure to avian
(indicator ceH culture method or culture method). material during production);
These guidelines will thus separate these 2 objectives by - M. arginini;
presenting first a guideline for the validation of the NAT
- S. citri (where there is use of or exposure to insect or plant
themselves, and second, a guideline for a comparability study
material during production).
between NAT and official methods.
Por each strain, at least 3 independent 10-fold dilution series
2. GUIDELINE POR MYCOPLASMA NAT VALIDATION should be tested, with a sufficient number of replicates at
3 parameters should be evaluated: specificity, detectionlimit each dilution to give a total number of 24 test results for each
and robustness. dilution, to enable a statistical analysis of the results.
2-1. Spedfidty. Specificity is the ability to unequivocaHy For example, a laboratory may test 3 dilution series on
assess target nucleic acid in the presence of components that different days with 8 replicates for each dilution, 4 dilution
may be expected to be presento series on different days with 6 replicates for each dilution,
The specificity of NAT is dependent on the choice of primers, or 6 dilution series on different days with 4 replicates for
the choice of probe (for analysis of the final product) and the each dilution. In order to keep the number of dilutions at a

182 See the information section on general monographs (ca ver pages)
EUROPEAN PHARMACOPOEIA 8.0 2.6.8. Pyrogens

manageable level, a preliminary test should be performed 0112008:20608


to obtain a preliminar y value for the positive cut -off point
(Le. the highest dilution giving a positive signal). The range 2.6.8. PYROGENS
of dilutions can then be chosen around the predetermined
preliminary cut -off point. The concentration of mycoplasmas The test consists of measuring the rise in body temperature
(CFUs or copies) that can be detected in 95 per cent of test evoked in rabbits by the intravenous injection of a sterile
runs can then be calculated using an appropriate statistical solution of the substance to be examined.
evaluation. Selection ofanimals. Use healthy, adult rabbits of either sex
weighing not less than 1.5 kg, fed a complete and balanced
These results may also serve to evaluate the variability of the
diet not containing antibiotics, and not showing loss of body
analytical procedure.
mass during the week preceding the test. A rabbit is not be
2-3. Robustness. The robustness of an analytical procedure used in a pyrogen test:
is a measure of its capacity to remain unaffected by small but a) if it has been used in a negative pyrogen test in the preceding
deliberate variations in method parameters, and provides an 3 days, or
indication of its reliability during normal usage.
b) if it has been used in the preceding 3 weeks in a pyrogen
The evaluation of robustness should be considered during test in which the substance under examination failed to pass
the development phase. It should show the reliability of the the test.
analytical procedure with respect to deliberate variations in Animals' quarters. Keep the rabbits individually in a quiet are a
method parameters. For NAT, small variations in the method with a uniform appropriate temperature. Withhold food from
parameters can be crucial. However, the robustness of the the rabbits overnight and until the test is completed; withhold
method can be demonstrated during its development when water during the test. Carry out the test in a quiet room where
small variations in the concentrations ofreagents (e.g. MgCI2 , there is no risk of disturbance exciting the animals and in
primers or deoxyribonucleotides) are tested. Modifications of which the room temperature is within 3 oC of that of the
extraction kits or extraction procedures as well as different rabbits' living quarters, or in which the rabbits have been kept
thermal cycler types may also be evaluated. for at least 18 h before the test.
Finally, robustness of the method can be evaluated through Materials. Glassware, syringes and needles. Thoroughly wash
collaborative studies. al! giassware, syringes and needles with water for injections
and heat in a hot-air oven at 250 oC for 30 min or at 200 oC
3. GOIDELINE POR COMPARABILITY STUDY
for 1 h.
NAT may be used instead of official methods (indicator
cell culture method and/or culture method). In this case a Retaining boxes. The retaining boxes for rabbits whose
comparability study should be carried out. This comparability temperature is being measured by an electrical device are made
study should indude mainly a comparison of the respective in such a way that the animals are retained only by loosely
detection limits of the alternative method and official fitting neck-stocks; the rest of the body remains relatively
methods. However, specificity (mycoplasma panel detected, free so that the rabbits may sit in a normal position. Theyare
putative false positive results) should also be considered. not restrained by straps or other similar methods which may
harm the animal. The animals are put into the boxes 110t less
For the detection limit, acceptability criteria are defined as
than 1 h before the first record of the temperature and remain
follows:
in them throughout the test.
- if the alternative method is proposed to replace the Thermometers. Use a thermometer or electrical device which
culture method, the NAT system must be shown to detect indicates the temperature with a precision of 0.1 oC and
10 CFU/mL for each mycoplasma test species described insert into the rectum of the rabbit to a depth of about 5 cm.
in paragraph 2-2; The depth of insertion is constant for any one rabbit in any
- if the alternative method is proposed to replace the one test. When an electrical device is used it may be left in
indicator ceH culture method, the NAT system must be position throughout the test.
shown to detect 100 CFU/mL for each mycoplasma test Preliminary test. After selection of the animals, one to
species described in paragraph 2-2. three days before testing the product to be examined, treat
those animals that have 110t been used during the previous
For both cases, suitable standards calibrated for the number 2 weeks by intravenous injection of 10 mL per kilogram
of nucleic acid copies and the number of CFUs may be used ofbody mass of a pyrogen-free 9 giL solution of sodium
for establishing that these acceptability criteria are reached. chloride R warmed to about 38.5 oc. Record the temperatures
The relation between CFUs and nucleic acid copies for the of the animals, beginning at least 90 min before injection and
reference preparations should be previously established to continuing for 3 h after the injection of the solution. Any
compare the performance of the alternative NAT method with animal showing a temperature variation greater than 0.6 oC
the performance of the official methods. is not used in the main test.
1 of the following 2 strategies can be used to perform this Main test. Carry out the test using a group of three rabbits.
comparability study: Preparation and injection of the product. Warm the liquid to
- perform the NAT alternative method in parallel with the be examined to approximately 38.5 oC before the injection.
official method(s) to evaluate simultaneously the detection The product to be examined may be dissolved in, or diluted
limit of both methods using the same samples of calibrated with, a pyrogen-free 9 giL solution of sodium chloride R or
strains; another prescribed liquido Inject the solution slowly into
the marginal vein of the ear of each rabbit over a period
- compare the performance of the NAT alternative method not exceeding 4 min, unless otherwise prescribed in the
using previously obtained data from official method monograph. The amount of the product to be injected varies
validation. In this case, calibration of standards used according to the product to be examined and is prescribed in
for both validations as well as their stabilities should be the mOl1ograph. The volume injected is not less than 0.5 mL
documented carefully. per kilogram and not more than 10 mL per kilogram ofbody
Comparability study reports should describe all the validation mass.
elements described in section 2 (specificity, limit of detection Determination of the initial and maximum temperatures.
and variability, as well as robustness) in order to assess al! The "initial temperature" of each rabbit is the mean of two
the advantages and/or disadvantages of the alternative NAT temperature readings recorded for that rabbit at an interval of
method compared to official methods. 30 min in the 40 min immediately preceding the injection of

General Notices (1) apply to all monographs and other texts 183
2.6.9. Abnormal toxidty EUROPEAN PHARMACOPOEIA 8.0

the product to be examined. The "maximum temperature" of The test must also be carried out on 2 healthy guinea-pigs
each rabbit is the highest temperature recorded for that rabbit weighing 250 g to 400 go Inject intraperitoneally into each
in the 3 h after the injection. Record the temperature of each animal 1 human dose but not more than 5.0 mL. The human
rabbit at intervals of not more than 30 min, beginning at least dose is that stated on the label of the preparation to be
90 min before the injection of the product to be examined and examined or on the accompanying leaflet. Observe the
continuing 3 h after the injection. The ditlerence between the animals for 7 days.
maximum temperature and the initial temperature of each The preparation passes the test if none of the animals shows
rabbit is taken to be its response. When this difference is signs of ill healtho If more than one animal dies the preparation
negative, the result is counted as a zero response. fails the test. Ir one of the animals die s or shows signs of ill
Rabbits showing a temperature variation greater than 0.2 oC health, repeat the test. The preparation passes the test if none
between two successive readings in the determination of of the animals in the 2 nd group die or shows signs of ill health
the initial temperature are withdrawn from the test. In any in the time interval specifiedo
one test, only rabbits having initial temperatures which do
not differ from one another by more than 1 oC are used. AH
rabbits having an initial temperature higher than 39.8 oC or 0112008:20610
less than 38.0 oC are withdrawn from the test.
Interpretation of results. Having carried out the test first on a 2.6.10. HISTAMINE
group of three rabbits, repeat if necessary on further groups
of three rabbits to a total of four groups, depending on the Euthanise a guinea-pig weighing 250 g to 350 g that has been
results obtained. If the summed response of the first group deprived of food for the preceding 24 ho Remove a portion of
does not exceed the figure given in the seeond eolumn of the the distal smaH intestine 2 cm in length and empty the isolated
Table 2.6.8.-1, the substance passes the test. If the summed part by rinsing carefully with solution B described be!ow using
response exceeds the figure given in the second column of the a syringe. Attach a fine thread to each end and make a small
table but does not exceed the figure given in the third eoIumn transverse incision in the middle of the piece of intestine.
of the tabIe, repeat the test as indicated aboye. If the summed Place it in an organ bath with a capacity of 10 111L to 20 mL,
response exceeds the figure given in the third column of the containing solutiol1 B maintained at a constant temperature
table, the product fails the test. (34 oC to 36 OC) and pass through the solution a current of a
mixture of 95 parts of oxygen and 5 parts of carbon dioxide.
Table 206.8.-1
Attaeh one of the threads near to the bottom of the organ bath.
Number of rabbits Product passes if smnrned Product fails if sumrned Attach the other thread to an isotonic myograph and record
response does not exceed response exceeds
the contractions of the organ on a kymograph or other suitable
1.15 oC 2.65 oC
means of giving a permanent record. If a lever is used, its
6 2.80 oC 4.30 oC length is such that the movements of the organ are amplified
about 20 times. The tension on the intestine should be about
9 4.45 oC 5.95 oC
9.8 mN (1 g) and it should be adjusted to the sensitivity
12 6.60 oC 6.60 oC of the organ. Flush out the organ bath with solution B.
Allow it to stand for 10 mino Flush 2 or 3 times more with
Rabbits used in a test for pyrogens where the mean rise in the solution B. Stimulate a series of contractions by the addition of
rabbits' temperature has exceeded 1.2 oC are permanently measured volumes between 002 mL and 0.5 mL of a solution of
excluded. histamine dihydrochloride R having a strength which produces
reproducible submaximal responses. This dose is termed the
"high do se". Flush the organ bath (preferably by overflow
01/2008:20609 without emptying the bath) 3 times with solution B before
each addition of histamine. The successive additions should
2.6.9, ABNORMAL TOXICITY be made at regular intervals allowing a complete re!axation
between additions (about 2 min). Add equal volumes of
GENERAL TEST a weaker dilutíon of histamine dihydrochloride R which
Inject intravenously into each of 5 healthy mice, weighing produces reproducible responses approximate!y half as great as
17 g to 24 g, the quantity of the substance to be examined the "high dose". This dose is termed the "Iow dose". Continue
prescribed in the monograph, dissolved in 0.5 mL of water for the regular additions of "high" and "low" doses of histamine
injections R or of a 9 giL sterile solution of sodium chloride R. solution as indicated aboye, and alternate each addition with
Inject the solution over a period of 15 sto 30 s, unless an equal volume of a dilution of the solutíon to be examined,
otherwise prescribed. adjusting the dilution so that the contraction of the intestine,
The substance pass es the test if none of the mice die within if any, is smaller than that due to the "high dose" ofhistamine.
24 h or within such time as is specified in the individual Determine whether the contraction, if any, is reproducible and
monograph. If more than one animal dies the preparation that the responses to the "high" and "low" doses of histamine
fails the test. If one of the animals dies, repeat the test. The are unchanged. Calculate the activity of the substance to
substance passes the test if none of the animals in the 2nd group be examined in terms of its equivalent in micrograms of
die within the time interval specified. histamine base from the dilution determined as aboye.
The quantity so determined does not exceed the quantity
IMMUNOSERA AND VACCINES POR HUMAN USE prescribed in the monograph.
Unless otherwise prescribed, injeet intraperitoneally 1 human If the solution to be examined does not produce a contraction,
dose but not more than 1.0 mL into each of 5 healthy prepare a fresh solution adding a quantity of histamine
mice, weighing 17 g to 24 g. The human dose is that stated corresponding to the maximum tolerated in the monograph
011 the ¡abe! of the preparation to be examined or on the and note whether the contractions produced by the
accompanying leaflet. Observe the animals for 7 days. preparation with the added histamine correspond to the
The preparation passes the test if none of the animals amount of histamine added. If this is not the case, or if the
shows signs of ill health. If more than one animal dies, the contractions caused by the substance to be examined are not
preparation fails the test. If one of the animals dies or shows reproducible 01' if subsequent responses to "high" and "low"
signs of ill health, repeat the test. The preparation passes the doses of histamine are diminished, the results of the tests are
test if none of the animals in the 2nd group die or shows signs invalid and the test for depressor substances (2.6.11) must be
of ill health in the time interval specified. carried out.

184 See the information section 011 general monographs (ca ver pages)
PHARMACOPOEIA 8.0 2.6.12. Microbial enurneration tests

Solution A after the administration of the substance to be examined is


Sodium chloride 160.0 g less than the mean response to the low doses of histamine
previously injected.
Polassiu111 chloride 4.0 g

Caiciull1 chloride, anhydrous 2.0 g


07/2010:20612
Níagnesium chloride, anhydrous 1.0 g

0.10 g
Disodillm hydrogen phosphate dodecahydrate 2.6.1 MICROBIOLOGICAL
1Vaterfor injections R to 1000 mL EXAMINATION OF NON-STERILE
Solution B PRODUCTS: MICROBIAL
Sollltion A 50.0 mL ENUMERATION TESTS(2)
1-\tropine sulfate 1. INTRODUCTION
Sodiu111 hydrogen carbonate 1.0 g The tests described hereafter will allow quantitative
enumeration of mesophilic bacteria and fungi that may grow
Glucose monohydrate 0.5 g
under aerobic conditions.
Water for injections R lo 1000 111L The tests are designed primarily to determine whether
a substance or preparation complies with an established
Solution B should be freshly prepared and used within 24 h. specification for microbiological quality. When used for such
purposes follow the instructions given below, including the
number of samples to be taken, and interpret the results as
0112008:20611 stated below.
The methods are not applicable to products containing viable
2.6.11. DEPRESSOR SUBSTANCES micro-organisms as active ingredients.
Carry out the test 011 a cat weighing 110t less thal1 2 kg and Alternative microbiological procedures, including automated
anaesthetised with chloralose OI with a barbiturate that methods, may be used, provided that their equivalence to the
allows the maintenance of uniform blood pressure. Protect Pharmacopoeia method has been demonstrated.
the animal from loss of body heat and maintain it so that 2. GENERALPROCEDURES
the rectal temperature remains within physiologicallimits.
Introduce a canl1ula into the trachea. Insert a cannula filled Carry out the determination under conditions designed to
with a heparinised 9 giL solution of sodium chloride into the avoid extrinsic microbial contamination of the product to be
examined. The pl'ecautions taken to avoid contamination
commol1 carotid artery and connect it to a device capable of
giving a continuous record of the blood pressure. Insert into must be such that they do not affect any micro-organisms that
the femoral vein another cal1nula, filled with a heparinised are to be revealed in the test.
9 giL solution of sodium chloride, through which can be If the product to be examined has antimicrobial activity, this
injected the solutions of histamine and of the substance to is insofar as possible removed 01' neutralised. If inactivators
be examined. Determine the sensitivity of the animal to are used fol' this purpose, their efficacy and their absence of
histamine by injecting intravenously at regular intervals, toxicity for micro-organisms must be demonstrated.
doses of histamíne solution R corresponding to 0.1 flg and If surface-active substances are used for sample preparation,
0.15 flg of histamine base per kilogram of body mass. Repeat their absence of toxicity for and their
the lower dose at least 3 times. Administer the second and compatibility with inactivators used muse be demonstrated.
subsequent injections not les s than 1 min after the blood
pressure has returned to the leve! it was at immediately befare 3. ENUMERATION METHODS
the previous injection. The animal is used for the test only if a Use the membrane filtration method or the plate-count
readily discernible decrease in blood pressure that is constant methods, as prescribed. The most-probable-number (MPN)
for the lower dose is obtained and if the higher dose causes method is gene rally the least accurate method for microbial
greater responses. Dissolve the substance to be examined coul1tS, however, for certain product groups with a very low
in sufficient of a 9 giL solution of sodium chloride or other bioburden, it may be the most appropriate method.
prescribed solvent, to give the prescribed concentration. The choice of method is based on factors such as the nature of
Inject intravenously per kilogram of body mass 1.0 mL of the product and the required limit of micro-organisms. The
histamine solution R, followed by 2 successive injections of chosen method must allow testing of a sufficient sample size
the prescribed amount of the solution to be examined and, to judge compliance with the specificatiol1. The suitability of
finally, 1.0 mL of histamine solutíon R. The second, third and the method chosen must be established.
fourth injections are given not les s than 1 min after the blood
pressure has returned to the leve! it was at immediately before 4. GROWTH PROMOTION TEST, SUITABILITY OF THE
the preceding injection. Repeat this series of injections twice COUNTING METHOD AND NEGATIVE CONTROLS
and condude the test by giving 1.5 mL of hístamine solution R 4-1. GENERAL CONSIDERATIONS
per kilogram of body mass.
The ability of the test to detect micro-organisms in the
If the response to 1.5 mL of histamine solution R per kilogram presence of product to be tested must be established.
of body mass is not greater than that to 1. O mL the test is
invalido The substance to be examined fails the test if the mean Suitability must be confirmed if a change in testing
performance, or the product, which may affect the outcome of
of the series of responses to the substance is greater than the
the test is introduced.
mean of the responses to 1.0 mL of hislamíne solution R per
kilogram of body mass or if any one dose of the substance 4-2. PREPARATION OF TEST STRAINS
causes a greater depressor response than the conduding dose Use standardised stable suspensions of test strains or
of the histamine solution. The test animal must not be used in prepare them as stated below. Seed lot culture maÍntenance
another test for depressor substances if the second criterion techniques (seed-lot systems) are used so that the viable
applies or if the response to the high dose of histamine given micro-organisms used for inoculation are not more than

(2) This chapler has undergone pharmacopoeial hJrmonisation. See chapter 5,8. PhrInllocopoci(/{ lumnonisation.

General Notices (1) apply to all monographs and other texts 185
2.6.12. Microbial enumeration tests EUROPEAN PHARMACOPOEIA 8.0

5 passages removed from the original master seed-Iot. Grow For solid media, growth obtained must not differ by a factor
each of the bacterial and fungal test strains separately as greater than 2 from the calculated value for a standardised
described in Table 2.6.12.-1. inoculum. For a freshly prepared inoculum, growth of the
micro-organisms comparable to that previously obtained with
Use buffered sodium chloride-peptone solution pH 7.0 or a previously tested and approved batch of medium occurs.
phosphate buffer solution pH 7.2 to make test suspensions; to Liquid media are suitable if clearly visible growth of the
suspend A. brasiliensis spores, 0.05 per cent of polysorbate 80 micro-organisms comparable to that previously obtained with
may be added to the buffer. Use the suspensions within a previously tested and approved batch of medium occurs.
2 h or within 24 h if stored at 2-8 oc. As an alternative to 4-5. SUITABILITY OF THE COUNTING METHOD IN THE
preparing and thendiluting a fresh suspension of vegetative PRESENCE OF PRODUCT
cells of A. brasiliensis or B. subtilis, a stable spore suspension
is prepared and then an appropriate volume of the spore 4-5-1. Preparation ofthe sample. The method for sample
suspension is used for test inoculation. The stable spore preparation depends upon the physical characteristics of the
suspension may be maintained at 2-8 oC for a validated period product to be tested. If none of the procedures described
oftime. below can be demonstrated to be satisfactory, an alternative
procedure must be developed.
4-3. NEGATIVE CONTROL Water-soluble products. Dissolve or dilute (usually a 1 in
To verify testing conditions, a negative control is performed 10 dilution is prepared) the product to be examined in
using the chosen diluent in place of the test preparation. There buffered sodium chloride-peptone solution pH 7.0, phosphate
must be no growth of micro-organisms. A negative control buffer solution pH 7.2 or casein soya bean digest broth.
is also performed when testing the products as described in rf necessary, adjust to pH 6-8. Further dilutions, where
section 5. A failed negative control requires an investigation. necessary, are prepared with the same diluent.
4-4. GROWTH PROMOTION OF THE MEDIA Non-fatty products insoluble in water. Suspend the product
Test each batch of ready-prepared medium and each batch of to be examined (usually a 1 in 10 dilution is prepared) in
medium, prepared either from dehydrated medium or from buffered sodium chloride-peptone solution pH 7.0, phosphate
the ingredients described. buffer solution pH 7.2 or casein soya bean digest broth. A
surface-active agent such as 1 giL of polysorbate 80 may be
Inoculate portions/plates of casein soya bean digest broth added to assist the suspension of poorly wettable substances.
and casein soya bean digest agar with a small number (not lf necessary, adjust to pH 6-8. Further dilutions, where
more than 100 CFU) of the micro-organisms indicated in necessary, are prepared with the same diluent.
Table 2.6.12.-1, using a separate portion/plate of medium for Fatty products. Dissolve in isopropyl myristate, sterilised
each. Inoculate plates of Sabouraud -dextrose agar with a small by filtration or mix the product to be examined with the
number (not more than 100 CFU) of the micro-organisms minimum necessary quantity of sterile polysorbate 80 or
indicated in Table 2.6.12.-1, using a separate plate of another non-inhibitory sterile surface-active agent, heated if
medium for each. Incubate in the conditions described in necessary to not more than 40 oc, or in exceptional cases to not
Table 2.6.12.-l. more than 45 oc. Mix carefully and if necessary maintain the
Table 2.6.12.-1. - Preparation and use oftest rnicro-organisrns
Micro-organism Preparation of test Growth promotion Suitability of couuting method in the
síraiu presence of the product
Total aerobic Total yeasts and Total aerobic Total yeasts aud
microbial count moulds count microbial couut moulds count
Staphylococcus aureus Casein soya bean Casein soya bean digest Casein soya bean digest
such as: digest agar or casein agar and casein soya agar/MPN casein soya
soya bean digest broth bean digest broth bean digest broth
ATCC 6538 - -
30-35 oC :5 100 CFU :5 100 CFU
NCIMB 9518
18-24 h 30-35 oC 30-35 oC
CIP 4.83
:5 3 days :5 3 days
NBRC 13276
Pseudo monas Casein soya bean Casein soya bean digest Casein soya bean digest
aeruginosa digest agar or casein agar and casein soya agar/MPN casein soya
such as: soya bean digest broth bean digest broth bean digest broth
ATCC 9027 30-35 oC :5 100 CFU :5 100 CFU -
NCIMB 8626 18-24 h 30-35 oC 30-35 oC
CIP 82.118 :5 3 days :5 3 days
NBRC 13275
Bacil/us subtilis Casein soya bean Casein soya bean digest Casein soya bean digest
such as: digest agar or casein agar and casein soya agar/MPN casein soya
ATCC 6633 soya bean digest broth bean digest broth bean digest broth
30-35 oC -
NCIMB 8054 :5 100 CFU :5 100 CFU
CIP 52.62 18-24 h 30-35 oC 30-35 oC
NBRC 3134 :5 3 days :5 3 days
Candida albicans Sabouraud -dextrose Casein soya bean Sabouraud -dextrose Casein soya bean Sabouraud-dextrose
such as: agar or Sabouraud- digest agar agar digest agar agar
dextros e broth :5 100 CFU :5 100 CFU :5 100 CFU :5 100 CFU
ATCC 10231 20-25 oC
NCPF 3179 30-35 oC 20-25 oC 30-35 oC 20-25 oC
2-3 days
IP 48.72 :5 5 days :5 5 days :5 5 days :5 5 days
NBRC 1594 MPN: not applicable
Aspergil/us brasiliensis Sabouraud -dextrose Casein soya bean Sabouraud -dextrose Casein soya bean Sabouraud -dextrose
such as: agar or potato-dextrose digest agar agar digest agar agar
agar :5 100 CFU :5 100 CFU :5 100 CFU :5 100 CFU
ATCC 16404
20-25 oC 30-35 oC 20-25 oC 30-35 oC 20-25 oC
IMI 149007
5-7 days, or until good :5 5 days :5 5 days :5 5 days :5 5 days
IP 1431.83 sporulation is achieved
NBRC 9455 MPN: not applicable

186 See the inforrnation section on general rnonographs (cover pages)


EUROPEAN PHARMACOPOEIA 8.0 2.6.12. Microbial ennmeration tests

temperature in a water-bath. Add sufficient of the pre-warmed Table 2.6.12.-2. - Common neutralising agents for interfering
chosen diluent to make a 1 in 10 dilution of the original substances
product. Mix carefully whilst maintaining the temperature for Interfering substance Potential neutralising
the shortest time necessary foy the formation of an emulsiono method
Further serial tenfold dilutions may be prepared using the Glutaraldehyde, mercurials Sodium hydrogensulfite
chosen diluent containing a suitable concentration of sterile (sodium bisulfite)
polysorbate 80 or another non-inhibitory sterile surface-active Phenolics, alcohol, aldehydes, sorbate Dilution
agent.
Aldehydes Glycine
Fluids or solids in aerosol formo Aseptically transfer the
product into a membrane filter apparatus or a sterile container Quaternary Ammonium Compounds Lecithin
for further sampling. Use either the total contents or a defined (QACs), parahydroxybenzoates (parabens),
bis-biguanides
number of metered doses from each of the containers tested.
QACs, iodine, parabens Polysorbate
Transdermal patches. Remove the protective cover sheets
('releas e liners') of the transdermal patches and place them, Mercurials Thioglycollate
adhesive side upwards, on sterile glass or plastic trays. Cover Mercurials, halogens, aldehydes Thiosulfate
the adhesive surface with a sterile porous material, for example
sterile gauze, to prevent the patches from sticking together, EDTA (edetate) Mg2+ ol' Ca2+ ions
and transfer the patches to a suitable volume of the chosen
diluent containing inactivators such as polysorbate 80 and/or 4-5-4. Recovery of mkro-organism in the presence oí
lecithin. Shake the preparation vigorously for at least 30 mino producto For each of the micro-organisms listed, separate
tests are performed. Only micro-organisms of the added test
4-5-2. Inoculation and dilution. Add to the sample prepared strain are counted.
as described aboye (4-5-1) and to a control (with no test
4-5-4-1. Membrane filtration. Use membrane filters having
material ineluded) a sufficient volume of the microbial
a nominal pore size not greater than 0.45 !lm. The type
suspension to obtain an irtoculum of not more than 100 CFU.
of filter material is chosen such that the bacteria -retaining
The volume of the suspension of the inoculum should not
efficiency is not affected by the components of the sample to
exceed 1 per cent of the volume of diluted product.
be investigated. For each of the micro-organisms listed, one
To demonstrate acceptable microbial recovery from the membrane filter is used.
product, the lowest possible dilution factor of the prepared Transfer a suitable amount of the sample prepared as described
sample must be used for the test. Where this is not possible under 4-5-1 to 4-5-3 (preferably representing 1 g of the
due to antimicrobial activity or poor solubility, further product, or less if large numbers of CFU are expected) to the
appropriate protocols must be developed. lf inhibition of membrane filter, filter immediately and rinse the membrane
growth by the sample cannot otherwise be avoided, the aliquot fiIter with an appropriate volume of diluent.
of the microbial suspension may be added after neutralisation,
For the determination of total aerobic microbial count
dilution or filtration.
(TAMC), transfer the membrane filter to the surface of casein
4-5-3. Neutralisation/removal of anHmicrobial activity. soya bean digest agar. For the determination of total combined
The number of micro-organisms recovered from the prepared yeasts/moulds count (TYMC), transfer the membrane to the
sample diluted as described in 4-5-2 and incubated following surface of Sabouraud-dextrose agar. Incubate the plates as
the procedure described in 4-5-4, is compared to the number indicated in Table 2.6.12.-1. Perform the counting.
of micro-organisms recovered from the control preparation. 4-5-4-2. Plate-count methods. Perform plate-count methods
If growth is inhibited (reduction by a factor greater than 2), at least in duplicate for each medium and use the mean count
then modify the procedure for the particular enumeration of the resulto
test to ensure the validity of the results. Modification of the 4-5-4-2-1. Pour-plate method
procedure may in elude, for example, (1) an increase in the For Petri dishes 9 cm in diameter, add to the dish 1 mL
volume of the diluent or culture medium, (2) incorporation of the sample prepared as described under 4-5-1 to
of specific or general neutralising agents into the diluent, 4-5-3 and 15-20 mL of casein soya bean digest agar or
(3) membrane filtration, or (4) a combination ofthe aboye Sabouraud-dextrose agar, both media being at not more
measures. than 45 oc. If larger Petri dishes are used, the amount of
Neutralising agents. Neutralising agents may be used to agar medium is increased accordingly. For each of the
neutralise the activity of antimicrobial agents (Table 2.6.12.-2). micro-organisms listed in Table 2.6.12.-1, at least 2 Petri dishes
They may be added to the chosen diluent or the medium are used. Incubate the plates as indicated in Table 2.6.12.-1.
preferably before sterilisation. If used, their efficacy and their Take the arithmetic mean of the counts per medium and
absence of toxicity for micro-organisms must be demonstrated calculate the number of CFU in the original inoculum.
by carrying out a blank with neutraliser and without product. 4-5-4-2-2. Surface-spread method
lf no suitable neutralising method can be found, it can be For Petri dishes 9 cm in diameter, add 15-20 mL of casein soya
assumed that the failure to iso late the inoculated organism is bean digest agar or Sabouraud-dextrose agar at about 45 oC
attributable to the microbicidal activity of the product. This to each Pe tri dish and allow to solidify. If larger Pe tri dishes
information serves to indicate that the product is not likely to are used, the volume of the agar is increased accordingly.
be contaminated with the given species of the micro-organismo Dry the plates, for example in a laminar-air-flow cabinet
However, it is possible that the product only inhibits some of or an incubator. For each of the micro-organisms listed
the micro-organisms specified herein, but does not inhibit in Table 2.6.12.-1, at least 2 Petri dishes are used. Spread
others not ineluded amongst the test strains or for which the a measured volume of not less than 0.1 mL of the sample
latter are not representative. Then, perform the test with the prepared as described under 4-5-1 to 4-5-3 over the surface
highest dilution factor compatible with microbial growth and of the medium. Incubate and count as prescribed under
the specific acceptance criterion. 4-5-4-2-1.

General Notices (1) apply to all monographs and other texts 187
2.6.12. Microbial enumeration tests EUROPEAN PHARMACOPOEIA 8.0

4-5-4-3. (MPN) method. The Table 2.6.12.-3. - Most-probable-number values of


precision and accuracy of the MPN method is less than micro-organisms
that of the membrane filtration method Ol' the plate-count Observed combinations of numbers {jf
method. Unreliable results are obtained partinilarly for the tubes showing grow1h in each set MPN per
enumeration of moulds. For these reasons the MPN method is 95 per cent
Nrnnber of grams 01' miHilitres of grammper
confidence
reserved for the enumeration of TAMC in situations where no poduct per tube mil!ilitre oí
limits
product
other method is available. If the use of the method is justified, 0.1 !1.O1 0.001
proceed as follows.
O O O <3 0-9.4
Prepare a series of at least 3 serial tenfold dilutions of the O O 1 3 0.1-9.5
product as described under 4-5-1 to 4-5-3. From each level of
dilution, 3 of 1 g or 1 mL are used to inoculate 3 tubes O 1 O 3 0.1-10
with 9-10 of casein soya bean digest broth. Ifnecessary, a O 1 1 6.1 1.2-17
surface-active agent such as polysorbate 80 or an inactivator
of antimicrobial agents may be added to the medium. Thus, if O 2 O 6.2 1.2-17
3 levels of dilution are prepared, 9 tubes are inoculated. O 3 O 9.4 3.5-35

Incubate a11 tubes at 30-35 oC for not more than 3 days. If 1 O O 3.6 0.2-17
reading of the results is difficult or uncertain owing to the 1 O J 7.2 1.2-17
nature of the product to be examined, sub culture in the same
broth, 01' in casein soya bean digest agar, foi' 1-2 days at the 1 O 2 11 4-35
same temperature and use these results. Determine the most 1 1 O 7.4 1.3-20
probable number of micro-organisms per gram or millilitre of
the to be examined from Table 2.6.12.-3. 1 1 1 11 4-35

4-6. RESULTS AND INTERPRETATION 1 2 O 11 4-35

When the suitability of the membrane filtration 1 2 1 15 5-38


plate-count method, a mean count of any of the
1 3 O 16 5-38
5aJeU'UIO not differing by a factor greater than 2 from

of the control defined in 4-5-2 in the absence of the 2 O O 9.2 1.5-35


must be obtained. When verifying the suitability of
2 O 1 14 4-35
MPN method the calculated value from the inoculum
must be within 95 per cent confidence limits of the results 2 O 2 20 5-38
obtained with the control.
2 1 O 15 4-38

If the aboye criteria cannot be met for one or more of the 2 1 1 20 5-38
)';"1"'.111' tested with any of the described methods, the 2 1 2 27 9-94
method and test condítiol1s that come closest to the criteria
are used to test the 2 2 O 21 5-40

2 2 1 28 9-94

2 2 2 35 9-94
5. TESTING OF PRODUCTS
2 3 O 29 9-94
5-1. AMOUNT USED FOR THE TEST
2 3 1 36 9-94
Unless otherwise prescribed, use 10 g or 10 mL of the product
to be examined taken with the precautio11s referred to aboye. 3 O O 23 5-94
For fluids or solids in aerosol for111, sample 10 containers. For 3 O 1 38 9-104
transdermal sample 10 patches.<
3 O 2 64 16-181
The amount to be tested may be reduced for active substances 3 1 O 43 9-181
that will be formulated in the following conditions: the
amount per dosage unit (e.g. tablet, capsule, injection) is less 3 1 1 75 17 -199
than 01' equal to 1 mg or the amount per gram or millilitre (for 3 1 2 120 30-360
preparatiol1s 110t presented in dose units) is les s than 1 mg. In
these cases, the amoul1t to be tested is not less than the amount 3 1 3 160 30-380
present in 10 dosage units or 10 g or 10 mL of the product. 3 2 O 93 18-360

3 2 1 150 30-380
For materials used as active substances where sample quantity
is limited or batch size isextremely small (Le. less tha11 3 2 2 210 30-400
1000 mL or 1000 the amount tested sha11 be 1 per cent of
3 2 3 290 90-990
the batch unless a amount is prescribed or justified and
authorised. 3 3 O 240 40-990

3 3 1 460 90-1980
For products where the total number of entities in a batch is
less than 200 (e.g. samples used in clinical trials), the sample 3 3 2 1100 200-4000
size rrIay be reduced to 2 units, or 1 unit if the size is less than
3 3 3 > llOO
100.
5-2. EXAMINATION OF THE PRODUCT
Select the at random from the bulk material or from
the available containers of the preparation. To obtain the 5-2-1. Membrane fiHration
mix the contents of a sufficient number of Use a filtration apparatus designed to allow the transfer of the
the filter to the medium. the sample using a method that

188 See the information section on (caver pages)


EUROPEAN PHARMACOPOEIA 8.0 2.6.13. Test for spedfied micro-organisms

has been shown suitable as described in section 4 and transfer 04/2010:20613


the appropriate amount to each of 2 membrane filters and
filter immediately. Wash each filter following the procedure 2.6.13. MICROBIOLOGICAL
shown to be suitable.
EXAMINATION OF NON -STERILE
For the determination ofTAMC, transfer one of the membrane
filters to the surface of casein soya bean digest agar. For the PRODUCTS: TEST FOR SPECIFIED
determination of TYMC, transfer the other membrane to MICRO-ORGANISMS(3J
the surface of Sabouraud-dextrose agar. lncubate the plate
of casein soya bean digest agar at 30-35 oC for 3-5 days and 1. INTRODUCTION
the plate of Sabouraud-dextrose agar at 20-25 oC for 5-7 days. The tests described hereafter will allow determination of the
Calculate the number of CFU per gram 01' per millilitre of absence or lil11ited occurrence of specifled micro-organisms
product. that may be detected under the conditions described.
When examining transdermal patches, filter 10 per cent of the The tests are designed primarily to determine whether
volume ofthe preparation described under 4-5-1 separately a substance or preparation complies with an established
through each of 2 sterile filter membranes. Transfer one specification for microbiological quality. When used for such
membrane to casein soya bean digest agar for TAMC and the purposes, follow the instructions given below, including the
other membrane to Sabouraud-dextrose agar for TYMC. number of samples to be taken, and interpret the results as
stated below.
5-2-2. Plate-count methods
Alternative microbiological procedures, including automated
5-2-2-1. Pour-plate method methods, may be used, provided that their equivalence to the
Prepare the sample using a method that has been shown to be Pharmacopoeia method has been demonstrated.
suitable as described in section 4. Prepare for each medium
at least 2 Petri dishes for each leve! of dilution. Incubate the 2. GENERAL PROCEDURES
plates of casein ,soya bean digest agar at 30-35 oC for 3-5 days The preparation of samples is carried out as described in
and the plates of Sabouraud-dextrose agar at 20-25 oC for general chapter 2.6.12.
5-7 days. Se!ect the plates corresponding to a given dilution If the product to be examined has antimicrobial activity, this
and showing the highest number of colonies less than 250 is insofar as possible removed or neutralised as described in
for TAMC and 50 for TYMC. Take the arithmetic mean per general chapter 2.6.12.
culture medium of the counts and calculate the number of If surface-active substances are used for sample preparation,
CFU per gram or per millilitre of product. their absence of toxicity for micro-organisms and their
5-2-2-2. Surface-spre¡td method compatibility with inactivators used must be demonstrated as
described in general chapter 2.6.12.
Prepare the sample using a method that has been shown
to be suitable as described in section 4. Prepare at least 2 3. GROWTH-PROMOTING AND INHIBITORY
Pe tri dishes for each medium and each leve! of dilution. For PROPERTIES OF THE MEDIA, SUITABILITY OF THE
incubation and calculatiol1 of the number of CFU proceed as TEST AND NEGATIVE CONTROLS
described for the pour-plate method.
The ability of the test to detect micro-organisms in the
5-2-30 Most-probable-number method presence of the product to be tested must be established.
Prepare and dilute the sample using a method that has been Suitability must be confirmed if a change in testing
shown to be suitable as described in section 4. Incubate all performance, or the product, which may affect the outcome of
tubes at 30-35 oC for 3-5 days. Sub culture if necessary, using the test is introduced.
the procedure shown to be suitable. Record for each level 3-1. PREPARATION OF TEST STRAINS
of dilution the number of tubes showing microbial growth. Use standardised stable suspensions of test strains or prepare
Determine the most probable number of micro-organisms them as stated below. Seed 10t culture maintenance techniques
per gram or millilitre of the product to be examined from (seed -lot systems) are used so that the viable micro-organisms
Table 2.6.12.-3. used for inoculation are not more than 5 passages removed
5-3. INTERPRETATION OF THE RESULTS from the original master seed-lot.
The total aerobic microbial count (TAMC) is considered to be 3-1-1. Aerobic micro-organisms. Grow each of the bacterial
equal to the number of CFU found using casein soya bean test strains separately in casein soya bean digest broth or
digest agar; if colonies of fungi are detected on this medium, on casein soya bean digest agar at 30-35 oC for 18-24 h.
they are counted as part of the TAMC. The total combined Grow the test strain for Candida albicans separately on
yeasts/mould count (TYMC) is considered to be equal to Sabouraud-dextrose agar or in Sabouraud-dextrose broth at
the number of CFU found using Sabouraud-dextrose agar; 20-25 oC for 2-3 days.
if colonies of bacteria are detected on this medium, they are - Staphylococcus aureus such as ATCC 6538, NCIMB 9518,
counted as part of the TYMC. When the TYMC is expected to CIP 4.83 or NBRC 13276;
exceed the acceptance criteriol1 due to the bacterial growth, - Pseudomonas aeruginosa such as ATCC 9027, NCIMB 8626,
Sabouraud-dextrose agar containing antibiotics may be used. CIP 82.118 or NBRC 13275;
lf the count is carried out by the MPN method the calculated
value is the TAMC. Escherichia coli such as ATCC 8739, NCIMB 8545,
CIP 53.126 or NBRC 3972;
When an acceptance criterion for microbiological quality is - Salmonella enterica subspo enterica serovar Typhimurium,
prescribed it is interpreted as follows: such as ATCC 14028 or, as an alternative, Salmonella
- 10 1 CFU: maximum acceptable count = 20; enterica subsp. enterica serovar Abony such as
NBRC 100797, NCTC 6017 or CIP 80.39;
- 10 2 CFU: maximul11 acceptable count = 200;
- Candida albicans such as ATCC 10231, NCPF 3179,
- 10 3 CFU: maximum acceptable count = 2000, and so forth. IP 48.72 or NBRC 15940
The recoml11ended solutions and media are described in Use buffered sodium chloride-peptone solution pH 7.0 or
general chapter 2.6.13. phosphate buffer solution pH 7.2 to make test suspensions.

(3) This chapter has undergone pharmacopoeial hannonisation. See chapter 5.8. Phannacopocial harmollisation.

General Natices (1) apply to al! monographs and ather texts 189
2.6.13. Test for sped:fied mkro-organisms EUROPEAN PHARMACOPOEIA 8.0

Use the suspensions within 2 h or within 24 h if stored at more than the shortest period of time specified in the test.
2-8 oc. Growth of the micro-organism comparable to that previously
3-1-2. Clostridia. Use elostridium sporogenes such as obtained with a previously tested and approved batch of
ATCC 11437 (NBRC 14293, NCIMB 12343, CIP 100651) medium occurs.
or ATCC 19404 (NCTC 532 or CIP 79.03) or NBRC 14293. Test for inhibitory properties, liquid or solid media: inoculate
Grow the clostridial test strain under anaerobic conditions in the appropriate medium with at least 100 CFU of the
reinforced medium for clostridia at 30-35 oC fOI 24-48 h. As appropriate micro-organismo Incubate at the specified
an alternative to preparing and then diluting down a fresh temperature for not less than the longest period of time
suspension of vegetative cells of el. sporogenes, a stable spore specified in the test. No growth of the test micro-organism
suspension is used for test inoculation. The stable spore occurs.
suspension may be maintained at 2-8 oC for a validated periodo
Test for indicative properties: perform the surface-spread
3-2. NEGATIVE CONTROL method, inoculating each plate with a small number (not more
To verify testing conditions, a negative control is performed than 100 CFU) of the appropriate micro-organismo Incubate at
using the chosen diluent in place of the test preparation. There the specified temperature for a period of time within the range
must be no growth of micro-organisms. A negative control specified in the test. Colonies are comparable in appearance
is also performed when testing the products as described in and indication reactions to those previously obtained with a
section 4. A failed negative control requires an investigation. previously tested and approved batch of medium.
3-3. GROWTH PROMOTION AND INHIBITORY 3-4. SUITABILITY OF THE TEST METHOD
PROPERTIES OF THE MEDIA For each product to be tested, perform the sample preparation
Test each batch of ready-prepared medium and each batch of as described in the relevant paragraph in section 4. Add each
medium prepared either from dehydrated medium or from test strain at the time of mixing, in the prescribed growth
ingredients. medium. Inoculate the test strains individually. Use a number
Verify suitable properties of relevant media as described in of micro-organisms equivalent to not more than 100 CFU in
Table 2.6.13.-l. the inoculated test preparation.
Test for growth promoting properties, liquid media: inoculate Perform the test as described in the relevant paragraph in
a portion of the appropriate medium with a small number section 4 using the shortest incubation period prescribed.
(not more than 100 CFU) of the appropriate micro-organismo The specified micro-organisms must be detected with the
Incubate at the specified temperature for not more than the indication reactions as described in section 4.
shortest period of time specified in the test. Clearly visible
growth of the micro-organism comparable to that previously Any antimicrobial activity of the product necessitates a
obtained with a previously tested and approved batch of modification of the test procedure (see 4-5-3 of general
medium occurs. chapter 2.6.12).
Test for growth promoting properties, solid media: perform lf for a given product the antimicrobial activity with respect
the surface-spread method, inoculating each plate with a to a micro-organism for which testing is prescribed cannot
small number (not more than 100 CFU) of the appropriate be neutralised, then it is to be assumed that the inhibited
micro-organismo Incubate at the specified temperature for not micro-organism will not be present in the producto

Table 2.6.13.-1 - Growth promoting, inhibitory and indicative properties of media


Medium Property Test strains

Test for bile-tolerant gram-negative Enterobacteria enrichment Growth promoting E. coli


bacteria broth-Mossel P. aeru;;inosa
Inhibitory S. aureus

Violet red bile glucose agar Growth promoting + indicative E. coli


P. aeru;;inosa
Test for Escherichia coli MacConkey broth Growth promoting E. colí

Inhibitory S. aureus

MacConkey agar Growth promoting + indicative E. coli

Test for Salmonella Rappaport Vassiliadis Salmonella Growth promoting Salmonella enterica subsp. enterica
enrichment broth serovar Typhimurium or Salmonella
enterica subsp. enterica serovar Abony
Inhibitory S. aureus

Xylase, lysine, deaxychalate agar Growth promating + indicative Salmonella enterica subsp. enterica
serovar Typhimurium ar Salmonella
enterica subsp. enterica serovar Abony
Test for Pseudo monas aeruginosa Cetrimide agar Growth promoting P. aeruginosa

Inhibitory E. coli

Test for Staphylococcus aureus Mannital salt agar Growth promating + indicative S. aureus

Inhibitory E. coli
Test for clostridia Reinforced medium for clostridia Growth promoting el. sporogenes
Columbia agar Growth promoting Cl. sporagenes

Test for Candida albicans Sabouraud dextro se broth Growth promoting C. albicans

Sabouraud dextrose agar Growth promoting + indicative C. albicans

190 See the information section on general monographs (cover pages)


EUROPEAN PHARMACOPOEIA 8.0 2.6.13. Test for specified micro-organisms

4. TESTING OF PRODUCTS 4-3-2. Selection and sub culture. Transfer 0.1 mL of casein
4-1. BILE-TOLERANT GRAM-NEGATIVE BACTERIA soya bean digest broth to 10 mL of Rappaport Vassiliadis
Salmonella enrichment broth and incubate at 30-35 oC for
4-1-1. Sample preparation and pre-incubation. Prepare 18-24 h. Sub culture on plates of xylose, lysine, deoxycholate
a sample using a 1 in 10 dilution of not less than 1 g of the agar. Incubate at 30-35 oC for 18-48 h.
product to be examined as described in general chapter 2.6.12,
but using casein soya bean digest broth as the chosen diluent, 4-3-3. Interpretation. The possible presence of Salmonella is
mix and incubate at 20-25 oC for a time sufficient to resuscitate indicated by the growth of well-developed, red colonies, with
the bacteria but not sufficient to encourage multiplication of or without black centres. This is confirmed by identification
the organisms (usually 2 h but not more than 5 h). tests.

4-1-2. Test for absence. Unless otherwise prescribed, use the The product complíes with the test if colonies of the types
volume corresponding to 1 g of the product, as prepared in described are not present or if the confirmatory identification
4-1-1, to inoculate enterobacteria enrichment broth-Mossel. tests are negative.
Incubate at 30-35 oC for 24-48 h. Sub culture on plates of violet 4-4. PSEUDOMONAS AERUGINOSA
red bile glucose agar. Incubate at 30-35 oC for 18-24 h. 4-4-1. Sample preparation and pre-incubation. Prepare
The product complíes with the test if there is no growth of a sample using a 1 in 10 dilution of not less than 1 g of the
colonies. product to be examined as described in general chapter
4-1- 3. Quantitative test 2.6.12, and use 10 mL or the quantity corresponding to
1 g or 1 mL to inoculate a suitable amount (determined as
4-1-3-1. Selection and subculture. Inoculate suitable described under 3-4) of casein soya bean digest broth and
quantities of enterQbacteria enrichment broth-Mossel with the mix. When testing transdermal patches, filter the volume of
preparation as described under 4-1-1 and/or dilutions of it sample corresponding to 1 patch of the preparation described
containing respectively 0.1 g, 0.01 g and 0.001 g (or 0.1 mL, under 4-5-1 in general chapter 2.6.12 through a sterile filter
0.01 mL and 0.001 mL) ofthe product to be examined. membrane and place in 100 I11L of casein soya bean digest
Incubate at 30-35 oC for 24-48 h. Sub culture each of the broth. Incubate at 30-35 oC for 18-24 h.
cultures on a plate of violet red bile glucose agar. Incubate at
30-35 oC for 18-24 h. 4-4-2. Selection and sub culture. Sub culture on a plate of
cetrimide agar and incubate at 30-35 oC for 18-72 h.
4-1-3-2. Interpretation. Growth of colonies constitutes a
positive result. Note the smallest quantity of the product that 4-4-3. Interpretation. Growth of colonies indicates the
gives a positive result and the largest quantity that gives a possible presence of P. aeruginosa. This is confirmed by
negative result. Determine from Table 2.6.13.-2 the probable identification tests.
number of bacteria. The product complíes with the test if colonies are not present
or if the confirmatory identification tests are negative.
Table 2.6.13.-2 - Interpretation of results
4-5. STAPHYLOCOCCUS AUREUS
Results fúr each quantity of product Probable
number of 4-5-1. Sample preparation and pre-incubation. Prepare
(U gor 0.01 g or 0.001 g or bacteria per a sample using a 1 in 10 dilution of not less than 1 g of the
0.1 rnL 0.01 rnL 0.001 rnL gram or product to be examined as described in general chapter
millilitre úf
product
2.6.12, and use 10 mL or the quantity corresponding to
1 g or 1 mL to inoculate a suitable amount (determined as
+ + + > lO'
described under 3-4) of casein soya bean digest broth and
+ + - mix. When testing transdermal patches, filter the volume of
< 10 and > 102
3

- sample corresponding to 1 patch of the preparation described


+ -
< !O' and > 10
under 4-5-1 in general chapter 2.6.12 through a sterile filter
- - - < 10 membrane and place in 100 mL of casein soya bean digest
broth. Incubate at 30-35 oC for 18-24 h.
4-2. ESCHERICHIA COLI 4-5-2. Selection and suoculture. Sub culture on a plate of
4-2-1. Sample preparation and pre-incubation. Prepare mannitol salt agar and incubate at 30-35 oC for 18-72 h.
a sample using a 1 in 10 dilution of not less than 1 g of the 4-5-3. Interpretation. The possible presence of S. aureus is
product to be examined as described in general chapter 2.6.12, indicated by the growth of yellow/white colonies surrounded
and use 10 mL or the quantity corresponding to 1 g or 1 mL to by a yellow zone. This is confirmed by identification tests.
inoculate a suitable amount (determined as described under
3-4) of casein soya bean digest broth, mix and incubate at The product complíes with the test if colonies of the types
30-35 oC for 18-24 h. described are not present or if the confirmatory identification
tests are negative.
4-2-2. Selection and subculture. Shake the container, transfer
1 mL of casein soya bean digest broth to 100 mL of MacConkey 4-6. CLOSTRIDIA
broth and incubate at 42-44 oC for 24-48 h. Sub culture on a 4-6-1. Sample preparation and heat treatment. Prepare a
plate of MacConkey agar at 30-35 oC for 18-72 h. sample using a 1 in 10 dilution (with a minimum total volume
4-2-3. Interpretation. Growth of colonies indicates the of 20 mL) of not less than 2 g or 2 mL of the product to be
possible presence of E. eolio This is confirmed by identification examined as described in general chapter 2.6.12. Divide the
tests. sample into 2 portions of at least 10 mL. Heat 1 portion at
80 oC for 10 min and cool rapidly. Do not heat the other
The product complies with the test if no colonies are present portion.
or if the identification tests are negative.
4-6-2. Selection and suoculture. Use 10 mL or the quantity
4-3. SALMONELLA corresponding to 1 g or 1 mL of the product to be examined
4-3 -1. Sample preparation and pre-incubation. Prepare of both portions to inoculate suitable amounts (determined
the product to be examined as described in general chapter as described under 3-4) of reinforced medium for clostridia.
2.6.12, and use the quantity corresponding to not less than Incubate under anaerobic conditions at 30-35 oC for 48 h.
10 g or 10 mL to inoculate a suitable amount (determined as After incubation, make sub cultures from each container on
described under 3-4) of casein soya bean digest broth, mix Columbia agar and incubate under anaerobic conditions at
and incubate at 30-35 oC for 18-24 h. 30-35 oC for 48-72 h.

General Notiees (1) apply to all monographs and other texts 191
2.6.13. Test for spedfied micro-organisms EUROPEAN PHARMACOPOEIA 8.0

4-6-3. Interpretation. The occurrence of anaerobic growth of Casein soya bean digest agar
rods (with or without endospores) giving a negative catalase Pancreatic digest of casein 15.0 g
reaction indicates the presence of clostridia. This is confirmed
by identification tests. Papaic digest of soya bean 5.0 g

Sodium chloride 5.0 g


The product complies with the test if colonies of the types
described are not present or if the confirmatory identiÍÍcation Agar 15.0 g
tests are negative.
Purified water 1000 mL
4-7. CANDIDA ALBICANS
4-7-1. Sample preparation and pre-incubation. Prepare the Adjust the pH so that after sterilisation it is 7.3 ± 0.2 at 25 oc.
product to be examined as described in general chapter 2.6.12, Sterilise in an autoclave using a validated cycle.
and use 10 mL Ol" the quantity corresponding to not less than
1 g or 1 mL to inoculate 100 mL of Sabouraud-dextrose broth
and mix. Incubate at 30-35 oC for 3-5 days.
4-7-2. Selection and subculture. Sub culture on a plate of Sabouraud-dextrose agar
Sabouraud-dextrose agar and incubate at 30-35 oC for 24-48 h. Dextrose 40.0 g
4-7-3. Interpretation. Growth ofwhite coJonies may indicate 10.0 g
Mixture of peptic digest of animal tissue and pancreatic
the presence of C. albicans. This is conÍÍrmed by identification digest of casein (1:1)
tests. Agar 15.0 g

The product complíes with the test if such coJonies are 110t Purified water 1000 mL
present or if the conflrmatory identiflcation tests are negative.
Adjust the pH so that after sterilisation it is 5.6 ± 0.2 at 25 oc.
Sterilise in an autoclave using a validated cyele.
The following section is given for information.

5. RECOMMENDED SOLUTIONS AND CULTURE MEDIA


Potato dextrose agar
lnfusion from patataes 200 g
The following SOlUtiOI1S and culture media have been found to
Dextrose 20.0 g
be satisfactory for the purposes fol' which they are prescribed
in the test for microbial contamination in the Pharmacopoeia. Agar 15.0 g
Other media may be used provided that their suitability can
Purified water 1000 mL
be demonstrated.

Stock buffer solution. Place 34 g of potassium dihydrogen


the pH so that after sterilisation it is 506 ± 0.2 at 25 oc.
phosphate in a 1000 mL volumetric flask, dissolve in 500 mL of
Sterilise in al1 autoclave using a validated
purified water, adjust to pH 7.2 ± 0.2 with sodium hydroxide,
dilute to 1000.0 mL with purified water and mix. Dispense
into containers and sterilise. Store at 2-8 oc.

Phosphate buffer solution pH 7.2. Prepare a mixture of stock Sabouraua-dextrose broth


buffer solution and purified water (1:800 V/V) and sterilise. Dextrose 20.0 g

Buffered sodium chloride-peptone solution pH 700 Mixture af peptic digest al' animal tissue amI pancreatic 10.0 g
diges! af casein (1: 1)
Potassium dihydrogen phosphate 3.6 g
Purified water 1000 mL
Disodium hydrogen phosphate 7.2 g, equivalent to 0.067 M phosphate
dihydrate
Sodium chloride 4.3 g Adjust the pH so that after sterilisation jt is 5.6 ± 0.2 at 25 oc.
Sterilise in an autoclave using a validated cyele.
Peptone (meat or easein) l.0 g

Purified water 1000 mL

Sterilise in an autoclave using a validated cycle. Enterobaderia enrichment broth-Mossel


Pancreatic digest of gelatin 10.0 g

Glucase monohydrate 5.0 g


Casein soya bean digest broth Dehydrated ox bile 20.0 g
Pancreatic digest of casein 17.0 g
Potassillm dihydrogen phosphate 2.0 g
Papaie digest of soya bean 3.0 g
Disodium hydrogen phasphate dibydrate 8.0 g
Sodium ehloride 5.0 g
Brilliant green 15 mg
Dipotassium hydrogen phosphate 2.5 g
Purified water 1000 mL
Glucose monohydrate 2.5 g

Purified water 1000 mL Adjust the pH so that after heating it is 7.2 ± 0.2 at 25 oc. Heat
at 100 oC for 30 min and cool immediately.
Adjust the pH so that after sterilisation it is 7.3 ± 0.2 at 25 oc.
Sterilise in an autoclave using a validated cycle.

192 See the monographs (cover pages)


EUROPEAN PHARMACOPOEIA 8.0 2.6.13. Test for spedfied micro-organisms

Violet red bile glucose agar Sueros e 7.5 g


Yeast extract 3.0 g Sodium chloride 5.0 g
Pancreatic digest of gelatin 7.0 g Yeast extraet 3.0 g
Bile salts 1.5 g 80 mg
Phenol red
Sodium chloride 5.0 g Agar 13.5g
Glucose monohydrate 10.0 g 2.5 g
Sodium deoxycholate
Agar 15.0 g Sodium thiosulfate 6.8 g
Neutral red 30 mg Ferric ammonium citrate 0.8 g
Crystal violet 2 mg 1000 mL
Purified water
Purified water 1000 mL
Adjust the pH so that after heating it is 7.4 ± 0.2 at 25 oc.
Adjust the pH so that after heating it is 7.4 ± 0.2 at 25 oc. Heat Heat to boiling, cool to 50 oC and pour into Petri dishes. Do
to boiling; do not heat in an autoclave. not heat in an autoclave.

MacConkey broth
Cetrimide agar
Pancreatic digest of gelatin 20.0 g
Pancreatic digest of gelatin 20.0 g
Lactose monohydrate 10.0 g
Magnesium chloride l.4 g
Dehydrated ox bile 5.0 g
Dipotassium sulfate 10.0 g
Bromocresol purple 10 mg
Cetrimide 0.3 g
Purified water 1000 mL
Agar 13.6 g
Adjust the pH so that after sterilisation it is 7.3 ± 0.2 at 25 oc. Purified water 1000 mL
Sterilise in an autoclave using a validated cycle.
Glyeerol 10.0 mL

MacConkey agar
Panereatic digest of gelaiín 17.0 g
Heat to boiling for 1 min with shaking. Adjust the pH so
Peptones (meat and casein) 3.0 g that after sterilisation it is 7.2 ± 0.2 at 25 oc. Sterilise in an
autoclave using a validated cycle.
Lactose monohydrate 10.0 g

Sodium chloride 5.0 g


Mannitol salt agar
Bile salts 1.5 g
Pancreatic digest of casein 5.0 g
Agar 13.5g
Peptic digest of animal tissue 5.0 g
Neutral red 30.0 mg
Beef extraet 1.0 g
Crystal violet 1 mg
D-Mannito! 10.0 g
Purified water 1000 mL
Sodium chloride 75.0 g
Adjust the pH so that after sterilisation it is 7.1 ± 0.2 at 25 oc. Agar 15.0 g
Boil for 1 min with constant shaking then sterilise in an
autoclave using a validated cycle. Phenol red 0.025 g

Purified water 1000 mL


Rappaport Vassiliadis Salmonella enrichment broth
Soya peptone 4.5 g Heat to boiling for 1 min with shaking. Adjust the pH so
that after sterilisation it is 7,4 ± 0.2 at 25 oc. Sterilise in an
Magnesium chloride hexahydrate 29.0 g autoclave using a validated cyele.
Sodium chloride 8.0 g

Dipotassium phosphate 0.4 g Rei.nforced medium for dostridia


Potassium dihydrogen phosphate 0.6 g Beef extraet 10.0 g

Malaehite green 0.036 g Peptone 10.0 g

Purified water 1000 mL Yeast extract 3.0 g

Dissolve, warming gently. Sterilise in an autoclave using a Soluble stareh 1.0 g


validated cycle, at a temperature not exceeding 115 oc. The Glucose monohydrate 5.0 g
pH is to be 5.2 ± 0.2 at 25 oC after heating and autoclaving.
Cysteine hydrochloride 0.5 g

Xylose, lysine, deoxycholate agar Sodium ehloride 5.0 g

Xylose 3.5 g Sodium acetate 3.0 g


L-Lysine 5.0 g Agar 0.5 g

Lactose monohydrate 7.5 g Purified water 1000 mL

General Natices (1) apply ta all managraphs and ather texts 193
2.6.14. Bacterial endotoxins EUROPEAN PHARMACOPOEIA 8.0

Hydrate the agar, dissolve by heating to boiling with pipette tips for automatic pipetters, use apparatus shown to
continuous stirring. If necessary, adjust the pH so that after be free of detectable endotoxin and which does not interfere
sterilisation it is 6.8 ± 0.2 at 25 0e. Sterilise in an autoclave in the test.
a validated NOTE: in this chapter, the tcrm 'tubc' includes all of
for micratitre plate wells.

Columbia agar 2. REAGENTS, TEST SOLUTIONS


Pancreatic digest of casein 10.0 g
Amoebocyte lysate
Mea! peptic diges! 5.0 g
Amoebocyte lysate is a lyophilised product obtained from
Heart pancreatic digest 3.0 g amoebocyte Iysate from the horseshoe crab (Limulus
polyphemus or Tachypleus tridentatus). This reagent refers
Yeast extraet 5.0 g only to a product manufactured in accordance with the
Maize starch 1.0 g regulations of the competent authority.
Sodium chloride 5.0 g NOTE: amoebocyte lysate reacts with some f3-glucans in
addition to endotoxins. Amoebocyte Iysate preparations whích
Agar, according to gelling power 10.0-15.0 g do not reaet with glucans are available; they are prepared by
Purified water 1000 mL removing fram amoebocyte lysate the which reacts
with glucans, or by inhibiting the G factor reacting system
of amoebocyte Iysate. These preparations may be used for
Hydrate the agar, dissolve by heating to boiling with
cndotoxin testing in the presence of glucans.
continuous stirring. If adjust the pH so that after
sterilisation it is 7.3 ± 0.2 at 25 Sterilise in an autoclave (2) Lysate solution
Allow to cool to 45-50 oC; add, where Dissolve amoebocyte lysate in water for BET or in a buffer, as
necessary, ¡SCHl<UlliUH sulfate corresponding to 20 mg of recommended by the lysate manufacturer, by gentle stirring.
gentamicin and pour into Petri dishes. Store the reconstituted lysate, refrigerated or frozen, as
indicated the manufacturero
Water for BET (water for bacterial endotoxins test)
Water for injections R or water produced by other procedures
that shows no reaction with the lysate employed at the
detection limit of the reagent.
01/2010:20614
correded 7.0 3. PREPARATION OF THE STANDARD ENDOTOXIN
STOCK SOLUTION
The standard endotoxin stock solution is prepared from
2.6. ENDOTOXINS an endotoxin reference standard that has been calibrated
against the International Standard, for '--"'''''I.m. endotoxin
The test Íor bacterial endotoxins (BET) is used to detect standard BRP.
endotoxins from gram -negative bacteria using Endotoxin is expressed in International Units (IU). The
from the horseshoe crab equivalence in IU of the International Standard is stated by
There are 3 techniques the World Health Organization.
test: the teclmique, which is based 011
gel formation; the turbidimetric technique, based 011 the NOTE: one fnternational Unit (fU) is equal to
development of after cleavage of a11 endogenous one Endotoxin Unit (E.
substrate; and the chromogenic technique, based 011 Follow the specifications in the package leaflet and 011 the
the of colour after of a synthetic label for preparation and storage of the standard endotoxin
VHiV",LH complexo stock solution.
The HHitJl'V n I " 6 methods are described in the present chapter:
4. PREPARATION OF THE STANDARD ENDOTOXIN
Method A. Gel-clot method: limit test SOLUTIONS
Method B. Gel-clot method: quantitative test After vigorously mixing the standard endotoxin stock solution,
prepare appropriate serial dilutions of this solution using
Method e. Turbidimetric kinetic method water for BET.
Method D. Chromogenic kinetic method Use the solutions as 500n as p05sible to avoid loss of activity
Method E. end-point method by adsorption.
Method F. Turbidimetric method 5. PREPARATION OF THE TEST SOLUTIONS
Proceed by of the 6 methods for the test. In the event Prepare the test solutions by dissolving or diluting active
of doubt or the final decision is made based upon substances or medicinal products using water for BET.
method A unless otherwise indicated in the monograph. Some substances or preparations may be more appropriately
dissolved or diluted in other aqueous solutions. If necessary,
The test is carried out in a manner that avoids endotoxin adjust the pH of the test solution dilution thereof) so that
contan1inatiol1. the pH of the mixture of the lysate and test solution falls within
the pH range specified the Iysate manufacturer, usually
1. APPARATUS 6.0 to 8.0. The pH may adjusted by the use of acid, base or
a suitable buffer, as recommended by the lysate manufacturero
and other heat-stable apparatus in Acids and bases may be prepared from concentrates or solids
a validated process. A commonly used with water for BET in containers free of detectable endotoxin.
temperature is 30 minutes at 250 0e. If Buffers must be validated to be free of detectable endotoxin
apparatus, such as microtitre plates and and interfering factors.

194 See the m'nr,,,,nTU' mnnn,CFrnlJi1, (ca ver pages)


EUROPEAN PHARMACOPOEIA 8.0 2.6.14. Bacterial endotoxins

6. DETERMINATION OF THE MAXIMUM VALID Confirmation of the Iysate sensitivity is carried out when a
DILUTION new lot of lysate is used or when there is any change in the test
The Maximum Valid Dilution (MVD) is the maximum conditions which may affect the outcome of the test.
allowable dilution of a sample at which the endotoxin limit Prepare standard solutions of at least 4 concentrations
can be detennined. Determine the MVD using the following equivalent to 211., A, O.SA and 0.2SA by diluting the standard
formulae: endotoxin stock solution with water for BET.

endotoxin linüt x concentration of test solution


Mix a volume of the Iysate solution with an equal volume
MVD= of 1 of the standard solutions (such as 0.1 mL aliquots) in
.\
each tube. When single test vials or ampoules containing
Endotoxín limit: the endotoxin limit for active substances Iyophilised Iysate are employed, add solutions of standard s
administered parenterally, defined on the basis of dose, is directly to the vial or ampoule. Incubate the reaction mixture
equal to: for a constant period according to the recommendations of
the Iysate manufacturer (usually at 37 ± loe for 60 ± 2 min),
K
avoiding vibration. Test the integrity of the gel: for tubes, take
M each tube in turn directly from the incubator and invert it
through approximately 180° in one smooth motion. If a firm
K threshold pyrogenic dose of endotoxin per gel has formed that remains in place upon inversion, record
kilogram of body mass, the result as positive. A result is negative if an intact gel is not
M maximum recommended bolus dose of product formed.
per kilogram of body mass. The test is considered valid when the lowest concentration of
When the product is to be injected at frequent intervals the standard solutions shows a negative result in al! replicate
or infused continuously, M is the maximum total dose tests.
administered in a single hour periodo
The end -point is the lowest concentration in the series
The endotoxin limit for active substances administe1'ed of decreasing concentrations of standard endotoxin that
parenterally is specified in units such as IU /mL, IU /mg, IU /Unit dots the Iysate. Determine the geometric mean end-point
of biological activity, etc., in monographs. concentration by calculting the mean of the logarithms of
Concentration of test solutíon: the end-point concentrations of the 4 dilution series, take
- mg/mL ifthe endotoxin limit is specined by mas s (IU/mg), the antilogarithm of this value, as indicated by the following
- Units/mL if the endotoxin limit is specified by unit of expression:
biological activity (IU IUnit) ,
Geometric mean end-point concentration = antilog ~ e
- m1!mL if the endotoxin limit is specified by volume
(IU/mL).
A the labelled lysate sensitivity in the gel-dot
Le sum of the 10g¡O end-point COl1centrations of the
dilution series used,
technique (IU/mL) or the lowest concentration
used in the standard curve of the turbidimetric or f number of replicates.
chromogenic techniques.
The geometric mean end-point concentration is the measured
7. GEL-CLOT TECHNIQUE (METHODS A AND B) sensitivity of the lysate solutiol1 (IU/mL). If this is 110t les s
than O.SA and not more than 2:\., the labelled sensitivity is
The gel-clot technique allows detection or quantification
confirmed and is used in the tests performed with this Iysate.
of endotoxins and is based on clotting of the Iysate in the
presence of endotoxins. The minimum concentration of (ii) Test for interfering factors
endotoxins required to cause the Iysate to clot under standard Prepare solutions A, B, e and D as shown in Table 2.6.14.-1,
conditions is the labelled Iysate sensitivity. To ensure both and use the test solutions at a dilution less than the MVD, not
the precision and validity of the test, confirm the labelled containing any detectable endotoxins, operating as described
Iysate sensitivity and perform the test fo1' interfering factors as under 1. Preparatory testing, (i) Confirmation of the labelled
described under 1. Preparatory testing. lysate sensitivity.
1. PREPARATORY TESTlNG
The geometric mean end-point concentrations of solutions B
(i) Confirmation of the labelled lysate sensitivity and C are determined using the expression described in
Confirm in 4 replicates the labelled sensitivity A, expressed l. Prepa1'atory testing, (i) Confirmation of the labelled lysate
in IU /mL, of the lysate solution prior to use in the test. sensitivity.
Table 2.6.14.-1
Solution Endotoxin concentration/Solution to Diluent Dilution factor Endotoxin Number of replicates
which endotoxin is added concentration
A None/Test solution 4

B 2A/Test solution Test solution I 2A 4


2 lA 4
4 0.5A 4
8 0.25A 4
e 2A/Water for BET Water for BET 2A 2
2 lA 2
4 0.5A 2
8 0.25A 2
D None/Water for BET 2

Solution A = solution of the preparation being examined that is rree of detectable endotoxins.
Solution B = test for interference.
Solution e = control of the labelled Iysate sensitivity.
Solution D = negative control (water for BET).

General Notíces (1) apply to all monographs and other texts 19S
2.6.14. Bacterial endotoxins EUROPEAN PHARMACOPOEIA 8.0

The test for interfering factors must be repeated when any (ii) Interpretation
changes are made to the experimental conditions that are The test is considered valid when both replicates of solution B
likely to influence the result of the test. and C are positive and those of solution D are negative.
The test is considered valid when all replicates of solutions A When a negative result is found for both replicates of
and D show no reaction and the result of solution C confirms solution A, the preparation being examined complies with
the labelled lysate sensitivity. the test.
If the sensitivity of the lysate determined with solution B is When a positive result is found for both replicates of
not less than 0.5A and not greater than 2A, the test solution solution A, the preparation being examined does not comply
do es not contain interfering factors under the experimental with the test.
conditions used. Otherwise, the test solution interferes with When a positive result is found for one replicate of solution A
the test. and a negative result is found for the other, repeat the test.
If the preparation being examined interferes with the test at In the repeat test, the preparation being examined complies
a dilution less than the MVD, repeat the test for interfering with the test if a negative result is found for both replicates of
factor s using a greater dilution, not exceeding the MVD. The solution A. The preparation does not comply with the test if a
use of a more sensitive lysate permits a greater dilution of the positive result is found for one or both replicates of solution A.
preparation being examined and this may contribute to the However, if the preparation does not comply with the test at a
elimination of interference. dilution less than the MVD, the test may be repeated using a
greater dilution, not exceeding the MVD.
Interference may be overcome by suitable validated treatment,
such as filtration, neutralisation, dialysis or heat treatment. 3. QUANTITATIVE TEST (METHOD B)
To establish that the treatment chosen effectively eliminates (i) Procedure
interference without loss of endotoxins, repeat the test for The test qllantifies bacterial endotoxins in the test solution by
interfering factors using the preparation being examined to titration to an end-point. Prepare solutions A, B, C and D as
which the standard endotoxin has been added and which has shown in Table 2.6.14.-3, and test these solutions according
then be en submitted to the chosen treatment. to the procedure described under 1. Preparatory testing, (i)
2. LIMIT TEST (METHOD A) Confirmation of the labelled lysate sensitivity.
(i) Procedllre (ii) Calculation and interpretation
Prepare solutions A, B, C and Das shown in Table 2.6.14.-2, The test is considered valid when the following 3 conditions
and perform the test on these solutions following the procedure are met:
described under 1. Preparatory testing, (i) Confirmation of (a) both replicates of solution D (negative control) are negative,
the labelled lysate sensitivity. (b) both replicates of solution B (positive product control)
are positive,
Table 2.6.14.-2
(c) the geometric mean end-point concentration of solution C
Solution EndotoXÍn conceutration/Solution Number of replicates is in the range of O.5A to 2A.
to which endotoXÍn is added
A 2
To determine the endotoxin concentration of solution A,
None/Diluted test solution
calculate the end-point concentration for each replicate, by
B 2A/Diluted test solution 2 multiplying each end-point dilution factor by A.
e 2A/Water for BET 2 The endotoxin concentration in the test solution is the
end-point concentration of the replicates. lf the test
D None/Water for BET 2 is conducted with a diluted test solution, calculate the
concentration of endotoxin in the original solution by
multiplying the result by the dilution factor.
Prepare solution A and soiution B (positive product control) lf none of the dilutions of the test solution is positive in a
using a dilution not greater than the MVD and treatments as valid test, report the endotoxin concentration as less than
described in 1. Preparatory testing, (ii) Test for interfering A (or, if a diluted sample was tested, report as less than the
factors. Solutions B and C (positive control5) contain the lowest dilution factor of the sample x A). lf all dilutions are
standard endotoxin at a concentration corresponding to twice positive, the endotoxin concentration is reported as equal to
the labelled lysate sensitivity. Solution D (negative control) or greater than the largest dilution factor multiplied by A (e.g.
consists of water for BET. in Table 2.6.14.-3, the initial dilution factor x 8 x A).
Table 2.6.14.-3
Solution Endotoxin concentration/Solution to which Diluent Dilution factor Endotoxin Number ofreplicates
endotoxin is added concentration
A None/Test solution Water for BET 2
2 2
4 2
8 2
B 2A/Test solution 2

e 2A/Water for BET Water for BET lA 2


2 lA 2
4 O.SA 2
8 O.2SA 2
D None/Water for BET 2

Solution A = test solution at the dillltion, not exceeding the MVD, with which the test for interfering factors was carried out. Subsequent dilution of
the test sollltion must not exceed the MVD. Use water for BET to make a dilution series of 4 tubes containing the test sollltion at concentrations of 1,
1/2, 1/4 and 1/8, relative to the dilution used in the test for interfering factors. Other dilutions up to the MVD may be used as appropriate.
Solution B = solution A containing standard endotoxin at a concentration of 2A (positive product control).
Solution e = a dilulion series of 4 tubes of water for BET containing the standard endotoxin at concentrations of 2A, A, O.SA and O.2SA.
Solution D = water for BET (negative control).

196 See the information section on general monographs (caver pages)


EUROPEAN PHARMACOPOEIA 8.0 2.6.14. Bacterial endotoxins

The preparation being examined meets the requirements of Table 2.6.14.-4.


the test if the endotoxin concentration in both replicates is
Solution Endotoxin Solutioll to which Number of
less than that specified in the monograph. collcentI"atioll elldotoxin is added rel'licates
A None Test solution Not less than 2
8. PHOTOMETRIC QUANTITATIVE TECHNIQUES
(METHODS C, D, E AND F) B Middle concentration Test solution Not les s than 2
of the standard curve
l. TURBIDIMETRIC TECHNIQUE (METHODS C AND F)
e At least 3 concentra- Water for BET Each concentration
This technique is a photometric test to measure the increase in tions (lowest coneen- not less than 2
turbidity. Based 011 the test principIe employed, this technique tration is designated A)
may be classified as being either the end-point-turbidimetric D None Water for BET Not les s than 2
test or the kinetic-turbidimetric test.
Solution A = test solution, that may be diluted not to exceed the MVD.
The end-point-turbidimetric test (Method F) is based on the Solution B = preparation to be examined at the same dilution as
quantitative relationship between the endotoxin concentration solution A, containing added endotoxin at a concentration equal to or
and the turbidity (absorbance or transmission) of the reaction near the middle of the standard curve.
mixture at the end of an incubation periodo Solution e = standard endotoxin solution at the concentrations used in
the validation of the method as described under 3. Preparatory testing,
The kinetic-turbidimetric test (Method C) is a method to (i) Assurance of criteria for the standard curve (positive controls).
measure either the time (onset time) needed for the reaction Solution D = water for BET (negative control).
mixture to reach a predetermined absorbance or transmission,
or the rate of turbidity development. The test is considered valid when the following conditions
are met:
The test is carried out at the incubation temperature
recommended by the lysate manufacturer (usually 37 ± 1°C). - the absolute value of the correlation coefficient of the
2. CHROMOGENIC TECHNIQUE (METHODS D AND E)
standard curve generated using solution C is greater than
or equal to 0.980;
This technique is used to measure the chromophore released
from a suitable chromogenic peptide by the reaction of - the result with solution D does not exceed the limit of the
endotoxins with the lysate. Depending on the test principIe blank value required in the description of the lysate reagent
employed, this technique may be classified as being either the employed, or it is less than the endotoxin detection limit of
end-point-chromogenic test or the kinetic-chromogenic test. the lysate reagent employed.
The end-point-chromogenic test (Method E) is based on the Calculate the mean recovery of the added endotoxin by
quantitative relationship between the endotoxin concentration subtracting the mean endotoxin concentration in the solution
and the quantity of chromophore released at the end of an (if any) (solution A, Table 2.6.14.-4) from that in the solution
incubation periodo containing the added endotoxin (solution B, Table 2.6.14.-4).
The kinetic-chromogenic test (Method D) measures either the The test solution is considered free of interfering factors if
time (onset time) needed for the reaction mixture to reach a under the conditions of the test, the measured concentration
predetermined absorban ce, or the rate of colour development. of the endotoxin added to the test solution is within 50-200 per
The test is carried out at the incubation temperature cent of the known added endotoxin concentration, after
recommended by the lysate manufacturer (usually 37 ± 1 OC). subtraction of any endotoxin detected in the solution without
added endotoxin.
3. PREPARATORY TESTING
To assure the precision or validity of the turbidimetric and When the endotoxin recovery is out of the specified range,
chromogenic techniques, preparatory tests are conducted to the test solution is considered to contain interfering factors.
show that the criteria for the standard curve are satisfied and Repeat the test using a greater dilution, not exceeding
that the test solution do es not interfere with the test. the MVD. Furthermore, interference of the test solution
or diluted test solution 110t to exceed the MVD may be
Validation of the test method is required when any changes eliminated by suitable validated treatment, such as filtration,
are made to the experimental conditions that are likely to neutralisation, dialysis or heat treatment. To establish that the
influence the result of the test. treatment chosen effectively eliminates interference without
(i) Assurance of criteria for the standard curve 10ss of endotoxins, repeat the test for interfering factors
The test must be carried out for each lot of lysate reagent. using the preparation being examined to which the standard
endotoxin has been added and which has then been submitted
Using the standard endotoxin solution, prepare at least to the chosen treatment.
3 endotoxin concentrations within the range indicated by the
lysate manufacturer to generate the standard curve. Perform 4. TEST
the test using at least 3 replicates of each standard endotoxin (i) Procedure
solution as recommended by the lysate manufacturer (volume
Follow the procedure described in 3. Preparatory testing,
ratios, incubation time, temperature, pH, etc.).
(ii) Test for interfering factors.
If the desired range is greater than 2 10g¡O in the kinetic
methods, additional standards must be included to bracket (ii) Calculation
each 10g¡O in crease in the range of the standard curve. Calculate the endotoxin concentration of each replicate of
The absolute value of the correlation coefficient, I r ¡, must solution A using the standard curve generated by the positive
be greater than or equal to 0.980, for the range of endotoxin control solution C.
concentrations set up. The test is considered valid when the following 3 requirements
(ii) Test for interfering factors are met:
Select an endotoxin concentration at or near the middle of (1) the results obtained with solution C comply with the
the endotoxin standard curve. requirements for validation defined under 3. Preparatory
testing, O) Assurance of criteria for the standard curve,
Prepare solutions A, B, e and Das shown in Table 2.6.14.-4.
Perform the test on at least 2 replicates of these solutions as (2) the endotoxin recovery, calculated from the endotoxin
recommended by the lysate manufacturer (volume of test concentration found in solution B after subtracting the
solution and lysate solution, v01ume ratio of test solution to endotoxin concentration found in solution A, is within the
lysate solution, incubation time, etc.). range of 50-200 per cent,

General Natices (1) apply ta all managraphs and ather texts 197
2.6.15. PrekaHikrein activator EUROPEAN PHARMACOPOEIA 8.0

(3) the result obtained with solution D (negative Carry out al! procedures from the beginning of the
does not exceed the limit of the blank value required in chromatography to freezing in portions during a single
the description of the lysate employed, or it is less than the day.
endotoxin detection limit of the lysate reagent employed.
METHOD
(iii) InterpretaHon
The assay may be carried out using an automated enzyme
The preparation being examined complies with the test if the
analyser or a suitable microtitre plate system allowing kinetic
mean endotoxin concentration of the replicates of solution A,
measurements, with appropriate software for calculation of
after correction for dilution and concentration, is less than the
results. Standards, samples and prekallikrein substrate may be
endotoxin limit for the product.
diluted as necessary using buffer B.
Guidelines on the test for bacterial endotoxins are given in Incubate diluted standards or samples with prekallikrein
general chapter 5.1.10. substrate for 10 mil1 such that the volume of the undiluted
sample do es 110t exceed l/lO of the total volume of the
incubation mixture to avoid errors caused by variation
01/2008:20615 in ionic strength and pH in the incubation mixture.
Incubate the mixture 01' a part thereof with at least an equal
2.6.15. PREKALLIKREIN ACTIVATOR volume of a solution of a suitable synthetic chromogenic
substrate, known to be specific for kallikrein (for example,
Prekallikrein activator (PKA) activates prekallikrein to N-benzoyl-L-prolyl-L-phenylalanyl-L-arginine 4-nitroanilide
kallikrein and may be assayed by its ability to cleave a acetate R or D-prolyl-L-phenylalanyl-L-arginine 4-nitroanilide
chromophore from a synthetic peptide substrate so that the dihydrochloride R), dissolved in buffer B. Record the rate
rate of cleavage can be measured spectrophotometrically and of change in absorbance per minute for 2-10 min at the
the concentration of PKA calculated by comparison with a wavelength specific fo1' the substrate used. Prepare a blank for
reference preparation calibrated in International Units. each mixture of sample or standard using buffer B instead of
The International Unit is the activity of a stated amount of prekallikrein substrate.
the International Standard which consists of freeze-dried Depending on the method used, L'.Almin has to be corrected
prekallikrein activator. The equivalence in International Units by subtracting the value obtained fOl" the corresponding
of the International Standard is stated by the World Health blank without the prekallikrein substrate. The results may be
Organization. calculated using a standard curve, a parallel-line or a slope
REAGENTS ratio assay or any other suitable statistical method. Plot
a calíbration curve using the values thus obtained for the
Prekallikrein activator in albumin BRP is calibrated in
reference preparation and the respective concentrations; use
International Units by comparison with the International
the curve to determine the PKA activity of the preparation to
Standard.
be examined.
Buffer A. Dissolve 6.055 g of tris(hydroxymethyl)aminome-
thane R, 1.17 g of sodium chloride R, 50 mg of hexadimethrine
bromide R and 0.100 g of sodium azide R in water R. Adjust
to pH 8.0 with 2 M hydrochloric acid R and dHute to 1000 mL 01/2011:20616
with water R.
Buffer B. Dissolve 6.055 g of tris(hydroxymethyl)aminome- 2.6.1 TESTS FOR EXTRANEOUS
thane R and 8.77 g of sodium chloride R in water R. Adjust to
pH 8.0 with 2 M hydrochloric acid R and dilute to 1000 mL AGENTS IN VIRAL VACCINES POR
with water R. HUlVIAN USE
PREPARATION OF PREKALLIKREIN SUBSTRATE In those tests that require prior neutralisation of the virus,
To avoid coagulation activation, blood or plasma used for the use specific antibodies ofnon-human, non-simian origin; if
preparation of prekallikrein must come into contact only with the virus has been propagated in avian tissues, the antibodies
plastics or silicone-treated glass surfaces. must also be of non-avian origino To prepare antiserum, use
Draw 9 volumes of human blood into 1 volume of an immunising antigen produced in cel! culture from a species
anticoagulant solution (ACD, CPD or 38 giL solution of different from that used fol' the production of the vaccine
sodium citrate R) to which 1 mg/mL of hexadimethrine and free from extraneous agents. Where tile use of SPF eggs
bromide R has been added. Centrifuge the mixture at 3600 g is prescribed, the eggs are obtained from a flock free from
for 5 mino Separate the plasma and centrifuge again at 6000 g specified pathogens (5.2.2).
for 20 min to sediment platelets. Separate the platelet-poor
VIRUS SEED LOT
plasma and dialyse against 10 volumes ofbuffer A for 20 h.
Apply the dialysed plasma to a chromatography column Take samples of the virus seed 10t at the time ofharvesting and,
containing agarose-DEAE for ion-exchange chromatography R if they are 110t tested immediately, keep them at a temperature
which has been equilibrated in buffer A and is equal to below - 40 oc.
twice the volume of the plasma. Elute from the column with Adult mice. Inoculate each of not fewer than 10 adult mice,
buffer A at 20 mL/cm 2/h. Collect the eluate in fractions and each weighing 15-20 g, intracerebrally with 0.03 mL and
record the absorbance at 280 nm (2.2.25). Pool the fractions intraperitoneally with 0.5 mL of the virus seed lot. Observe
containing the first protein peak so that the volume of the the mice for at least 21 Carry out an autopsy of al! mice
pool is about 1.2 times the volume of the platelet-poor plasma. that die aÍter the first 24 of the test OY that show signs of
Test the substrate pool for absence of kallikrein activity by illness, and examine for evidence of viral infection, both by
mixing 1 part with 20 parts of the pre-warmed chromogenic direct macroscopical observation and by subinoculation
substrate solution to be used in the assay and incubate at of appropriate tissue suspensions by the intracerebral and
37°C for 2 mino The substrate is suitable if the increase in intraperitoneal routes into not fewer than 5 additional
absorbance is less than 0.001 per minute. Add to the pooled mice, which are observed for 21 days. The virus seed 10t
solution 7 giL of sodium chloride R and filter through a complíes with the test if no mouse shows evidence of infection
membrane filter (nominal pore size 0.45 ¡'Ul1). Freeze the attributable to the seed lot. The test is not valid unless at
filtrate in portions and store at - 25 oC; the substrate may be least 80 per cent of the original inoculated mice survive the
freeze-dried before storage. observation periodo

198 See the (cover pages)


EUROPEAN PHARMACOPOEIA 8.0 2.6.16. Tests f(jr extraneous agents in viral vacdnes

Sudding mke. Inoculate each of not fewer than 20 mice, Insect viruses (onIy requl.red for virus propagated in insect
each less than 24 h old, intracerebrally with 0.01 mL and cells). Neutralised samples equivalent, unless otherwise
intraperitoneally with at 1east 0.1 mL of the virus seed loto prescribed, to 500 human doses of vaccine or 50 mL, whichever
Observe the mice daily for at least 14 days. Carry out an is the greater, are tested for the presence of extraneous agents
autopsy of all mice that die after the first 24 h of the test or by inoculation into at least one ceH culture different [rom
that show signs of illness, and examine for evidence of viral that used in production and permissible to insect viruses,
infection, both by direct macroscopical observation and and that allows detection of human arboviruses. The choice
by subinoculation of appropriate tissue suspensions by the of ceUs is approved by the competent authority and takes
intracerebral and intraperitoneal routes into not fewer than into account the origin of the production ceUs and the likely
5 additional suckling mice, which are observed daily for contaminants that may be detected by the chosen ceUs. The
14 days. The virus seed lot passes the test if no mouse shows cells are incubated at 27 ± 1 oC and observed for a period
evidence of infection attributable to the seed lot. The test is of 14 days. The virus seed lot or harvest passes the tests if
not valid unless at least 80 per cent of the original inoculated none of the ceH cultures show evidence of the presence of any
mice survive the observation periodo extraneous agents. The test is not valid unless at least 80 per
cent of the ceH cultures remain viable.
Guinea-pigs. Inoculate intraperitoneally into ea eh of not
fewer than 5 guinea pigs, each weighing 350-450 g, 5.0 mL of
PRODUCTION CELL CULTURE: CONTROL CELLS
the virus seed loto Observe the animals for at least 42 days
for signs of disease. Carry out an autopsy of all guinea-pigs Examine the control cells microscopically for freedom from
that die after the first 24 h of the test or that show signs of any virus causing cytopathic degeneration throughout the
illness, and examine macroscopically; examine the tissues time of incubation of the inoculated production ceH cultures
both microscopically and cultural1y for evidence of infection. or for not less than 14 days beyond the time of inoculation of
Euthanise animals that survive the observation period and the production vessels, whichever is the longer. The test is
examine in a similar manner. The virus seed lot pass es the test not valid unless at least 80 per cent of the control ceH cultures
if no guinea-pig shows evidence of infection attributable to survive to the end of the observation periodo
the seed 10t. The test is not valid unless at least 80 per cent of At 14 days or at the time of the last virus harvest, whichever is
the guinea-pigs survive the observation periodo the longer, carry out the tests described below.
Spiroplasmas. Virus seed 10ts produced in insect ceUs Haemadsorbing vl.ruses. Examine not fewer than 25 per cent
are demonstrated by a validated method approved by the of the control cultures for the presence of haemadsorbing
competent authority to be free of spiroplasmas. virus es by the addition of guinea-pig red blood ceUs. If the test
for haemadsorbing viruses is not feasible, carry out a test for
haemagglutination viruses. If the guinea-pig red blood cells
VIRUS SEED LOT AND VIRUS HARVESTS have been sto red, they shall have been stored at 5 ± 3 oC for not
more than 7 days. Read half of the cultures after incubation
Take samples at the time of harvesting and, if not tested at 5 ± 3 oC for 30 min and the other half after incubation at
immediately, keep them at a temperature below - 40 oc. 20-25 oC Íor 30 mino No evidence of haemadsorbing agents is
Bacterial and fungal sterility. A 10 mL sample complies with found.
the test for sterility (2.6.1). Tests in ceH cultures for other extraneous agents. Pool the
Mycoplasmas. A 10 mL sample complies with the test for supernatant fluids from the control cells and examine for the
mycoplasmas (2.6.7). presence of extraneous agents by inoculation of simian kidney
and human ceH cultures. If the virus is grown in a mammalian
Mycobacteria (2.6.2). AS mL samp1e is tested for the ceH system other than simian or human, cells of that species,
presence of Mycobacterium spp. by culture methods known to but from a separate batch, are also inoculated. In each cen
be sensitive for the detection of these organisms. system, at least 5 mL is tested. Incubate the inoculated cultures
Test in ceH culture fo1" other extraneous agents. Neutralised at 36 ± 1 oC and observe for a period of 14 days. No evidence
samples equivalent, un1ess otherwise prescribed, to 500 human of extraneous agents is found.
doses of vaccine or 50 mL, whichever is the greater, are tested Jf the production cell culture is maintained at a temperature
for the presence of extraneous agents by inoculation into different from 36 ± 1°C, a supplementary test for extraneous
continuous simian kidney and human ceH cultures. If the agents is carried out at the production temperature using the
virus is grown in simian or human cells, the neutralised virus same type of ceUs as used for growth of the virus.
harvest is tested on a separate culture of these cells. If the
virus is grown in a mammalian or avian ceu system other than If the virus is grown in insect cells the pooled supernatant is
simian or human, cells of that species, but from a separate also inoculated into at least one ceH culture different from
batch, are also inoculated. The cells are incubated at 36 ± 1 oC that used in production and permissible to insect viruses,
and observed for a period of 14 days. The virus seed lot and that allows detection of human arboviruses. The cells are
or harvest passes the tests if none of the ceH cultures show incubated at 27 ± 1 oC for 14 days. No evidence of extraneous
evidence of the presence of any extraneous agents. The test is agents is found.
not valid unless at least 80 per cent of the ceH cultures remain Avian leucosis viruses (required only if the virus is
viable. propagated in primary avian tissues). Carry out a test for
Avían viruses (only required for virus seed lot propagated avían leucosis virus es using 5 mL of the supernatant fluid
in avían tissues and for virus harvest propagated in from the control ceUs.
primary avían tissues). Neutralise a sample equivalent to
100 human doses ofvaccine or 10 mL, whichever is the CONTROL EGGS
greater. Using 0.5 mL per egg, inoculate a group of fertilised Haemagglutinating agents. Examine 0.25 mL of the allantoic
SPF eggs, 9-11 days old, by the allantoic route and a second fluid from each egg for haemagglutinating agents by mixing
group, 5-7 days old, into the yolk saCo Incubate for 7 days. dírectly with chicken red blood cells and after a passage in
The virus seed lot or harvest complies with the test if the SPF eggs carried out as follows: inoculate a 5 mL sample
allantoic and yolk sac fluids show no sign of the presence of the pooled amniotic fluíds fram the control eggs in
of any haemagglutinating agent and if all embryos and 0.5 mL volumes into the allantoic cavity and into the amniotic
chorio-allantoic membranes, examined for gross pathology, cavity of SPF eggs. The control eggs comply with the test if no
are normal. The test is not valid unless at least 80 per cent of evidence of the presence of haemagglutinating agents is found
the inoculated eggs survive for 7 days. in either test.

General Notices (1) apply to all monographs and other texts 199
2.6.17. Test fmr anticomplementary acHvHy of immunoglobulin EUROPEAN PHARMACOPOEIA 8.0

Avian leucosis viruses. Use a 10 mL sample of the pooled METHOD


amniotic fluids from the control eggs. Carry out amplification Preparation oí standardised 5 per cent red blood (eH
by 5 passages inleucosis-free chick-embryo cel! cultures; carry suspension. Separate sheep red blood cells by centrifuging an
out a test for avían leucosis using cells from the 5 passage.
th
appropriate volume of stabilised sheep bIood and wash the
The control comply with the test if no evidence of the cells at least 3 times vvith gelatin barbitai buffer solution and
presence of leucosis virus es is found. prepare a 5 per cent V/V suspension in the same solution.
Other extraneous agents. Inoculate 5 mL samples of the Measure the cel! density of the suspension as follows: add
amniotic fluid s from the control eggs into human and 0.2 mL to 2.8 111L of water R and centrifuge the lysed solution
cel! cultures. Observe the cell cultures for 14 days. for 5 min at 1000 g; the ceH density is suitable if the absorbance
The control eggs with the test if no evidence of the (2.2.25) of the supernatant at 541 nm is 0.62 ± 0.01. Correet
presence of extraneous agents is found. The test is not valid the cel! density by adding gelatin barbital buffer solution
unless 80 per cent of the inoculated cultures survive to the according to the following equation:
end of the observation periodo
v,_VixA
f - 0.62

01/2010:20617 final adjusted volume;


corrected 7.6 the initial volume;
A absorbance of the original suspension at 541 nm.
2,6,17. TEST FOR
ANTICOMPLEMENTARY ACTIVITY The adjusted suspension contains about 1 x 10 9 cells/mL.
OF IMMUNOGLOBULIN Haemolysin titraüon. Prepare haemolysin diIutions as shown
in TabIe 2.6.17.-1.
For the measurement of anticomplementary activity (ACA)
of immunoglobulin, a defined amount of test material (10 mg Table 2.6.17.-1
of immunoglobulin) is incubated with a defined amount
of guinea-pig complement (20 CH so ) and the remaining Required Prepared usil1lg
dilution of
complement is titrated; the anticomplementary activity is nacmo/yeín
eXjJresst:a as the percentage cOl1sumption of complement
to the complement control considered as 100 per cent. Gel"tin barbital
Haemolysin
buffer solution
The unit of complement activity (CH so ) is the
amount of complement that, in the given reaction eonditions, Volume Dilution Volume
will the of 2.5 x 10 8 out of a total of 5 x lOs (mi) (l/ ... ) (mi)
0'-"0H'0'"'''' red bIood cells.
7.5 0.65 undiluted 0.1
and calcium stock so/ution. Dissolve 1.103 g of
10 0.90 undiluted 0.1
eh/oride R and 5.083 g of magnesium chloride R in
water R and dilute to 25 mL with the same solvent. 75 1.80 7.5 0.2
stock solutiono Dissolve 207.5 g of sodium 100 1.80 10 0.2
25.48 g of barbital sodium R. in 4000 mL of
to pH 7.3 using 1 M hydrochloric acid. Add 150 LOO 75 1.0
and caicium stock solution and dilute 200 1.00 100 1.0
to 5000 mL water R.. Filter through a membrane filter
Hvuu.,m, pore size 0.22 I-1m). Store at 4 oC in glass containers. 300 1.00 150 1.0

Celatín solution. Dissolve 12.5 g of gelatín R in about 800 mL 400 1.00 200 1.0
of water R and heat to boiling in a water-bath. Cool to 20 oC
600 1.00 300 1.0
and dilute to 10 L with water R.. Filter through a membrane
filter pore size 0.22 I-1m). Store at 4 oc. Use clear 800 1.00 400 1.0
solutions on!y.
1200 1.00 600 1.0
Citrate solutiol1o Dissolve 8.0 g of sodium citrate R, 4.2 g of
sodium chloride R and 20.5 g of g/ucose R in 750 mL of water R. 1600 1.00 800 1.0
to 6.1 using a 100 gil solution of citric acid R and 2400 1.00 1200 1.0
to mL with water R..
3200* 1.00 1600 1.0
Ce/atin barbital buffer solutiol1. Add 4 volumes of gelatin
solution to 1 volul11e ofbarbital buffer stock solution and mix. 4800* 1.00 2400 1.0
7.3, if necessary, using 1 M sodium hydroxide
Oc discard l.O mi of the mixture.
acid. Maintain at 4 oc. Prepare fresh
Add 1.0 mL of 5 per cent sheep red bIood cell suspension to
Stabilised sheep blood. Collect 1 volume of sheep blood into each tube of the haemolysin dilution series, starting at the
1 volume of citrate solution and mix. Stofe at 4 oC for 110t 1/75 dilution, and mix. Incubate at 37 oC for 30 mino
less than 7 and not more than 28 days. (Stabilised sheep
blood and red blood cells are available from a nUl11ber Transfer 0.2 mL of each of these incubated mixtures to new
tubes and add 1.10 mL oE gelatin barbital buffer solution and
0.2 mL of diluted guinea-pig complement (for example, 1/150).
Antiserum against sheep red blood cells prepared Perform this in duplicate.
antisera are available from a number of
As the unhaemolysed ceH control, prepare 3 tubes with 1.4 mL
of gelatin barbital buffer solution and 0.1 mL of 5 per cent
Prepare a pool oí serum from the
sheep red blood ceH suspension.
blood not Íewer than 10 guinea-pigs. Separate the serum
from the clotted blood centrifugation at about 4 oc. Stofe As the fully haemolysed control, prepare 3 tubes with 1.4 mL
the serum in sl11all amounts below - 70 oc. of water R and 0.1 mL of 5 per cent sheep red cell suspension.

200 See the sectiol1 on general (caver pagcs)


EUROPEAN PHARMACOPOEIA 8.0 2.6.17. Test for anticomplementary activity ofirnmunoglobulin

Incubate al! tubes at 37 oC for 60 min and centrifuge at 1000 g Add 0.2 mL of sensitised sheep red blood cells to each tube,
for 5 mino Measure the absorbance (2.2.25) of the supernatants mix well and incubate at 37 oC for 60 mino Cool the tubes in
at 541 11m a11d calculate the percentage degree of haemolysis an ice-bath and centrifuge at 1000 g for 5 mino Measure the
ineach tube using the following expression: absorbance of the supernatant at 541 nm and calculate the
degree of haemolysis (Y) using the following expression:
Aa - Al X 100
Ab - Al Ac - Al
Ab - Al
absorbance of tubes with haemolysin dilution;
mean absorbance of the 3 tubes with full absorbance of tubes 1 to 12;
haemolysis;
mean absorbance of tubes with 100 per cent
mean absorbance of the 3 tubes with no haemolysis. haemolysis;
Plot the percentage degree of haemolysis as the ordinate Al mean absorbance of cel! controls with O per cent
against the corresponding reciprocal value of the haemolysin haemolysis.
dilution as the abscissa on linear graph papero Determine
the optimal dilution of the haemolysin from the graph by Plot Y/(l- Y) as the abscissa against the amount of diluted
inspection. Select a dilution such that further increase in the complement in millilitres as the ordinate 011 log-log graph
amount of haemolysin do es not cause appreciable change in papero Fit the best line to the points and determine the
the degree of haemolysis. This dilution is deflned as 1 minimal ordinate for the 50 per cent haemolytic complement dose
haemolytic unit (l MHU) in 1.0 mL. The optimal haemolytic where Y/(l- Y) = 1.0. Calculate the activity in haemolytic units
haemolysin dilution for preparatiol1 of sensitised sheep red (CHso/mL) using the following expression:
blood cells contains 2 MHU/mL.
Cd
The haemolysin titration is not valid unless the maximum
degree of haemolysis is 50 per cent to 70 per cent. rf the Ca X 5

maximum degree of haemolysis is not in this range, repeat the


titration with more or less diluted complement solution. Cd reciprocal value of the complement dilution;
Preparation of optimised sensitised sheep red blood ceUs Cn volume of diluted complement resulting in 50 per
(haemolytic system). Prepare an appropriate volume of cent haemolysis, in millilitres;
diluted haemolysin containing 2 MHU/mL and an equal
5 scaling factor to take account of the number of red
volume of standardised 5 per cent sheep red blood ceH
blood cells.
suspension. Add the haemolysin dilution to the standardised
cel! suspension and mix. Incubate at 37 oC for 15 min, store at
The test is not valid unless the plot is a straight line between
2 oC to 8 oC and use within 6 h.
15 per cent and 85 per cent haemolysis and the slope is 0.15 to
Titration of complemento Prepare an appropriate dilution of 0.40, and preferably 0.18 to 0.30.
complement (Íor example 1/250) with gelatin barbital buffer
solution and perform the titration in duplicate as shown in Test for anticomplementary activity. Prepare a complement
TabIe 2.6.17.-2. dilution having 100 CH 50 /mL by diluting titrated guinea-pig
complement with gelatin barbital buffer solution. Depending
Table 2.6.17.-2 on the immunoglobulin to be examined and based on
Tube number Volnme oí dilnted complement Volume of gelatin barbital validation data, a pH adjustment to 7 may be necessary.
(fol" example 1/250) buffer solution Prepare incubatio11 mixtures as follows for an immunoglobulin
(mL) (mL) containing 50 mg/mL:
0.1 1.2

2 0.2 1.1 Table 2.6.17.-3


0.3 1.0 Immunoglobulin Complement control
to be examined (in duplicate)
4 0.4 0.9
lmmunoglobulin (50 mg/mL) 0.2 mL
0.5 0.8
Gelatin barbital buffer 0.6 mL 0.8 mL
6 0.6 0.7
Complement 0.2 mL 0.2 mL
7 0.7 0.6

8 0.8 0.5 Carry out the test on the immunoglobulin to be examined


and prepare ACA negative and positive contro1s using human
9 0.9 0.4
immunoglobulin (ACA and molecular size) BRP, as indicated
10 1.0 0.3 in the leaflet accompanying the reference preparation. Higher
or lower volumes of sample and of gelatin barbital buffer
11 1.1 0.2
solution are added if the immunoglobulin concentration varies
12 1.2 0.1 from 50 mg/mL; for example, 0.47 mL of gelatin barbital
buffer solution i5 added to 0.33 mL of immunoglobulin
3 tubes as 1.3
containing 30 mg/mL to give 0.8 mL. Close the tubes and
cel! control
al O per cent incubate at 37 oC for 60 mino Add 0.2 mL of each incubation
haemolysis mixture to 9.8 mL of gelatin barbital buffer solution to dilute
3 tubes at 1.3 mL of water the complemento Perform complement titrations on each tube
lOO per cent as described aboye to determine the remaining complement
haemolysis activity (Table 2.6.17.-2). Calculate the anticomplementary

General Notices (1) apply to al! monographs and other texts 201
2.6.18. Test for neurovirulence afHve virus vaccines EUROPEAN PHARMACOPOElA 8.0

activity of the preparation to be examined relative to the with the vaccine to be examined and the referel1ce preparation.
complement control considered as 100 per cent, using the The animals are allocated randomly to treatment groups
following express ion : and cages and their identity is coded so that the treatment
received by each animal is concealed from the observers
a - b x 100 and the evaluators of the sections. The l1umber of monkeys
a il10culated is such that in the evaluation of both the vaccine
and the reference preparation not fewer than eleven positive
a mean complement activity (CH 50 /mL) of monkeys are included for type 1 and type 2 virus and not
complement control; fewer than eighteen positive monkeys for type 3 virus (positive
b complement activity (CHso/mL) of tested sample. monkeys are those that show specific neuronallesions of
poliovirus in the centralnervous system). More than one
The test is not valid unless:
batch of vaccine may be tested with the same homotypic
- the anticomplementary activities found for ACA negative reference. Monkeys from the same quarantine group are used
control and ACA positive control are within the limits stated wherever possible, otherwise monkeys from two groups are
in the leaflet accompanying the reference preparation; used and equalnumbers from each group are treated with the
- the mean complement activity of complement control (a) is vaccine and the reference preparation. lf the test is carried out
in the range 80 CH 50 /mL to 120 CHso/mL. on two working days, an equal number of monkeys from each
group are inoculated on each day with the vaccine and the
0112008:20618 homotypic reference preparation.
Virus content. The virus contents of the vaccine and
2.6.18. TEST FOR NEUROVIRULENCE the homotypic reference preparation are adjusted so as
OF LIVE VIRUS VACCINES to be as near as possible equal and between 1055 and
1065 CCID so /0.1 mL.
Por each test, use not fewer than ten monkeys that are Observation. AH monkeys are observed for 17 to 22 days
seronegative for the virus to be tested. Por each monkey, for signs of poliomyelitis or other virus infection. Monkeys
inject not more than 0.5 mL of the material to be examined that survive the first 24 h but die before the 11 th day after
into the thalamic region of each hemisphere, unless otherwise inoculation are autopsied to determine whether poliomyelitis
prescribed. The total amount of virus inoculated in each was the cause of death. Animals that die from causes
monkey must be not less than the amount contained in the other than poliomyelitis are excluded from the evaluation.
recommended single human dose of the vaccine. As a check Animals that become moribund or are severely paralysed are
against the introduction of wild neurovirulent virus, keep a euthanised and autopsied. Al! animals that survive until the
group of not fewer than four control monkeys as cage-mates or end of the observation period are autopsied. The test is not
in the immediate vicinity of the inoculated monkeys. Observe valid if more than 20 per cent of the animals show intercurrent
the inoculated monkeys for 17 to 21 days for symptoms of infection during the observation periodo
paralysis and other evidence of neurological involvement;
observe the control monkeys for the same period plus 10 days. Number of sections examined. The lumbar cord, the cervical
Animals that die within 48 h of injection are considered to cord, the lower and upper medulla oblongata, the midbrain,
have died from non-specinc causes and may be replaced. The the thalamus and the motor cortex of each monkey, as a
test is not valid if: more than 20 per cent of the inoculated mínimum, are subjected to histological examination. Sections
monkeys die from nonspecific causes; serum samples taken are cut with a thickness of 15 11m and stained with gallocyanin.
from the control monkeys at the time of inoculation of the The minimum number of sections examined is as follows:
test animals and 10 days after the latter are euthanised show (a) 12 sections representative of the whole of the lumbar
evidence of infection by wild virus of the type to be tested enlargement,
or by measles virus. At the end of the observation period,
(b) 10 sections representative of the whole of the cervical
carry out autopsy and histopathological examinations of
enlargement,
appropriate areas of the brain for evidence of central nervous
system involvement. The material complies with the test if (c) 2 sections from the medulla oblongata,
there is no unexpected clínical or histopathological evidence (d) 1 sectiol1 from the pons and cerebellum,
of involvement of the central nervous system attributable to
the inoculated virus. (e) 1 section from the midbrain,
(f) 1 section from the left and the right of the thalamus,
0112008:20619 (g) 1 section from the left and the right motor cerebral cortex.
Scoring of virus activity. For the evaluation of virus activity
2.6.19. TEST FOR NEUROVIRULENCE in the hemisections of the spinal cord and brain-stem, a
OF POLIOMYELITIS VACCINE (ORAL) score system for the severity of lesions is used, differentiating
cellular infiltration and destruction of neurons as follows:
Monkeys used in the neurovirulence test comply with the
requirements given in the monograph on Poliomyelitis vaccine 1. Cellular infiltration only (the monkey is not counted as
oral (0215) and weigh not les s than l.5 kg. The pathogenicity positive),
for Macaca or Cercopithecus monkeys is tested in comparison 2. Cellular infiltration with minimal neuronal damage,
with that of a reference virus preparation for neurovirulence
testing by inoculation into the lumbar region of the central 3. Cellular infiltration with extensive neuronal damage,
nervous system after sedation with a suitable substance, for 4. Massive neuronal damage with or without cellular
example, ketamine hydrochloride. A sample of serum taken infiltration.
before the injection shal! be shown not to contain neutralising The scores are recorded on a standard form(4). A monkey with
antibody at a dilution of 1:4 when tested against not more neuronallesions in the sections but that shows no needle tract
than 1000 CCID 50 of each of the three types of poliovirus. is counted as positive. A monkey showing a needle tract in the
Number of monkeys. The vaccine and the appropriate sections, but no neuronallesions is not regarded as positive. A
homotypic reference virus are tested concurrently in the same section that shows damage from trauma but no specific virus
group of monkeys. Equal numbers of animals are inoculated lesions is not included in the score.

(4) A suitable form is shown in lhe Requirements for PoliomyeHtis Vaccine (Oral) (Requiremenls for Biological Substances No. 7, World Health Organization).

202 See the information sectian an general monographs (ca ver pages)
EUROPEAN PHARMACOPOEIA 8.0 2.6.20. Anti-A and anti-B haemagglutinins

Severity scores are based on hemisection readings of the The constants Cl' C2 and CJ are calculated from the
lumbar (L), cervical (C) and brain (B) histological sections. expressions:
The les ion score (LS) for each positive monkey is calculated
as follows:
Sumof Sumof Sum of
L score escore B score
Number 01' Numberol'
+
NOillÓCC o; 1
LS~
hemisections hemisections hernlsectIons
3

A mean lesion score is calculated for each group of positive N¡ number of positive monkeys per vaccine test,
monkeys.
N2 number of positive monkeys in the two tests,
Evaluation. The comparison of the virus activity in the
vaccine and the reference preparation is based on the activity 2.3 normal deviate at the 1 per cent leve!,
in the lumbar enlargement of the cord and the degree of 2.6 normal deviate at the 0.5 per cent leve!,
spread of activity from this regio n to the cervical enlargement
and the brain. Acceptance or rejection is based on the total 1.6 normal deviate at the 5 per cent level.
score of aH the test animals. Individual animals showing A neurovirulence test in which the mean lesion score for the
evidence of unusuaHy high activity, either in the lumbar regio n reference (Xref) is not compatible with previous experience
or as the result of spread from this region, are also taken into is not used for assessing a test vaccine. If the test is valid,
consideration in the final evaluation. The monovalent bulk the mean lesion score for the vaccine to be tested (Xtest ) is
passes the test if the required number of animals is positive calculated and compared with that of the homotypic reference
and if none of the clinical and histopathological examinations vaccine.
shows a significant difference in pathogenicity between
the vaccine virus and the reference material. Criteria for
acceptance are given below.
Criteria. A suitable number of neurovirulence qualifying 0712011:20620
tests (for example, four tests) is carried out on each reference
vaccine (types 1, 2 and 3) to provide data on the activity of 2.6.20. ANTI-A AND ANTI-B
such vaccines that will serve as the basis of the criteria for
vaccines to be tested. The overall mean lesion score (M) for
HAEMAGGLUTININS
the replicate tests on each reference virus is calculated together METHOD A: INDIRECT METHOD
with the pooled estimate of the within-test variance (S2) and
Prepare in duplicate serial dilutions of the preparation to
the within-test deviation (s).
be examined in a 9 giL solution of sodium chloride R. To
Validity criteria for the results of a test on a reference each dilution of 1 series add an equal volume of a 5 per
cent V/V suspension of group A¡ red blood cells previously
preparation are established on the basis of the cumulative
data from the qualifying tests. No generally applicable criteria washed 3 times with the sodium chloride solution. To each
can be given; for laboratories with limited experience, the dilution of the other series add an equal volume of a 5 per
cent V/V suspension of group B red blood ceUs previously
following empirical method for setting acceptable limits for
washed 3 times with the sodium chloride solution. Incubate
the mean lesion score for the reference preparation (X,cf) may
the suspensions at 37 oC for 30 min then wash the cells
be he!pful (see Table 2.6.19.-1):
3 times with the sodium chloride solution. Leave the ceUs in
contact with a polyvalent anti-human globulin reagent for
Table 2.6.19.-1 30 mino Without centrifuging, examine each suspension for
agglutination under a microscope.
Lower Iimit Upper limit

Types 1 and 2 M - s M + s METHOD B: DIRECT METHOD

M - s/2
MATERIALS
Type 3 M+s
Phosphate-buffered saline (PBS). Dissolve 8.0 g of sodium
chloride R, 0.76 g of anhydrous disodium hydrogen phosphate R,
If the mean les ion score for the vaccine to be tested is X'est and 0.2 g of potassium chloride R and 0.2 g of potassium dihydrogen
Cl' Cz and CJ are constants determined as described below, phosphate R in water R and dilute to 1000 mL with the same
then: solvent. If the solution has to be kept for several days, 0.2 g
of sodium azide R may be added in order to avoid microbial
the vaccine is not acceptable if: contamination.
PBS-BSA so/ution. PBS containing 2 giL of bovine albumin R
(Cohn Fraction V, for ELISA). Store the solution at 2-8 oC but
allow it to reach 19-25°C before use.
the vaccine may be retested once if: Papain solution. Use serological-grade papain from a
commercial source, the activity of which has been validated.
Red b/ood ce/ls. Use pooled D-negative A¡ (A¡rr),
D-negative B (Brr) and D-negative O (Orr) red blood cells
from preferably 3 donors. When Immunoglobulin for anti-A
lf the vaccine is retested, the means of the lesion scores for the and anti-B antibodies limit test BRP is used, 3 donors are to
vaccine to be tested and the reference vaccine are recalculated. be used. Az red blood cells are not recommended as they give
The vaccine is not acceptable if: weaker reactions.
X(test l+test 2) - X(ref l+ref 2) > C3 Wash the cells 4 times with PBS or until the supernatant is
2 cIear. Each wash consists of suspending the cells in a minimum
of 2 volumes of PBS, centrifuging the cells at 1800 g for 5 min

General Notices (1) apply to all monographs and other texts 203
2.6.21. Nudeic add amplification techniques EUROPEAN PHARMACOPOEIA 8.0

to pack, and discarding the supernatant. Treat the packed of the preparation to be examined, the positive control and
cells with papain solution according to the manufacturer's the negative controL Mix by shaking the plate on a shaker for
instructions and wash the cells 4 times with PBS. 10 s (or until the cells are resuspended).
Red blood ceUs may be sto red for not more than 1 week in a Centrifuge the plate at 80 g at room temperature for 1 min to
preservative solution at 2-8 oc. A preparation of the following pack the cells. Place the plate at an angle oI' approximately 70 0

composition is appropriate: Read after 4-5 min or when the negative controls (D-negative
Trisodiu111 citrate 8 giL O red blood cells and negative control solution) have streamed.
A cel! button at the bottom of the well indicates a positive
D-glucose 20 giL result. A stream of cells represents a result.
Citric acid 0.5 giL Record the endpoint titre as the reciproca! of the highest
dilution that rise to a positive result.
Sodiu111 chloride 4.2 giL
The positive control has nominal anti-A and anti-B titres
Inosine 0.938 giL of 32 (range 32-64 for anti-A; range 16-32 for anti-B) and
Adenosine triphosphate (ATP) 0.4 giL the negative controls (D-negative O red blood cells and
negative control solution) must not show agglutination at the
Chlora111phenicol 0.34 giL starting dilution of 1 in 2. Users must validate their own test
N e0111ycin sulfate 0.1 giL conditions, and investigate their assay conditions and reagents
in the event of results being significantly different from
those expected. Failure to obtain negative reactions with the
If using stored cells, wash the ceUs at least twice in PBS or negative controls may indicate that, for example, insufficient
until the supernatant is clear before proceeding. time has elapsed for the cells to stream, or that reagents have
Microtitre plates. Use V -bottomed rigid microtitre plates. been used directly from cold storage.
Reference standards. Immunoglobulin (anti-A, anti-B
If the anti -A or anti -B titre of the preparation to be examined
antibodies test positive control) BRP and Immunoglobulin
is greater than the titre of the positive control when both
(anti-A, anti-B antibodies test negative control) BRP are preparations are titrated from 25 giL the test preparation is
suitable for use as the positive control and negative control, to be compared with Immunoglobulin for anti-A and anti-B
antibodies limit test BRP.
respectively, and should be used as guides for operators
establishing and performing the direct method fo1' anti-A and The maximum allowable titre is 64 when the preparations are
anti-B haemagglutinins. titrated from 25 giL
Immunoglobulin for anti-A and anti-B aníibodies limit test BRP
defines the recommended maximum limits pel'missible for
07/2010:20621
batches of human immunoglobulin and must be used only
for comparison with batches of human immunoglobulin that
have higher titres than the positive controL 2,6,21. NUCLEIC ACID
METHOD Al\1PLIFICATION TECHNIQUES
The test described in this chapter is at room 1. INTRODUCTION
temperature on the positive control sollltions, the negative
control solutions and the test solutions at the same time and Nucleic acid amplification techniques are based on 2 different
llnder identical conditions. V¡henever necessary, a further approaches:
test is performed with Immunoglobulin for anti-A and anti-B L amplification of a target nucleic acid sequence using, for
antibodies limit test BRP. example, chain reaction (PCR), ligase chain
reaction or isothermal ribonucleic acid (RNA)
Reference solutions. Reconstitute the positive control and amplification;
the negative control according to the instructions. The
immunoglobulin G (IgG) cOl1centration is 50 giL in each of 2. amplification of a hybridisation signal using, fol' example,
the recol1stituted preparations. Make a 2-fold dilution of each for deoxyribonucleic acid (DNA), the branched DNA
reconstituted preparation with PBS-BSA solutiol1 to obtain (bDNA) method; in this case signa! amplification is
solutions containing IgG at 25 giL Prepare 7 further serial achieved without subjecting the nucleic acid to repetitive
2-fold dilutions of each preparation using PBS-BSA solution of amplification.
to extend the dilutiol1 range to 1/256 (0.195 giL IgG). Add In this general chapter, the PCR method is described as the
20 I1L of each dilution of each preparation in triplicate to the reference technique. Alternative methods may be used, if they
microtitre plateo comply with the quality requirements described below.
Test solutions. Dilute the preparation to be examined with 2. SCOPE
PBS-BSA solution to obtain a starting IgG COl1cel1tration This section establishes the reqllirements for sample
of 25 giL For 50 giL preparations, this is a 2-fold dilution; preparation, in vitro amplification of DNA sequences and
adjust the dilutiol1 factor accordingly for preparations with detection of the specific PCR product. With the aid of PCR,
an IgG concentratiol1 other than 50 giL to obtain a starting defined DNA sequences can b"e detected. RNA sequences can
COl1centration of 25 giL for testing. This 25 giL solution is also be detected following reverse transcription of the RNA to
assigned a nominal2-fold dilution factor for comparison with complementary DNA (cDNA) and subsequent amplification.
the reference solutions, even if this does not reflect the true
dilution factor used to achieve 25 giL Prepare 7 further serial 3. PRINCIPLE OF THE METHOD
2-fold dilutions of the preparation using PBS-BSA solution to PCR is a procedure that allows specific in vitro amplification
extend the nominal dilution l'ange to 1/256 (0.195 giL IgG) for of segments of DNA or of RNA after reverse transcription
comparison with the reference preparations over the same IgG into cDNA.
concentration range. Add 20 I1L of each dilution in tripEcate
Following denaturation of double-stranded DNA into
to the microtitre plateo
single-stranded DNA, 2 synthetic oligonuc!eotide primers of
Prepare 3 per cent V/V suspensions of papain-treated opposite polarity anneal to their respective complementary
D-negative Al' B and O red blood cells in PBS/BSA solution. sequences in the DNA to be amplified. The short
Add 20 I1L of D-negative Al' B and O red blood cells double-stranded chat form as a result of specific base
respectively to the 1st, the 2nd and the 3,,1 dilution series of each pairing between the primers and the complementary DNA

204 See ¡he scction 011 general ''''''''"''<Yrn (cover pages)


EUROPEAN PHARMACOPOEIA 8.0 2.6.21. Nudeic add amplification techniques

sequence border the DNA segment to be amplified, and serve - the type of DNA polymerase, buffer composition and
as starting positions for in vitro DNA synthesis by means of a reaction volume used for the amplification;
heat-stable DNA polymerase. - the type of thermocycler used and the thermal conductivity
Amplification of the DNA occurs in cyeles consisting of: rate between the apparatus, reaction tube and reaction fluid.
- heat denaturation of the nueleie aód (target sequence) into 5.4. Detection
2 single strands; The amplicon generated by PCR may be identified by
- specific annealing of the primers to the target sequence size, sequence, chemieal modification or a combination
under suitable reaction conditions; of these parameters. Detection and characterisation by
- extension of the primers, which are bound to both single size may be achieved by gel electrophoresis (using agarose
strands, by DNA polymerase at a suitable temperature or polyacrylamide slab geIs or capillary electrophoresis)
(DNA synthesis). or column chromatography (for example, liquid
Repeated cyeles of heat denaturation, primer annealing and chromatography). Detection and characterisation by sequence
DNA synthesis results in an exponential amplification of the composition may be achieved by the specific hybridisation
DNA segment limited by the primers. of probes having a sequence complementary to the target
The specific PCR product known as an amplicon can be sequence or by eleavage of the amplified material reflecting
detected by a variety of methods of appropriate specificity and target-specific restrietion-enzyme sites. Detection and
sensitivity. characterisation by chemical modification may be achieved by,
for example, incorporation of a fluorophore into the amplicons
Multiplex PCR assays use several primer pairs designed for and subsequent detection of fluorescence following excitation.
simultaneous amplification of different targets in one reaction.
Detection of amplicons may also be achieved by using
4. TEST MATERIAL probes labelled to permit a subsequent radioisotopic or
Because of the high sensitivity of PCR, the samples must immuno-enzyme-coupled detection.
be protected against external contamination with target 6. EVALUATION AND INTERPRETATION OF RESULTS
sequences. Sampling, storage and transport of the test material
are performed under conditions that minimise degradation A valid result is obtained within a test only if the positive
of the target sequence. In the case of RNA target sequences, control(s) is unambiguously positive and the negative
special precautions are necessary sin ce RNA is highly sensitive control(s) is unambiguously negative. Due to the very high
to degradation by ribonueleases. Care must be taken since sensitivity of the PCR method and the inherent risk of
sorne added reagents, such as anticoagulants or preservatives, contamination, it is necessary to confirm positive results by
may interfere with the test procedure. repeating the complete test procedure in duplicate, where
possible on a new aliquot of the sample. The sample is
5. TEST METHOD considered positive if at least one of the repeat tests gives a
5.1. Prevention of contamination positive result. As soon as a measurable target threshold is
defined, a quantitative test system is required.
The risk of contamination requires a strict segregation of the
are as depending on the material handled and the technology 7. QUALITY ASSURANCE
used. Points to consider inelude movement of personnel, 7.1. Validation ofthe PCR assay system
gowning, material flow and air supply and decontamination
procedures. The validation programme must inelude validation of
instrumentation and the PCR method employed. Reference
The system should be sub-divided into compartments such as:
should be made to the ICH guidelines (topie Q2B) Validation
- master-mix are a (area where exelusively template-free of Analytieal Method: Methodology.
material is handled, e.g. primers, buffers, etc.);
Appropriate official working reference preparations
- pre-PCR (area where reagents, samples and controls are or in-house reference preparations calibrated against
handled); International Standards for the target sequences for whieh the
- PCR amplification (amplified material is handled in a test system will be used are indispensable for validation of a
elosed system); PCR test.
- post-PCR detection (the only area where the amplified 7.1.1. Determination oi the positive cut-off point
material is handled in an open system). During validation of qualitative tests, the positive cut-off point
5.2. Sample preparation must be determíned. The positive cut-off point is defined
When preparing samples, the target sequence to be amplified as the mínimum number of target sequen ces per volume
needs to be efficiently extracted or liberated from the test sample that can be detected in 95 per cent of test runs. The
material in a reproducible manner and in such a way that positive cut -off point depends on interrelated factors such as
amplification under the selected reaction conditions is the volume of the sample extracted and the efficacy of the
possible. A variety of physico-chemical extraction procedures extraction methodology, the transcription of the target RNA
and/or enriehment procedures may be employed. into cDNA, the amplification process and the detection.
Additives present in test material may interfere with PCR. To define the detection limit of the assay system, reference
The procedures described under 7.3.2. must be used as a must be made to the positive cut-off point for each target
control for the presence of inhibitors originating from the sequence and the test performance aboye and below the
test material. positive cut -off point.
In the case of RNA-templates, care must be taken to avoid 7.1.2. Quantitative assay systems
ribonuelease activity. Por a quantitative assay, the following parameters are
5.3. Amplification determined during validation: accuracy, precision, specificity,
quantitation limit, linearity, range and robustness.
PCR amplification of the target sequence is conducted
under defined cyeling conditions (temperature profile 7.2. Quality control of reagents
for denaturation of double-stranded DNA, annealing AH reagents crucial for the methodology used have to
and extension of primers; incubation times at selected be controlled prior to use in routine applications. Their
temperatures; ramp rates). These depend on various acceptance/withdrawal is based on pre-defined quality criteria.
parameters such as: Primers are a crucial component of the PCR assay and as
- the length and base composition of primer and target such their design, their purity and the validation of their use
sequences; in a PCR assay require careful attention. Primers may be

General Notices (1) apply fo all monographs and other texts 205
2.6.21. Nudeic acid amplification techniqucs EUROPEAN PHARMACOPOEIA 8.0

modifled (for example, by conjugation with a fluo1'Ophore ol' user. Nevertheless, the performance ofthe kit with respect
UHC'h,.Hjin order 'lo permit a speciflc method of detection to its intended use has to be demonstrated by the user (e.g.
5uch modifications do not inhibit detection limit, 1'Obustness, cross-contamination).
accurate and efficient amplification oí the target sequence.
2, SPECIFICITY
7.3. Run wntrols
Specificity is the ability to assess unequivocally nudeic acid
7.3.1. External controls
in the presence of components that may be expected to be
In order to minimise the risk of contamination and to ensure presenL
external controls are
The specificity oí nucleic acid amplification analytical
11l\.lUU<::U in each assay:
p1'Ocedures is dependent on the choice of primers, the choice
control: this contains a defined number oí of probe (for analysis oí the final product) and the stringency
target-sequence copies, the number being close to the of the test conditions (for both the amplincation and the
positive cut-off value, and determined individually for each detection steps).
and indicated as a multiple of the positive
When designing primers and probes, the specificity of
oí the assay system;
the primers and pro bes to detect only HCV RNA should
- negative control: a sample of a suitable matrix already be investigated by comparing the chosen sequences with
proven to be free of the target sequences. sequences in published data banks. For HC\!, primers
7.3.2. Internal control (and p1'Obes) will normally be chosen from areas of the 5'
Internal controls are denned nucleic acid sequences non-coding region of the HCV genome which are highly
containing, unless otherwise prescribed, the primer binding conserved for all genotypes.
sites. Internal control s must be amplified with defined efficacy, The amplified p1'Oduct should be unequivocally identified by
and the must be clearly discernible. Internal using one of a number of methods such as amplification with
cont1'Ols must be of the same type of nucleic acid (DNA/RNA) nested primers, restriction enzyme analysis, sequencing, or
as the material to be tested. The internal control is preferably hybridisation with a specific probe.
added to the test material before isolating the nucleic acid In order to validate the specificity of the analytical procedure,
and therefore acts as an overall control (extraction, reverse at least 100 HCV RNA-negative plasma pools should be
transcription, detection). tested and shown to be non-reactive. Suitable samples of
7.3.3. Threshold control non-reactive pools are available from the European Directorate
The threshold control for quantitative assays is a test sample for the Quality of Medicines & HealthCare (EDQM),
with the at a concentration that is defined as the The ability of the analytical procedure to detect al! HCV
threshold not to be exceeded. It contains the analyte suitably genotypes will again depend on the choice of primers, probes
calibrated in International Units and is analysed in parallel in and method parameters. This ability should be demonstrated
each run of a quantitative assay. using characterised reÍerence paneis. However, in view of
7,4, External assessment the difficulty in obtaining samples of some genotypes (e.g.
assessment programl11es genotype 6), the most prevalent genotypes (e.g. genotypes 1
assurance procedure for each and 3 in Europe) should be detected at a suitable leve!.
I,,,r~,,r;n, and each operator. 3. DETECTION LIMIT
The detection limit 01' an individual analytical procedure is the
lowest amount of nucleic acid in a sample that can be cletected
of l1udeíc acid amplification but not necessarily quantitated as an exact value.
the detection of The nucleic acid amplification analytical used for
the detection 01' HCV RNA in plasma yields
hepatitis C virus (HCV) RNA in plasma qualitative results. The number 01' possible results limited
pools: guidelines to 2: either positive or negative. Although the determination
of the detection limit is recommended, fOl' practical purposes,
L SCOPE a positive cut-off point should be determined for the nucleic
The l11ajority oí nucleic acid amplification analytical acid amplificabon analytical procedure. The positive cut-ofÍ
p1'Ocedures are qualitative (quantal) tests for the presence of point (as defined in the general chapter 2.6.21) is the minimum
nucleic acid with sorne quantitative tests (either in-house or number 01' target sequences per volume sample that can be
commercial) being available. For the detection of HCV RNA detected in 95 per cent 01' test runs. This positive cut-off
contamination of plasma pools, qualitative tests are adequate point is influenced by the distribution 01' viral genomes in the
and may be considered to be a ¡¡mit test for the control individual samples being tested and by factors such as enzyme
of impurities as described in the Pharmeuropa Technical efficiency, and can result in different 95 per cent cut -off values
Guide for the elaboration oí monographs, December 1999, for individual analytica! test runs,
Chapter III 'Validation of analytical procedures'. These In order to determine the positive cut -off point, a dilutiol1
guidelines describe methods to validate only qualitative series of a working reagent or of the hepatitis e virus BRP,
nucleic acid amplification analytical procedures for assessing which has been calibrated against the WHO HCV International
HCV RNA contamination of plasma pools. Therefore, the
Standard 96/790, shoulcl be tested 011 different days to examine
2 characteristics regarded as the m05t important for validation variation between test fUns. At least 3 independent dilution
oÍ the p1'Ocedure are the specificity and the detection series should be tested with a sufficient number oí replicates at
limito In the robustness of the analytical procedure each dilution to give a total number of 24 test results for each
should be evaluated.
dilution, to enable a statistical analysis of the results.
However, this document may also be used as a basis for the
For example, a laboratory could test 3 dilution series on
validation of nucleic acid amplification in generaL
different days with 8 replicates Íor each clilution, 4 dilution
For the purpose of this document, an analytical procedure is series 011 different days with 6 replicates for each dilution,
defined as the complete f1'Om extraction 01' nucleic or 6 dilution series on different days with 4 replicates for
acid to detection of the amplified products, each dilution. In order to keep the number of dilutions at
VJhere commercial kit s are used Íor part or all of the analytical a manageable leve!, a preliminary test (using, fOl' example,
documented validation points already covered by 10glO dilutions of the plasma pool sample) should be carried
kit manufacturer can substitute for the validation the out in order to obtain a prelil11inary value for the positive

206 See the information section on monographs (ca ver pages)


EUROPEAN PHARMACOPOEIA 8.0 2.6.21. Nudeic acid amplification techniques

cut-off point (i.e. the highest dilution giving a positive be documented by conducting a parallel test on 8 samples
signal). The range of dilutions can then be chosen around the of a plasma pool that is spiked with HCV RNA to a final
predetermined preliminary cut -off point (using, for example, concentration of 3 times the previously determined 95 per
a dilution factor of 0.5 10g¡O or less and a negative plasma pool cent cut-off value. Al! results should be positive.
for the dilution matrix). The concentration of HCV RNA
Operator qual~fication. An appropriate qualification
that can be detected in 95 per cent of test runs can then be
programme should be implemented for each operator involved
calculated using an appropriate statistical evaluation.
in the testing. To confirm successful training each operator
These results may also serve to demonstrate the intra-assay should test at least 8 replicate samples of a plasma pool
variation and the day-to-day variation of the analytical spiked with HCV RNA to a final concentration of 3 times the
procedure. previously determined 95 per cent cut-off va1ue. This test (8
replicate samples) should be repeated twice on 2 separate days,
4. ROBUSTNESS i.e. a total of 24 tests performed on 3 different days. AH results
The robustness of an analytical procedure is a measure of should be positive.
its capacity to remain unaffected by small but deliberate
variations in method parameters and provides an indication
of its reliability during normal usage.
Validation of nudeic acid amplification
The evaluation of robustness should be considered during
techniques (NAT) for the quantification of
the development phase. 1t should show the reliability of the B19 virus (B19V) DNA in plasma pools:
analytical procedure with respect to deliberate variations in guidelines
method parameters. For NAT, small variations in the method
parameters can be crucial. However, the robustness of the
1. SCOPE
method can be demonstrated during its development when
sma11 variations in the concentrations ofreagents (e.g. MgCl2 , The European Pharmacopoeia requires that plasma pools used
primers or dNTP) are tested. To demonstrate robustness, at for manufacture of certain products are tested for the presence
least 20 HCV RNA negative plasma pools (selected at random) of B19 virus (B19V) DNA with a threshold concentration
spiked with HCV RNA to a final concentration of 3 times that must not be exceeded. In order to comply with these
the previously determined 95 per cent cut -off value should requirements, quantitative NAT tests are preferred. The
be tested and found positive. characteristics regarded as the most important for validation
Problems with robustness may also arise with methods that of the quantitative NAT procedure are accuracy, precision,
use an initial ultracentrifugation step prior to extraction of the specificity, quantitation limit, linearity and range. In addition,
viral RNA. Therefore, to test the robustness of such methods, the robustness of the analytical procedure should be eva1uated.
at least 20 plasma pools containing varying levels of HCV This guideline describes methods to validate NAT analytical
RNA, but lacking HCV -specific antibodies, should be tested procedures for assessing B19V DNA contamination of plasma
and found positive. pools based on the 1CH guidelines. However, this document
Cross-contamination prevention should be demonstrated may a1so be used as a basis for the validation of quantitative
by the accurate detection of a panel of at least 20 samples NAT in general.
consisting of alternate samples of negative plasma pools and For the purpose of this document, an analytical procedure is
negative plasma pools spiked with high COl1centrations of defined as the complete procedure from extraction of nucleic
HCV (at least 10 2 times the 95 per cent cut-off value or at acid to detection of the amplified products.
least 104 IU/mL).
Where commercial kits are used for part or all of the analytical
5. QUALITY ASSURANCE procedure, documented validation points already covered by
the kit manufacturer can substitute for the validation by the
For biological tests such as NAT, specific problems may arise user. Nevertheless, the performance of the kit with respect
that influence both the validation and the interpretation of to its intended use has to be demonstrated by the user (e.g.
results. The test procedures must be described precisely in the precision, accuracy, range, robustness).
form of standard operating pro ce dures (SOPs). These should
cover:
2. ACCURACY
- the mode of sampling (type of container, etc.);
Accuracy expresses the closeness of agreement between the
- the preparation of mini -pools (where appropriate); value that is accepted as either a conventional true value
- the conditions of storage before analysis; or an accepted reference value and the value found. The
accuracy of an assay is dependent on the calibration of
- the exact description of the test conditions, including
the assay and on the variance of the different assay steps.
precautions taken to prevent cross-contamination or Though it is recommended to establish the accuracy across
destruction of the viral RNA, reagents and reference
the specified range of the analytical procedure, the most
preparations used;
important assessment of accuracy is in the range of the
- the exact description of the apparatus used; threshold concentration. In the case of B19V NAT assays for
- the detailed formulae for calculation of results, including investigation of plasma pools it is recommended to assess
statistical evaluation. the accuracy of the calibrated assay by assaying at least
5 concentrations (dilution factor ofO.5log¡o) of B19 virus DNA
The use of a suitable run control (for example, an appropriate jor NAT testing BRP or another material, suitably calibrated
dilution of hepatitis e virus BRP or plasma spiked with an in International Units against the actual WHO B19 DNA
HCV sample calibrated against the WHO HCV International International Standard, covering the range of the currently
Standard 96/790) can be considered a satisfactory recommended thresho1d concentration of 10.0 IU/flL B19V
system -suitability check and ensures that the reliability of the DNA (e.g. lOs IU/mL, 1045 IU/mL, 10 4 IU/mL, 10 35 IU/mL
analytical procedure is maintained whenever used. and 10 3 IU/mL), with at least 3 replicates for each dilution.
Technical qualification. An appropriate installation and Accuracy should be reported for the different concentrations
operation qualification programme should be implemented in terms of percentage determined compared with the known
for each critical piece of the equipment used. For confirmation amount of B19V DNA. It should reflect the level of technology
of analytical procedure performance after a change of of the respective assays, which should also be defined, for
critical equipment (e.g. thermocyclers), the change should example in collaborative studies.

General Notices (1) apply ta all monographs and other íexts 207
2.6.21. Nudeic add amplification techniques EUROPEAN PHARMACOPOEIA 8.0

3. PRECISION intermediate-precision studies by limiting dilution analysis.


Precision expresses the closeness of agreement between a The lowest concentration of target nucleic acids that is
series of measurements, obtained from multiple sampling of quantitated with suitable precision and accuracy is defined.
the same homogenous sample. The precision is defined at 6. LINEARITY
3 levels:
The linearity of an assay is its ability to obtain test results that
- repeatability expresses the precision under the same are directly proportional to the concentration of the nucleic
operating conditions over a short interval of time acid. The linearity of the B19V NAT assay is assessed during
(intra-assay precision); it is assessed by using 1 assay and the repeatability and intermediate-precision studies by testing
testing 3 replicates of appropriate dilutions of a B19V replicates of diluted samples with the concentrations covering
DNA-positive sample suitably calibrated in International the whole quantitative range. The interval between the upper
Units and covering the whole quantitative range of the and the lower concentration of the target nucleic acid where
assay; the coefficient of variation for the individual samples test results are directly proportional to the concentrations is
is calculated (intra-assay variability); defined.
- intermediate precision expresses the intra-laboratory
variations (inter-assay precision); it is established by 7. RANGE
assaying replicates (as routinely used for the assay) of The range of an assay is the interval between the upper and
appropriate dilutions of a Bl9V DNA-positive sample the lower concentration of nucleic acid in the sample for
suitably calibrated in International Units covering the which it has been demonstrated that the procedure has a
whole quantitative range of the assay under different suitable level of precision, accuracy and linearity. The range
circumstances (e.g. different days, different analysts, of the B19V NAT assay is assessed during the repeatability
different equipment, different reagents); the coefficient of and intermediate-precision studies by testing replicates of
variation for the individual samples is calculated; diluted samples. The interval between the upper and the lower
- reproducibility expresses the precision between different concentration that can be expressed with an acceptable degree
laboratories (inter-laboratory precision); it is assessed of accuracy and precision is defined.
by participation in quantitative collaborative studies on
8. ROBUSTNESS
B19V DNA-NAT assays, e.g. under the Proficiency Testing
Scheme (PTS), including the comparative analysis of the The robustness of an analytical procedure is a measure of its
obtained quantitative results, where appropriate. capacity to remain unaffected by small but deliberate variations
in method parameters and provides an indication of its
4. SPECIFICITY reliability during normal usage. The evaluation of robustness
should be considered during the development phase. It should
Specificity expresses the ability to assess unequivocally nucleic
show the reliability of the analytical procedure with respect
acid in the presence of components that may be expected to
to deliberate variations in method parameters. For NAT,
be presento The specificity of NAT analytical procedures is
small variations in the method parameters can be crucial.
dependent on the choice of primers, the choice of probe (for
Nonetheless, the robustness of NAT can be demonstrated
analysis of the final product) and the stringency of the test
during the development of the method when small variations
conditions (for both the amplification and the detection steps).
in the concentrations of reagents, for example MgCl 2, primers
When designing primers and probes, the specificity of the or dNTP, are tested. To demonstrate robustness, at least 20
primers and probes to detect only human B19V DNA should B19V DNA-negative plasma pool samples spiked with B19V
be investigated by comparing the chosen sequen ces with DNA at the threshold concentration should be tested and
sequences in published data banks. There should be no major found to have acceptable quantitative values.
homology found with sequences unrelated to B19Y. Cross-contamination prevention should be demonstrated
The amplified product should be unequivocally identified by by the accurate detection of a panel of at least 20 samples
using one of a number of methods such as amplification with consisting of alternate samples of plasma pools without
nested primers, restriction enzyme analysis, sequencing, or B19V DNA or with levels below the threshold concentration
hybridisation with a specific probe. (lO samples) and plasma pools spiked with high
In order to examine the specificity of the analytical procedure, concentrations ofB19V DNA (at least 10 times the threshold
2

at least 20 B19V DNA-negative plasma pools should be tested leve!, 10 samples).


and shown to be non-reactive. 9. QUALITY ASSURANCE
Parvovirus B19 genotypes. The International Committee on For biological tests such as NAT, specific problems may arise
Taxonomy of Virus es (ICTV) has classified representatives that may influence both the validation and the interpretation
of the 3 genotypes as strains of human parvovirus B19. oí results. The test procedures must be described precisely
Genotype 1 represents prototype B19Y, genotype 2 represents in the form oí standard operating procedures (SOPs). These
viral sequences like A6, and genotype 3 represents V9-like should cover:
sequences. By performing sequence alignment with respective
B 19V genotype sequences available from nucleic acid sequence - the mode of sampling (type of container, etc.);
databases, primers and probes should be designed to detect - the preparation of mini-pools by manufacturers (where
and quantify consistently the different human parvovirus B19 appropriate) ;
genotypes. Reference materials should be used to check the - the conditions of storage before analysis;
approach chosen. Since biological reference preparations
reflecting sorne genotypes might be difficult to obtain, - the exact description of the test conditions including
respective plasmid preparations or synthetic nucleic acids may precautions taken to prevent cross-contamination or
also serve as a characterised target sequence source. However, destruction of the viral nucleic acids, reagents and reference
those cannot be used to validate the extraction procedure. preparations used;
- the exact description of the apparatus used;
5. QUANTITATION LIMIT - the detailed formulae for calculation of results, including
The quantitation limit is the lowest amount of nucleic acid statistical evaluation.
in a sample that can be determined quantitatively with The inclusion of an appropriate threshold control (for
suitable precision and accuracy. The quantitation limit of example, plasma spiked with a B19V DNA sample suitably
the B19V NAT assay is assessed during the repeatability and calibrated in International Units, such as B19 virus DNA

208 See the information section on general monographs (caver pages)


EUROPEAN PHARMACOPOEIA 8.0 2.6.24. Avían viral vaccines: tests for extraneous agents in seed lots

for NA T testing BRP) is considered to be a satisfactory d) For a freeze-dl'ied preparation, reconstitute using a suitable
system-suitability check and en sures that the reliability of the liquido Unless otherwise stated or justified, the test substance
analytical procedure is maintained whenever used. must contain a quantity of virus equivalent to at least 10 doses
Technical qualification. An appropriate instaUation and of vaccine in 0.1 mL of inoculum,
operation qualificatiol1 programme should be implemented e) lf the virus of the seed 10t would interfere with the
for each critical piece of the equipment used. For confirmation conduct and sensitivity of the test, neutralise the virus in the
of analytical procedure performance after a change of critical preparation with a monospecific antiserum.
equipment (e.g. thermocyclers), the change should be f) Monospecific antiserum and serum of avian origin used
documented by conducting a parallel test on 8 samples of a for cell culture 01' any other purpose, in any of these tests,
plasma pool that is spiked with a COl1centration ofB19V DNA shall be free from antibodies against and free from inhibitory
around the threshold concentration. Al! results should be effects on the organisms listed hereafier under 7. Antibody
acceptable and reflect the features of the assay as determined specifications for sera used in extraneous agents testing.
during the validation phase. g) Where specified in a monograph or otherwise justified,
Operator qualification. An appropriate qualification if neutralisation of the virus of the seed 10t is required but
programme should be implemented for each operator involved difficult to achieve, the in vitro tests described below are
in the testing. To confirm successful training, each operator adapted, as required, to provide the necessary guarantees of
should test, on 3 separate days, at least 8 replicate samples of a freedom from contamination with an extraneous agent.
plasma pool that is spiked with a concentration of B19V DNA h) Other types of tests than those indicated may be used
around the threshold concentration (i.e. a total of 24 samples). provided they are at least as sensitive as those indicated and of
AH results should be acceptable and reflect the features of the appropriate specificity. Nucleic acid amplification techniques
assay as determined during the validation phase. (2.6.21) give specific detection for many agents and can be
used afier validation fOI sensitivity and specificity.
0112008:20622 1. TEST FOR EXTRANEOUS AGENTS USING
EMBRYONATED HENS' EGGS
2.6.22. ACTIVATED COAGULATION Use a test substance, diluted if necessary, containing a quantity
FACTORS of neutralised virus equivalent to at least 10 doses of vaccine
in 0.2 mL of inoculum. Suitable antibiotics may be added.
Whel'e applicable, determine the amount of heparin Inoculate the test substance into 3 groups of 10 embryonated
pl'esent (2.7.12) and neutralise the heparin, for example hens' eggs as follows:
by addition of protamine sulfate R (10 flg of protamine - group i: 0.2 mL into the allantoic cavity of each
sulfate neutralises 1 IU ofheparin). Prepare 1 to 10 and 1 9- to ll-day-old embryonated egg;
to 100 dilutions of the preparation to be examined using
- group 2: 0.2 mL onto the chorio-allantoic membrane of
tris(hydroxymethyl)aminomethane buffer solution pH 7.5 R.
Place a series of polystyrene tubes in a water-bath at 37 oC each 9- to ll-day-old embryonated egg;
and add to each tube 0.1 mL of platelet-poor plasma R and - group 3: 0.2 mL into the yolk sac of each 5- to 6-day-old
0.1 mL of a suitable dilution of a phospholipid preparation to embryonated egg.
act as a platelet substitute. Allow to stand for 60 s. Add to Candle the eggs in groups 1 and 2 daily for 7 days and the
each tube either 0.1 mL of 1 of the dilutions or 0.1 mL of the eggs in group 3 daily for 12 days. Discard embryos that die
buffer solution (control tube). To each tube add immediately during the first 24 h as non -specific deaths; the test is not
0.1 mL of a 3.7 giL solution of calcium chloride R previously valid unless at least 6 embryos in each group survive beyond
heated to 37 oC, and measure, within 30 min of preparing the the first 24 h after inoculation. Examine macroscopically
original dilution, the time that elapses between addition of the for abnormalities al! embryos that die more than 24 h after
calcium chloride solution and the formation of a clot. The inoculation, or that survive the incubation periodo Examine
test is not valid unless the coagulation time measured for the also the chorio-allantoic membranes of these eggs for any
control tube is 200 s to 350 s. abnormality and test the al!antoic fluids for the presence of
haemagglutinating agents.
Carry out a further embryo passage. Pool separately material
0712009:20624 from live and from the dead and abnormal embryos. Inoculate
each pool into 10 eggs for each route as described aboye,
2.6.24. AVIAN VIRAL VACCINES: chorio-allantoic membrane material being inoculated onto
TESTS POR EXTRANEOUS AGENTS IN chorio-allantoic membranes, allantoic fluids into the allantoic
cavity and embryo material ¡nto the yolk sac. For eggs
SEED LOTS inoculated by the allantoic and chorio-allantoic routes, candle
the eggs daily for 7 days, proceeding and examining the
GENERAL PROVISIONS
material as described aboye. Por eggs inoculated by the yolk
a) In the following tests, chickens and/or chicken material sac route, candle the eggs daily for 12 days, proceeding and
such as eggs and ceH cultures shall be derived from chicken examining the material as described aboye.
flocks free from specified pathogens (SPF) (5.2.2).
The seed lot complíes with the test if no test embryo shows
b) Cell cultures for the testing of extraneous agents comply macroscopic abnormalities or dies from causes attributable
with the requirements for the master ceH seed of chapter 5.2.4. to the seed lot and if examination of the chorio-allantoic
Cell cultures for the production of veterinary vaccines, with the membranes and testing of the allantoic fluids show no
exception of the karyotype test and the tumorigenicity test, evidence of the presence of any extraneous agent.
which do not have to be carried out.
c) In tests using ceH cultures, precise specifications are given 2. TEST IN CHICKEN KIDNEY CELLS
for the number of replicates, monolayer surface are as and Prepare 7 monolayers of chicken kidney ceUs, each monolayer
minimum survival rate of the cultures. Alternative numbers of having an area of about 25 cm 2 . Maintain 2 monolayers
replicates and cel! surface areas are possible as well, provided as negative controls and treat these in the same way as the
that a minimum of 2 replicates are used, the total surface area 5 monolayers inoculated with the test substance, as described
and the total volume of test substance applied are not less below. Remove the culture medium when the cells reach
than that prescribed here and the survival rate requirements confluence. Inoculate 0.1 mL of the test substance onto each
are adapted accordingly. of the 5 monolayers. Allow adsorption fol' 1 h, add culture

General Notices (1) apply to all monographs and other texts 209
2.6.24. Avian viral vacdl1es: tests for extraneous agents in seed 10ts EUROPEAN PHARMACOPOEIA 8.0

medium and incubate the cultures for a total of at least The test is 110t valid if group-specific antigen is detected in
21 days, subculturing at 4- to 7-day intervals. Each passage is fewer than 5 of the 6 positive control replicate monolayers or
made with pooled cells and fiuids from aH 5 monolayers after if a positive result is obtained in any of the negative control
carrying out a freeze-thaw cyele. Inoculate 0.1 mL of po oled monolayers, or if the results for both of the 2 negative control
material onto each of 5 recently prepared monolayeIs of about are inconclusive. If the results for more than 1 of
25 cm 2 each, at each passage. For the last passage, grow the the test replicate monolayers are inconclusive, then further
cells 0.150 on a suitable substrate so as to obtain an area of sub cultures of reserved portions of the fibroblast monolayers
about 10 cm 2 of cells from each of the monolayers for test A. shall be made and tested until an unequivocal result is
The test is not valid if les s than 80 per cent of the monolayers obtained, If a positive result is obtained for any of the test
survive after any passage. monolayers, then the presence of avian leucosis virus in the
test substance has been detected.
Examine microscopically al! the cel! cultures frequently
throughout the entire incubation period for any signs The seed lot complies with the test if there is no evidence of
of cytopathic effect or other evidence of the presence of the presence of any avian leucosis virus.
contaminating agents in the test substance. At the end of the
total incubation period, carry out the following procedures. 4. TEST FOR AVIAN RETICULOENDOTHELIOSIS VIRUS
A. Fix and stain (with Giemsa or haematoxylin and Prepare 11 monolayers oE primary or secondary chick
eosin) about 10 cm 2 of confiuent cells from each of the embryo fibroblasts from the tissues of 9- to ll-day old chick
5 monolayers. Examine the cells microscopical!y for any embryos or duck embryo fibroblasts from the tissues of
cytopathic effect, inelusion bodies, syncytial formation, 13- to 14-day-old embryos, each monolayer having an are a
or other evidence of the presence of contaminating agents of about 25 cm 2 ,
from the test substance. Remove the culture medium when the cells reach confluence.
R Drain and wash about 25 cm 2 of cells from each of the Inoculate 0,1 mL of the test substance onto each of 5
5 monolayers. Cover these cells with a 0.5 per cent of the monolayers, Allow adsorption for 1 h, and add
suspension of washed chicken erythrocytes (using at least culture medium, Inoculate 4 of the monolayers with avian
I mL of suspension far each 5 cm 2 of cells). Incubate the reticuloendotheliosis virus as positive controls (not more than
cells at 4 oC for 20 min and then wash gentIy in phosphate 10 CCID so in 0,1 mL), Maintain 2 110n-inoculated monolayers
buffered saline pH 7.4. Examine the ceUs microscopically as negative controls.
for haemadsorption attributable to the presence of a Incubate the cells for a total of at least 10 days, subculturing
haemadsorbing agent in the test substance. twice at 3- to 4-day intervals. The test is not valid if fewer
C. Test individual samples of the t1uids from each cell than 3 of the 4 positive controls or fewer than 4 of the 5 test
culture using chicken erythrocytes for haemagglutination monolayers or neither of the 2 negative controls survive after
attributable to the presence of a haemagglutinating agent any passage.
in the test substance. For the last sub culture, grow the fibroblasts on a suitable
substrate so as to obtain an area of about 10 cm 2 of
The test is not valid if there are any signs of extraneous agents
confluent fibroblasts from each of the original 11 monolayers
in the negative control cultures, The seed lot complies with the
for the subsequent test: test about 10 cm 2 of confinent
test if there is no evidence of the presence of any extraneous
fibroblasts derived from each of the original 11 monolayers by
agent.
immunostaining for the presence of avian reticuloendotheliosis
virus. The test is not valid if avian reticuloendotheliosis virus
3. TEST FOR AVIAN LEUCOSIS VIRUSES is detected in fewer than 3 of the 4 positive controlmonolayers
Prepare at least 13 replicate monolayers of either D F -1 cells or in any of the negative control OI if the results

or primary or secondary chick embryo fibroblasts from for both of the 2 negative control monolayers are inconclusive.
the tissues of 9- to ll-day-old embryos that are known to If the results for more than 1 of the test monolayers are
be genetically susceptible to subgroups A, B and J of avian inconclusive then further sub cultures of reserved portions of
leucosis viruses and that support the growth of exogenous but the fibroblast shall be mOlde and tested until an
not endogenous avian leucosis viruses (cells from C/E strain unequivocal result is
chickens are suitable), Each replicate shall have an are a of The seed lot complies with the test if there is no evidence of
about 50 cm 2 . the presence of avian reticuloendotheliosis virus.
Remove the culture medium when the cells reach confiuence,
Inoculate 0,1 mL of the test substance onto each of 5 of the 5. TEST FOR CHICKEN ANAEMIA VIRUS
replicate monolayers. Allow adsorption for 1 h, and add Prepare eleven 20 mL suspensions of the MDCC-MSBI cel!
culture medium. Inoculate 2 of the replicate monolayers with line or another cellline of equivalent sensitivity in 25 cm 2 cel!
subgroup A avían leucosis virus (not more than 10 CCID so in culture fiasks containing about 5 x lO' cells/mL. Inoculate
0.1 mL), 2 with subgroup B avian leucosis virus (not more 0.1 mL of the test substance into each of 5 flasks. Inoculate
than 10 CCID so in 0.1 mL) and 2 with subgroup J avian 4 of the suspensions with 10 CCID sll chicken anaemia virus as
leucosis virus (not more than 10 CCID 50 in 0.1 mL) as positive positive controls. Maintain 110t fewer than 2 110n-inoculated
controls. Maintain not fewer than 2 non-inoculated replicate suspensions. Maintain al! the cell cultures for a total of at least
monolayers as negative controls. 24 days, subculturing 8 times at 3- to Ll-day intervals. During
Incubate the cells for a total of at least 9 days, subculturing the subculturing the presence of chicken anaemia virus may
at 3- to 4-day intervals, Retain cells from each passage leve! be indicated by a metabolic colour change in the infected
and harvest the cells at the end of the total incubation periodo cultures, the culture fluids becoming red in comparison with
Wash cells from each passage level from each replicate and the control cultures. Examine the cells microscopically for
resuspend the cells at 10 7 ceUs per millilitre in barbital-buffered cytopathic effect. At this time or at the end of the incubation
saline for subsequent testing by a Complement Fixation for period, centrifuge the ce]]s from each fiask at low speed
Avian Leucosis (COFAL) test or in phosphate buffered saline and resuspend at about 10 6 cells/mL and place 25 f-lL in
for testing by Enzyme-Linked Immunosorbent Assay (ELlSA). each of 10 wells of a multi-well slide. Examine the cells by
Then, carry out 3 cycles of freezing and thawing to release immunostaining.
group-specific antigen and perform a COFAL test or an The test is not valid if chicken anaemia virus is detected
test on each extract to detect group-specific avian leucosis in fewer than 3 of the 4 positive controls or in any of the
antigen if present. non-inoculated controls. rf the results for more than 1 of the

210 See the information section on (ca ver pages)


EUROPEAN PHARMACOPOEIA 8.0 2.6.24. Avían viral vaccines: tests for extraneous agents in seed lots

test suspensions are inconclusive, then further sub cultures of B. Addmonal tests for turkey extraneous agents
reserved portions of the test suspensions shall be made and lf the seed virus is of turkey origin or was propagated in
tested until an unequivocal result is obtained. turkey substrates, tests fOI antibodies against the following
agents are also carried out.
The seed lot complies with the test if there is no evidence of
the presence of chicken anaemia virus. Agent Type oftest

Chlanzydia spp. ELA


6. TEST POR EXTRANEOUS AGENTS USING CHICKS Avian infectious haen10rrhagic enteritis virus AGP

Inoculate each of at least 10 chicks with the equivalent of Avian paramyxovirus 3 HI


100 doses of vaccine by the intramuscular route and with
Avian infectious bursal disease virus type 2 SN
the equivalent of 10 doses by eye-drop. Chicks that are
2 weeks of age are used in the test except that if the seed A test for freedom from turkey lympho-proliferative
virus is pathogenic for birds of this age, older birds may disease virus is carried out by intraperitoneal inoculation
be used, if required and justified. In exceptional cases, for of twenty 4-week-old turkey poults. Observe the poults for
inactivated vaccines, the virus may be neutralised by specific 40 days. The test is not valid if more than 20 per cent of the
antiserum if the seed virus is pathogenic for birds at the poults die from non-specific causes. The seed 10t complies
age of administration. Repeat these inoculations 2 weeks with the test if sections of spleen and thymus taken from
later. Observe the chicks for a period of 5 weeks from the 10 poults 2 weeks after inoculation show no macroscopic
day of the first inoculatiol1. No antimicrobial agents shall be or microscopic lesions (other than those attributable to the
administered to the chicks during the test periodo The test is seed 10t virus) and no poult die s from causes attributable to
not valid if fewer than 80 per cent of the chicks survive to the the seed 10t.
end of the test periodo
C. Additional tests for duck extraneous agents
Collect serum from each chick at the end of the test periodo lf the seed virus is of duck origin or was propagated in
Test each serum sample fo1' antibodies against each of the duck substrates, tests for antibodies against the following
agents listed below (with the exception of the virus type of agents are also carried out.
the seed 10t) using one of the methods indicated for testing Agent Type oftest
for the agent.
Chlamydia spp. ELA
Clinical signs of disease in the chicks during the test period
(other than signs attributable to the virus of the seed 10t) and Duele and goose parvoviruses SN, ErA
the detection of antibodies in the chicks after inoculation Duck enteritis virus SN
(with the exception of antibodies to the virus of the seed 10t),
are classed as evidence of the presence of an extraneous agent Duck hepatitis virus type r SN
in the seed loto
The seed 10t complies with the test if there is no evidence
lt is recommended that sera from these birds is retained so that of the presence of any extraneous agent.
additional testing may be carried out if requirements change. D. Additional tests fOl" goose extraneous agents
A. Standard tests lf the seed virus is of goose origin or was prepared in goose
substrates, tests for the following agents are also caniea out.
Agent Type oftest Agent Type oÍtest
Avian adenoviruses, group 1 SN, EIA, AGP Duck and goose parvovirus SN, EIA
Avian encephalomyelitis virus AGP, ELA Duek enteritis virus SN
Avian infectious bronchitis virus EIA, HI Goose haemorrhagic polyomavirus test in goslings shown below
or another suitable test
Avian infectious laryngotracheitis virus SN, ElA, IS
Inoculate subcutaneously the equivalent of at least 10 doses
Avian leucosis viruses SN, ElA
to each often l-day-old susceptible goslings. Observe the
Avian nephritis virus IS goslings for 28 days. The test is not valid if more than
20 per cent of the goslings die from non -specific causes.
Avian orthoreoviruses IS, EIA The seed virus complies with the test if no gosling dies
Avian reticllloendotheliosis virus AGP, IS, ErA from causes attributable to the seed 101.
Chicken anaemia virus IS, EIA, SN 7. ANTIBODY SPECIFICATIONS FOR SERA USED IN
EXTRANEOUS AGENTS TESTING
Egg drop syndrome virus HI, ELA
All batches of serum to be used in extraneous agents testing,
Avian infectious bursal disease virus Serotype 1: AGP, ELA, SN either to neutralise the vaccine virus (seed lot or batch of
Serotype 2: SN finished product) or as a supplement for culture media used
Influenza A virus AGP, ErA, HI for tissue culture propagation, shall be shown to be free fram
AGP
antibodies against and free from inhibitory effects on the
Marek's disease virus
following micro-organisms by suitably sensitive tests.
Newcastle disease virus HI, EIA Avian adenoviruses
Turkey rhinotracheitis virus EIA Avian encephalomyelitis virus
Salmonella pullorum Agg
Avian infectious bronchitis viruses
Avian infectious bursal disease virus types 1 and 2
Agg: agglutination
Avían infectious haemorrhagic enteritis virus
AGP: agar gel precipitation
EIA: enzyme immunoassay (e.g. EUSA)
Avian infectious laryngotracheitis virus
HI: haemagglutination inhibition Avian leucosis viruses
IS: immunostaining (e.g. tluorescent antibody) Avian nephritis virus
SN: serUI11 neutralisation Avian paramyxoviruses 1 to 9

General Notices (1) apply to all monographs and other texts 211
2.6.25. Avian Uve virus vaccines: exÍfaneous agents in finished pwduct EUROPEAN PHARMACOPOEIA 8.0

Avian orthoreoviruses from testing the batch of vaccine, as decribed under 6. Test
Avian reticu10endotheliosis virus for extraneous agents using chicks of chapter 2.6.24. Testfor
extraneous agents in seed ¡ots.
Chicken anaemia virus
g) Monospecific antiserum and serum of avian origin used for
Duck enteritis virus
cell culture and any other purpose, in any of these tests, shall
Duck hepatitis virus type 1 be free of antibodies against and Íree from inhibitory effects
Egg drop syndrome virus on the organisms listed under 7. Antibody specifications for
Fowl pox virus sera used in extraneous agents testing (2.6.24).
Influenza viruses h) Other types oí tests than those indicated may be used
Marek's disease virus provided they are at least as sensítive as those indicated and of
appropriate specificity. Nucleic acid amplificatíon techniques
Turkey herpesvirus (2.6.21) give specific detection for many agents and can be
Turkey rhinotracheitis virus used after validation for sensitivity and specificity.
Non-immune serum for addition to culture media can be
assumed to be free from antibodies against any of these 1. TEST FOR EXTRANEOUS AGENTS USING
virus es if the agent is known not to infect the species of originEMBRYONATED HENS' EGGS
of the serum and it is not necessary to test the serum for such Prepare the test vaccine, diIuted if necessary, to contain
antibodies. Monospecific antisera for virus neutralisation can neutralised virus equivalent to 10 doses of vaccine in 0.2 mL
be assumed to be free from the antibodies against any of these of inoculum. Suitable antibiotics may be added. Inoculate
virus es if it can be shown that the immunising antigen could the test vaccine into 3 groups of 10 embryonated hens' eggs
not have been contaminated with antigens derived from that as follows:
virus and if the virus is known not to infect the species of - group 1: 0.2 mL into the allantoic cavity of each 9- to
origin of the serum; it is not necessary to test the serum for ll-day-old embryonated egg,
such antibodies. It is not necessary to retest sera obtained - group 2: 0.2 mL onto the chorio-allantoic membrane of
from birds from SPF chicken flocks (5.2.2). each 9- to ll-day-old embryonated egg,
Batches of sera prepared for neutralising the vaccine virus - group 3: 0.2 mL into the yolk sac of each 5- to 6-day-old
must not be prepared from any passage level derived from embryonated egg.
the virus isolate used to prepare the master seed 10t 01' from
an isolate cultured in the same cellline. Candle the eggs in groups 1 and 2 daily for 7 days and the
eggs in group 3 for 12 days. Discard embryos that die during
the first 24 h as non-specific deaths; the test is not valid
0112008:20625 unless at least 6 embryos in each group survive beyond the
first 24 h after ínoculation. Examine macroscopically for
abnormalities al! embryos which die more than 24 h after
2.6.25. AVIAN LIVE VIRUS VACCINES: inoculation, or which survive the incubation periodo Examine
TESTS FOR EXTRANEOUS AGENTS IN also the chorio-allantoic membranes of these eggs for any
BATCHES OF FINISHED PRODUCT abnormality and test the allantoic fluids fol' the presence of
haemagglutinating agents.
GENERAL PROVISIONS Carry out a further embryo passage. Pool separately material
a) In the following tests, chickens and/or chicken material from live and from the dead and abnormal embryos. Inoculate
such as eggs and ceH cultures shall be derived from chicken each pool into 10 eggs for each route as described aboye,
flocks free from specified pathogens (SPF) (5.2.2). chorio-allantoic membrane material being inoculated onto
b) Cel! cultures for the testing of extraneous agents comply chorio-allantoic membranes, allantoic fluids into the allantoic
with the requirements for the master cel! seed of chapter 5.2.4. cavity and embryo material into the yolk sac. For eggs
Cell cultures for the production of veterinary vaccines, with the inoculated by the allantoic and chorio-allantoic routes, candle
exception of the karyotype test and the tumorigenicity test, the eggs daily for 7 days, proceeding and examining the
which do not have to be carried out. material as described aboye. For eggs inoculated by the yolk
sac route, candle the eggs daily fol' 12 days, proceeding and
c) In tests using ceH cultures, precise specifications are given examining the material as described aboye.
Íor the number of replicates, monolayer surface areas and
minimum survival rate of the cultures. Alternative numbers of The batch of vaccine complíes with the test if no test
replicates and cel! surface areas are possible as well, provided embryo shows macroscopic abnormalities or dies from
that a minimum of 2 replicates are used, the total surface are a causes attributable to the vaccine and if examination of the
and the total volume of vaccine test applied are not less than chorio-allantoic membranes and testing of the allantoic fluid s
that prescribed here and the survival rate requirements are show no evidence of the p1'esence of extraneous agents.
adapted accordingly. 2. TEST IN CHICKEN EMBRYO FIBROBLAST CELLS
d) In these tests, use the liquid vaccine or reconstitute a
Prepare 7 monolayers of primary or secondary chieleen
quantity ofthe freeze-dried preparation to be tested with the
embryo fibroblasts, from the tissues of 9- to ll-day-old
liquid stated on the label or another suitable diluent such as
embryos, each monolayer having an are a of about 25 cm 2 •
water for injections. Unless otherwise stated or justified, the
Maintain 2 monolayers as negative controls and treat these
test substance contains the equivalent of 10 doses in 0.1 mL
in the same way as the 5 monolayers inoculated with the test
of inoculum. vaccine, as described below. Remove the culture mediu111
e) If the vaccine virus would interfere with the conduct and when the cells reach confluence. Inoculate 0.1 mL of test
sensitivity of the test, neutralise the virus in the preparation vaccine onto each of 5 of the monolayers. Allowadsorption
with a monospecific antiserum. for 1 h and add culture medium. Incubate the cultures
f) Where specified in a monograph or otherwise justified, if for a total of at least 21 days, subculturing at 4- to S-day
neutralisation of the vaccine virus is required but difficult intervals. Each passage is made with po oled cells and fluids
to achieve, the in vitro tests described below are adapted, as from all 5 monolayers after carrying out a freeze-thaw cyele.
required, to provide the necessary guarantees of freedom from Inoculate 0.1 mL of pooled material onto each of 5 recently
contamination with an extraneous agent. Alternatively, or in prepared monolayers of chicken embryo fibroblast ceUs, each
addition to in vitro tests conducted on the batch, a test for monolayer having an are a of about 25 cm l each as before. For
extraneous agents may be conducted on chíck sera obtained the last passage; grow the cells also on a suitable substrate so

212 See the information sectian on general monographs (cover pages)


EUROPEAN PHARMACOPOEIA 8.0 2.6.25. Avian live virus vacdnes: extraneous agents in finished product

as to obtain an area of about 10 cm 2 of cells from each of the The batch of vaccine complies with the test if there is no
monolayers, for test A. The test is not valid if less than 80 per evidence of the presence of egg drop syndrome virus or any
cent of the test monolayers, or neither of the 2 negative control other extraneous agent.
monolayers survive after any passage.
Examine microscopically aH the ceH cultures frequently 4. TEST POR MAREK'S DISEASE VIRUS
throughout the entire incubation period for any signs Prepare 11 monolayers of primary or secondary chick embryo
of cytopathic effect or other evidence of the presence of fibroblasts from the tissues of 9- to 11-day-old embryos,
contaminating agents in the test vaccine. At the end of the each monolayer having an area of about 25 cm 2 • Remove the
total incubation period, carry out the following procedures. culture medium when the cells reach confluence. Inoculate
A. Fix and stain (with Giemsa or haematoxylin and eosin) 0.1 mL of test vaccine onto each of 5 of the monolayers (test
about 10 cm2 of confluent ceUs from each of the 5 original monolayers). Allow adsorption for 1 h, and add culture
monolayers. Examine the cells microscopically for any medium. Inoculate 4 of the monolayers with a suitable strain
cytopathic effect, inclusion bodies, syncytial formation, of Marek's disease virus (not more than 10 CCID so in 0.1 mL)
or any other evidence of the presence of a contaminating to serve as positive controls. Maintain 2 non-inoculated
agent from the test vaccine. monolayers as negative controls.
B. Drain and wash about 25 cm 2 of cells from each of the Incubate the cultures for a total of at least 21 days, subculturing
5 monolayers. Cover these cells with a 0.5 per cent at 4- to S-day intervals. Each passage is made as follows:
suspension of washed chicken red blood cells (using at least trypsinise the cells, prepare separate pools of the ceUs from
1 mL of suspension for each 5 cm 2 of cells). Incubate the the test monolayers, from the positive control monolayers and
ceUs at 4 oC for 20 min and then wash gently in phosphate from the negative control monolayers. Mix an appropriate
buffered saline pH 7.4. Examine the cells microscopically quantity of each with a suspension of freshly prepared primary
for haemadsorption attributable to the presence of a or secondary chick embryo fibroblasts and prepare 5, 4 and
haemadsorbing agent in the test vaccine. 2 monolayers, as before. The test is not valid if fewer than 4 of
C. Test individually samples of the fluid from each cell culture the 5 test monolayers or fewer than 3 of the 4 positive controls
using chicken red blood cells for haemagglutination or neither of the 2 negative control monolayers survive after
attributable to the presence of a haemagglutinating agent any passage.
in the test vaccine. Examine microscopically all the ceH cultures frequently
The test is not valid if there are any signs of extraneous agents throughout the entire incubation period for any signs of
in the negative control cultures. The batch of vaccine complies cytopathic effect or other evidence of the presence of a
with the test if there is no evidence of the presence of any contaminating agent in the test vaccine.
extraneous agent. For the last sub culture, grow the cells on a suitable substrate
so as to obtain an area of about 10 cm 2 of confluent ceUs fr0111
3. TEST FOR EGG DROP SYNDROME VIRUS each of the original 11 monolayers for the subsequent test:
Prepare 11 monolayers of chicken embryo liver cells, from the test about 10 cm 2 of confluent cells derived from each of the
tissues of 14- to 16-day-old embryos, each monolayer having original 11 monolayers by immunostaining for the presence of
an are a of about 25 cm 2 • Remove the culture medium when Marek's disease virus. The test is 110t valid if Marek's disease
the cells reach confluence. Inoculate 0.1 mL of test vaccine virus is detected in fewer than 3 of the 4 positive control
onto each of 5 of the monolayers (test monolayers). Allow monolayers or in any of the negative control monolayers, or
adsorption for 1 h, add culture medium. Inoculate 4 of the if the results for both of the 2 negative control monolayers
monolayers with a suitable strain of egg drop syndrome virus are inconclusive.
(not more than 10 CCID so in 0.1 mL) to serve as positive The batch of vaccine complies with the test if there is no
control monolayers. Maintain 2 non-inoculated monolayers
evidence of the presence of Marek's disease virus or any other
as l1egative control monolayers.
extraneous agent.
Incubate the cells for a total of at least 21 days, subculturing
every 4-5 days. Each passage is made as follows: carry out 5. TESTS FOR TURKEY RHINOTRACHEITIS VIRUS
a freeze- thaw cycle; prepare separate pools of the cells plus
A. In chicken embryo fibroblasts
fluid from the test monolayers, from the positive control
monolayers and from the negative control monolayers; NOTE: this test can be combined with Test 2 by using the
inoculate 0.1 mL of the pooled material onto each of 5, 4 and 2 same test mono/ayers and negative controls, for al/ stages up
recently prepared monolayers of chicken embryo liver cells, to the final specific test for turkey rhinotracheitis virus on
each monolayer havirig an are a of about 25 cm 2 as before. cel/s prepared from the last subculture.
The test is not valid if fewer than 4 of the 5 test monolayers Prepare 11 monolayers of primary or secondary chick
or fewer than 3 of the 4 positive controls or neither of the embryo fibroblasts from the tissues of 9- to ll-day-old
2 negative control monolayers survive after any passage. embryos, each monolayer having an are a of about
Examine microscopically al! the cell cultures at frequent 25 cm 2• Remove the culture medium when the cells reach
intervals throughout the entire incubation period for any confluence. Inoculate 0.1 mL of test vaccine onto each of 5
signs of cytopathic effect or other evidence of the presence of a of the monolayers (test monolayers). Allowadsorption
contaminating agent in the test vaccine. At the end of the total for 1 h, and add culture medium. Inoculate 4 of the
incubation period, carry out the following procedure: test monolayers with a suitable strain of turkey rhinotracheitis
separately, cen culture fluid from the test monolayers, positive virus as positive control s (not more than 10 CCID 50 in
control monolayers and negative control monolayers, using 0.1 mL). Maintain 2 non-inoculated monolayers as negative
chicken red blood ceHs, for haemagglutination attributable to controls.
the presence of haemagglutinating agents. Incubate the cultures for a total oí at least 21 days,
The test is not valid if egg drop syndrome virus is detected subculturing at 4- to S-day intervals. Each passage is made
in fewer than 3 of the 4 positive control monolayers OI in as follows: carry out a freeze- thaw cycle; prepare separate
any of the negative control monolayers, or if the results for pools of the cells plus fluid from the test monolayers,
both of the 2 negative control monolayers are inconclusive. from the positive control monolayers and from the
If the results for more than 1 of the test monolayers are negative control monolayers; inoculate 0.1 mL of the
inconclusive then further sub cultures of reserved portions of pooled material onto each of 5, 4 and 2 recently prepared
the monolayers shall be made and tested until a11 unequivocal monolayers of chicken embryo fibroblasts cells, each
result is obtained. monolayer having an are a of about 25 cm 2 as before. The

General Notices (1) apply to all monographs and other texts 213
2.6.25. Avían Uve virus vaccines: extraneous agents in finished product EUROPEAN PHARMACOPOEIA 8.0

test is not valid if fewer than 4 of the 5 test mon01ayers or cultures, the culture fiuids becoming red in comparison with
fewer than 3 of the 4 positive contr01s or neither of the the control cultures. Examine the cells microscopically for
2 negative control monolayers survive after any passage. cytopathic effect. At this time or at the end of the incubation
For the last sub culture, grow the cells on a suitable substrate period, centrifuge the ceUs from each fiask at low speed,
so as to obtain an area of about 10 cm 2 of confiuent ceUs resuspend at about 10 6 cells per millilitre and place 25 ~lL in
from each of the original 11 monolayers for the subsequent each of 10 wells of a multi-well slide. Examine the cells by
test: test about 10 cm 2 of confiuent ceUs derived from each immunostaining.
of the original 11 monolayers by immunostaining for the The test is not valid if chicken anaemia virus is detected
presence of turkey rhinotracheitis virus. The test is not in fewer than 3 of the 4 positive controls or in any of the
valid if turkey rhinotracheitis virus is detected in fewer non-inoculated eontrols. Ifthe results for more than 10fthe
than 3 of the 4 positive control monolayers or in any of the test suspensions are inconclusive then further sub cultures of
negative control monolayers, or if the results for both of reserved portions of the test suspensions shaU be made and
the 2 negative control monolayers are inconelusive. rf the tested until an unequivocal result is obtained.
results for both of the 2 test monolayers are inconelusive The batch of vaccine complies with the test if there is no
then further sub cultures of reserved portions of the evidence of the presence of chicken anaemia virus.
fibroblasts shall be made and tested until an unequivocal
result is obtained. 7. TEST FOR DUCK ENTERITIS VIRUS
The batch of vaccine complies with the test if there is no This test is carried out for vaccines prepared on duck or goose
evidence of the presence of turkey rhinotracheitis virus or substrates.
any other extraneous agent.
Prepare 11 monolayers of primary or secondary Muscovy
B. In Vero cells
duck embryo liver cells, from the tissues of 21- or 22-day-old
Prepare 11 monolayers ofVero ceUs, each monolayer having embryos, each monolayer having an area of about 25 cm z.
an are a of about 25 cm 2 . Remove the culture medium when Remove the culture medium when the cells reach confiuenee.
the cells reach confiuence. Inoculate 0.1 mL of test vaccine Inoculate 0.1 mL of test vaccine onto each of 5 of the
onto each of 5 of the monolayers (test monolayers). AUow monolayers (test monolayers). Allow adsorption for 1 h
adsorption for 1 h, and add culture medium. Inoculate and add culture medium. Inoculate 4 of the monolayers
4 of the monolayers with a suitable strain of turkey with a suitable strain of duek enteritis virus (not more than
rhinotracheitis virus (not more than 10 CCID so in 0.1 mL) 10 CCID sll in 0.1 mL) to serve as positive controls. Maintain
to serve as positive controls. Maintain 2 non-inoculated 2 non-inoculated monolayers as negative controls.
monolayers as negative controls.
Incubate the cultures for a total of at least 21 days, subculturing
Incubate the cultures for a total of at least 21 days, at 4- to S-day intervals. Each passage is made as follows:
subculturing at 4- to S-day intervals. Each passage is made trypsinise the ceUs and prepare separate pools of the ceUs from
as follows: carry out a freeze- thaw cyele. Prepare separate the test monolayers, from the positive control monolayers
pools of the cells plus fiuid from the test monolayers, and from the negative control monolayers. Mix a portion
from the positive control monolayers and from the of each with a suspension of freshly prepared primary or
negative control monolayers. Inoculate 0.1 mL of the secondary Muscovy duck embryo liver ceUs to prepare 5, 4 and
pooled material onto each of 5, 4 and 2 recently prepared 2 mOl1olayers, as before. The test is not valid if fewer than 4 of
monolayers ofVero ceUs, each monolayer having an are a of the 5 test monolayers or fewer than 3 of the 4 positive cOl1trols
about 25 cm 2 as before. The test is not valid if fewer than 4 or neither of the 2 negative control s survive after any passage.
of the 5 test monolayers or fewer than 3 of the 4 positive
c011trols or neither of the 2 negative controls survive after For the last sub culture, grow the cells 011 a suitable substrate
any passage. so as to obtain an area of about 10 cm z of confiuent cells from
each of the original 11 monolayers for the subsequent test:
For the last sub culture, grow the ceUs on a suitable substrate test about 10 cm 2 of confiuent cells derived from each of the
so as to obtain an are a of about 10 cm z of confiuent ceUs original 11 monolayers by immunostaining for the presel1ce of
from each of the original 11 monolayers for the subsequent duck enteritis virus. The test is not valid if duele enteritis virus
test: test about 10 cm 2 of confiuent cells derived from each is detected in fewer than 3 of the 4 positive control monolayers
of the original 11 monolayers by immunostaining for the or in any of the negative control monolayers, or if the results
presence of turkey rhinotracheitis virus. The test is not for both of the 2 negative control monolayers are inconclusive.
valid if turkey rhinotracheitis virus is detected in fewer If the results for more than 1 of the test monolayers are
than 3 of the 4 positive control monolayers or in any of inconclusive then further sub cultures of reserved portions of
the negative control monolayers, or if the results for both the monolayers shall be made and tested until an unequivocal
of the 2 negative control monolayers are inconelusive. result is obtained.
If the results for more than 1 of the test monolayers are
inconelusive then further sub cultures of reserved portions The batch of vaccine complies with the test if there is no
of the monolayers shall be made and tested until an evidence of the presence of duck enteritis virus or any other
unequivocal result is obtained. extraneous agent.
The batch of vaccine complies with the test if there is no 8. TEST FOR DUCK AND GOOSE PARVOVIRUSES
evidence of the presence of turkey rhinotracheitis virus or
any other extraneous agent. This test is carried out for vaccines prepared on duck or goose
substrates.
6. TEST FOR CHICKEN ANAEMIA VIRUS Prepare a suspension of sufficient primary or secondary
Prepare eleven 20 mL suspensions of the MDCC-MSBI cell Muscovy duck embryo fibroblasts from the tissues of 16- to
line or another cellline of equivalent sensitivity in 25 cm 2 18-day-old embryos, to obtain not fewer than 11 monolayers,
fiasks containing about 5 x lOs cells/mL. Inoculate 0.1 mL each having an are a of abant 25 cm 2 • Inoculate 0.5 mL of test
of test vaccine into each of 5 of these fiasks. Inoculate vaccine into an aliquot of eells for S monolayers and seed into
4 other suspensions with 10 CCID so chicken anaemia virus as 5 replicate containers to form 5 test monolayers. Inoculate
positive controls. Maintain not fewer than 2 non-inoculated 0.4 mL of a suitable strain of duck parvovirus (not more than
suspensions. Maintain all the cell cultures for a total of at least 10 CCID so in 0.1 mL) into an aliquot of cells for 4 monolayers
24 days, subculturing 8 times at 3- to 4-day intervals. During and seed into 4 replicate containers to form 4 positive control
the subculturing the presence of chicken anaemia virus may monolayers. Prepare 2 non-inoculated monolayers as negative
be indicated by a metabolic colour change in the infected contr01s.

214 See the information section on general monographs (ca ver pages)
EUROPEAN PHARMACOPOEIA 8.0 2.6.26. Test for anti-D antibodies in human immunoglobulin

Incubate the cultures for a total of at least 21 days, subculturing Adenosine triphosphate (ATP) 0.4 giL
at 4- to 5-day intervals. Each passage is made as follows: Chloramphenicol 0.34 giL
carry out a freeze-thaw cyele. Prepare separate pools of the
cells plus fluid from the test monolayers, from the positive Neomycin sulfate 0.1 giL
control monolayers and from the negative control monolayers.
Inoculate 0.5 111L, 0.4 mL and 0.2 mL of the pooled materials If using stored cells, wash the ceUs at least twice in PBS Ol"
into aliquots of a fresh suspension of sufficient primary or until the supernatant is clear before proceeding.
secondary Muscovy duck embryo fibroblast cells to prepare 5, Microtitre plates. Use V-bottomed rigid microtitre plates.
4 and 2 monolayers, as before. The test is 110t valid if fewer Reference standards. Immunoglobulin (anti-D antibodies
than 4 of the 5 test monolayers or fewer than 3 of the 4 positive test) BRP and Immunoglobulin (anti-D antibodies test negative
controls or neither of the 2 negative controls survive after control) BRP are suitable for use as the positive control and
any passage. negative control, respectively.
For the last sub culture, grow the cells on a suitable substrate
so as to obtain an area of about 10 cm1 of confluent cells from METHOD
each of the original 11 monolayers for the subsequent test: The test described in this chapter is performed at room
test about 10 cm 2 of confluent cells derived from each of the temperature on the positive control solutions, the negative
original 11 monolayers by immunostaining for the presence control solutions and the test solutions at the same time and
of duck Ol" goose parvovirus. The test is not valid if duck under identical conditions.
parvovirus is detected in fewer than 3 of the 4 positive control Reference solutions. Reconstitute the positive control and
monolayers or in any of the negative control monolayers, or the negative control according to the instructions. The
if the results for both of the 2 negative control monolayers immunoglobulin G (IgG) concentration is 50 giL in each of
are inconclusive. the reconstituted preparations. Make a 2-fold dilution of each
The batch of vaccine complies with the test if there is no reconstituted preparation with PBS-BSA solution to obtain
evidence of the presence of duele (or goose) parvovirus or any solutions containing IgG at 25 giL. Prepare 7 further serial
other extraneous agent. 2-fold dilutions of each preparation using PBS-BSA solution
to extend the dilution range to 1/256 (0.195 giL IgG). Add
20 ¡.tL of each dilution of each preparation in duplicate to the
07/2011:20626 microtitre plateo
Test solutions. Dilute the preparation to be examined with
2.6.26. TEST FOR ANTI-D ANTIBODIES PBS-BSA solution to obtain a starting IgG concentration
IN HUMAN IMMUNOGLOBULIN of 25 giL. For 50 giL preparations, this is a 2-fold dilution;
adjust the dilution factor accordingly for preparations with
MATERIALS an IgG concentration other than 50 giL to obtain a starting
Phosphate-buffered saline (PBS). Dissolve 8.0 g of sodium concentration of 25 giL for testing. This 25 giL solution is
chloride R, 0.76 g of anhydrous disodium hydrogen phosphate R, assigned a nominal 2-fold dilutiOl1 factor for comparison with
0.2 g of potassium chloride R and 0.2 g of potassium dihydrogen the reference solutions, even if this does not reflect the true
phosphate R in water R and dilute to 1000 mL with the same dilution factor used to achieve 25 giL. Prepare 7 further serial
solvent. If the solution has to be kept for several days, 0.2 g 2-fold dilutions of the preparation using PBS-BSA solution to
of sodium azide R may be added in order to avoid microbial extend the nominal dilution range to 1/256 (0.195 giL IgG) for
contaminatiol1. comparison with the reference preparations over the same IgG
PBS-BSA solution. PBS containing 2 giL of bovine albumin R concentration range. Add 20 ¡.tL of each dilution in duplicate
(Cohn Fraction V, for ELISA). Sto re the solution at 2-8 oC but to the microtitre plateo
allow it to reach 19-25 oC before use. Prepare 3 per cent V/V suspensions of papain-treated
Papain solution. Use serological-grade papain from a D-positive (preferably OR 1 R 2 , but OR¡R¡ or OR¡Rl may also
commercial source, the activity of which has been validated. be used) and D-negative (Orr) red blood cells in PBS-BSA
solution. Add 20 ¡.tL of D-positive red blood cells to 1 dilution
Red blood ce/ls. Use pooled D-positive red blood cells from 110t
series of each of the preparation to be examined, the positive
fewer than 3 donors, preferably of group OR 2R2• D-positive
control and the negative control, and 20 ¡.tL of D-negative red
red blood cells may also be obtained from OR¡R¡ or OR¡R2
blood cells to the other dilution series. Mix by shaking the
donors. Mixing phenotypes has not been tested and is plate on a shaker for 10 s (or until the cells are resuspended).
therefore not recommended.
Centrifuge the plate at 80 g at room temperature for 1 min to
Use pooled D-negative red blood cells, preferably from
pack the cells. Place the plate at an angle of approximately 70 0

3 donors of group Orr. When only 1 donor of group Orr is
Read after 4-5 min or when the negative controls (D-negative
available, D-negative red blood cells from only 1 donor may
red blood ceUs and negative control solution) have streamed.
be used.
A ceH button at the bottom of the well indicates a positive
Wash the cells 4 times with PBS or until the supernatant is result. A stream of cells represents a negative result.
clear. Each wash consists of suspending the ceUs in a minimum
Record the endpoint titre as the reciprocal of the highest
of 2 volumes of PBS, centrifuging the cells at 1800 g for 5 min
dilution that gives rise to a positive resulto
to pack, and discarding the supernatant. Treat the packed
cells with papain solution according to the manufacturer's The positive control has a nominal titre of 8 and the negative
instructions and wash the cells 4 times with PBS. contrals (D-negative red blood ceUs and negative control
solution) must 110t show agglutination at the starting dilution
Red blood cells may be stored for not more than 1 week in a
of 1 in 2. Users must validate their own test conditions, and
preservative solution at 2-8 oc. A preparation of the following
investigate their assay conditions and reagents in the event
composition is appropriate:
of results being significantly different fram those expected.
Trisodium citrate 8 giL Failure to obtain negative reactions with the negative controls
D-glucose 20 giL may indicate that, for example, insufficient time has elapsed
for the cells to stream, or that reagents have been used directly
Citric acid 0.5 giL
fram cold storage.
Sodium chloride 4.2 giL The titre of the preparation to be examined must not be greater
Inosine 0.938 giL
than the titre of the positive control when both preparations
are titrated fram 25 giL.

General Notices (1) apply to all monographs and o/her texts 215
2.6.27. Microbiological control of ceUular products EUROPEAN PHARMACOPOEIA 8.0

0112011:20627 carried out using the preparatíon deliberately contaminated


to different degrees with the following micro-organisms,
chosen for the likelihood of contamination and their growth
2.6.27. MICROBIOLOGICAL CONTROL requirements:
OF CELLULAR PRODUCTS - Aspergillus brasiliensis, for example, ATCC 16404,
IP 1431.83, 1MI 149007;
This test has been shown to be preferable to the test for
sterility (2.6.1) for certain cellular products, since it has better - Bacillus subtilis, for example, ATCC 6633, CIP 52.62,
sensitivity, has a broader range, and is more rapid. It is applied NCIMB 8054;
instead of the test for sterility (2.6.1) where prescribed in - Candida albicans, for example, ATCC 10231, IP 48.72,
a monograph. It may be carried out manually or using an NCPF 3179;
automated system.
- Clostridium sporogenes, for example, ATCC 19404,
C1P 79.3, NCTC 532 or ATCC 11437;
GENERAL PRECAUTIONS
- Propionibacterium acnes, for example, ATCC 11827;
The test is carried out under aseptic conditions according to
current regulations for potentially infective material. - Pseudomonas aeruginosa, for exal11ple, ATCC 9027,
NCIMB 8626, CIP 82.118;
The precautions taken to avoid contamination are such that
they do not affect any micro-organisms that are to be revealed - Staphylococcus aureus, fOI example, ATCC 6538, CIP 4.83,
in the test. The test is performed under working conditions NCTC 10788, NCIMB 9518;
that are monitored regularly by appropriate sampling of the - Streptococcus pyogenes, for example, ATCC 19615,
working area and by carrying out appropriate controls. CIP 1042.26, NCIMB 13285;
- Yersinia enterocolitica, for example, ATCC 9610, CIP 80.27,
GROWTH PROMOTION TEST
NCTC 12982.
Use at least 2 suitable enriched culture media (for example, It may be necessary to modify the list of micro-organisms
blood culture media) intended for detection of fungi and
depending on the origin of the ceUs and any l11icro-organisms
aerobic and anaerobic bacteria. previously found or associated with the particular type of cells.
Confirl11 the sterility of each batch of medium by the Other approaches to validation may also be used, for example,
incubation of representative containers at 35-37 oC for not interlaboratory comparison.
less than 7 days.
Each batch of medium is tested by the supplier and/or the TESTING OF THE PREPARATION TO BE EXAMINED
user for its growth-prol110ting capacities by inoculating
Sample. A representative sample including cells and/or
duplicate test containers of each medium with 10-100 viable medium is tested. The sample is added to the culture medium
l11icro-organisms of each of the strains listed in Table 2.6.27.-1, as soon as possible after collection. lf it is not added promptly
and incubating for either 7 days for automated detection or
after collection, it is stored at 5 ± 3 oC to avoid phagocytosis of
14 days for visual detection of l11icrobial growth at 35-37 oc.
micro-organisms by cells present in certain types of products
The test media are satisfactory if there is clear evidence of (for example, neutrophils).
growth in all inoculated media containers within this periodo
For haematopoietic products, the minimum al110unt to be
Table 2.6.27.-1. - Micro-organisms used for growth promotion used for the test depending 011 the total volul11e of the product
(V mL) is shown below.
Aerobic mediurn
Total product volume Inoculum volume
Staphylococcus aureus for example, ATCC 6538, CIP 4.83,
NCTC 10788, NCIMB 9518 (millilitres)

Bacillus subtilis for example, ATCC 6633, CIP 52.62, V Ce> 10 1 per cent of total volume
NCIMB 8054
! <:; V < 10 100 flL
Pseudomol1as aeruginosa for example, ATCC 9027, NCIMB 8626,
CIP 82.118 V<l Not applicable

Candida albicans for examp!e, ATCC 10231, IP 48.72, For haematopoietic products that require dilution before
NCPF 3179 freezing, the inoculum volume must be il1creased by the
Aspergillus brasiliensis for example, ATCC 16404, IP 143l.83, dilution factor. For other cellular products, suitable minimum
IMI 149007 amounts are defined in terms of volume or number of doses.
Anaerobic medium Analysis. Sal11ples are inoculated into containers of culture
medium as 500n as possible after coHection and incubated
Clostridiwn sporogenes for example, ATCC 19404, CIP 79.3, at 35-37 oC for not less than 7 or 14 depending on the
NCTC 532 or ATCC 11437 detection system used. A suitable proportion of the inoculum
is added to the medium to be incubated in aerobic conditions
Bacteroides fragilis for example, ATCC 25285, CIP 77.16, and the remainder of the inoculum to the medium to be
NCTC 9343
incubated in anaerobic conditions.

METHOD VALIDATION OBSERVATION AND INTERPRETATION OF RESULTS


Depending on the type of product, its method of preparation, Examine media, visually or with automated systems at ¡east
the inoculum volume used and the type of test system, the daily, and at the end of the observation period for evidence
need for validation in the presence of the type of preparation of microbial growth. If no growth is observed during or at
to be examined must be considered. Unless otherwise justified the end of the observation period, the product is 'culture
and authorised, the test system is validated with respect negative' at the limit of detection. lf growth is observed in a
to specificity (absence of false positive results), sensitivity valid test, the product is 'culture positive'; the contaminant is
(limit of detection) and reproducibility. During validatíon, identified to a suitable taxonomic level (genus, species) and
particularly to determine the limit of detection, the test is an antibiogral11 is established.

216 See the information section on general monographs (cave!" pages)


EUROPEAN PHARMACOPOEIA 8.0 2.6.30. Monocyte-activation test

04/2010:20630 Endotoxin equivalents are values for the contaminant


concentration read off the standard endotoxin dose-response
curve (Method A) or estimated by comparison with responses
2.6.30. MONOCYTE-ACTIVATION to standard endotoxin solutions (Method B). The standard
TEST endotoxin stock solution is prepared from an endotoxin
reference standard that has been calibrated against the
1. INTRODUCTION International Standard, for example endotoxin standard BRP.
The monocyte activation test (MAT) is used to detect or The cut -off value is calculated using the following expression:
quantify substances that activate human monocytes or
monocytic cells to release endogenous mediators: such as x+ 35
pro-inflammatory cytokines, for example tumour necrosis
factor alpha (TNFa), interleukin-l beta (IL-l~) and mean of the 4 replicates for the responses to the
interleukin-6 (IL-6). These cytokines have a role in fever blank (Ro);
pathogenesis. Consequently, the MAT will detect the presence s standard deviation of the 4 replicates of the
of pyrogens in the test sample. The MAT is suitable, after a responses to the blank (Rol.
product -specific validation, as a replacement for the rabbit
pyrogen test. The cut -off value is expressed in units appropriate to the
Pharmaceutical products that contain non-endotoxin read-out.
pyrogenic or pro-inflammatory contaminants often show very The limit of detection (LOD) is determined using the
steep dose-response curves in comparison with endotoxin endotoxin standard curve. The LOD is the concentration of
dose-response curves. Frequently the greatest response to such endotoxin corresponding to the cut-off value. For the purpose
contaminated products is obtained with undiluted solutions of the test, the LOD is expressed as endotoxin equivalents per
of the preparations being examined or small dilutions of the millilitre.
preparations being examined. For this reason preparations
that contain or may contain non-endotoxin contaminants have 3. GENERALPROCEDURE
to be tested at a range of dilutions that includes minimum A solution of the preparation being examined is incubated with
dilution. a source of human monocytes or human monocytic cells, e.g.
from human heparinised peripheral bIood that is preferably
The following 3 methods are described in the present chapter.
not more than 4 h old, or a monacyte-containing fraction of
Method A. Quantitative test that blood, such as human peripheral blood mononuclear cells
Method B. Semi-quantitative test (PBMe) isolated, e.g. by density-gradient centrifugation, or
Method C. Reference lot comparison test a human monocytic cellline. Human heparinised peripheral
blood is usually diluted with culture medium or saline e.g. to
The test is carried out in a manner that avoids pyrogen 2-50 per cent V/V (final concentration). PBMC ar monocytic
contamination. celllines, in culture medium and with either the donor's own
2. DEFINITIONS plasma or AB serum, are typically used at a final cell density
of 0.1-1.0 x 10 6 cells per well, tube or other receptacle. For
The maximum valid dilutíon (MVD) is the maximum monocytic celllines, heat-inactivated foetal bovine serum
allowable dilution of a sample at which the contaminant limit may be substituted for AB serum. The cel! culture is carried
can be determined. Determine the MVD using the following out at 37 ± 1 oc, in an atmosphere appropriate for the culture
expression: medium, e.g. 5 per cent CO 2 in humidified airo The duration
CLCxC of the culture is sufficient to allow accumulation of the
LOD chosen read-out. The responses of the chosen read-out, e.g. a
pro-inflammatory or pyrogenic cytokine, to a solution of the
CLC contaminant limit concentration; preparation being examined are compared with responses to
standard endotoxin or to a reference 10t of the preparation
C concentration of test solution; being examined. The chosen read-out method is calibrated
LOD limit of detection. using the appropriate standard.

The acceptance criterio n for a passlfail decision is the 4. APPARATUS


contaminant limit concentration (CLC), which is expressed in Depyrogenate all glassware and other heat-stable apparatus
endotoxin equivalents per milligram or millilitre, or in units in a hot-air oven using a validated process. A commonly
of biological activity of the preparation being examined. used minimum time and temperature is 30 min at 250 oc. lf
The CLC is calculated using the following expression: employing pI as tic apparatus, such as microtitre pI ates and
pipette tips for automatic pipetters, use apparatus shown to be
J(
free of detectable pyrogens and which do not interfere with
M the test.

K threshold pyrogenic dose of endotoxin per 5. CELL SOURCES AND QUALIFICATION


kilogram of body mas s ; 5-1. WHOLE BLOOD
M maximum recommended bolus dose of product Whole blood is obtained from single donors or from pooled
per kilogram of body mass. whole blood which are qualified according to the requirements
described under section 5-3 and section 5-4, respectively.
When the product is to be injected at frequent intervals
or infused continuously, M is the maximum total dose 5-2. PERIPHERAL BLOOD MONONUCLEAR CELLS (PBMC)
administered in a single hom periodo PBMC are isolated from blood obtained from single donors
or from pooled whole blood which are qualified according to
Where an endotoxin limit concentration (ELC) has been
the requirements described under section 5-3 and section 5-4,
specified for a product, the CLC is the same as the ELC,
respectively.
unless otherwise prescribed. In this case, the concentration of
test solution is expressed in mg/mL if the endotoxin limit is 5-3. QUALIFICATION OF BLOOD DONORS
specified by mass (IU Img), in Units/mL if the endotoxin limit Blood donors are to satisfy the following qualification criteria,
is specified by unit of biological activity (IU IUnit), in mL/mL together with other requirements in force that relate to
if the endotoxin limit is specified by volume (IU/mL). consent, health and safety and ethical considerations. Blood

General Notices (1) apply lo all monographs and other texts 217
2.6.30. Monocyte-activation test EUROPEAN PHARMACOPOEIA 8.0

donors are to describe themselves as being in good health, 6-1. ASSURANCE OF CRITERIA FOR THE STANDARD
as not to be suffering from any bacterial or viral infections CURVE
and to have been free from the symptoms of any such Using the standard endotoxin solution, prepare at least
infection for a period of at least 1 week prior to the donation 4 endotoxin concentrations to generate the standard
of blood. Blood donors are not to have taken non-steroidal curve. Perform the test using at least 4 replicates of each
anti-inflammatory drugs during the 48 h prior to donating concentration of standard endotoxin.
blood and steraidal anti-inflammatory drugs during the 7 days
prior to donating blood. Individuals who have been prescribed The basal releas e of the chosen read-out (blank) in the absence
immunosuppressant or other drugs known to influence the of added standard endotoxin is to be optimised to be as low
production of the chosen readout are not to serve as blood as possible.
donors. Blood donation are to be tested for infection markers
There are 2 acceptance criteria for the standard curve:
according to national requirements for transfusion medicine.
5-4. QUALIFICATION OF CELLS PO OLED FROM A - the regression of responses (appropriately transformed
NUMBER OF DONORS if necessary) on log dose sball be statistically significant
(p<O.Ol);
Pools (of whole blood or blood fractions, e.g. PBMC), must
consist of donations from a minimum of 4 individual donors - the regression of responses on log dose must not deviate
but preferably 8 or more donors, where practicable, taking significantly from linearity (p > 0.05). If analysis for
fram each donation an approximately equal volume of blood, a 4-parameter logistic curve is performed, then the
or cells from an approximately equal volume of blood. For observed curve must not deviate significantly fram the
the qualification of po oled cells praceed as follows: within theoretical curve as calculated by using the usual statistical
4 hours of collection of blood, generate dose- response methods (see chapter 5.3. Statistical analysis).
curves from the pool using standard endotoxin with at least
6-2. TEST POR INTERFERING PACTORS
4 geometrically diluted endotoxin concentrations, e.g. in the
range ofO.01 IU/mL to 4 IU/mL. The dose-response curves To assure the validity of the test, preparatory tests are
are to meet the 2 criteria for the standard curve described conducted to assure that the test solution do es not interfere
under sectiol1 6- L with the test. Validation of the test method is required when
any changes are made to the experimental conditions that are
5-5. QUALIFICATION OP CRYO-PRESERVED CELLS likely to influence the result of the test. Using an apprapriate
The ceH source intended for use in a MAT, e.g. human whole diluent, dilute the preparation to be examined in geometric
blood, blood fractions, such as PBMC or monocytic cell steps, with all dilutions not exceeding the MVD. Make the
lines, may be cryo-preserved. Pools of cryo-preserved ceUs same dilutions of the preparation to be examined and add
are obtained by pooling before freezing, 01' by pooling single endotoxin at a concentration equal to or near the middle of
cryo-preserved donations immediately after thawing. Pools the standard curve (Method A) or equal to twice the LOD
must consist of donations from a minimum of 4 individual (Method B), or use a diluent containing added endotoxin at
donors but preferably 8 or more donors where practicable, a concentration equal to or near the middle of the standard
taking fram each donation an approximately equal volume of curve (Method A) or equal to twice the LOD (Method B).
blood, or cells from an approximately equal volume of blood. Test these dilution series in parallel in the same experiment.
Qualification of cryo-preserved blood or cells is performed Use the standard curve to calculate the concentration of
immediately after thawing (and pooling if necessary): endotoxin-equivalents in each solution. Calculate the mean
dose-response curves for cryo-preserved blood or cells are to recovery of the added endotoxin by subtracting the mean
comply with the 2 criteria fol' the standard curve as described concentration of endotoxin equivalents in the solution (if any)
undel' section 6-1. from that in the solutiol1 containing the added endotoxin. The
test solution is considered free of interfering factors if, under
5-6. MONOCYTIC CONTINUOUS CELL LINES
the conditions of the test, the measured endotoxin equivalents
A human monocytic cellline is continuously cultured in order in the test solution to which endotoxin is added is within
to warrant a sufficient supply fOl" the MAT. To optimise the 50-200 per cent of the added concentration, after subtraction
method, clones derived fram the cellline can be used. of any endotoxin equivalents detected in the solution without
added endotoxin. When this criterion is not met, Method C is
Cells must be maintained under aseptic conditions and to be preferred over Methods A and B.
regularly tested for the presence of mycoplasma contamination.
Additionally, cells must be regularly checked for identity (e.g. In Method C, a solution of the preparation being examined
doubling time, morphology, and funcHon) and stability. The is tested at 3 dilutions: the highest concentration (lowest
functional stability of a cellline is assessed by monitoring its dilution) that stimulates the greatest release of the chosen
performance in relation to the number of passages during read-out and the 2-fold dilutions immediately below and
routine testing. Criteria for functional stability are to be aboye the chosen dilution. Since the concentration that
established and may include grawth criteria, maximum signal stimulates the greatest releas e of the chosen read-out
obtained in the test, background noise and receptor expression. may be donor-dependent as well as batch-dependent, the
The receptor expression may be tested with specific ligands product -specific validation is to be performed in at least
e.g. lipopolysaccharide (LPS) for toll-like receptor 4 (TLR4), 3 independent tests, each using cells from different donors.
lipoteichoic acid (LTA) for toll-like receptor 2 (TLR2), The highest concentration (lowest dilution) that stimulates
synthetic bacteriallipoprotein for TLR2-TLRl or synthetic the greatest release of the chosen read-out in the majority of
bacteriallipoprotein for TLR2-TLR6. danors, and the 2-fold dilutions immediately below and aboye
that dilution are deemed to be validated for further testing.
rf undiluted test solution stimulates the greatest release of
the chosen read-out, subsequent testing is to be performed
6. PREPARATORY TESTING using undiluted test solution and also test solution diluted
in the ratios 1:2 and 1:4 before its addition to the PBMC.
To ensure both the precision and validity of the test, The 3 dilutions to be used in subsequent testing are not to
preparatory tests are conducted, to assure that the criteria for exceed the MVD; the dilution factors for these 3 solutions
the standard curve are satisfied, that the solution does 110t are designatedfl'h andj,. Following the product-specific
interfere with the test, that the test detects endotoxins and validation, the test is routinely performed with ceUs from
non-endotoxins contaminants and that the solution do es 110t 4 individual donors or a single pool or with cells from
interfere in the detection system. 1 passage of a human monocytic cellline.

218 See the information section on general monographs (caver pages)


EUROPEAN PHARMACOPOEIA 8.0 2.6.30. MOllocyte-activation test

6-3. METHOD VALIDATION FOR NON-ENDOTOXIN 7-1-2. Calculation and interpretation


MONOCYTE-ACTIVATING CONTAMINANTS Al! data to be included in the data analysis are to relate to ceUs
The preparatory testing is also to show that the chosen for which the 2 criteria for the standard curve are satisfied.
test system detects, in addition to bacterial endotoxins, The endotoxin equivalents recovery calculated from the
non-endotoxin pro-inflammatory or pyrogenic contaminants. endotoxin equivalents concentration found in solution D after
This can be achieved using historie batches that have been subtracting the endotoxin equivalents concentration found
found to be contaminated with non-endotoxin contaminants in solution A, is within the range of 50-200 per cent. For
that caused positive responses in the rabbit pyrogens test or each different cell source, e.g. individual donation, donor
adverse drug reactions in mano Where such batches are not pool, 01' cellline, use the endotoxin standard curve R¡"R4 to
available, the preparatory testing is to inelude validation of calculate the concentration of endotoxin equivalents in each
the test system using specific ligands for toU-like receptors, of the replicates of solutions A, B and C. The preparation
e.g. peptidoglycans, lipoteichoic acids or synthetic bacterial being examined complies with the requil'ements of the test for
lipoproteins. a given cell source if the mean COl1centrations of endotoxin
6-4. INTERFERENCE IN THE DETECTION SYSTEM equivalents measured in the replicates of solutions A, B and C,
Once the optimum dilution of the solution of the preparation after correction for dilution and concentration, are aUless
being examined for further testing has been identified, this than the CLC specified for the preparation being examined.
dilution is tested for interference in the detection system 7-1-3. Pass/fail criteda ofthe preparation
(e.g. ELISA) for the chosen read-out. The agreement between When cells from individual donors are used, the preparation
a dilution series of the standard for the chosen read -out, in being examined is required to comply with the test with the
the presence and absence of the test preparation, is to be ceUs from each of 4 different donors. If the preparation being
within ± 20 per cent. examined passes the test with cells from 3 of the 4 donors
(l donor excluded for failing to comply with test performance
7. METHODS
criteria or showing a positive reaction), the test is continued
with cells from a further 4 donors, non e of whom provided
7-1. METHOD A: QUANTITATIVE TEST ceUs for the 1,t test, and the preparation being examined is
Method A involves a comparison of the preparation being required to pass the test with cells from 7 of the 8 different
examined with a standard endotoxin dose-response curve. donors (i.e. a maximum of 1 positive reaction in 8 donors
The contaminant concentration of the preparation being is aUowed). When the source of monocytes consists of cells
examined is to be less than the CLC to pass the test. pooled from a number of individual donoIs, the preparation
being examined is required to pass the test with 1 pool of cells.
7-1-1. Test procedure
Where a human monocytic cellline is used for the test, the
Using the validated test method, prepare the solutions shown preparation being examined is required to pass the test with
in Table 2.6.30.-1 and culture 4 replicates of each solution with 1 passage of the cellline.
ceUs from each of 4 individual donol's ol' a single pool or with 7-2. METHOD B. SEMI-QUANTITATIVE TEST
cells from 1 passage of a human monocytic ceUline. Method B involves a comparison of the preparation being
examined with standard endotoxin. The contaminant
Table 2.6.30.-1 concentration of the test preparation it to be less than the CLC
Added to pass the test. Solution A must be chosen for the release
Number of decision, unless otherwise justified and authorised.
Solution Solution endotoxin
replicates
(IV/mL)
7-2-1. Test pro ce dure
A Test solution/f None 4
Using the validated test method, prepare the solutiol1s shown
B Test solution/2 x f None 4 in Table 2.6.30.-2 and culture 4 replicates of each solution with
cells from each of 4 individual donors or a single pool or with
e Test solution/4 x f None 4
ceUs from 1 passage of a human monocytic cellline.
Middle dose
D Test solution/f
from endotoxin 4 Table 2.6.30.-2
stan dard curve
(R) Added
enaotoxin Number of
Pyrogen-free Solution Solutiol1
None (negative replicates
Ro saline or test 4 (IV/rnL)
control)
diluent A Test solution/f None 4
Pyrogen -free 4 concentrations
4 of each B Test solution/f, None 4
R]-R, saline or test of standard
concentration
diluent endotoxin
e Test solution/j, None 4
Solution A == Solution of the preparation being examined at Standard
the dilution, here designated 1, at which the test for interfering D Test solution/f
endotoxin at 4
factors was carried out, i.e. the highest concentration 2 x LOD for the
test system
(Iowest dilution) for which the endotoxin recovery is within
Standard
50-200 per cent. endotoxin at
E Test solution/}; 4
2 x LOD for the
Solution B == 2-fold dilution of solution A, not exceeding the test svstem
MVD.
Standard
Solution C == 2-fold dilution of solution B, not exceeding the endotoxin at 4
F Test solution/J,
2 x LOD for the
MVD. test system
Solution D == solution A spiked with standard endotoxin: the Pyrogen -free
None (negative 4
middle dose from endotoxin standard curve (R 3 ). Ro saline or test
control)
dilnent
Solution Ro == negative control. Pyrogen-free
Standard
endotoxin al
R] saline or test 4
Solutions R¡-R4 == solutions of standard endotoxin at the diluent
0.5 x LOD for
concentrations used in the test for interfering factors. the test system

General Natices (1) apply ta all managraphs and ather texts 219
2.6.30. Monocyte-activation test EUROPEAN PHARMACOPOEIA 8.0

Pyrogen -free
Standard MVD to overcome the interference because it contains or is
endotoxin at believed to contain non-endotoxin contaminants. Responses
R2 saline or test 4
1 x LOD for the
diluent
test system
to non-endotoxin contaminants may dilute out more rapidly
than responses to endotoxin, which makes it necessary to
Standard
Pyrogen -free
endotoxin at
perform the test at a range of dilutions that indude minimum
R3 saline or test 4 dilution. The test procedure is described below and in dudes
2 x LOD for the
diluent
test system an example for the comparison of a test lot with the reference
Standard 10t.
Pyrogen -free
endotoxin at
R4 saline or test 4 x LOD for the
4 7-3-1. Test procedure
diluent
test system Using the validated test method, prepare the solutions shown
Solution A = solution of the preparation being examined at in Table 2.6.30.-3 and culture 4 replicates of each solution with
the dilution, here designated f, at which the test for interfering cells from each of 4 individual donors or a single pool or with
factor s was completed. cells from 1 passage of a human monocytic cellline.
Solution B = solution of the preparation being examined at a Table 2.6.30.-3
dilution, here designated};, not exceeding the MVD, chosen
after a review of data from the product-specific validation, e.g. Solutionl di/utioll
Solution Number of replicates
factor
1:2 x MVD (i.e. a 2-fold dilution aboye the MVD).
Solution of reference
A 4
Solution C = solution of the preparation being examined at a lot/~
dilution, here designated};, not exceeding the MVD, chosen Solution of reference
B 4
after a review of data from the product -specific validation, lotlt;
e.g. MVD. Solution of reference
e lotlj,
4
Solution D = solution A spiked with standard endotoxin at
2 x LOD for the test system (as determined in preparatory Solution of test
D 4
preparatiol1/{¡
testing).
Solution of test
Solution E = solution B spiked with standard endotoxin at E 4
preparation/f,
2 x LOD fo1' the test system. Solution of test
F 4
Solution F = solution C spiked with standard endotoxin at preparationlj;
2 x LOD for the test system. G
Positive control
4
(standard endotoxin)
Solution Ro = negative control.
Diluent (negative
Ro 4
Solution R¡ = standard endotoxin at 0.5 x LOD for the test control)
system.
Solutions A, B and C are solutiol1s of the reference 10t diluted
Solution R2 = standard endotoxin at 1 x LOD for the test by the dilution factors, 11' 12 and jv determined in the test for
system. interfering factors.
Solution R, = standard endotoxin at 2 x LOD for the test Solutions D, E and F are solutions of the preparation
system. being examined diluted by the dilutiol1 faetors,!1' 12 and 13'
Solution R4 = standard endotoxin at 4 x LOD for the test determined for the reference lot in the test for interfering
system. factors.
7 -2- 2. Calculation and interpretation Solution G is the positive test control for the viability of the
AH data to be induded in the data analysis are to relate to cells and is a standard endotoxin concentration that gives a
cells for which mean responses to solutions Ro-R4 in crease dear positive response.
progressively. The mean response to Ro may be equal to Solution Ro is the diluent used to dilute the preparation being
the mean response to R¡. For each different ceH source, examined and serves as the test blank.
e.g. individual donation, donor pool, or cellline, the mean
7-3-2. Calculation and interpretation
response to solution R2 is to be greater than a positive cut-off
value. Data below this cut-off value are considered negative. All data to be included in the data analysis are to relate to
rf the mean response to R¡ or R2 exceeds the cut -off value, cells for which solution G and at least one of solutions A, B
the response to the solution chosen for the passlfail decision and C give a response that is greater than the basal releas e of
must be negative (= pass). For each negative solution of the the read-out (Solution Ro)' For each different ceH source, e.g.
test preparation (A, B and C), the mean response to the individual donation, donor pool, or cellline, use the standard
corresponding spiked solution (D, E or F respectively) is curve for the read-out (a calibration curve in duplicate with
compared with the mean response to R, to determine the a blank and at least 4 geometrically diluted concentrations
percentage spike recovery. The contaminant concentration of the standard for the chosen read-out) and calculate the
of the preparation being examined is less than the CLC for mean responses of the replicates of solutions A-F. Sum the
a given cell source if the solution of the test preparation mean responses to solutions A, B and C and sum the mean
designated for the pass/fail-decision and the dilutions below responses to solutions D, E and F. Divide the sum of the mean
all give negative results and the endotoxin spike recovery is responses to solutions D, E and F by the sum of the mean
within the range of 50-200 per cent. responses to solutions A, B and C. The preparation being
7-2-3. Pass/fan criteria ofthe preparation examined complies with the test for a given ceH source if the
resulting value complies with a defined acceptance criterion
The criteria are the same as for method A (see 7-1-3). not exceeding 2.5.
7-3. METHOD C: REFERENCE LOT COMPARISON TEST 7 -3-3. Pass/faiJ criteria of the preparation
Method C involves a comparison of the preparation being
The criteria are the same as for method A (see 7-1-3).
examined with a validated reference lot of that preparation.
The reference lot is selected according to criteria that have To quantify more dosely the level of contamination,
been justified and authorised. The test is intended to be Methods A, B and e may be performed using other dilutions
performed in cases where a preparation being examined of the solution of the preparation being examined not
shows marked interference but cannot be diluted within the exceeding the MVD.

220 See the in10rmation section on general mOl1ographs (caver pages)


EUROPEAN PHARMACOPOEIA 8.0 2.6.30. Monocyte-activation test

The following section is published for information only. Where an endotoxin limit concentration (ELC) has been
specified for a product, the CLC is the same as the ELC,
unless otherwise prescribed. The CLC is expressed in terms
Guidance notes of endotoxin equivalents. The CLC is calculated using the
1. INTRODUCTION following expression:
The monocyte activation test (MAT) is primarily intended K
to be used as an alternative method to the rabbit pyrogen M
test. The MAT detects pyrogenic and pro-inflammatory
contaminants, including endotoxins from gram-negative K threshold pyrogenic dose of endotoxin per
bacteria and 'non -endotoxin' contaminants, including kilogram of body mass;
pathogen-associated molecular patterns (PAMPs), derived IvI maximum recommended bolus dose of product
from gram-positive and gram-negative bacteria, virus es and per kilogram of body mass.
fungi, and product-related and process-related biological or When the product is to be injected at frequent intervals
chemical entities. or infused continuously, M is the maximum total dose
Since non-endotoxin contaminants are a physico-chemically administered in a single hour periodo
diverse class of molecules, and usually the nature of The endotoxin limits depends on the product and its route of
the contaminant in a preparation being examined is administration and is stated in monographs.
unknown, the leve! of contamination is expressed either in
Values fOl' K are suggested in Table 2.6.300-4.
endotoxin-equivalent units, derived by comparison with
responses to standard endotoxin, or by comparison with a Table 2.6.30.-4
reference lot of the test preparation. Route of administration K (IV of endotoxin per kilogram
ofbody mass)
In the MAT, responses to standard endotoxin usually dilute lntravenous 5.0
out over approximatively 1 10g¡O and responses to products
contaminated with non-endotoxin contaminants (alone lntravenous, for radiopharmaceuticals 2.5
or in combination with endotoxins) often show very steep
Intrathecal 0.2
dose-response curves, usually over only 1 or 2 dilution steps
when tested for their capability to stimulate monocytes.
For other routes, the acceptance criterion for bacterial
Frequently, the largest response to 5uch contaminated
endotoxins is generally determined on the basis of results
products is obtained with undiluted solutions of preparations
obtained during the development of the preparation.
being examined or small dilutions of the preparations being
examined. For this reason test solutions of preparations 2-3. INFORMATION REGARDING CRYO-PROTECTANTS
being examined that contain or may contain non-endotoxin The influence of cryo-protectants, eog. dimethyl
contaminants have to be tested at a range of dilutions that sulfoxide (DMSO), and their residues in thawed cells, is to be
includes minimum dilution. considered: DMSO is toxic to cells in culture and, even when
cells have been washed thoroughly, cryo-preservation may
2. METHODS have altered cell properties, e.g. cell membrane permeability.
2-1. INFORMATION REGARDING THE CHOICE OF 2-4. INTERFERENCE TESTING
METHODS Where practicable, interference testing is performed 011 at
Methods A, B and C, are not normally applied where a test least 3 different 10ts of the preparation being examined.
preparation has the intrinsic activity of stimulating the Preparations being examined that show marked batch-to-batch
release of the chosen read-out or where the test preparation variation, that effectively renders each batch unique for
is contaminated with the chosen read-out. In both cases, this the purposes of interference testing, are to be subjected
fact is to be addressed by modifying and validating the chosen to interference testing within each individual test, i.e.
method accOl'dinglyo The product-specific validation of the concomitant validation.
chosen method would be expected to identify the frequency Interference testing is preferably performed 011 batches of the
of non-responders to a particular product/contaminant(s) preparation being examined that are free of en do toxin s and
combination and to identify steps to address this, e.go other pyrogenic/pro-inflammatory contaminants and, where
screening of donors, increasing the number of donors per this is not practicable, none of the batches are to be heavily
test, and setting pass/fail criteria of appropriate stringency to contaminated. If only 1 batch is available the validation has to
maximise the likelihood of detecting contaminated batches. be performed on that batch in 3 independent tests. Precision
Methods A and B are appropriate when responses to dilutions parameters for reproducibility, e.g. ± 50 per cent, are to be
of a preparation being examined are broadly parallel to fulfilled.
responses to dilutions of standard endotoxin. Method B is a
semi-quantitative test that can also be applied when responses 30 REPLACEMENT OF THE RABBIT PYROGEN TEST BY
to dilutions of a test preparation are not parallel to responses THE MONOCYTE ACTIVATION TEST
to dilutions of standard endotoxin. As noted aboye, the monocyte activation test (MAT) is
Method C, the reference lot comparison test, was developed primarily intended to be used as an alternative method to the
to address extreme donOl' variability in responses to certain rabbit pyrogen test. Monographs on pharmaceutical products
product/contaminant(s) combinationso In this regard, intended for parenteral administration that may contain
it should be noted that, while monocytes from most pyrogenic contaminants require either a test fOl' bacterial
donors respond in a broadly similar manner to bacterial endotoxins or a monocyte activation test.
endotoxin, responses of monocytes from different donors As a general policy:
to non -endotoxin contaminants can differ markedly, so - in any individual monograph, when a test is required, only
that it is possible to identify non-responders to certain 1 test is included, either that for bacterial endotoxins or
product/contaminant(s) combinations. the MAT. Before including the MAT in a monograph,
2-20 CALCULATION OF CONTAMINANT LIMIT evidence is required that 1 of the 3 methods described
CONCENTRATION in the MAT chapter can be applied satisfactorily to the
The acceptance criterion for a pass/fail decision is the product in question;
contaminant limit concentration (CLC), which is expressed in - the necessary information is sought from manufacturers.
endotoxin equivalents per milligram or millilitre or in units Companies are invited to provide any validation data
of biological activity of the preparatíon being examined. that they have concerning the applicability of the MAT

General No/ices (1) apply to all monographs and other texts 221
2.6.31. Mkrobiological examination ofherbal products and extracts EUROPEAN PHARMACOPOEIA 8.0

to the substances and formulations of interest. Such data microbiological quality. When used for such purposes, follow
include details of sample preparation and of any procedures the instructions given belovv, including the number of samples
necessary to eliminate interfering factors. In addition, any to be taken, and interpret the results as stated below.
available parallel data for rabbit pyrogen testing that would Alternative microbiological procedures, including automated
contribute to an assurance that the replacement of a rabbit methods, may be used, provided that their equivalence to the
pyrogen test by the MAT is appropriate, is to be provided. Pharmacopoeia method has been demonstrated.
2-2. GENERAL PROCEDURES
4. VALIDATION OF ALTERNATIVE METHODS
The preparation of samples is carried out as described in
Replacement of a rabbit pyrogen test by a MAT, or replacement general chapter 2.6.12.
of a method for detecting pro-inflammatory/pyrogenic lf the product to be examined has antimicrobial activity, this
contaminants by another method, is to be regarded as is as far as possible removed or neutralised as described in
the use of an alternative method in the replacement of a general chapter 2.6.12.
pharmacopoeial test, as described in the General N otices:
If surface-active substances are used for sample preparation,
'The test and assays described are the official methods upon their absence of toxicity for micro-organisms and their
which the standards of the European Pharmacopoeia are compatibility with inactivators used must be demonstrated as
based. With the agreement of the competent authority, described in general chapter 2.6.12.
alternative methods of analysis may be used for control 2-3. GROWTH-PROMOTING AND INHIBITORY
purposes, provided that the methods used enable an PROPERTIES OF THE MEDIA, SUITABILITY OF THE TEST
unequivocal decision to be made as to whether compliance AND NEGA TIVE CONTROLS
with the standards of the monographs would be achieved if the
official methods were used. In the event of doubt or dispute, The ability of the test to detect micro-organisms in the
the methods of analysis of the European Pharmacopoeia are presence of the product to be examined must be established.
alone authoritative: Suitability must be confinned if a change in testing
performance, or the product, which may affect the outcome of
The following pro ce dures are suggested for validating a the test is introduced.
method for the MAT other than the one indicated in the 2-3-1. PREPARATION OF TEST STRAINS
monograph:
Use standardised stable suspensions of test strains or prepare
- the procedure and the materials and reagents used in them as stated below. Seed lot culture maintenance techniques
the method should be validated as described for the test (seed-Iot systems) are used so that the viable micro-organisms
concerned; used for inoculation are 110t more than 5 passages removed
- the presence of interfering factors (and, if needed, the from the original master seed lot.
procedure for removing them) should be tested 011 samples 2-3-1-1. Aerobic micro-organisms. Grow each of the
of at least 3 production batches. bacterial test strains separately in casein soya bean digest broth
or on casein soya bean digest agar at 30-35 oC for 18-24 h.
MKf should be applied to al! new products intended for
parenteral administration that have to be tested for the - Staphylococcus aureus such as ATCC 6538, NCIMB 9518,
presence of monocyte-activating contaminants according to CIP 4.83 or NBRC 13276;
the requirements of the European Pharmacopoeia. - Pseudomonas aeruginosa such as ATCC 9027, NCIMB 8626,
CIP 82.118 or NBRC 13275;
- Escherichia coli such as ATCC 8739, NCIMB 8545,
CIP 53.126 or NBRC 3972;
Ol/2014:20631 - Salmone/la enterica subsp. enterica serovar Typhimurium
such as ATCC 14028 or, as an alternative, S. enterica subsp.
enterica serovar Abony such as NBRC 100797, NCTC 6017
2.6.31. MICROBIOLOGICAL or CIP 80.39;
EXAMINATION OF HERBAL - Bacillus subtilis such as ATCC 6633, NCIMB 8054,
CIP 52.62 or NBRC 3134.
MEDICINAL PRODUCTS FOR ORAL
Use buffered sodium chloride-peptone solution pH 7.0 or
USE AND EXTRACTS USED IN THEIR phosphate buffer solution pH 7.2 to make test suspensions.
PREPARATION Use the suspensions within 2 h, or within 24 h if sto red at
2-8 oc. As an alternative to preparing and then diluting a
1. MICROBIAL ENUMERATION TESTS fresh suspension of vegetative cells of B. subtilis, a stable spore
suspension is prepared and then an appropriate volume is
Total aerobic microbial count (TAMC). Perform as used for test inoculation. The stable spore suspension may be
described in general chapter 2.6.12. maintained at 2-8 oC for a validated period of time.
Total combined yeasts/moulds count (TYMC). Perform as 2-3-2. NEGATIVE CONTROL
described in general chapter 2.6.12. Due to the natural high
bioburden in the products covered by general chapter 5.1.8, To verify testing conditions, a negative control is performed
use of Sabouraud-dextrose agar containing antibiotics is using the chosen diluent in place of the test preparation. There
suitable. must be 110 growth of micro-organisms. A negative control
is also performed when testing the products as described in
section 2-4. A failed negative control requires an investigation.
2. TEST FOR SPECIFIED MICRO-ORGANISMS
2-3-3. GROWTH-PROMOTING AND INHIBITORY PROPERTIES
2-1. INTRODUCTION
OFTHEMEDIA
The tests described hereafter will allow determination of the
absence or limited occurrence of specified micro-organisms Test each batch of ready-prepared medium and each batch of
that may be detected under the conditions described. medium prepared either from dehydrated medium or from
The tests are designed primarily to determine whether a the ingredients described.
product, substance or preparation (hereinafter referred to as Verify suitable properties of relevant media as described in
'the product') complies with an established specification for Table 2.6.31.-1.

222 See the information scction on general monographs (cover pages)


EUROPEAN PHARMACOPOEIA 8.0 2.6.31. Microbiologkal examination ofherbal products and extracts

Table 2.6.31.-1. - Growth-promoting, inhibitory and indicative the prescribed growth medium (casein soya bean digest
properties of media broth or buffered peptone medium). For the enumeration
method for bile-tolerant gram-negative bacteria, inoculate
Medium Pl'operty Test strains
E. coli and P. aeruginosa individually. For the tests for E. coli
S. (/ureus
Casein soya bean Growth and Salmonella, inoculate the specified micro-organism
P. ¡¡eruginosa individually.
digest broth promoting
B. subtilis
Test for E. coli Any antimicrobial activity of the product necessitates a
Growth
bile-tolerant Enterobacteria modification of the test procedure (see section 4-5-3 of general
promoting P. aeruainosa
gran1-negative enrichment
broth -Mossel
chapter 2.6.12).
bacteria Inhibitory S. aureus
If for a given product the antimicrobial activity with respect
Growth E. coli
Violet red bile
promoting to a micro-organism for which testing is prescribed cannot
glucose agar P. aeruginosa be neutralised, then it is to be assumed that the inhibited
+ indicative
S. aureus micro-organism will not be present in the product.
Casein soya bean Grov.c:h
P. aeruginosa
digest broth promoting 2-3-4-1. Test for absence. Use a l1umber of micro-organisms
B. subtilis
equivalent to not more than 100 CFU in the inoculated test
Growth
Test for promoting
E. coli preparation. Perform the test as described in the relevant
MacConkey broth
Escherichia coli paragraph in section 2-4 using the shortest incubation period
Inhibitory S. aureus
prescribed. The specified micro-organisms must be detected
Growth with the indication reactions as described in section 2-4.
MacConkey agar promoting E. coli
+ indicative 2-3-4-2. Enumeration test. Semi-quantitative test
S. ellterica subsp. (probable-number method).
ente rica serovar
Buffered peptone Growth Typhimurium 01' Use a number of micro-organisms equivalent to not more
ll1ediUlTI promoting S. enterica subsp. than 100 CFU per gram or millilitre of product. Perform
enterica serovar the test as described in the relevant paragraph in section 2-4
Abony
using the shortest incubation period prescribed. The dilution
S. enterica subsp.
enterica serovar corresponding to 0.1 g or 0.1 mL of product must be positive.
Rappaport Growth Typhimurium 2-4. TESTING OF PRODUCTS
Vassiliadis promoting or S. enterica
Test for Salmollella 2-4-1. BILE- TOLERANT GRAM -NEGATIVE BACTERIA
Salmonella subsp. enterica
enrichment broth serovar Abony
2-4-1-1. Enumeration test. Semi-quantitative test
lnhibitory (probable-number method).
S. aureus
S. ente rica subsp. 2-4-1-1-1. Sample preparation and pre-incubation. Prepare
entedea serovar
Xylose. lysine.
Growth
Typhimurium
a sample using a lO-fold dilution of not less than 1 g of the
promoting product to be examined as described in general chapter 2.6.12,
deoxycholate agar or S. enterica
+ indicative
subsp. enterica but using casein soya bean digest broth as the chosen diluent,
serovar Abony mix and incubate at 20-25 oC for a time sufficient to resuscitate
Test for gl"owth-promoting properties, liquid media: the bacteria but not sufficient to encourage multiplication of
inoculate a portion of the appropriate medium with a the organisms (2-3 h).
small number (not more than 100 CFU) of the appropriate 2-4-1-1-2. Selection and subculture. Inoculate suitable
micro-organismo Incubate at the specified temperature for quantities of enterobacteria enrichment broth-Mossel with
110t more than the shortest period of time specified in the the preparation as described aboye and/or, depending 011
test. Incubate the casein soya bean digest broth at 30-35 oC the limit applied for the particular product, with 3 of the 4
for not more than 3 days. Clearly visible growth of the dilutions of the preparation, which contain respectively 0.1 g,
micro-organism comparable to that obtained with a previously 0.01 g, 0.001 g and 0.0001 g (or 0.1 mL, 0.01 mL, 0.001 mL
tested and approved batch of medium occurs. and 0.0001 mL) of the product to be examined. Incubate at
30-35 oC for 24-48 h. Sub culture each of the cultures on a plate
Test for growth-promoting properties, solid media: perform
of violet red bile glucose agar. Incubate at 30-35 oC for 18-24 h.
the surface-spread method, inoculating each plate with a
small number (not more than 100 CFU) of the appropriate 2-4-1-1-3. Interpretation. Growth of colonies constitutes a
micro-organismo Incubate at the specified temperature for not positive result. Note the smallest quantity of the product that
more than the shortest period of time specified in the test. gives a positive result and the largest quantity that gives a
Growth of the micro-organism comparable to that obtained negative resulto
with a previously tested and approved batch of medium occurS. Determine from Table 2.6.31.-2 the probable number of
Test for inhibitory properties, liquid or salid media: bacteria.
inoculate the appropriate medium with at least 100 CFU of Table 2.6.31.-2. - Interpretation oÍ results
the appropriate micro-organismo Incubate at the specified
temperature for not less than the longest period of time Results for each quantity of product Probable
number of
specified in the test. No growth of the test micro-organism bacteria
occurs. 0.1 g 01' 0.01 g or 0.001 g or 0.0001 g 0'- per gram
0.1 mL 0.01 mL 0.001 mL 0.0001 mL millilitre
01'
Test for indkative properties: perform the surface-spread ofproduct
method, inoculating each plate with a small number (not
more than 100 CFU) of the appropriate micro-organismo + + + + > !O'
Incubate at the specified temperature for a period of time < lO' and
within the range specified in the test. Colonies are comparable + + +
> !O'
in appearance and indication reactions to those obtained with
< lO' and
a previously tested and approved batch of medium. + + -
> 10'
2-3-4. SUITABILITY OF THE TEST METHOD
< 10 2 and
+
For each product to be examined, perform the sample >10
preparation as described in the relevant paragraph in - <10
section 2-4. Add each test strain at the time of mixing, in

General Notices (1) apply to all monographs and other texts 223
2.6.33. Residual pertussis toxin and irreversibility of pertussis toxoid EUROPEAN PHARMACOPOEIA 8.0

2-4-2. ESCHERICHIA COL! The product complies with the test if colonies of the types
2-4-2-1. Test for absence described are not present or if the identification tests are
negative.
2-4-2-1-1. Sample preparation and pre-incubation. Prepare
a sample using a lO-fold dilution of not less than 1 g of the The following section is given for information.
product to be examined as described in general chapter 2.6.12, RECOMMENDED SOLUTIONS AND CULTURE MEDIA
and use 10 mL or the quantity corresponding to 1 g or 1 mL The solutions and culture media mentioned in this chapter
to inoculate a suitable amount (determined as described in and described in general chapter 2.6.13 and the following
section 2-3-4) of casein soya bean digest b1'oth, mix and buffered peptone medium have been found to be satisfactory
incubate at 30-35 oC for 18-24 h. for the purposes for which they are prescribed in this chapter.
2-4-2-1-2. Selection and subculture. Shake the container, Other media may be used provided that their suitability can
transfer 1 mL of casein soya bean digest broth to 100 mL be demonstrated.
of MacConkey broth and incubate at 42-44 oC for 24-48 h. Buffered peptone medium
Subculture on a plate of MacConkey agar at 30-35 oC for Potassium dihydrogen phosphate l.5 g
18-72 h.
Disodium hydrogen phosphate dodecahydrate 9.0 g
2-4-2-1-3. Interpretation. Growth of colonies indicates the
possible presence of E. eoli. This is confirmed by identification Sodium chloride 5.0 g
tests.
Peptone 10.0 g
The product complies with the test if no colonies are present
01' if the identification tests are negative. Purified water 1000 mL

2-4-2-2. Enumeration test. Semi-quantitative test


(probable-number method). Adjust the pH so that after sterilisation it is 7.0 ± 0.2 at 25 oc.
Sterilise in an autoclave using a validated cycle.
2-4-2-2-1. Sample preparation and pre-incubation. Prepare
a sample using a 10-fold dilution of not less than 1 g of the
product to be examined as described in general chapter 2.6.12, 0712013:20633
and use the quantities corresponding respectively to 0.1 g,
0.01 g and 0.001 g (or 0.1 mL, 0.01 mL and 0.001 mL) to 2.6.33. RESIDUAL PERTUSSIS TOXIN
inoculate a suitable amount (determined as described in
section 2-3-4) of casein soya bean digest broth, mix and
AND IRREVERSIBILITY OF PERTUSSIS
incubate at 30-35 oC for 18-24 h. TOXOID
2-4-2-2-2. Selection and subculture. Shake the container, This test is not necessary for the product obtained by genetic
transfer 1 mL of casein soya bean digest broth to 100 mL modification.
of MacConkey broth and incubate at 42-44 oC for 24-48 h.
Pertussis toxin BRP or an in-house toxin preparation calibrated
Sub culture on a plate of MacConkey agar at 30-35 oC for
in International Units against the BRP are suitable for use as
18-72 h.
a reference pertussis toxin preparation.
2-4-2-2-3. Interpretation. Growth of colo ni es indicates the
f.1ouse strain. A suitable mouse strain has a toxin LDso
possible presence of E. eolio This is confirmed by identification
between 6 IU and 50 ID.
tests.
Use equal groups of not fewer than 10 histamine-sensitive
Note the smallest quantity of the product that gives a positive
l11ice of suitable strain, age and weight. For the test for residual
result and the largest quantity that gives a negative resulto
pertussis toxin, inject intraperitoneally into the 1,t group
Determine from Table 2.6.31.-3 the probable number of between 1 and 2 human doses of the vaccine stored at 2-8 oc.
bacteria. For the test for irreversibility of pertussis toxoid, inject
Table 2.6.31.-3. - Interpretation of results intraperitoneally into the 2nd group between 1 and 2 human
doses of the vaccine incubated at 37 oC for 4 weeks.
Resnlts for each qnantity of prodne! Probable
number of The histamine sensitivity criteria must be set during a
bacteria per validation study using multiple doses of a reference toxin; a
0.01 g or 0.001 g or gram or
0.1 g or 0.1 roL
0.01 mL 0.001 mL
significant dose-response must be demonstrated. A suitable
millilitre of
prodnet dos e of the reference toxin, chosen in the linear region of the
dose- response curve and giving a positive response considered
+ + + > 10 3 appropriate by the competent authority, is subsequently
+ + < 10 and > 10
3 2 included in each assay as the positive control to demonstrate
assay sensitivity.
+ - < lO' and > la
The positive control group of mice is injected with an
< 10 equivalent volume of reference pertussis toxin preparation
(e.g. pertussis toxin BRP) at adose that has been defined in
2-4-3. SALMONELLA the validation stage as demonstrating the assay sensitivity, for
2-4-3-1. Test for absence example adose causing death in at least 30 per cent of the mice.
2-4-3-1-1. Sample preparation and pre-incubation. Use 25 g or The negative control group of mice is injected intraperitoneally
25 mL of the product to be examined to inoculate 225 mL of with an equivalent volume of diluent.
buffered peptone medium and mix (e.g. homogenise in a filter lf a reference group of mice is used, it may be injected with a
bag by using a blender). Incubate at 30-35 oC for 18-24 h. reference pertussis toxin preparation (e.g. pertussis toxin BRP)
2-4-3-1-2. Selection and subculture. Transfer 0.1 mL of at adose previously set as the allowable upper limit of pertussis
buffered peptone medium to 10 mL of Rappaport Vassiliadis toxin in the product according to historical safety data.
Salmonella enrichment broth and incubate at 30-35 oC Alternatively, a reference vaccine with established clinical
for 18-24 h. Sub culture on plates of xylose, Iysine and safety may be used instead of the reference toxin preparation.
deoxycholate agaf. Incubate at 30-35 oC for 18-48 h. After 5 days, inject intraperitoneally into each mouse of each
2-4-3-1-3. Interpretation. The possible presence of Salmonel/a group the equivalent of 2 mg ofhistamine base in a volume not
is indicated bythe growth ofwell-developed, red colonies, with exceeding 0.5 mL and observe for 24 h. The test is not valid if:
or without black centres. This is confirmed by identification - 1 or more mice in the negative control group die following
tests. histamine challenge;

224 See the information section on general monographs (eover pages)


EUROPEAN PHARMACOPOEIA 8.0 2.6.33. Residual pertussis toxin and irreversibiHty of pertussis toxoid

- the histamine sensitivity does not meet the defined limit If a reference group is included, the vaccine complies with the
(e.g. at least 30 per cent of the mice die in the positive test for residual pertussis toxin if the percentage of deaths in
control group). the 1st group is not greater than that in the reference group. If
a vaccine 10t fails the test, the test may be repeated once with
If a reference group is not included in the assay, the vaccine the same number of mice or with a greater number and the
complies with the test for residual pertussis toxin if no mice results of the tests combined; the vaccine complies with the
die in the 15t group. If a vaccine 10t fails the test, the test may test if the percentage of deaths in the groups given the vaccine
be repeated once with the same number of mice or with a does not exceed the percentage of deaths in the reference
greater number and the results of the tests combined; the groups. The vaccine complies with the test for irreversibility of
vaccine complies with the test if the percentage of deaths in pertussis toxoid if the 2nd group, given the vaccine incubated
the group given the vaccine do es not exceed 5 per cent. The at 37 oC, also complies with these criteria.
vaccine complies with the test for irreversibility of pertussis Alternatively, after validation, a histamine-sensitisation test
toxoid if the 2nd group, given the vaccine incubated at 37 oC, based on body temperature measurements as end-points may
also complies with these criteria. be used instead of the letha! end-point test in mice.

General Notices (1) apply to all monographs and other texts 225
EUROPEAN PHARMACOPOEIA 8.0

226 See the information section on general monographs (ca ver pages)
EUROPEAN PHARMACOPOEIA 8,0

2.7. Biological assays


2,7, Biological assays" """,,",""""""""""""""""""""""""'" 229 2,7,18, Assay ofhuman coagulation factor IL """""""""",254
2.7, L lmmunochemical methods" """""""""""""""""""",229 2.7,19, Assay ofhuman coagulation factor X"""""""""""" 255
2,7,2, Microbiological assay of antibiotics" """"""""""""",230 2,7,20, In vivo assay ofpoliomyelitis vaceine (inactivated)" 255
2.7A, of human coagulation factor VIIL """"""""",236 2,7.21. Assay ofhuman von Willebrand factor" """"""""",257
2,7,5, Assay of heparin" """"""""""""""""""""""""""""",237 2,7,22, Assay ofhuman coagulation factor XL """""""""" 258
2,7,6, Assay of diphtheria vaccine (adsorbed)" """""""""" 237 2.7,23, Numeration of CD34/CD45+ cells in
2,7.7, Assay of pertussis vaccine (whole ceH)" '''''''" """" "'" 242 haematopoietic products" '"'''''''''''''''''''''''''''''''''''''''''''''''''' 258
2,7,8, Assay of tetanus vaccine (adsorbed)" """",,,,,,,,,,,,,,,,,,242 2.7.24, Flow cytometry, """"""""""""""""""""""""""""",259
2.7.9, Test for Fe function of immunoglobulin"""""""""", 246 2,7.25, Assay ofhuman plasmin inhibitoL ",,'"'''''''''''''''''''' 261
2,7,10, Assay of human coagulation factor VIL """"""""" 247 2.7.27, Flocculation value (Lf) of diphtheria and tetanus toxins
2,7,1 L Assay of human coagulation factor IX" """""""""" 248 and toxoids (Ramon assay)" """""""",,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,261
2,7,12, Assay of heparin in coagulation factors" """'"'',,''''' 249 2,7.28, Colony-forming ceH assay for human
2.7.13, Assay ofhuman anti-D immunoglobuliIL """"""'" 249 haematopoietic progenitor cells"""""""",,,,,,,,,,,,,,,,,,,,,,,,,,,, 262
2,7,14, Assay ofhepatitis A vaccine" """""""""""""""""",251 2,7,29, Nucleated cell count and viability, """"""",,"""""" 263
2.7,15, Assay of hepatitis B vaccine (rDNA)" """",,",,"""'" 252 2,7,30, Assay ofhuman protein c" """""""""""""""""""" 265
2,7,16, Assay of pertussis vaccine (acellular)" """"""""""" 252 2.7,31. Assay ofhuman protein S" """""""""'"'' """ """ " " " 266
2,7,17, Assay of human antithrombin IIL """""""""""""" 254 2,7,32, Assay ofhuman a-l-proteinase inhibitoL """"""",266

General Notices (1) apply ID al! monographs and other texts 227
EUROPEAN PHARMACOPOEIA 8.0

228 See the 1n1",",-""n,,n section on general monographs (cover pages)


EUROPEAN PHARMACOPOEIA 8.0 2.7.1. Irnmunochernical rnetnods

2.7. BIOLOGICAL ASSAYS Where the immunoprecipitating system consists of one


antigen combining with its corresponding antibody, the
system is referred to as simple; when it involves related but
110t serologically identical reactants, the system is complex and
where several serologically unrelated reactants are involved,
0112008:20701 the system is multiple.
In simple diffusion methods, a concentration gradient is
2.7,}, IMMUNOCHEMICAL METHODS established for only one of the reactants diffusing from
an external source into the gel medium containing the
lmmunochemical methods are based on the selective, corresponding reactant at a comparatively low concentratiol1.
reversible and non-covalent binding of antigens by antibodies. Single radial immunodiffusion (SRID) is a simple quantitative
These methods are employed to detect or quantify either immunodiffusion technique, When the equilibrium between
antigens or antibodies. The formation of an antigen-antibody the external and the internal reactant has been established,
complex may be detected, and the amount of complex formed the circular precipitation area, originating from the site of
may be measured by a variety of techniques. The provisions of the external reactant, is directly proportional to the amount
this general method apply to immunochemical methods using of the antigen applied and inversely proportional to the
labelled or unlabelled reagents, as appropriate. concentration of the antibody in the gel.
The results of immunochemical methods depend on the In double diffusion methods, concentration gradients are
experimental conditions and the nature and quality of the established for both reactants. Both antigen and antibody
reagents used. It is essential to standardise the components of diffuse from separate sites into an initially immunologically
an immunoassay and to use, wherever available, international neutral gel.
reference preparations for immunoassays. Comparative double diffusion methods are used for qualitatively
The reagents necessary for many immunochemical methods comparing various antigens versus a suitable antibody or vice
are available as commercial assay kits, that is, a set including versa. The comparison is based on the presence or absence of
reagents (particularly the antigen or the antibody) and interaction between the precipitation patterns. Reactions of
materials intended for the in vitro estimation of a specified identity, non-identity or partial identity of antigens/antibodies
substance as well as instructions for their proper use. can be distinguished,
The kits are used in accordance with the manufacturers' Immuuoelectmphoretic rnethods
instructions; it is important to ascertain that the kits are
Immunoelectrophoresis (IE) is a qualitative technique
suitable for the analysis of the substance to be examined, with
combining 2 methods: gel electrophoresis followed by
particular reference to selectivity and sensitivity. Guidance immunodiffusion,
concerning immunoassay kits is provided by the World Health
Organization, Technical Report Series 658 (1981). Crossed immunoelectrophoresis is a modification of the IE
method. It is suitable both for qualitative and quantitative
METHODS IN WHICH A LABELLED ANTIGEN OR A analysis. The first part of the procedure is an ordinary gel
LABELLED ANTIBODY IS USED electrophoresis, after which a longitudinal gel strip, containing
the separated fractions to be determined, is cut out and
Methods using labelled substances may employ suitable transferred to another plate, The electrophoresis in the
labels such as enzymes, fluorophores, luminophores and second direction is carried out perpendicular to the previous
radioisotopes. Where the label is a radioisotope, the method electrophoretic run in a gel containing a comparatively low
is described as a "radio-immunoassay". The recommendations concentration of antibodies corresponding to the antigens.
for the measurement of radioactivity given in the monograph For a given antibody concentration and gel thickness, the
on Radiopharmaceutical Preparations (0125) are applicable relationship between the area of the respective precipitatíon
to immunoassays involving radioisotopes. Al! work with peaks and the amount of the corresponding antigen is linear.
radio active materials must be carried out in conformity with
Electroimmunoassay, often refened to as rocket
nationallegislation and internationally accepted codes of
immuno-electrophoresis is a rapid quantitative method for
practice for protection against radiation hazards.
determining antigens with a charge differing from that
of the antibodies or vice versa. The electrophoresis of the
METHODS IN WHICH AN UNLABELLED ANTIGEN OR antigen to be determined is carried out in a gel containing
ANTIBODY 1S USED a comparatively lower concentration of the corresponding
Irnmunopredpitation methods antibo dy. The test material and dilutions of a standard antigen
Immunoprecipitation methods include flocculation and used for calibration are introduced into different wells in
precipitation reactions. When a solution of an antigen is mixed the geL During electrophoresis, migrating peak-shaped
with its corresponding antibody under suitable conditions, the precipitation zones originating from the wells are developed.
reactants form flocculating or precipitating aggregates. The The front of the precipitate becomes stationary when
ratio of the reactants which gives the shortest flocculation time the antigen is no longer in excess. For a given antibody
or the most marked precipitation is called the optimal ratio, concentration, the relationship between the distance travelled
and is usually produced by equivalent amounts of antigen and by the precipitate and the am~unt of antigen applied is linear.
antibo dy. Immunoprecipitation can be assessed visually or by Counter-immunoelectrophoresis is a rapid quantitative
light-scattering techniques (nephelometric or turbidimetric method allowing concentration gradients of external antigen
assay). An increase in sensitivity can be obtained by using and external antibody to be established in an electric field
antigen- or antibody-coated particles (e.g. latex) as reactants. depending on the different charges. Dilutions of a standard
for calibration and dilutions of the test material are introduced
In flocculation methods, stepwise dilutions of one of the
into a row of wells in a gel and a fixed amount of the
reactants is usually used whereas, in immunodiffusion (ID)
corresponding reactant is introduced into an opposite row of
methods, the dilution is obtained by diffusion in a gel medium:
wells. The titre of the test material may be determined as the
concentration gradients of one or both of the reactants are
highest dilution showing a precipitation lineo
obtained, thus creating zones in the gel medium where the
ratio of the reactants favours precipitation, While flocculation A number of modifications of crossed immul1oelectrophoresis
methods are performed in tubes, immunodiffusion methods and electroimmunoassay methods exist.
may be performed using different supports such as tubes, Other techniques combine separation of antigens by molecular
plates, slides, cel!s or chambers, size and serological properties.

General Notices (1) apply lo all monographs and other texts 229
2.7.2. Microbiological assay of antibiotics EUROPEAN PHARMACOPOEIA 8.0

Visualisation and chal'acterisation of immunoprecipitation 0112009:20702


Hnes correcíed 7.6
These may be performed by selective or non-selective stains,
by fluorescence, by enzyme or isotope labelling or other 2.7.2. MICROBIOLOGICAL ASSAY
relevant techniques. Selective staining methods are usually OF ANTIBIOTICS
performed fOI characterisation of non -protein substances in
the precipitates. The potency of an antibiotic is estimated by comparing the
inhibition of growth of sensitive micro-organisms produced
In translucent gels such as agar or agarose, the precipitation by known eoncentrations of the antibiotic to be examined and
line becomes clearly visible in the gel, provided that the a reference substance.
concentration of each of the reactants is appropriate. The reference substances used in the assays are substances
whose activity has been precisely determined with reference
VALIDATION OF THE METHOD to the corresponding international standard or international
reference preparation.
Validation criteria
The assay must be designed in a way that will permit
A quantitative immunochemical method is not valíd unless: examination of the validity of the mathematical model on
1) The antibody or antigen do es not significantly discriminate
which the potency equation is based. If a parallel-line model
between the test and standard. For a labelled reactant, the is chosen, the 2 log dose-response (or transformed response)
corresponding reactant does not significantly discriminate lines of the preparation to be examined and the reference
between the labelled and unlabelled compound, preparation must be parallel; they must be linear over the
range of doses used in the calculation. These conditions must
2) The method is not affected by the assay matrix, that is, any be verified by validity tests for a given probability, usually
component of the test sample or its excipients, which can vary P = 0.05. Other mathematical models, such as the slope
between samples. These may include high concentrations ratio mode!, may be used provided that proof of validity is
of other proteins, salts, preservatives or contaminating demonstrated.
proteolytic activity, Unless otherwise stated in the monograph, the confidence
3) The limit of quantitation is below the acceptance criteria limits (P = 0.95) of the assay for potency are 110t less than
stated in the individual monograph, 95 per cent and not more than 105 per cent of the estimated
potency.
4) The precision of the assay is such that the variance of Carry out the assay by method A or method B.
the results meets the requirements stated in the individual
monographs, A. DIFFUSION METHOD
5) The order in which the assay is performed do es not give Liquefy a medium suitable for the conditions of the assay
rise to systematic errors. and inoculate it at a suitable temperature, for example 48 oC
to 50 oC for vegetative forms, with a known quantity of a
Validation methods suspension of micro-organisms sensitive to the antibiotic to
In order to verify these criteria, the validation design indudes be examined, such that clearly defined zones of inhibition of
the following elements: suitable diameter are produeed with the concentrations of
the antibiotic used for the assay. Immediately pour into Petri
1) The assay is performed at least in triplicate, dishes or large rectangular dishes a quantity of the inoculated
2) The assay includes at least 3 different dilutions of the medium to form a uniform layer 2-5 mm thick. Alternatively,
standard preparation and 3 dilutions of sample preparations the medium may consist of 2 layers, only the upper layer
of presumed activity similar to the standard preparation, being inoculated.
Store the dishes so that no appreciable growth or death of the
3) The assay layout is randomised, micro-organisms occurs before the dishes are used and so that
4) lf the test sample is presented in serum or formulated with the surface of the medium is dry at the time of use.
other components, the standard is likewise prepared, Using the solvent and the buffer solution indicated in
Table 2.7.2.-1, prepare solutions of the reference substance and
5) The test includes the measurement of non -specific binding of the antibiotic to be examined having known concentrations
of the labelled reactant, and presumed to be of equal activity. Apply the solutions to
6) For displacement immunoassay: the surface of the medium, for example, in sterile cylinders
of porcelain, stainless steel or other suitable material, 01' in
(a) maximum binding (zero displacement) is determined, cavities prepared in the agar. The same volume of solution
(b) dilutions cover the complete response range from must be added to each cylinder or cavity. Alternatively, use
values close to non-specific binding to maximum binding, sterile absorbent paper dises of suitable quality; impregnate
preferably for both standard and test preparations. the discs with the solutions of the reference substance or the
solutions of the antibiotic to be examined and place on the
surface of the agar.
STATISTICAL CALCULATION
In order to assess the validity of the assay, use not Íewer than
To analyse the results, response curves for test and standard 3 doses of the reference substance and 3 closes of the antibiotic
may be analysed by the methods described in 5.3. Statistical to be examined having the same presumed activity as the
Analysis of Results of Biological Assays and Tests. doses of the reference substance. It is preferable to use a series
of doses in geometric progressiol1. In routine assays when
Significant non-parallelism indicates that the antibody or the linearity of the system has been demonstrated over an
antigen discriminates between test and standard, and the adequate number of experiments using a three-point assay, a
results are 110t valido two-point assay may be sufficient, subject to agreement by
In displacement immunoassays, the values for non-specific the competent authority. However, in al! cases of dispute, a
binding and maximum displacement at high test or standard three-point assay as described aboye must be applied.
concentration must not be significantly different. Differences Arrange the solutions on eaeh Petri dish or on each rectangular
may indicate effects due to the matrix, either inhibition of dish accorcling to a statistically suitable design, except for small
binding or degradation of tracer. Petri dishes that cannot accommodate more than 6 solutions,

230 See the information section on general monographs (cover pages)


EUROPEAN PHARMACOPOEIA 8.0 2.7.2. Microbiological assay of antibiotics

arrange the solutions of the antibiotic to be examined and the In order that the validity of the assay may be assessed, use not
solutions of the reference substance in an alternate manner to fewer than 3 doses of the reference substance and 3 doses of the
avoid interaction of the more concentrated solutions. antibiotic to be examined having the same presumed activity
as the doses of the reference substance. It is preferable to use
Incubate at a suitable temperature for about 18 h. A period
of diffusion prior to incubation, usually 1-4 h, at room a series of doses in geometric progression. In order to obtain
temperature or at about 4 oC, as appropriate, may be used the required linearity, it may be necessary to select from a
large number 3 consecutive doses, using corresponding doses
to minimise the effects of the variation in time between the
for the reference substance and the antibiotic to be examined.
application of the solutions and to improve the regression
slope.
Distribute an equal volume of each of the solutions into
Measure the diameters with a precision of at least 0.1 mm or identical test-tubes and add to each tube an equal volume of
the areas of the circular inhibition zones with a corresponding inoculated medium (for example, 1 mL of the solution and
precision and calculate the potency using appropriate 9 mL of the medium). For the assay of tyrothricin add 0.1 mL
statistical methods. of the solution to 9.9 mL of inoculated medium.
Use in each assay the number of replications per dose
sufficient to ensure the required precision. The assay may be Prepare at the same time 2 control tubes without antibiotic,
repeated and the results combined statistically to obtain the both containing the inoculated medium and to one of which is
required precision and to ascertain whether the potency of added immediately 0.5 mL of farmaldehyde R. These tubes are
the antibiotic to be examined is not less than the minimum used to set the optical apparatus used to measure the growth.
required.
Place all the tubes, randomly distributed or in a Latin square
B. TURBIDIMETRIC METHOD or randomised block arrangement, in a water-bath or other
Inoculate a suitable medium with a suspension of the chosen suitable apparatus fitted with a means of bringing al! the
micro-organism having a sensitivity to the antibiotic to be tubes rapidly to the appropriate incubation temperature
examined such that a sufficiently large inhibition of microbial and maintain them at that temperature for 3-4 h, taking
growth occurs in the conditions of the test. Use a known precautions to ensure uniformity of temperature and identical
quantity of the suspension chosen so as to obtain a readily incubation time.
measurable opacity after an incubation period of about 4 h.
After incubation, stop the growth of the micro-organisms
Use the inoculated medium immediately after its preparation.
by adding 0.5 mL of formaldehyde R to each tube or by heat
Using the solvent and the buffer solution indicated in treatment and measure the opacity to 3 significant figures
Table 2.7.2.-2 prepare solutions of the reference substance and using suitable optical apparatus. Alternatively use a method
of the antibiotic to be examined having known concentrations which allows the opacity of each tube to be measured after
presumed to be of equal activity. exactly the same period of incubation.

Table 2.7.2.-1. - Diffusian assay

Solvellt to be used
Buffer solution Medium and final Incuhation
Antibiotic Reference substance in preparing the Micro-organism
(pH) pH (± 0.1 pH unít) temperature
stock solution
Saccharomyces
Amphotericin B cerevisiae
Amphotericin B fo1' microbiological Dimethyl sulfoxide R pH 10.5 (0.2 M) F pH 6.1 35-37 oC
ATCC 9763
assay CRS
IP 1432-83
Micrococcus luteus
0.01 M hydrochloric NCTC 7743
Bacitracin zinc Bacitracin zinc CRS pH 7.0 (0.05 M) A - pH 7.0 35-39 oC
acid CIP 53.160
ATCC 10240
Mycobacterium
Bleomycin sulfate Bleomycin sulfate CRS Water R pH 6.8 (0.1 M) smegmatis G - pH 7.0 35-37 oC
ATCC 607
Bordetella B - pH 7.3 35-39 oC
bronchiseptica
NCTC 8344
CIP 53.157
Colistimethate Colistimethate ATCC 4617
Water R pH 6.0 (0.05 M)
sodium sodium CRS
Escherichia coZ¡ B - pH 7.3 35-39 oC
NCIMB 8879
CIP 54.127
ATCC 10536
Bordetella B - pH 7.3 35-39 oC
bronchiseptica
NCTC 8344
CIP 53.157
Colistin sulfate for
Colistin sulfate microbiological Water R pH 6.0 (0.05 M) ArCC 4617
assay CRS Escherichia coZ¡ B - pH 7.3 35-39 oC
NCIMB 8879
CIP 54.127
ATCC 10536

General Natices (1) apply ta all manographs and ather texts 231
2.7.2. Mkrobiological assay of antibiotics EUROPEAN PHARMACOPOEIA 8.0

Solvent to be used
Buffer solution Medium and final Incubation
Aniibiotic Reference substance in preparing the Micro-organism
(pH) pH (± 0.1 pH unit) temperatnre
stock solution
Bacillus subtilis E - pH 7.9 30-37 oC
NCTC 10400
CIP 52.62
Framycetin
Framycetin sulfate Water R pH 8.0 (0.05 M) ATCC 6633
sulfate CRS
Bacillus pumilus E - pH 7.9 30-37 oC
NCTC 8241
CIP 76.18
Bacillus pumilus A - pH 7.9 35-39 oC
NCTC 8241
CIP 76.18
Gentamicin Staphylococcus A - pH 7.9 35-39 oC
Gentamicin sulfate Water R pH 8.0 (0.05 M)
sulfate CRS epidermidis
NCIMB 8853
CIP 68.21
ATCC 12228
Bacillus subtilis
Methanol R (see the CIP 52.62
Josamycin Josamycin CRS pH 5.6 A - pI-I 6.6 35-37 oC
monograph) ATCC 6633
NCTC 10400
Bacillus subtilis
Josamycin Josamycin Methanol R (see the CIP 52.62
pI-I5.6 A - pH 6.6 35-37 oC
prapionate propionate CRS ll1onograph) ATCC 6633
NCTC 10400
Bacillus subtilis A - pH 7.9 30-37 oC
Kanmnycin NCTC 10400
ll1onosulfate CIP 52.62
Kanamycin ATCC 6633
Water R pH 8.0 (0.05 M)
monosulfate CRS Staphylococcus aureus A - pH 7.9 35-39 oC
Kanall1ycin acid NCTC 7447
sulfate CIP 53.156
ATCC 6538 P
Bacillus pumilus E - pH 7.9 30-37 oC
NCTC 8241
CIP 76.18
Neomycin sulfate
Neoll1ycin sulfate for microbiological Water R pH 8.0 (0.05 M) Bacillus subtilis E - pI-I 7.9 30-37 oC
assay CRS
NCTC 10400
CIP 52.62
ATCC 6633
Staphylococcus Ilureus
Netilmicin sulfate Netilmicin sulfate CRS Water R pH 8.0 ± 0.1 ATCC 6538 P A - pH 7.9 32-35 oC
CIP 53.156
Candida tropicalis F - pH 6.0 30-37 oC
CIP 1433-83
NCYC 1393
pH 6.0 (0.05 M)
Dimethylforma- containing 5 per Saccharomyces F - pI-I 6.0 30-32 oC
Nystatin Nystatin CRS
mide R cent V/V of dime- cerevisiae
thylformamide R
NCYC87
CIP 1432-83
ATCC 9763
Micrococcus ¡uteus
Rifamycin NCTC 8340
Rifall1ycin sodiul11 Methanol R pI-I 7.0 (0.05 M) A - pI-I 6.6 35-39 oC
sodium CRS C!P 53.45
ATCC 9341
Bacillus subtilis
NCTC 10400
Spirall1ycin Spiramycin CRS Methanol R pH 8.0 (0.05 M) A -pI-I 7.9 30-32 oC
CIP 52.62
ATCC 6633

232 See the informatíon section on general monographs (cover pages)


EUROPEAN PHARMACOPOEIA 8.0 2.7.2. Micmbiological assay oi antibiotics

Solvent to be used
Buffer solution Medium and final lncubation
Antibiotic Refel'ence substance in preparing the Micro-ol'ganism
(pH) pH (± 0.1 pH unit) temperature
stock solutioll
Bacillus subtilis A - pH 7.9 30-37 oC
NCTC 8236
CIP 1.83
Streptomycin
Streptomycin sulfate Water R pH 8.0 (0.05 M) Bacillus subtilis A - pH 7.9 30-37 oC
sulfate CRS
NCTC 10400
CIP 52.62
ATCC 6633
Bacillus sllbtilis
NCTC 10400
Teicoplanin Teicoplanin CRS pH 6.0 (0.05 M) pH 6.0 (0.05 M) H - pH 7.8-8.0 35-37 oC
CIP 52.62
ATCC 6633
Tylosin [or A mixture of
2.5 per cent VIV IVIicrococcus luteus
Yeterinary use 40 yolumes of
501ution of NCTC 8340
methanol R and 32-35 oC
Tylosin CRS methanol R in 0.1 M A - pH 8.0
60 yolumes o[ 0.1 IVI CIP 53.45
Tylosin tartrate for phosphate buffer
phosphate buJler ATCC 9341
veterinary use solution pH 7.0 R
solution pH 8.0 R
Bacillus subtilis
Vancomycin Vancomycin NCTC 10400
Water R pH 8.0 A - pH 8.0 37-39 oC
hydrochloride hydrochloridc CRS CIP 52.62
ATCC 6633

Calculate the potency using appropriate statistical methods. two-point assay may be sufficient, subject to agreement by
Linearity of the dose-response relationship, transformed or the competent authority. However, in all cases of dispute, a
untransformed, is often obtained only over a very limited three-point assay must be applied.
range. It is this range which must be used in calculating the
activity and it must include at least 3 consecutive doses in Use in each assay the number of replications per dos e
order to permit linearity to be verified. In routine assays when sufficient to ensure the required precision. The assay may be
the linearity of the system has been demonstrated over an repeated and the results combined statistically to obtain the
adequate number of experiments using a three-point assay, a required precision and to ascertain whether the potency of
the antibiotic to be examined is not les s than the minimum.
required.

Table 2.7.2.-2. - Turbidimetric assay

Salvent ta be used
Buffer salutian Medium and final Incubatian
AntibioHc Reference substunce in preparing the Micro-organism
(pH) pH (± 0.1 pH nnit) temperature
stock solution

Escherichia coli
Colistimethate Colistimethate NCIMB 8666
Water R pH 7.0 C - pH 7.0 35-37 oC
sodium sodiu111 CRS CIP 2.83
ATCC 9637

Eschcrichia coli
Colistin sulfate for NCIMB 8666
Colistin sulfate microuiological Water R pH 7.0 C - pH 7.0 35-37 oC
assay CRS CIP 2.83
ATCC 9637
Staphylococcus aureus

I~~'ij~iecg~~
NCTC 7447
Framycetin sulfate Water R pH 8.0 C - pH 7.0 35-37 oC
CIP 53.156
ATCC 6538 P
Staphylococcus aureus
Gentamicin NCTC 7447
Gentamicin sulfate Water R pH 7.0 C - pH 7.0 35-37 oC
sulfate CRS CIP 53.156
ATCC 6538 P
Enterococcus hirae
CIP 58.55
Gramicidin CRS Methanol R pH 7.0' ATCC 10541 C- pH 7.0 35-37 oC
Gran1icidin Staphylococcus aureus
ATCC 6538 P
* Addition of a detergent may be necessary to avoid adsorption on the material during the dilutions, for example 0.1 mg/mL
of polysorbate 80 R

General Notices (1) apply to all monographs and other texts 233
2.7.2. Microbiological assay of antibiotks EUROPEAN PHARMACOPOEIA 8.0

Solvent to be used
Buffer solution Micro-organism Medium and final Incubation
Antibiotic Reference substance in preparing the
(pH) pH (± 0.1 pH unit) temperature
stock solution
Staphylococcus aureus
lvlethanol R (see lhe CIP 53.156
)osamycin Josamycin CRS pH 5.6 C - pH 8.0 35-37 oC
monograph) ATCC 6538 P
NCTC 7447
Staphylococcus aureus
)osamycin Josamycin Metha1101 R (see the CIP 53.156
pH 5.6 C - pH 8.0 35-37 oC
propionate propiol1ate CRS monograph) ATCC 6538 P
NCTC 7447
Kanamycin Staphylococcus aureus
monosulfate
Kanamycin NCTC 7447
Water R pH 8.0 C - pH 7.0 35-37 oC
monosulfate CRS CIP 53.156
Kanamycin acid
sulfate ATCC 6538 P

Staphylococcus aureus
Neomycin sulfate NCTC 7447
Neomycin sulfate for núcrobiological Water R pH 8.0 C - pH 7.0 35-37 oC
assay CRS CIP 53.156
AfCC 6538 P
Escherichia coli
Rifamycin NCIMB 8879
Rifan1ycin sodiun1 Methanol R pH 7.0 C - pH 7.0 35-37 oC
sodium CRS CIP 54.127
ATCC 10536
Stap/¡ylococcus aureus
NCTC 7447
Spiramycin Spiramycin CRS Methanol R pH 7.0 C - pH 7.0 35-37 oC
CIP 53.156
ATCC 6538 P
Klebsiella pneumoniae
Streptomycin NCTC 7427
Streptomycin sulfate Water R pH 8.0 C - pH 7.0 35-37 oC
sulfate CRS CIP 53.153
ATCC 10031
Tylosin for 2.5 per cent VIV Staphylococcus aureus
veterinary use solution of NCTC 6571
Tylosin CRS methanol R in 0.1 M pH 7.0 C - pH 7.0 37 oC
TyIosin tartrate for phosphate buffer AfCC 9144
veterinary use solution pH 7.0 R CIP 530154
Enterococcus hirae
Tyrothricin Gramicidin CRS Alcohol R Alcohol R C - pH 7.0 37 oC
ATCC 10541
Staphylococcus aureus
Vanco111ycin Vancomycin
Water R pH 8.0 CIP 53.156 C - pH 7.0 37-39 oC
hydrochloride hydrochloride CRS
ATCC 6538 P

The following section is published for information. Alternatively, spore suspensions may be prepared by
cultivating the organisms in medium e at 26 oc for 4-6 days,
then adding, aseptically, sufficient manganese sulfate R to give
a concentration of 0.001 giL and incubating for a further
Recommended micro-organisms 48 h. Examine the suspension microscopically to ensure
that adequate spore formation has taken place (about 80 per
cent) and centrifuge. Re-suspend the sediment in sterile
The following text details the recommended micro-organisms water R to give a concentration of 10 x 10 6 to 100 X 106 spores
and the conditions of useo Other micro-organisms may be
per millilitre, and then heat to 70 oC for 30 mino Stofe the
used provided that they are shown to be sensitive to the suspension at a temperature not exceeding 4 oc.
antibiotic to be examined and are used in appropriate media
and appropriate conditions of temperature and pH. The Bordetella bronchiseptica. Grow the test organism on
concentrations of the solutions used should be chosen so as to medium B at 35-37 oC foy 16-18 h. Wash off the bacterial
ensure that a linear relationship exists between the logarithm growth with sterile water R and dilute to a suitable opacity.
of the dose and the response in the conditions of the test.
Staphylococcus aureus; Klebsiella pneumoniae; Escherichia
Preparation of inocula. Bacillus cereus varo mycoides; Bacillus coli; Micrococcus luteus; Staphylococcus epidermidis. Prepare
subtilis; Bacillus pumilus. Spore suspensions ofthe organisms as described aboye for B. bronchiseptica but using medium A
to be used as inocula are prepared as follows. and adjusting the opacity to one which has been shown to
produce a satisfactory dose-response relationship in the
Grow the organism at 35-37 oC for 7 days on the surface of turbidimetric assay, or to produce clearly defined zones of
a suitable medium to which has been added 0.001 giL of inhibition of convenient diameter in the diffusion assay, as
manganese sulfate R. Using sterile water R, wash off the growth, appropriateo
which consists mainly of spores. Heat the suspension at 70 oC
for 30 min and d¡lute to give an appropriate concentration of Saccharomyces cerevisiae; Candida tropicaliso Grow the test
spores, usually 10 x 106 to 100 x 10 6 per millilitre. The spore organism on medium F at 30-37 oC for 24 h. Wash off the
suspensions may be stored for long periods at a temperature growth with a sterile 9 giL solution of sodium chloride R.
not exceeding 4 oc. Dilute to a suitable opacity with the same solution.

234 See the information section on (cover pages)


EUROPEAN PHARMACOPOEIA 800 2.7.2. Microbiological assay of antibiotics

Buffer solutionso Buffer solutions having a pH between 508 Medium C


and 800 are prepared by mixing 5000 mL of 0.2 M potassium Peptone 6 g
dihydrogen phosphate R with the quantity of 002 M sodium
Beef extraet 1.5 g
hydroxide indicated in Table 207020-30 Dilute with freshly
prepared distilled water R to produce 20000 mL Yeast extraet 3 g

Sodium ehloride 305 g


Table 2.7.20-30
Glucose monohydrate 1g
pH 0.2 !vI Sodium hydmxide (mL)
Dipotassium hydrogen phosphate 3068 g
508 3072
Potassium dihydrogen phosphate 1.32 g
600 5.70
Water to 1000 mL
6.2 8060

6,4 12060 Medium D


606 17080 Heart extraet 1.5 g

608 23065 Yeast extraet 1.5 g

700 29063 Peptone-casein 5 g

702 35000 Glucose monohydrate 1g

7,4 39.50 Sodium ehloride 3.5 g

706 42080 Dipotassium hydrogen phosphate 3068 g

708 45020 Potassium dihydrogen phosphate 1.32 g

800 46080 Potassium nitrate 2 g

Water to 1000 mL
These buffer solutions are used for all microbiological assays
shown in Table 2.7020-1 with the exception of bleomycin Medium E
sulfate and amphotericin Bo Peptone 5g

For bleomycin sulfate, prepare the buffer solution pH 608 as Meat extraet 3 g
follows: dissalve 6.4 g of potassium dihydrogen phosphate R
2609 g
and 1809 g of dísodium hydrogen phosphate R in water R and Disodium hydrogen phosphate,12H,O
dilute to 1000 mL with water R. Agar 10 g

For amphotericin B, prepare the 002 M phosphate buffer Water to 1000 mL


solution pH 10.5 as follows: dissolve 35 g of dipotassium
hydrogen phosphate R in 900 mL af water R, add 20 mL of 1 M The disodium hydrogen phosphate is added as a sterile
sodium hydroxide and dilute to 100000 mL with water R. solutiol1 after sterilisation of the mediumo
Culture mediao The following media or equivalent media Medium F
may be usedo Peptone 9.4 g

MediumA Yeast extraet 407 g

Peptone 6g Beef extraet 2.4 g

Pancreatic digest oÍ casein 4g Sodium ehloride 1000 g

Beef extraet 1.5 g Glucose monohydrate 1000 g

Yeast extraet 3g Agar 23.5g

Glucose monohydrate 1g Water to 1000 mL

Agar 15 g
Medium G
Water to 1000 mL Glyceroi lOg

Peptone lOg
Medium B
Meat extraet lOg
Pancreatic digest of casein 17 g
Sodium ehloride 3 g
Papaie digest of soya bean 3g
Agar 15 g
Sodium chloride 5g
Water to 1000 mL
Dipotassium hydrogen phosphate 2.5 g

Glueose monohydrate 2.5 g pH 700 ± 001 after sterilisationo


Agar 15 g MediumH
Peptone 500 g
Polysorbate 80 10 g
Agar 1500 g
Water to 1000 mL
Beef extraet powder 300 g
The polysorbate 80 is added to the hot solution of the other Water to 1000 mL
ingredients after boiling, and immediately befare adjusting
to volumeo pH 708 - 800 adjusted with 001 M sodium hydroxideo

General Notices (1) apply fo all monographs and other texts 235
2.7.4. fadorVIH EUROPEA1\[ PHARMACOPOEIA 8.0

0112008:20704 the divided into at least 2 separate


reagents, each to generate factor Xa 011
its own. One of the reagents contain5 calcium ions. Arter
reconstitution, the reagents be combined
that no substantial amounts Xa are 1;"'"''''''''''''
the absence of Íactor VIII. In the final mixture,
by its biological factor VIII must be the rate-limiting component.
as a cofactor in the activation factor X by activated
of calcium ions and of the formed
factor Xa, a substrate that is
a factor VIII preparation is
specific for factor Generally this con5i5t5 of a derivatised
necessary to achieve a
short peptide of between 3 and 5 amino acids, to a
certain rate Xa in a test mixture containing
the substances that take part in the activation of factor X, and chromophore group. On of this group from the
peptide substrate, its shift to a
the quantity of the International Standard, or oí a reference
wavelength its sp(;ctr01Jl1'Jtclmetr
preparation calibrated in International Units, required to
The substrate must also contain
produce the same rate of factor Xa formation.
stop further factor Xa
The International Unit is the factor VIII activity oí a stated to suppress thrombin
amount of the International Standard, which consists of a
freeze-dried human coagulation factor VIII concentrate.
The equivalence in International Units of the International ASSAY PROCEDURE
Standard is stated by the World Health Organization.
Reconstitute the entire contents oí 1 of the reference
VIII BRP is calibrated in nn'n'1r~l"ir.n aud of the nn'mlr~i"irl11
with the International Add OUL'''.-''.-H

Standard.
The Ol" oE an
activation oí Íactor X in a that contains sufficient von
reagent composed of purified components, results that do not differ
cleavage of a chromogenic Íactor Xa significantly [rom those obtained haemophilia
a that can be quantified plasma. The materials must stable beyond the
assay conditions, time the assay.
rate oí factor Xa
formation and the factor VIII concentration. The assay is Prepare further dilutions of the reference and test
summarised the scheme. using a buffered solution, fol'
or imidazole,
1 pe!" cent oí human or bovine albumino
Step 1 UHUUVH series of at least 3 fmther dilutiol1s Íor each

the dilutions such t11at the final factor VIII


factor X (act!vated) factor VIII ~ factor Xa
cOl1centration in the reaction mixture is below
factor IXa, phospholipid, Ca 2 + 0.01 IU/mL, during the step offactor Xa
a control solution that includes all components except
Step :2 factor VIII.

chromogenic substrate factor Xa .... peptide + chromophore

be obtained commercially
from a sources. HL"VU,," the composition of
individual reagents to some variation, their
essential Íeatures are the

factor VIII concentrate as the


obtained do not ddIer signific2.ntly. maximal
usually between 2 min
It is important to demonstrate validation the suitability oí
the kit used, checking the time course of factor Xa Step 2. Terminate the activation
the time taken to reach reagent a
the rate of substrate cleavage, which must be linear
the concentration of factor Xa formed, measuring the
REAGENTS absorbance change at an ~nn1",~,nn
either HHJJUlVJ.
The factor reagent purified proteins
derived human or bovine sources. These include
factor X, factor IXa, and a factor VIII activator, usually
thrombin. These are partIy purified, preferably to at
least 50 per cent, and do not contain impurities that interfere
with the activation of factor VIII or Íactor X. Thrombin
may be present in its precursor Íorm prothrombin, provided
[hat its activation in the reagent is sUÍficiently rapid to give
almost instantaneous activation of factor VIII in the
may be obtained f1'om natural sources or
and must, to a substantial extent, the usual
The of

236 See the mrnnww


EUROPEAN PHARMACOPOEIA 8.0 2.7.6. Assay of diphtheria vacdne (adsorbed)

01/2008:20705 Repeat the procedure using fresh dilutions and carrying out
the incubation in the order TI' T 2, T 3, SI' S2' S3' Calculate the
results the usual statistical methods (5.3).
2,7,5. ASSAY OF HEPARIN
Carry out not fewer than 3 independent assays. For each such
The anticoagulant activity ofheparin is determined in vitro by ass~y prepare [resh solutions of the reference preparation
comparing its ability in given conditions to delay the clotting and the preparation to be examined and use another, freshly
of recalcified citrated sheep plasma with the same ability of a thawed portion of plasma substrate.
reference preparation of heparin calibrated in International Calculate the potency of the preparation to be examined,
Units. combining the results of these assays, by the usual statistical
The International Unit is the activity contained in a stated methods (5.3). 'vVhen the variance due to differences between
amount of the International Standard, which consists assays is significant at P = 0.01, a combined estimate of
of a quantity of freeze-dried heparin sodium from pork potency may be obtained by calculating the non-weighted
intestinal mucosa. The equivalence in International Units mean of potency estimates.
of the International Standard is stated by the World Health
Organization. 01/2008:20706
Heparin sodium BRP is calibrated in International Units by correded 6.0
comparison with the International Standard by means of the
assay given below. 2,7.6. ASSAY OF DIPHTHERIA
Carry out the assay using one of the following methods for VACCINE (ADSORBED)
determining the onset of clotting and using tubes and other
equipment appropriate to the chosen method: The potency of diphtheria vaccine is determined by
administration of the vaccine to guinea-pigs followed either
a) direct visual inspection, preferably using indirect by challenge with diphtheria toxin (method A ar B) or by
illumination and viewing against a matt black background; determination of the titre of antibodies against diphtheria
b) spectrophotometric recording of the change in optical toxin or toxoid in the serum of guinea-pigs (method C).
density at a wavelength of approximately 600 nm; In both cases, the potency of the vaccine is calculated by
comparison with a reference preparation, calibrated in
c) visual detection of the change in fluidity on manual tilting
International Units.
of the tubes;
The International Unit is the activity contained in a stated
d) mechanical recording of the change in fluidity on stirring, amount of the International Standard, which consists of
care being taken to cause the minimum disturbance of the a quantity of diphtheria toxoid adsorbed on aluminium
solution during the earliest phase of clotting. hyd1'Oxide. The equivalence in International Units of
the International Standard is stated by the World Health
ASSAY PROCEDURE Organization (WHO).
The valumes in the text are given as examples al1d Diphtheria vaecine (adsorbed) BRP is suitable for use as a
adapted to the apparatus used provided that the ratios reference preparatiol1.
the different vo/umes are
The method chosen for the assay of diphtheria vaccine
Dilute heparin sadium BRP with a 9 giL solution of depends 011 the intended purpose. Method A or
sodium eh/oride R to contain a precisely known number Bis used:
of International Units per millilitre and prepare a similar 1. during development of a vaccine, to assay batches produced
solntion of the preparation to be examined which is expected to validate the productiol1;
to have the same activity. Using a 9 giL solution of sodium
2. wherever revalidation is needed following a significant
eh/aride R, prepare f1'Om each solution a series of dilutions in
change in the manllfacturing process.
geometric p1'Ogression such that the clotting time obtained
with the lowest concentration is not less than 1.5 times Method A or B may also be used for the 1'Outine assay of
the blank recalcification time, and that obtained with the batches of vaccine, but in the interests of animal welfare,
highest concentration is such as to give a satisfactory log method C is used wherever possible.
dose-response curve, as determined in a preliminary test. Method C may be used, except as specified under 1 and 2
Place 12 tubes in a bath of iced water, labeiling them in aboye, after verification of the suitability of the method for
duplicate: Tl' T 2 and T} for the dilutions ofthe preparation the p1'Oduct. For this purpose, a suitable number of batches
to be examined and S" S2 and S3 for the dilutions of the (usua!ly 3) are assayed by method C and method A or B.
reference preparation. To each tube add 1.0 mL of thawed Where different vaccines (monovalent or combinations) are
plasma substrate Rl and 1.0 mL ofthe appropriate dilution of
prepared from diphtheria toxoid of the same origin, and with
the preparation to be examined or the reference preparation. comparable levels (expressed in Lf/mL) of the same diphtheria
After each addition, mix but do not allow bubbles to formo toxoid, suitability demonstrated for the combination with the
highest number of components can be assumed to be valid
Treating the tubes in the order S)' S2' S3' Tl' T 3, transfer
each tube to a water-bath at 37 oC, allow to equilibrate at 37 oC for combinations with fewer components and for monovalent
for about 15 min and add to each tube 1 mL of a suitable vaccines. Any combinations containing a whole-cell pertussis
APTT (Activated Partial Thromboplastin Time) reagent component or containing haemophilus type b conjugate
containing phospholipid and a contact activator, at a dilution vaccine with diphtheria toxoid or CRM 197 diphtheria p1'Otein
giving a suitable blank recalcification time not exceeding as carrier in the same vial must always be assessed separately.
60 S. After exactly 2 min add 1 mL of a 3.7 giL solution of For combinations containing diphtheria and tetanus
caleium eh/aride R previously heated to 37 oC and record as components, the serological assay (method e) can be
the clotting time the interval in seconds between this last performed with the same group of animals used for the
addition and the onset of clotting determined by the chosen se1'Ological assay of the tetanus vaccine (adsorbed) (2.7.8)
technique. Determine the blank recalcification time at the when the common immunisation conditions for the diphtheria
beginning and at the end of the procedure in a similar manner, and the tetanus components example, doses, duration)
using 1 mL of a 9 giL solution of sodium chloride R in Dlace have been demonstrated to be valid for the combined vaccine.
of one of the heparin dilutions; the 2 blank values obt;ined The design of the assays described below uses multiple
should not differ significantly. Transfonn the clotting times dilutions for the test and reference preparations. Once the
to logarithms, using the mean value for the duplicate tubes. has sufficient experience with this method for a given

General Natiees (1) apply ta all manographs and ather texts 237
2.7.6. Assay of diphtheria vaedne (adsorbed) EUROPEAN PHARMACOPOEIA 8.0

vaeeine, it is possible to applya simplified model sueh as DETERMINATION OF THE ACTIVITY OF THE
a single dilution for both test and rderenee preparations. CHALLENGE TOXIN
Such a model enables the analyst to determine whether If necessary, injeet the unvaccinated control animals with
the potency of the test preparation is signifieantly higher dilutions containing 80, 40, 20, 10 and 5 x 10 6 Lf of the
than the minimum required, but do es not give information challenge toxin.
on linearity, parallelism and the dose-response curve. The
READING AND INTERPRETA TION OF RESULTS
simplified model allows for a considerable reduction in the
l1umber of animals required and must be eonsidered by eaeh Examine al! injection sites 48 h after injection of the challenge
analyst in accordance with the provisions of the European toxin and record the incidenee of specific diphtheria erythema.
Convention for the Proteetion of Vertebrate Animals Used for Record also the number of sites free from such reaetions as
Experimental and Other Seientific Purposes. the intra-dermal challenge score. Tabulate the intradermal
challenge scores for all the animals receiving the same dilution
Where a single-dilution assay is used, produetion and test of vaecine and use those data with a suitable transformation,
eonsisteney over time are monitored via suitable indicators such as (score)2 or arcsin ((seore/6)2), to obtain an estimate
and by carrying out a fullmultiple-dilution assay periodically, of the relative poteney for each of the test preparations by
for example every 2 years. For serological assays, suitable parallel-line quantitative analysis.
indieators to monitor test eonsistency are:
REQUIREMENTS FOR A VALID ASSAY
- the mean and standard deviation of relative antitoxin
titres or scores of the serum samples obtained after The test is not valid unless:
administration of a fixed dose of the vaccine referenee - for both the vaecine to be examined and the reference
preparation; preparation, the mean seore obtained at the lowest dose
- the antitoxin titres or scores of run controls (positive and level is less than 3 and the mean seore at the highest dose
negative serum samples); level is more than 3;
the ratio of antitoxin titres or seo res for the positive - where applicable, the toxin dilution that contains
serum control to the serum samples corresponding to the 40 x 10 6 Lf gives a positive erythema in at least 80 per
referenee vaccine. cent of the control guinea-pigs and the dilution containing
20 x 10-6 Lf gives a positive erythema in less than 80 per
METHOD A: INTRADERMAL CHALLENGE TEST IN cent of the guinea-pigs (if these eriteria are not met a
GUINEA-PIGS different toxin has to be seleeted);
SELECTION AND DISTRIBUTION OF THE TEST ANIMALS - the eonfidenee limits (P = 0.95) are not less than 50 per cent
Use in the test healthy, white guinea-pigs from the same stock and not more than 200 per eent of the estimated potency;
and of a size suitable for the prescribed number of ehallenge
- the statistieal analysis shows no deviation from linearity
sites, the differenee in body mass between the heaviest
and parallelism.
and the lightest animal being not greater than 100 g. Use
guinea-pigs of the same sex or with males and females equally The test may be repeated but when more than 1 test is
distributed between the groups. Distribute the guinea-pigs performed the results of al! valid tests must be combined in
in not fewer than 6 equal groups; use groups containing the estimate of potency.
a number of animals sufficient to obtain results that fulfil
the requirements for a valid assay prescribed below. If the METHOD B: LETHAL CHALLENGE TEST IN
challenge toxin to be used has not been shown to be stable or GUINEA-PIGS
has not been adequately standardised, indude 5 guinea-pigs SELECTION AND DISTRIBUTION OF THE TEST ANIMALS
as unvaccinated controls. Use in the test healthy guinea-pigs from the same stock, each
SELECTION OF THE CHALLENGE TOXIN weighing 250-350 g. Use guinea-pigs of the same sex or with
Select a preparation of diphtheria toxin containing 67 to males and females equally distributed between the groups.
1331r/100 in 1 Lf and 25 000 to 50 000 minimal reacting doses Distribute the guinea -pigs in not fewer than 6 equal groups;
for guinea-pig skin in 1 Lf. If the ehallenge toxin preparation use groups eontaining a number of animals sufficient to obtain
has been shown to be stable, it is not necessary to verify the results that fulfil the requirements for a valid assay prescribed
aetivity for every assay. below. If the challenge toxin to be used has not been shown
to be stable or has not been adequately standardised, indude
PREPARATION OF THE CHALLENGE TOXIN SOLUTION
4 further groups of 5 guinea-pigs as unvaccinated controls.
Immediately befare use, dilute the challenge toxin with a
suitable diluent to obtain a ehallenge toxin solution eontaining SELECTION OF THE CHALLENGE TOXIN
about 0.0512 Lf in 0.2 mL. Prepare from this a further series of Seleet a preparation of diphtheria toxin containing not less
5 four-fold dilutions eontaining about 0.0128,0.0032,0.0008, than 100 LD 50 per millilitre. If the ehallenge toxin preparation
0.0002 and 0.00005 Lf in 0.2 mL. has been shown to be stable, it is not neeessary to verify the
DILUTION OF THE TEST AND REFERENCE
lethal dose for every assay.
PREPARATIONS PREPARATION OF THE CHALLENGE TOXIN SOLUTION
Using a 9 giL solution of sodium chloride R, prepare Immediately before use, dilute the challenge toxin with a
dilutions of the vaccine to be examined and of the referenee suitable diluent to obtain a challenge toxin solution containing
preparation, such that for eaeh, the dilutions form a series approximately 100 LD 50 per millilitre. If neeessary, use
differing by not more than 2.5-fold steps and in which the portions of the challenge toxin solution diluted 1 to 32, 1
intermediate dilutions, when injected subeutaneously at a to 100 and 1 to 320 with the same diluent.
dose of 1.0 mL per guinea-pig, will result in an intradermal DILUTION OF THE TEST AND REFERENCE
seore of approximately 3 when the animals are challenged. PREPARATIONS
IMMUNISATION AND CHALLENGE Using a 9 giL solution of sodium chloride R, prepare dilutions
Allocate the dilutions, 1 to each of the groups of guinea-pigs, of the vaccine to be examined and of the reference preparation,
and injeet subeutaneously 1.0 mL of each dilution into eaeh sueh that for each, the dilutions form a series differing by
guinea-pig in the group to which that dilution is allocated. not more than 2.5-fold steps and in which the intermediate
After 28 days, shave both flanks of eaeh guinea-pig and injeet dilutions, when injected subcutaneously at adose of 1.0 mL
0.2 mL of eaeh of the 6 toxin dilutions intradermally into per guinea-pig, protect approximately 50 per cent of the
6 separate sites on each of the vaccinated guinea-pigs in sueh animals from the lethal effeets of the subeutaneous injection
a way as to minimise interference between adjacent sites. of the quantity of diphtheria toxin prescribed for this test.

238 See the information section on general monographs (ca ver pages)
EUROPEAN PHARMACOPOEIA 8.0 2.7.6. Assay of diphtheria vaccine (adsorbed)

IMMUNISATION AND CHALLENGE IMMUNISA TION


Alloeate the dilutions, 1 to eaeh of the groups of guinea-pigs, Inject subcutaneously to each guinea-pig 1.0 mL of the
and injeet subcutaneously l.0 mL of eaeh dilution into eaeh dilution allocated to its group.
guinea-pig in the group to which that dilution is alloeated. BLOOD SAMPLING
After 28 days, injeet subeutaneously into each animal 1.0 mL 35-42 days after immunisation, take a blood sample from each
of the ehallenge toxin solution (lOO LD so ).
vaccinated and control guinea-pig using a suitable method.
DETERMINATION OF THE ACTIVITY OF THE
PREPARATION OF SERUM SAMPLES
CHALLENGE TOXIN
Avoid frequent freezing and thawing of serum samples. To
If neeessary, alloeate the challenge toxin solution and the avoid microbial eontamination, it is preferable to carry out
3 dilutions made from it, 1 to each of the 4 groups of manipulations in a laminar-flow cabinet.
5 guinea-pigs, and injeet subcutaneously l.0 mL of eaeh
solution into eaeh guinea-pig in the group to which that DETERMINA TION OF ANTIBODY TITRE
solution is alloeated. Determine the relative antibody titre or score of each serum
sample by a suitable immunochemical method (2.7.1). The
READING AND INTERPRETATION OF RESULTS
methods shown below (enzyme-linked immunosorbent assay
Count the number of surviving guinea-pigs 4 days after (ELISA) and Vera cel! assay) have been found lo be suitable.
injeetion of the ehallenge toxin. Calculate the poteney of
the vaccine to be examined relative to the potency of the CALCULATION OF POTENCY
reference preparation on the basis of the proportion of animals Calculate the potency of the vaccine to be examined in
surviving in each of the groups of vaccinated guinea -pigs, International Units relative to the reference preparation, using
using the usual statistical methods (for example, 5.3). the usual statistical methods (for example, 5.3).
REQUIREMENTS FOR A VALID ASSAY REQUIREMENTS FOR A VALID ASSAY
The test is not valid unless: The test is not valid unless:
for both the vaccine to be examined and the reference - the confidence limits (P = 0.95) are not less than 50 per cent
preparation, the 50 per cent protective dose líes between and not more than 200 per cent of the estimated potency;
the largest and smallest doses of the preparations given to - the statistical analysis shows a signifieant slope and
the guinea-pigs; no deviation fram linearity and parallelism of the
- where applicable, the number of animals that die in the dose-response curves (chapter 5.3 describes possible
4 groups of 5 injected with the challenge toxin solution alternatives if significant deviations are observed).
and its 3 dilutions indicates that the challenge dose was The test may be repeated but when more than 1 test is
approximately 100 LDso; performed the results of all valid tests must be combined in
- the confidence limits (P = 0.95) are not less than 50 per eent the estimate of poteney.
and not more than 200 per cent of the estimated potency; The following section is published for information.
- the statistical analysis shows no deviation from linearity
and parallelism. Assay of diphtheria vaccine (adsorbed):
The test may be repeated but when more than 1 test is guidelines
performed the results of all valid tests must be combined in
the estimate of potency. METHOD c. DETERMINATION OF ANTIBODIES IN
GUINEA-PIGS
METHOD C. DETERMINATION OF ANTIBODIES IN PREPARATION OF SERUM SAMPLES
GUINEA-PIGS For the preparation of serum samp1es, the following technique
SELECTION AND DISTRIBUTION OF THE TEST ANIMALS has been found to be suitable. Invert the tubes containing
Use in the test healthy guinea-pigs from the same stock, eaeh blood samp1es 6 times and allow to stand at 37 oC for 2 h,
weighing 250-350 g. Use guinea-pigs of the same sex or with then at 4 oC for 2 ho Centrifuge at room temperature at 800 g
males and females equally distributed between the groups. for 20 mino Transfer the serum to sterile tubes and store at
Distribute the guinea -pigs in not fewer than 6 equal groups; a temperature below - 20 oc. At least a 40 per cent yield of
use graups containing a number of animals sufficient to obtain serum is obtained by this procedure.
results that fulfil the requirements for a valid assay prescribed DETERMINATION OF ANTIBODY TITRE
below. Use a further group of non-vaccinated guinea-pigs The ELISA and Vera cell assays shown below are given as
of the same origin to provide a negative serum control. lf examples of immunochemical methods that have been found
test consistency has been demonstrated, a reference negative to be suitable for the determination of antibody titre.
serum control may be used.
Determination of antibody titre in guinea-pig serum by
REFERENCE PREPARATION enzyme-linked immunosorbent assay (ELISA). Dilutions of
Use a suitable reference preparation such as diphtheria vaccine test and reference sera are made on ELISA plates coated with
(adsorbed) BRP or a batch of vaccine shown to be effective in diphtheria toxoid. A positive guinea-pig serum control and a
clinical studies, or a batch representative thereof, and which negative guinea-pig serum control are included on each plate
has been calibrated in International Units with reference to monitor the assay performance. Peroxidase-conjugated
to diphtheria vaccine (adsorbed) BRP or the International rabbit or goat antibody directed against guinea-pig-IgG is
Standard for diphtheria toxoid (adsorbed). added, followed by a peroxidase substrate. Optical density is
DILUTION OF THE TEST AND REFERENCE measured and the relative antibody titre is calculated using the
PREPARA TIONS usual statistical methods (for example, 5.3).
Using a 9 giL solution of sodium chloride R as diluent, prepare Reagents and equipment
serial dilutions of the vaccine to be examined and the reference - ELISA plates: 96 wells, columns 1-12, raws A -Ho
preparation; series differing by 2.5- to 5- fold steps have been
- Diphtheria guinea-pig antiserum (for vaccines-human use)
found to be suitableo Use not fewer than 3 dilutions within the
(positive control serum), obtained by immunisation of
range of, for example, 0.5-16 IU/mL for the reference vaccine
guinea-pigs using diphtheria vaccine (adsorbed) BRP.
and within the range of, for example, 1:2 to 1: 125 for the
vaccine to be examined. Use the dilutions for immunisation - Peroxidase co njuga te. Peroxidase-conjugated rabbit or goat
preferably within 1 h of preparation. Allocate 1 dilution to antibody directed against guinea-pig IgG.
each group of guinea-pigs. - Diphtheria toxoid.

General Notices (1) apply to all monographs and other texts 239
2.7.6. of diphtheri.a vaccine (adsorbed) EUROPEAN PHARMACOPOEIA 8.0

- Carbonate coating bujfer pH 9.6. Dissolve 1.59 g of and


anhydrous sodium carbonate R and 2.93 g of sodium - Flat-bottomed tissue culture plates: 96 wells, columns 1-12,
hydrogen carbonate R in 1000 mL of water R. Distribute rows A-H.
into 150 mL bottles and sterilise by autoclaving at 121 oC
- 75 cm 2 tissue
for 15 mino
- Diphtheria toxin.
Phosphate-bujfered saline pH 7.4 (PES). Dissolve with
stirring 80.0 g of sodium chloride R, 2.0 g of potassium - Diphtheria guinea-pig antiserum (for vaccines-human use)
dihydrogen phosphate R, 14.3 g of disodiu11l hydrogen (positive control serum), obtained by immunisation of
phosphate dihydrate R and 2.0 g chloride R in with vaccine (adsorbed) BRP.
1000 mL of water R. Store at room temperature to prevent - Vero cells (African Green Monkey kidney cells). Cell
crystallisation. Dilute to 10 times its volume with water R passages from P2 to PI5 are suitable for use.
before use. Method 1. The diphtheria toxin causes a cytopathogenic effect
- Citrie acid solution. Dissolve 10.51 g of eitrie acid R in on Vero cells leading to cellular lysis. Antibodies directed
1000 mL of water R and adjust the solution to pH 4.0 with against diphtheria toxin may inhibit this cytopathogenic
a 400 giL solution of sodium hydroxide R. effect. Consequently, the potency of a diphtheria vaccine may
be indirectly determined with the help of this ceH culture
- Washing buffer. PBS containing 0.5 giL of polysorbate 20 R.
system if different serum dilutions from immunised animals
- Diluent blocking buffer. PBS containing 0.5 giL of are cultured with a constant toxin concentration. In the Vero
polysorbate 20 R and 25 giL of dried skimmed milk. ceH assay, colour indicates viable cells, red colour dead
- Peroxidase substrate. Shortly before use, dissolve 10 mg cells. When only part of the cells are dead, the colour may
of diammanium 2,2 ~azinobis(3-ethylbenzothiazoline- be orange.
6-sulfonate) R (ABTS) in 20 mL of citric acid solution. Reagents and
Immediately before use add 5 flL of strong hydrogen MEM. Minimum Essential Medium (MEM) with
peroxide solution R. Salts, without L-glutamine and sodium bicarbonate.
Method medium 199. Medium 199, with Hanks' Solution
The description below is given as an example of a suitable and L-glutamine, without sodium bicarbonate.
plate layout but others may be used. Wells lA-H are for - Foetal bovine serU11l.
negative control serum and wells 2A-H and 12A-H are for
- Sodium bicarbonate 7.5 per cent solutian.
positive control serum for assay monitoring. Wells 3-11A -H
are for 'test samples. solution: 2.5 per cent solutiol1.
Coat each well of the ELISA pi ates with 100 flL of diphtheria - EDTA solution: EDTA 0.02 per cent 1:5000)
toxoid solution (0.5 Lf/mL in carbonate coating buffer pH 9.6). solutioI1.
Allow to stand overnight at 4 oC in a hUl11id atl110sphere. To D-PBS. Dulbecco's phosphate buffered saline
avoid tel11perature gradient effects, do 110t stack more than without calcium, or magnesium.
4 plates high. On the following day, wash the plates thoroughly _ 200mlVI solLltion.
with washing buffer. Block the plates by addition of 100 liL
solution.
of diluent block buffer to each well. Incubate in a hum id
atl110sphere at 37 oC for 1 h. Wash the plates thoroughly culture medium. To 50 mL of modified MEM add
with washing buffer. Place 100 flL of diíuent block buffer of water R, 5 mL of L-glutamine 200 mM solution,
in each well of the plates, except those of row A. Prepare and 10 mL of sodium bicarbonate 7.5 per cent solution. To
suitable dilutions of negative controi serum, positive control 25 mL of this medium add 1.25 mL o(foetal bovine serum.
serum (from about 0.01 IU/mL) and test sera. Allocate the - Maintenance culture medium. Similar to the primary
negative control serUl11 to column 1, positive control serum culture medium except that 0.5 mL instead of 1.25 mL of
to columns 2 and 12 and test sera to columns 3-11 and add foetal bovine serum is added to 20 mL of the enriched
100 flL of each serum to the first 2 wells of the column to MEMmedium.
which it is allocated. Using a multichannel micropipette, make - Medium A. To 50.0 mL 01' modified medium 199 add
twofold serial dilutions from row B, down the plate to row H, 440.0 mL of water R, 5.0 mL of L-glutamine 200 mM
by transferring 100 IlL from one well to the neit well. Discard solution and 10.0 mL of sodium bicarbonate 7.5 per cent
100 IlL from the last row so that all wells contain 100 ~lL. solution.
Incubate at 37 oC for 2 h. Wash thoroughly with washing
- Medium B. To 150.0 mL of medium A add 3.0 mL of
buffer. Prepare a suitable dilution (a 2000-fold dilution has
foetal bovine serum and 0.3 mL of penicillin/streptomycin
been found to be suitable) of po"oxidase conjugate in diluent
solution.
block buHer and add 100 flL to each well. Incubate at 37 oC in
a humid atmosphere for 1 h. Wash the plates thoroughly with - Medium C. To 22.0 mL of medium A add 0.44 mL of foetal
washing buffer. Add 100 flL of pero xi das e substrate to each bovine serum and 0.44 mL of penicillin/streptomycin
well. Allow to stand at room temperature, protected from solution.
light, for 30 mino Read the plates at 405 nm in the same order Vero cells are cultured in tissue culture flasks (for example
as addition of substrate was made. 75 cm 2/250 mL) in an incubator at 36 ± 1 oC, 5 per cent CO 2
Determination of antibody titre in guinea-pig serum by and 90 per cent reiative humidity. Vero cells are first grown
Vero ceH assay. The method used relies either on metabolic in the primary culture medium. After 2-3 days of growth,
inhibition (method 1) or 011 cytotoxicity (method 2) as the the primary culture medium is replaced by the maintenance
end point, and on either microscopic (cel! morphology) or culture medium. When a confluent monolayer is obtained,
visual (colour) inspection ofthe cells. the culture supernatant is discarded and the celllayer washed
gently with modified D- PBS. Add a mixture of 1 volume of
The limit of detection is specific for each antitoxin and is trypsin solution and 1 volume of EDTA solutiol1 to the flask.
usually between 0.015 IulmL (method 1) and 0.05 IV/mL Swirl the flask gently and incubate in the CO 2 incubator for
(method 2). abont 3 min until the cells start to break from the monolayer.
The endpoint is taken as the highest serum dilution protecting tap the side of the flask to make the cells fall.
cells from the diphtheria toxin effect. The antitoxin activity the cells in 5-6 mL of fresh medium e to obtain
is calculated with respect to guinea-pig or WHO reference a homogeneous suspension. Prepare a cel! suspension in
standard, and expressed in International Units per millilitre. medium C containing 1 x 10 5 cells/mL.

240 See the section on general monographs (cover pages)


EUROPEAN PHARMACOPOEIA 8,0 2.7.6. Assay of diphtheria vacdne (adsorbed)

Place 25 flL of medium B in each well except those of potassium dihydrogen phosphate R in water R, and dilute
column L Place 25 flL of the diphtheria guinea-pig antiserum to 1000.0 mL with the same solvent, Adjust the pH if
(for vaccines-human use) (positive control serum, working necessary. Autoclave at 120 oC for 15 min,
dilution in medium B oí 0.40 IU/mL) in wells Al, A2 and Al L - Thiazolyl blue MTT solution, Dissolve 0,1 g of thiazolyl
Place 25 flL of guinea-pig serum samples in wells B-G of blue MTT in 20 mL of PBS, Sterilise by filtration (0,2 [lm)
columns 1, 2 and 11, Place 25 ¡.¡L of negative control serum and store in dark bottle,
in row H oí columns L 2 and 1 L Using a multichannel
- pH adjuster solution, Mix 40 mL of acetic acid R with
micropipette, make twoÍold serial dilutions across the plate
1.25 mL of 1 M hydrochlorie acid and 8,75 mL of water R.
(from column 2 up to column 10 for rows A-G and up
to column 8 for row H), Discard 25 flL from the wells in - Extraetíon buffer pH 4.7. Dissolve 10 g of sodium
column 10 in rows A-G, and from well H8, laurilsulfate R in water R and add 50 mL of
dimethylformamide R, and dilute to 100 mL with
Reconstitute the diphtheria toxin with saline solution to give water R. Adjust the pH with an appropriate volume of pH
a solution of 50 IU/mL Prepare a 50-fold dilution of this adjuster solution,
diphtheria toxin dilution in medium B to obtain a working
solution of LO IU /mL Add 25 flL of this working solution Vero cells are cultured in tissue culture flasks (for example
to wells A12 and B12 (toxin control), Make twoÍold serial 75 cm 2 /250 111L) in an incubator at 36 ± 1 oc, 5 per cent CO 2
dilutions by tranferring 25 flL from one well to the next, Írom and 90 per cent relative humidity. Vero cells are grown in
well B12 down to HIL Change the tip between each dilution. the complete culture medium, After 6-7 days of growth, a
Discard 25 flL from well H12, Add 25 flL of medium B to confluent monolayer is obtained, the culture supernatant
wells B12-H12, Then, place 25 flL of the working dilution of is discarded and the celllayer is washed 3 times with
the diphtheria toxin (LO IU/mL) in each well of rows A-H, trypsin-EDTA: gently pipette out the medium, add 0.5-1 mL
from column 1-10, except in wells H9 and HI0 (ceUs only, of trypsin -EDTA, swirl the flask and tip the trypsin out Do
without serum and without toxin), this twice, and the 3'd time, place the flask in the incubator
for 5 mil1 until the ceUs start to break from the monolayer.
Cover the plates with lids or sealer and shake gently, Incubate Vigorously tap the side of the flask to make the cells fal!.
the plates for at least 2 h in a humid container in a CO 2 Resuspend the cells in 6-25 mL offresh complete culture
incubator at 37 oC, Add 200 I1L of ceH suspension containing medium to obtain a homogeneous suspension, Prepare a
1 x 10 5 cells/mL to al! the wells, Cover the plates with cel! suspension in complete culture medium containing
sealer. Incubate at 37 oC for 5 days, Check for microbial approximately 4 x 10 5 cells/mL
contamination by microscopic examination,
Place 50 flL of complete culture medium in each well except
Yellow wells are recorded as negative and red wells indicate those of column L Place 100 I1L of diphtheria guinea pig
dead cells and are recorded as positive, A colour between antiserum (for vaccines-human use) (positive control serum,
yellow and red indicates a mixture of viable and dead cells working dilution in complete culture medium of O, 12 IU /mL)
and is recorded as positive/negative, The results based on the in well Al and 50 [1L in well Al!. Place 100 I1L of guinea
change in colour can be confirmed by reading viable and dead pig test serum samples, diluted if necessary, in wells Bl-G 1.
cells under the microscope, Add 50 flL ofthe same sample to wells Bll-GIl in the
The potency of the guinea-pig antiserum samples is obtained corresponding row, Place 100 flL oí negative control serum
by comparing the last well of the standard preparation in well Hl and 50 ¡.¡L in well Hl L Using a multi-channel
showing complete neutralisation of the toxin, with the last well micropipette, make twofold serial dilutions by transferring
of the sample demonstrating the same effect For calculations 50 flL Írom one well to the next working across the plate (from
of potency, it must be remembered that the endpoint may be column 1-10 for rows A-G and from column 1-8 for row H),
between a negative weH and a positive/negative welL Diphtheria toxin of known activity and Lf content is diluted
Method 2: Thiazoly! blue MTT is reduced to a blue/black to a suitable working stock containing at least 4 mínimum
formazan product by the mitochondrial dehydrogenase of cytopathic doses in complete culture medium, Add 50 I.lL of
viable ceUs, and thus serves as a quantitative measure of living the diluted toxin to each well except H9 and HIO (cel! control),
cells present, indicating when the toxin has been neutralised All-Hll (serum control) and A12-H12 (toxin control), Add
by the antitoxin, White or colourless wells indicate absence of 100 flL oí diluted toxin to well A12 and make twofold serial
viable cells due to insufficient antitoxin to neutralise the toxin, dilutions by transferring 50 I1L from one well to the next
working down the plate (from weli AI2-HI2), Discard 50 flL
Reagents and equipment
from well H12, Add 50 flL of complete medium 10 wells H9
- MEM (Minimal Essential Media), and HIO,
- Newborn calf serum, Cover the pIates with a lid or sealer and leave for 1 h at room
- Antibiotic solution (containing 10 000 units of penicillin, temperature to allow toxin neutralisation to occur, 50 flL of
10 mg of streptomycin and 25 flg of amphotericin B per ceH suspension containing approximately 4 x 10 5 cells/mL
millilitre), is added to each welL The plates are sealed and incubated
at 37 oC for 6 days, Check for microbial contamination by
- L-glutamine 200mM solution, microscopic examination, 10 flL of thyazolyl bIue MTT
- Trypsin-EDTA, solution is added to each we!L The pIates are incubated at
- Thiazolyl blue MTT [3-( 4,5-dimethylthiazol-2-yl)-2,5- 37 oC Íor a further 2-4 h, Then, the medium is removed and
diphenyltetrazolium bromide l, 100 flL of extraction buffer pH 4,7 is added to each welL
The plates are incubated at 37°C and left overnight to aid
- 1 M HEPES buffer pH 8,1, Dissolve 18,75 g ofHEPES in
the extraction process, Once extraction and solubilisation is
825 mL of water R and 30,0 mL of 2 M sodium hydroxide R.
complete, plates are visually examined or read at 570 nm,
- Glucase solution (lO per eent), Blue/black wells are recorded as negative (al! the ceUs are alive,
- Complete culture medium, Mix 200 mL of MEM with toxin neutralisation by antitoxin) and white or colourless wells
10 mL of newborn calf serum, 3,0 mL of 1 M HEPES buffer indicate dead ceHs (no toxin neutralisation) and are recorded
pH 8,1, 2.0 mL oí glucose solution (lO per cent), 2,0 mL as positive,
of antibiotic solution and 2,0 mL of L-glutamine 200mM The potency of the test antitoxin is obtained by comparing
solution, the last well of the reference antitoxin prepara tía n showing
- Phosphate-buffered saline pH 7.4 (PBS) , Dissolve 10,0 g neutralisation of the taxin, with the last well of the antitoxin
of sodium chloride R, 0.75 g of potassium chloride R, preparation demonstrating the same effect The neutralising
1.44 g of disodium hydrogen phosphate R, and 0,125 g of antibody titre of the sample being examined can be calculated

General Notices (1) apply to all mnnnl7raIJhs and other texts 241
2,7,7, of pel'tussis vaccine ceH) EUROPEAN PHARMACOPOEIA 8.0

multiplication oí the dilution factor with totalnumber of potency of the vaccine to be examined relative to the
International Units per millilitre of the reference preparabon oE the reference preparation on the basis of the
at the end point. The test is valid if all the cells in the toxin animals in each of the groups of not fewer than 16.
control are dead and reference antitoxin a neutralisation The test is not valícl unless:
in at least the first 2 dilutions tested. - for D01h the vaecine to be examined and the reference
preparation, the 50 pe!" cent dos e líes between
the largest and the smallest to the mielO;
07/20 H :20707 - the l1umber of animals that die in the 4
with the LH,",H;H~;C
2.7,7. PERTUSSIS VACCINE that the challenge close is
('VHOLE CELL) - the statistical shows no deviation from linearity
01' parallelism.
is determined The test may be but when more thall one test is
to protect mice against the the of al! valid tests must be combined.
pertussis, administered
of a reference preparation,
01/2008:20708
ntí:rIlatlOl¡lal Units, needed to give the same
correcíed 6.0
The International Unít is the aetivity eontained in a stated
amount of the International Standard which consists of
2.7.8. OF VACCINE
a quantity of dried pertussis vaecine. The equivalenee in (ADSORBED)
International Units oI' the International Standard is stated
the Worlcl Health Organization.
Selection and distribution of the test animals. Use in the
test mi ce les s than 5 weeks old of a suitable strain f1'om
the same stock, the difference in mass between the heaviest toxoid in the serum of the
and the being not greater than 5 g. Distribute the cases, the potency of the vaecine is
mice in 6 groups of not I'ewer than 16 and 4 groups of 10. The with a reference vaccine, calibrated in
miee must al! be of the same sex or the males and females methods A and B, in countries where the
clistributed between the groups. is not obligatory, the LDso method may be
Selection oí the chaHenge sírain and metlI0d, the number of animals and the are
challenge Select a suitable identical to those deseribed for the method, but the
eapable of the death oí mice within 14 is the death of the rather than
intracerebral If more than 20 per cent The International Unit is the containecl in a statecl
die within 48 of the the strain is not suitable. Make amount oI' the International for tetanus toxoid
one sub culture from the strain and the harvested B. The in International Units of
in a solution 10 R the International is stated by the World Health
and 6 giL of sodium chioride R and a pH to
7.2 or in another suitable solution. Determine the opacity Tetanus vaccine BRP is calibrated in International
oí" the a series of dilutions in the same Units with reference to the International Standard.
solution and allocate each dilution to a of 10 mice, The method chosen far the assay of tetanus vaccine
into eaeh mouse a mL or UC;I-'<;;lH~LO on the intended purpose. Method A or B is used:
allocated to its group, After 14 clays,
LH'I-"H~HC of a vaccine, to assay batches produced
eount the number of mice in each group. Prom
the results, ealculate the expectecl opaeity of a suspension to
eontaining 100 in each challenge close. Por the test of 2. wherever revalidation is needed
the vaccine to be examined make a fresh sub culture from the in the lH,\W,,"Ci,-" process.
and prepare a suspension of the Methocl A or B also be used I'or the routine
h'U"HJH'J with an opacity eorresponding to about batches of vaccine, in the interests of animal
100 LD 50 in eaeh close. Prepare 3 dilutions oí" the method C is used wherever
challenge suspension. Method e may be usecl, as specified uncler 1 and 2
Deíermination of potency. Prepare 3 serial dilutions of the aboye, after verificatiol1 of suitability of the method for
vaccine to be examined and 3 similar dilutions of the reference the produet. Por this a suitable number oí" batches
preparation such that in each the intermediate dilution (usually 3) are method Cana method A ol' B.
may be expectecl to protect about 50 per eent of the mice Where different vaccines
from the lethal effeets of the close of B. pertussis.
doses are 1/8, 1/40 and of the human dose of
the vaecine lo be examined and 0.5 IU, 0.1 IU and 0.02 IU
of the reference preparation, each close being contained in a
,"V.Ul¡JVJlH;;JL1l0 and for monovalent
volume not exeeeding 0.5 mL. Alloeate the 6 dilutions, one
to each of the groups oI' not fewer than 16 miee, and vaccines. Any combinations
intraperitoneally into eaeh mouse one close of the dilution eomponent or containing llaCll.lU,'H"U'
allocated to its group. After 14 - 17 days injeet intracerebrally vaccine with tetanus toxoid in be
into eaeh animal in the groups of not fewer than 16, one assessed separately.
dose of the suspension. Allocate the challenge Por combinations containing diphtheria and tetanus
and the dilutions made from it, one to each of components, the serological C) can be
the groups of 10 rnice, and inject intraeerebrally one dose of performed with the same group animals used for the
each into each mouse in the group to which that ""Jn>',",",m assay of the diphtheria vaccine

OU'IJ'Ci¡;".V'U is allocated. Exclude frarn consideration mice common immunisation conditions for the tetal1us
that die within 48 h of ehallenge. Count the number mice doses,
in eaeh of the groups aÍter 14 Calculate the to be valid for tbe combined vaccine.

242 See the


EUROPEAN PHARMACOPOEIA 8.0 2.7.8. oí tetanus vacdne (adsorbed)

The design of the assays described below uses multiple After 28 days, inject subcutaneously into each animal 1.0 mL
dilutions for the test and reference preparations. Based on of the challenge toxin solution (containing 50 times the 50 per
the potency data obtained in multiple-dilution assays, it cent paralytic dosel.
may be possible to reduce the number of animals needed to DETERMINATION OF THE ACTIVITY OF THE
obtain a statistically significant result by applying a simplified CHALLENGE TOXIN
model such as a single dilution for both test and reference
If necessary, allocate the 3 dilutions made from the challenge
preparations. Such a model enables the analyst to determine
toxin solution, 1 to each of the 3 groups of 5 guinea-pigs,
whether the potency of the test preparation is significantly
and inject subcutaneously 1.0 mL of each solution into each
higher than the minimum required, but does not give
guinea-pig in the group to which that solution is allocated.
information on the dose-response curves and their linearity,
The activity and stability of the challenge toxin are determined
parallelism and significant slope. The simplified model allows
by carrying out a suitable number of determinations of the
for a considerable reduction in the number of animal s required
50 per cent paralytic do se. It is then not necessary to repeat
and must be considered by each analyst in accordance with
the determinatiol1 for each assay.
the provisions of the European Convention for the Protection
of Vertebrate Animals Used for Experimental and Other READING AND INTERPRETATION OF RESULTS
Scientific Purposes. Examine the guinea-pigs twice daily. Remove and euthanise
Where a single-dilution assay is used, production and test al! animals showing definite signs of tetanus paralysis. Count
consistency over time are monitored via suitable indicators the number of guinea-pigs without paralysis 5 days after
and by carrying out a full multiple-dilution assay periodically, injection of the challenge toxin. Calculate the potency of the
for example every 2 years. For serological assays, suitable vaccine to be examined relative to the potency of the reference
indicators to monitor test consistency are: preparation on the basis of the proportion of challenged
animals without paralysis in each group of vaccinated
- the mean and standard deviation of relative antitoxin guinea-pigs, using the usual statistical methods (for example,
litres or scores of the serum samples obtained after 5.3).
administration of a fixed dose of the vaccine reference
preparation; REQUIREMENTS FOR A VALID ASSA y
The test is not valid unless:
- the antitoxin titres or scores of run control s (positive and
negative serum samples); - for both the vaccine to be examined and the reference
preparation, the 50 per cent protective dose líes between
- the ratio of antitoxin titres or scores for the positive
the largest and smallest doses of the preparations given to
serum control to the serum samples corresponding to the
the guinea -pigs;
reference vaccine.
- where applicable, the number of paralysed animals
METHOD A. CHALLENGE TEST IN GUINEA-PIGS in the 3 groups of 5 injected with the dilutions of the
challenge toxin solution indicates that the challenge was
SELECTION AND DISTRIBUTION OF THE TEST ANIMALS
approximately 50 times the 50 per cent paralytic dose;
Use in the test healthy guinea-pigs from the same stock,
each weighing 250-350 g. Use guinea-pigs of the same sex - the confidence lirnits (P", 0.95) are not less than 50 per cent
or with males and females equaliy distributed between the and not more than 200 per cent of the estimated potency;
groups. Distribute the guinea-pigs in 110t fewer than 6 equal - the statistical analysis shows a significant slope and
groups; use groups containing a number of animals sufficient no deviation from linearity and parallelism of the
to obtain results that fulfil the requirements for a valid assay dose-response curves (chapter 5.3 describes possible
prescribed below. If the activity of the challenge toxin has to alternatives if significant deviations are observed).
be determined, include 3 further groups of 5 guinea-pigs as The test may be repeated but when more than 1 test is
unvaccinated controls. performed the results of al! valid tests must be combined in
SELECTION OF THE CHALLENGE TOXIN the estimate of potency.
Select a preparation of tetanus toxin containing not less than
50 times the 50 per cent paralytic dose per millilitre. If the METHOD B. CHALLENGE TEST IN MICE
challenge toxin preparation has been shown to be stable, it is SELECTION AND DISTRIBUTION OF THE TEST ANIMALS
not necessary to verify the paralytic dose for every assay.
Use in the test healthy mice from the same stock, about
PREPARATION OF THE CHALLENGE TOXIN SOLUTION 5 weeks old and from a strain shown to be suitable. Use mice
Immediately before use, dilute the challenge toxin with a of the same sex or with males and females equally distributed
suitable diluent (for example, peptone buffered saline solution between the groups. Distribute the mice in 110t fewer than
pH 7.4) to obtain a stable challenge toxin solution containing 6 equal groups; use groups containing a number of animals
approximately 50 times the 50 per cent paralytic dose per sufficient to obtain results that fulfil the requirements for a
millilitre. If necessary, use portiol1s of the challenge toxin valid assay prescribed below. If the challenge toxin to be used
solution diluted 1 to 16, 1 to 50 and 1 to 160 with the same has not been shown to be stable or has not been adequately
diluent to determine the activity of the toxin. standardised, include 3 further groups of 110t fewer than
5 mice to serve as unvaccinated contro15.
DILUTION OF THE TEST AND REFERENCE
PREPARATIONS SELECTION OF THE CHALLENGE TOXIN
Using a 9 giL solution of sodium chloride R, prepare dilutions Select a preparation of tetanus toxin containing 110t less than
of the vaccine to be examined and of the reference preparation, 100 times the 50 per cent paralytic dose per millilitre. lf the
such that for each, the dilutions form a series differing by challenge toxin preparation has been shown to be stable, it is
not more than 2.5-fold steps and in which the intermediate not necessary to the paralytic dose for every assay.
dilutions, when injected subcutaneously at adose of 1.0 mL per PREPARATION OF THE CHALLENGE TOXIN SOLUTION
guinea-pig, protect approximately 50 per cent of the animals
Immediately before use, dilute the challenge toxin with a
from the paralytic effects of the subcutaneous injection of the
suitable diluent (for example, peptol1e buffered saline solution
qual1tity of tetanus toxin prescribed for this test.
pH 7.4) to obtain a stable challenge loxin solution containing
IMMUNISA TION AND CHALLENGE approximately 50 times the 50 per cent paralytic dose in
Allocate the dilutions, 1 to each of the groups of guinea-pigs, 0.5 mL. If necessary, use portions of the challenge toxin
and inject subcutaneously 1.0 mL of each dilution into each solution diluted 1 to 16, 1 to 50 and 1 lo 160 with the same
guinea-pig in the group to which that dilution is allocated. diluent to determine the activity of the toxin.

General Notices (1) apply lo all monographs and other texts 243
2.7.8. Assay of tetanus vaccine (adsorbed) EUROPEAN PHARMACOPOEIA 8.0

DILUTION OF THE TEST AND REFERENCE REFERENCE PREPARATION


PREPARATIONS Use a suitable reference preparation such as tetanus vaccine
Using a 9 giL solution of sodium chloride R, prepare dilutions (adsorbed) BRP or a batch of vaccine shown to be effective in
of the vaccine to be examined and of the reference preparation, clinical studies, or a batch representative thereof, and which
such that for each, the dilutions form a series differing by has been calibrated in International Units with reference to
not more than 2.5-fold steps and in which the intermediate tetanus vaccine (adsorbed) BRP or the International Standard
dilutions, when injected subcutaneously at adose of 0.5 mL for tetanus toxoid (adsorbed).
per mouse, protect approximately 50 per cent of the animals DILUTION OF THE TEST AND REFERENCE
from the paralytic effects of the subcutaneous injection of the PREPARATIONS
quantity of tetanus toxin prescribed for this test. Using a 9 giL solution of sodium chloride R as diluent,
IMMUNISATION AND CHALLENGE prepare serial dilutions of the vaccine to be examined and the
Allocate the dilutions, 1 to each of the groups of mice, and reference preparation; series differing by 2.5- to 5-fold steps
inject subcutaneously 0.5 mL of each dilution into each mouse have been found to be suitable. Use 110t fewer than 3 dilutions
in the group to which that dilution is allocated. After 28 days, withil1 the range of, for example, 0.5-16 IU/mL for each series.
inject subcutaneously into each animal 0.5 mL of the challenge Use the dilutions for immunisation preferably within 1 h of
toxin solution (containing 50 times the 50 per cent paralytic preparation. Allocate 1 dilution to each group of guinea-pigs.
dosel. IMMUNISATION
DETERMINATION OF THE ACTIVITY OF THE Inject subcutaneously to each guinea-pig 1.0 mL of the
CHALLENGE TOXIN dilution allocated to its group.
If necessary, allocate the 3 dilutions made from the challenge BLOOD SAMPLING
toxin solution, 1 to each of the 3 groups of not fewer than 35-42 days after immunisation, take a blood sample from each
5 mice, and inject subcutaneously 0.5 mL of each solution into vaccinated and control guinea-pig using a suitable method.
each mouse in the group to which that solution is allocated.
PREPARATION OF SERUM SAMPLES
READING AND INTERPRETATION OF RESULTS Avoid frequent freezing and thawing of serum samples. To
Examine the mice twice daily. Remove and euthanise all avoid microbial contamination, it is preferable to carry out
animals showing definite signs of tetanus paralysis. Count the manipulations in a laminar-flow cabinet.
number of mice without paralysis 4 days after injection of the
DETERMINATION OF ANTIBODY TITRE
challenge toxin. Calculate the potency of the vaccine to be
examined relative to the potency of the reference preparation Determine the relative antibody titre or score of each serum
on the basis of the proportion of challenged animals without sample by a suitable immunochemical method (2.7.1). The
paralysis in each group of vaccinated mice, using the usual methods shown below (enzyme-linked immunosorbent assay
statisticalmethods (for example, 5.3). (ELISA) and toxin-binding inhibition (ToBI)) have been
found to be suitable.
REQUIREMENTS FOR A VALID ASSAY
CALCULA TION OF POTENCY
The test is not vaJid un les s :
Calculate the potency of the vaccine to be examined in
- for both the vaccine to be examined and the reference International Units relative to the reference preparation, using
preparation, the 50 per cent protective dose hes between the usual statistical methods (for example, 5.3).
the largest and smallest doses of the preparations given to REQUIREMENTS FOR A VALID ASSAY
the mice; The test is 110t valid unless:
where applicable, the number of paralysed animals in the - the confidence limits (P = 0.95) are 110t less than 50 per cent
3 groups of not fewer than 5 injected with the dilutions of and not more than 200 per cent of the estimated potency;
the challenge toxin solution, indicates that the challenge - the statistical analysis shows a significant slope and
dose was approximately 50 times the 50 per cent paralytic no deviation from lil1earity and parallelism of the
dose; dose-response curves (chapter 5.3 describes possible
alternatives if significant deviations are observed).
- the confidence limits (P = 0.95) are not less than 50 per cent
and not more than 200 per cent of the estimated potency; The test may be repeated but when more than 1 test is
performed the results of al! valid tests must be combined in
- the statistical analysis shows a significant slope and the estimate of potency.
no deviation from linearity and parallelism of the
dose-response curves (chapter 5.3 describes possible The following section is published for information.
alternatives if significant deviations are observed).
The test may be repeated but when more than 1 test is Assay of tetanus vaccine (adsorbed):
performed the results of all valid tests must be combined in
the estimate of potency. guidelines
METHOD A. CHALLENGE TEST IN GUINEA-PIGS
METHOD C. DETERMINATION OF ANTIBODIES IN READING AND INTERPRETATION OF RESULTS
GUINEA-PIGS In order to minimise suffering in the test animaIs, it is
SELECTION AND DISTRIBUTION OF THE TEST ANIMALS
recommended to note the degree of paralysis on a scale
such as that shown beJow. The scale gives typical signs
Use in the test healthy guinea-pigs from the same stock, each when subcutaneous injection of the challenge toxin is made
weighing 250-350 g. Use guinea-pigs of the same sex or with mid-ventrally, directly behind the sternum with the needIe
males and females equally distributed between the groups. pointing towards the neck of the guinea-pig. Grade T3 is taken
Distribute the guinea -pigs in not fewer than 6 equal groups; as the end-point, but with experience grade T2 can be used
use groups containing a number of animals sufficient to obtain instead. Tetanus toxin produces in at least 1 of the forelimbs
results that fulfil the requirements for a valid assay prescribed paralysis that can be recognised at an early stage. The tetanus
below. Use a further group of non-vaccinated guinea-pigs grades in guinea-pigs are characterised by the following signs:
of the same origin to provide a negative serum control. If
test consistency has been demonstrated, a reference negative - Tl: slight stiffness of 1 forelimb, but difficult to observe;
serum control may be used. - T2: paresis of 1 forelimb which still can function;

244 See the information section on general monographs (caver pages)


EUROPEAN PHARMACOPOEIA 8.0 2.7.8. Assay of tetanus vaccine (adsorbed)

- T3: paralysis of 1 forelimb. The animal moves reluctantly, phosphate dihydrate R and 2.0 g of potassium chloride R in
the body is often slightly banana -shaped owing to scoliosis; 1000 mL of water R. Sto re at room temperature to prevent
- T 4: the forelimb is completely stiff and the toes are crystallisation. Dilute to 10 times its volume with water R
immovable. The muscular contraction of the forelimb is before use.
very pronounced and usually scoliosis is observed; - Citric aeid solution. Dissolve 10.51 g of citrie aeid R in
- T5: tetanus seizures, continuous tonic spasm of muscles; 1000 mL of water R and adjust the solution to pH 4.0 with
- D: death. a 400 giL solution of sodium hydroxide R.
- Washing buffer. PES containing 0.5 giL of polysorbate 20 R.
METHOD B. CHALLENGE TEST IN MICE
- Diluent block buffer. PBS containing 0.5 giL of
READING AND INTERPRETATION OF RESULTS polysorbate 20 R and 25 giL of dried skimmed milk.
In order to minimise suffering in the test animals, it is - Peroxidase substrate. Shortly before use, dissolve 10 mg
recommended to note the degree of paralysis on a scale such as of diammonium 2,2 '-azinobis(3-ethylbenzothiazoline-
that shown below. The scale gives typical signs when injection 6-sulfonate) R (ABTS) in 20 mL of citric acid solution.
of the challenge toxin is made in the dorsal region, close to Immediately before use add 5 flL of strong hydrogen
one of the hind legs. Grade T3 is taken as the end-point, but peroxide solution R.
with experience grade T2 can be used instead. Tetanus toxin
produces in the toxin-injected hind leg paresis followed by Method
paralysis that can be recognised at an early stage. The tetanus The description below is given as an example of a suitable
grades in mice are characterised by the following signs: plate layout but others may be used. Wells 1A-H are for
- TI: slight stiffness of toxin-injected hind leg, only observed negative control serum and wells 2A-H and 12A-H are for
when the mouse is lifted by the tail; positive control serum for assay monitoring. Wells 3-11A-H
are for test samples.
- T2: paresis of the toxin-injected hind leg, which still can
function for walking; Coat each well of the ELISA plates with 100 flL of tetanus
toxoid solution (0.5 Lf/mL in carbonate coating buffer pH 9.6).
- T3: paralysis of the toxin-injected hind leg, which does not
AlIow to stand overnight at 4 oC in a humid atmosphere. To
function for walking;
avoid temperature gradient effects, do not stack more than
- T4: the toxin-injected hind leg is completely stiffwith 4 plates high. On the following day, wash the plates thoroughly
immovable toes; with washing buffer. Block the plates by addition of 100 flL
- T5: tetanus seizures, continuous tonic spasm of muscles; of diluent block buffer to each well. Incubate in a humid
- D: death. atmosphere at 37 oC for 1 h. Wash the plates thoroughly
with washing buffer. Place 100 flL of diluent block buffer
METHOD e. DETERMINATION OF ANTIBODIES IN in each well of the plates, except those of row A. Prepare
GUINEA-PIGS suitable dilutions of negative control serum, positive control
PREPARATION OF SERUM SAMPLES serum (from about 0.01 IU/mL) and test sera. Allocate the
For the preparation of serum samples, the following technique negative control serum to column 1, positive control serum
has been found to be suitable. Invert the tubes containing to columns 2 and 12 and test sera to columns 3-11 and add
blood samples 6 times and allow to stand at 37 oC for 2 h, 100 flL of each serum to the first 2 wells of the column to
then at 4 oC for 2 h. Centrifuge at room temperature at 800 g which it is allocated. Using a multichannel micropipette, make
for 20 mino Transfer the serum to sterile tubes and sto re at twofold serial dilutions from row B down the plate to row H,
a temperature below - 20 De. At least a 40 per cent yield of by transferring 100 flL from one well to the next. Discard
serum is obtained by this pro ce dure. 100 flL from the last row so that al! wells contain 100 flL.
Incubate at 37 oC for 2 h. Wash thoroughly with washing
DETERMINATION OF ANTIBODY TITRE
buffer. Prepare a suitable dilution (a 2000-fold dilution has
The ELISA and ToBI tests shown below are given as examples
been found to be suitable) of peroxidase conjugate in diluent
of immunochemical methods that have been found to be
block buffer and add 100 flL to each well. Incubate at 37 oC in
suitable for the determination of antibody titre. a hum id atmosphere for 1 h. Wash the plates thoroughly with
Determinatlon of antibody titre in guinea-pig serum by washing buffer. Add 100 flL of peroxidase substrate to each
enzyme-linked immunosorbent assay (EUSA). Dilutions of well. Allow to stand at room temperature, protected from
test and reference sera are made on ELISA plates coated with Iight, for 30 mino Read the plates at 405 nm in the same order
tetanus toxoid. A positive guinea-pig serum control and a as addition of substrate was made.
negative guinea-pig serum control are included on each plate
Determination of antibody titre in gui.nea-pig serum by
to monitor the assay performance. Peroxidase-conjugated
toxin- or toxoid-binding inhibition (ToBI). Tetanus toxin
rabbit or goat antibody directed against guinea-pig-IgG is
or toxoid is added to serial dilutions of test and reference
added, followed by a peroxidase substrate. Optical density is
sera; the serum/antigen mixtures are incubated overnight.
measured and the relative antibody titre is calculated using the
To determine unbound toxin or toxoid, the mixtures are
usual statistical methods (for example, 5.3).
transferred to an ELISA plate coated with tetanus antitoxin.
Reagents and equipment Peroxidase-conjugated equine anti-tetanus IgG is added
- ELISA plates: 96 wells, columns 1-12, rows A-H. followed by a peroxidase substrate. Optical density is
- Clostridium tetani guinea-pig antiserum (for measured and the antibody titre is calculated using the usual
vaecines-human use) BRP (positive control serum). statistical methods (for example, 5.3). A positive control
- Peroxidase conjugate. Peroxidase-conjugated rabbit or goat serum and a negative control serum are included on each plate
antibody directed against guinea-pig IgG. to monitor assay performance.
- Tetanus toxoid. Reagents and equipment
Carbonate coating buffer pH 9.6. Dissolve 1.59 g of - Round-bottomed, rigid polystyrene mierotitre plates.
anhydrous sodium carbonate R and 2.93 g of sodium - Flat-bottomed ELISA plates.
hydrogen carbonate R in 1000 mL of water R. Distribute - Tetanus toxin or tetanus toxoid.
into 150 mL bottles and sterilise by autodaving at 121 oC
for 15 mino - Clostridium tetani guinea-pig antiserum (for
vaecines-human use) BRP (positive control serum).
- Phosphate-buffered saline pH 7.4 (PBS). Dissolve with
stirring 80.0 g of sodium ehloride R, 2.0 g of potassium - Equine anti-tetanus IgG.
dihydrogen phosphate R, 143 g of disodium hydrogen - Peroxidase-conjugated equine anti-tetanus IgG.

General Notices (1) apply to all monographs and other texts 245
2.7.9. Test for Fe funetion of immunoglobulin EUROPEAN PHARMACOPOEIA 8.0

- Carbonate buffer pH 9.6. Dissolve 1.5 g oE anhydrous 07/2009:20709


sodium carbonate R, 2.39 g of sodium hydrogen carbonate R corrected 7.6
and 0.2 g of sodium azide R in 1000 mL of water R, adjust
to pH 9.6 and autoclave at 121 oC for 20 mino 2.7.9. TEST FOR Fe FUNCTION OF
- Sodium acetate buffer pH 5.5. Dissolve 90.2 g of anhydrous IMMUNOGLOBULIN
sodium acetate R in 900 mL of water R, adjust to pH 5.5
using a saturated solution of citric acid monohydrate R and The test for Fe function of immunoglobulin is carried out using
dilute to 1000 mL with water R method A or B. Method B is an adaptation of the procedure of
method the use of microtitre plates for the measurement
- Phosphate-buffered saline pH 7.2 (PBS). Dissolve 135.0 g of of complement-mediated haemolysis. Differences in the test
sodium chloride R, 20.55 g oí disodium hydrogen phosphate procedures between methods A and B are addressed in the test.
dihydrate R and 4.80 g of sodium dihydrogen phosphate
monohydratc R in water R and dilute to 15 L with the same REAGENTS
solvent. Autoclave at 100 oC for 60 mino
Stabilised human blood. Colleet group O human blood into
- Diluent buffer. PBS containing 5 giL of bovine albumin R ACD anticoagulant solution. Sto re the stabilised bIood at 4 oC
and 0.5 giL oí polysorbate 80 R for not more than 3 weeks.
- Block buffer. PBS containing 5 giL of bovine albumin R Phosphate-buffered saline pH 7.2. Dissolve 1.022 g of
anhydrous disodium hydl'Ogen phosphate R, 0.336 g of
- Tetramethylbcnzidillc solution. 6 giL solution oí anhydrous sodium dihydrogen phosphate R and 8.766 g of
tetramethylbenzidinc R in ethanol (96 per cent) R The sodium chloride R in 800 mL of water R and dilute to 1000 mL
substance dissolves within 30-40 min at room temperature. with the same solvento
- Peroxidase substrate. Mix 90 mL of water R, 10 mL oí sodium Magnesium and calcium stock solution. Dissolve 1.103 g oí
acetate buffer pH 5.5, 1.67 mL oí tetramethylbenzidine calcium chloride R and 5.083 g of chloride R in
solution and 20 ~lL of strong hydrogen peroxide solutíon R. water R and dilute to 25 mL with the same solvent.
- Washing solution. Tap water containing 0.5 giL of Barbital bu.ffer stock so/ution. Dissolve 207.5 g of sodium
chloride R and 25.48 g of barbital sodium R in 4000 mL of
polysorbate 80 R.
water R and adjust to pH 7.3 using 1 M hydroch/oric acid.
Method Add 12.5 mL of magnesium and calciul11 stock solution and
dilute to 5000 mL with water R. Store at 4 oC in transparent
Block the microtitre pIates by placing in each well 150 flL of containers.
block buffer. Cover the plates with a lid or sealer. Incubate
in a humid atmosphere at 37 oC for 1 h. Wash the plates Albumin barbita/ buffer so/u/ion. Dissolve 0.150 g of bovine
thoroughly with washing solution. Place 100 ~lL of PBS albumin R in 20 mL ofbarbital buffer stock solution and dilute
in each well. Place 100 ~lL of reference guinea -pig tetanus to 100 mL with water R. Prepare immediately before use.
antitoxin in the first well of a roW. Place 100 ~lL oí undiluted Tan71ic acid solutio71. Dissolve 10 mg oí tannic acid R in
test sera in the first well of the required number of rowS. Using 100 111L of phosphate-buffered saline pH 7.2. Prepare
a multichannel micropipette, make twofold serial dilutions immediately before use.
across the plate (up to column 10), by transferring 100 I1L from Guinea-pig Prepare a pool oí serum from the
one well to the next. Discard 100 flL from the last column so blood of not fewer than 10 guinea-pigs. the serum
that al! wells contain 100 flL. Prepare a 0.1 Lf/mL solution of from the clotted blood by centrifugation at about 4 oc. Sto re
tetanus toxin 01' toxoid using PBS as diluent. Add 40 flL of the serum in small amounts below - 70 oc. Immediately
this solution to each well except those of column 12. The wells before starting complement-initiated haemolysis, dilute to
of column 11 are a positive control. Add 40 I1L of PBS to the 125-200 CH so per millilitre with albumin barbital buffer
wells of column 12 (negative control). Shake the pI ates gently solution and store in an ice-bath during the test.
and cover them with lids. Coat the ELISA plates: immediately Rubella antígeno Suitable rubella antigen Íor
before use make a suitable dilution of equine anti-tetanus haemagglutination-inhibition titre (HIT). Titre > 256 HA units.
IgG in carbonate buÍfer pH 9.6 and add 100 [lL to each well.
Incubate the 2 series of plates overnight in a hum id atmosphere Preparation of tanned human red blood ceUs. Separate
at 37 oc. To avoid temperature gradient effects, do not stack human red blood cells by centrifuging an appropriate volume
more than 4 plates high. Cover the plates with lids. On the of stabilised human blood, wash the cells at least 3 times
following day, wash the ELISA plates thoroughly with washing with phosphate-bufÍered saline pH 7.2 and suspend at 2 per
solution. Block the plates by placing in each well125 1110 of cent V/V in phosphate-buffered saline pH 7.2. Add 0.2 mL oí
block buffer. Incubate at 37 oC in a humid atmosphere for 1 h. tannic acid solution to 14.8 mL of phosphate-buffered saline
Wash the plates thoroughly with washing solution. Transfer pH 7.2. Mix 1 volume of the freshly prepared dilution with
100 I1L of the pre-incubation mixture from the polystyrene 1 volume of the human red blood cel! suspension and incubate
plates to the corresponding wells of the ELISA plates, starting at 37 oC for 10 mino Collect tIle cells by centrifugation (800 g
with column 12 and then continuing from 1 to 11. Cover the for 10 min), discard the supernatant and wash the cells once
plates with a lid. Incubate at 37 oC in a humid atmosphere for with phosphate-buffered saline pH 7.2. Resuspend the tanned
2 h. Wash the ELISA plates thoroughly with washing solution. cells at 1 per cent V/V in phosphate-buffered saline pH 7.2.
Make a suitabIe dilution (a 4000-fold dilution has been Antigen coating of tanned human red blood ceUs. Take a
found to be suitable) of the peroxidase-conjugated equine suitable volume (VJ of tanned cells, add 0.2 mL of rubella
anti-tetanus IgG in diluent buffer. Add 100 flL ofthe dilution antigen per 1. O mL of tanned cells and incubate at 37 oC for
to each well and cover the plates with a lid. Incubate at 37 oC 30 mino Collect the cells by centrifugation (800 g for 10 min)
in a humid atmosphere Íor 1.5 h. Wash the ELISA plates and discard the supernatant. Add a volume of albumin barbital
thoroughly with washing solution. Add 100 ~lL of peroxidase buffer solution equivalent to the discarded supernatant,
substrate to each well. A bIne colour develops. Incubate the resuspend and collect the cells as described and repeat the
plates at room temperature. Stop the reaction at a given time washing procedure. Resuspend with albumin barbital buffer
(within 10 min) by the addition of 100 ~lL of 2 !vI sulfuric acid solution using a volume equivalent to 3/4 of Vs' thereby
to each well in the same order as the addition oí substrate. The obtaining the initial volume (VJ Mix 900 flL of albumin
colour changes from blue to yellow. Measure the absorbance barbital buffer solutiOl1 with 100 flL of Vi' which is thereby
at 450 11m immediately after addition of the sulfuric acid 01' reduced to the residual volume (VJ, and determine the initial
maintain the plates in the dark until reading. absorbance at 541 nm (A). Dilute V r by a factor equal to A

246 See the monographs (ca ver pages)


EUROPEAN PHARMACOPOEIA 8.0 2.7.10. Assay ofhuman coagulation factor VII

using albumin barbital buffer solution, thereby obtaining the largest value for S serves as Sexp' In addition, determine the
final adjusted volume V f '" V,. x A of sensitised human red absorbance at the start of measurement (A) by extrapolating
blood cells and adjusting Ato 1.0 ± 0.1 for a tenfold dilution. the curve, which is almost linear and parallel to the time axis
Antibody binding of antigen-coated tanned human red within the first few minutes. Correct Sexp using the expression:
blood cells. Prepare the following solutions in succession and S' = Sexp
in duplicate, using for each solution a separate half-micro As
cuvette (for example, disposable type) 01' test-tube.
(1) Test solutions. If necessary, adjust the immunoglobulin to Calculate the arithmetic mean of the values of S' for each
be examined to pH 7. preparation (test and reference solution). Calculate the index
of Fe functiol1 (I F) from the expression:
Where method A is performed, dilute volumes of the
preparation tobe examined with albumin barbital buffer to
obtain 30 mg and 40 mg of immunoglobulin and adjust the
volume to 900 I1L with albumin barbital buffer.
Where method B is performed, dilute volumes of the arithmetic mean of the corrected slope for the
preparation to be examined with albumin barbital buffer to S'
preparation to be examined;
obtain 15 mg and 30 mg of immunoglobulin and adjust the
volume to 1200 ¡..tL with albumin barbital buffer. S's arithmetic mean of the corrected s10pe for the
reference preparation;
(2) ReÍerence solutions. Prepare as for the test solutions using
human immunoglobulin (Fe Íunction and molecular size) BRP. arithmetic mean of the corrected s10pe for the
(3) Complement control. Albumin barbital buffer solution. complement controL
Where method A is performed, add to each cuvette/test-tube Calculate the index of Fc function for the preparation to be
100 I1L of sensitised human red blood cells and mix well. examined: the value is not less than that stated in the leaflet
Allow to stand for 15 min, add 1000 I1L of albumin barbital accompanying the reference preparation.
buffer solution, collect the cells by centrifugation (l000 g for
10 min) of the cuvette/test-tube and remove 1900 I1L of the
supernatant. Replace the 1900 IlL with albumin barbital buffer
solution and repeat the whole of the washing procedure, o1/2008:2071 O
finally leaving a volume of 200 ¡..tL Test samples may be stored
in sealed cuvettes/test-tubes at 4 oC for not longer than 24 h. 2.7.10. ASSAY OF HUMAN
Where method B is performed, add to each test-tube
300 ¡..tL of sensitised human red blood cells and mix well
COAGULATION FACTOR VII
(the final immunoglobulin concentration is in the range Human coagulation factor VII is assayed by its biological
of 10-20 mg/mL). Allow to stand for 15 min, add 1500 ¡..tL activity as a factor VIIa-tissue factor complex in the activation
of albumin barbital buffer solution and stir gently until of factor X in the presence of calcium ions and phospholipids.
homogeneous. Collect the cells by eentrifugation (l000 g The potency of a factor VII preparation is estimated by
for 10 min) of the test-tube, remove the supernatant and comparing the quantity necessary to achieve a certain rate of
add approximately 3 mL of albumin barbital buffer solution. factor Xa formation in a test mixture containing the substances
Repeat this operation up to 4 times in total, leaving a final that take part in the activation of factor X, and the quantity
volume of 300 I1L Test samples may be stored in sealed of the International Standard, or of a reference preparation
test-tubes at 4 oC for not longer than 24 h. calibrated in International Units, required to produce the
Complement-initiated haemolysis. same rate of factor Xa formation.
To measure haemolysis where method A is performed, add The International Unit is the factor VII activity of a stated
600 I1L of albumin barbital buffer solution warmed to 37 oC amount of the International Standard, which consists of
to the test sample, resuspend the cells carefully by repeated freeze-dried plasma. The equivalence in International Units
pipetting (11ot fewer than 5 times) and place the cuvette in of the International Standard is stated by the World Health
the thermostatted cuvette holder of a spectrophotometer. Organization.
After 2 min, add 200 ¡..tL of diluted guinea-pig complement Human coagulation Íactor VII concentrate BRP is calibrated
(125-200 CHso/mL), mix thoroughly by pipetting twice in International Units by comparison with the International
and start immediately after the second pipetting the Standard.
time-dependent recording of absorbance at 541 nm, using
The chromogenic assay method consists of 2 consecutive
albumin barbital buffer solution as the compensation liquido
steps: the factor VII -dependent activation of factor X reagent
Stop the measurement if absorbance as a function of time has
mixture containing tissue factor, phospholipids and calcium
clearly passed the inflexion point.
ions, followed by enzymatic cleavage of a chromogenic
To measure haemolysis where method B is performed, add factor Xa substrate into a chromophore that can be quantified
900 flL of albumin barbital buffer solution warmed to 37 oC spectrophotometrically. Under appropriate assay conditions,
to each test-tube and resuspend the cells carefully by repeated there is a linear relation between the rate of factor Xa
pipetting (not fewer than 5 times). The microtitre pIate must formation and the factor VII concentration. The assay is
be prewarmed to 37 oC before starting the test. Transfer 240 ¡..tL summarised in the following scheme.
of each solution into 4 microtitre plate wells then incubate
the micropIate at 37 oC for 6 min, stirring gentIy every 10 S. Step 1
To each microtitre plate well add 60 fJ.L of diluted guinea-pig tissue factor, Ca 2+
complement (150 CHso/mL). Mix Íor 10 s and immediately a) factor VII --------.:.~----)\>1\Io_ factor Vlla
start recording the absorbance at 541nm at 37 oC, measuring
every 20 S. Stop the measurement if the absorbance as a
factor Vlla, Ca 2+
function of time has clearly passed the inflexiol1 point. b) factor X ______________________-)ol\lo_faclorXa
Evaluation. For each cuvette/test-tube/well, determine the tissue faclor/phospholipid
slope (5) of the haemolysis curve at the approximate inflexion
point by segmenting the steepest section in suitable time Step 2
intervals (for example, lit '" 1 min), and calculate S between
chromogenic substrate factor xa)\> peptide + chromophore
adjaeent intersection points, expressed as L'l.A per minute. The

General Notices (1) to all monographs and other texts 247


2.7.11. of human coagulation factor IX EUROPEAN PHARMACOPOEIA 8.0

be obtained commercially
from a sources. the composition of
individual reagents may be subject to some variation, their
essential features are described in the following specification.

REAGENTS a spectrophotometer, either


The coagulation factor reagent purified proteins continuously, thus the initial rate of substrate c1eavage
derived from human or bovine sources. These include to be calculated, or terminating the reaction after
factor X and tissue factor/phospholípid as a suitable interval the addition of a
factor VII activator. proteins are partly purified and suitable
do not contain that interfere with the activation citrate time
of factor VII or factor X. Factor X is present in amounts to achieve a linear
a final concentration during the first step of the assay Appropriate times are
14-70 nmol/L. Thromboplastin 15 min, but deviations are permissible
from natural sources (bovine or rabbit brain) or synthetic dose- response relationship is thus obtained.
may be used as the tissue factor/phospholipid Check the of the assay and calculate the potency of the
component. suitable for use in prothrombin test the usual statistical methods (for example,
time determination is 1:5 to 1:50 in buffer such that 5.3).
the final concentration of Ca2+ i5 15-25 mmol!L. The final
factor Xa generation is perÍormed in a solution containing
human or bovine albumin at a concentration such that
Ol!2008:20711
adsorption losses do not occur and which is appropriately
buffered at pH 7.3-8.0. In the final incubation mixture,
factor VII must be the rate-limiting component and each 2,7.11. ASSAY OF HUMAN
reagent component must the ability to generate factor Xa
on its own.
The principIe of the assay is to measure the
factor IX preparation to reduce the
time offactor IX-deficient
between three and five amino acids, bound to a by addition of a reagent
On of this group from the peptide substrate, activator, e.g. kaolin, silica or
maximum shifts to a wavelength its is assessed by the
n1·,'n'H~tir", to be 'CA,""Hl'.'-"
The substrate is usually
in water R and used at a final concentration of in International Units.
0.2-2 mmol!L. The substrate also contain ",,,wnnn The International Unit is the factor IX of a stated
inhibitors to stop Íurther factor amount of the International Standard, which consists of a
edetate). freeze-dried concentrate of human factor IX.
The in International Units of the International
ASSAY PROCEDURE Standard is stated
Reconstitute the entire contents of one of the
and the preparation to be examined in with the International
of water R; use within 1 h. Standard.
0W'H~'U" ""._'HH"-iH to the reconstituted to
between 0.5 IU

Prepare further dilutions oÍ reference and test


using al1 isotonic non-chelating buffer containing 1 per cent
of bovine or human albumin, buffered preferably between
7.3 and 8.0. Prepare at least three separate, independent
l'or each material, preferably in duplicate. Prepare
the dilutions 5uch that the final factor VII concentration is
below 0.005 IU/mL.
Prepare a control solution that includes al! components except
factor VII.
all dilutions in tubes and use within 1 h.
Use an apparatus suitable for measuremel1t of
Step 1. Mix dilutions of the factor VII reference preparation out the with incubatiol1 tubes maintained
and the to be examined with an appropriate ina at37 oc.
¡-p,M~rn-IPn coagulation factor reagent or a
factor IX -deficient
constituents, and incubate the and 0.1 mL of one
or wells at 37 oc. The or ofthe
of the various components during the factor Xa of a
must be as specified aboye under the description ol' reagent containing
reagents. incubate the mixture for a recommended time at 37 oc.
Allow the activation of factor X to proceed for a suitable To each tube, add 0.1 mL of a 3.7 giL solution of calcium
time, usually terminating the reaction before the factor Xa chloride R previously heated to 37 oc. Using a timer, measure
concentration has reached its maximallevel in order to obtain the coagulation time, í.e. the interval between the moment of
a linear relationship. The activation the addition of the calcium chioride and the first indication
to achieve linear production of factor Xa of the formation oÍ fíbrin. The volumes aboye may be
activation times are between to the APTT and used. Calculate
2 min 5 min, but deviatiol1s are permissible acceptable
linearity of the is thus obtained.

248 See the pages)


EUROPEAN PHARMACOPOEIA 8.0 2.7.13. ofhuman anti-D immunoglobulin

Ol/200S:20712 01/2008:20713
correded 7.5

2.7.12. ASSAY OF HEPARIN IN 2.7,13. ASSAY OF HUMAN ANTI-D


COAGULATION FACTORS IMMUNOGLOBULIN
METHODA
Heparin is assayed as a complex with antithrombin III (AT) via
its inhibition of coagulation factor Xa (anti-Xa activity). An The potency ofhumal1 anti-D immunoglobulin is determined
excess of AT is maintained in the reacHon mixture to ensure a by comparing the quantity necessary to produce agglutination
constant concentration ofthe heparin-AT complexo Factor Xa of D-positive red blood cells with the quantity of a reference
is neutralised by the heparin-AT complex and the residual preparation, calibrated in International Units, required to
factor Xa hydrolyses a specific chromogenic peptide substrate produce the same effect.
to release a chromophore. The quantity of chromophore is The International Unit is the activity contained in a stated
inversely proportional to the activity of the heparin. amount of the International Reference Preparation. The
equivalen ce in International Units of the International
Factor Xa chromogenic substrate. Specific chromogenic Reference Preparation is stated by the World Health
substrate for factor Xa such as: N-benzoyl-L-isoleucyl-L- Organization.
glutamyl-glycyl-L-arginine-4-nitroaniJide hydrochloride.
Reconstitute according to the manufacturer's instructions. Human anti-D imrnulloglobulin BRP is calibrated in
International Units by comparison with the International
Dilution buffer. 6.05 giL solution of tris(hydroxymethyl)ami- Standard and intended for use in the assay ofhuman anti-D
nomethane R. Adjust to pH 8.4 if necessary using hydrochloric immunoglobulin.
acid R. Use pooled D-positive red blood cells, collected not more than
7 days earlier and suitably stored, obtained fr0111 not fewer
Test solution. Dilute the preparation to be examined with than 4 group O R¡R¡ donors. To a suitable volume of the cells,
dilution buffer to obtain a solution expected to contain 0.1 IU previously washed 3 times with a 9 giL solution of sodium
of heparin per millilitre. chloride R, add an equal volume of bromelains solutiol1 R,
allow to stand at 37 oC for 10 min, centrifuge, remove the
Reference solution. Dilute the heparin reference preparation supernatant and wash 3 times with a 9 giL solution oí sodium
with dilution buffer to obtain a solution containing 0.1 IU of chloride R. Suspend 20 volumes of the red blood cells in a
heparin per millilitre. mixture of l5 volumes of inert serum, 20 volumes of a 300 giL
solution of bovine albumin R and 45 volumes of a 9 giL
The following working conditions apply to microtitre plates. solution of sodium chloride R. Stand the resulting suspensiol1
If the assay is carried out in tubes, the volumes are adjusted in iced water, stirring continuously.
while maintaining the proportions in the mixture.
Using a calibrated automated dilutor, prepare suitable dilutions
Warm all solutions to 37 oC in a water-bath shortly befOl'e the of the preparation to be examined and oí the reference
test. preparation using as diluent a solution containing 5 giL of
bovine albumin R and 9 of sodium chloride R.
Distribute in a series of wells, 20 !lL of normal human plasma Use a suitable apparatus for automatic continuous analysis.
and 20 !lL of antithrombin JII solution Rl. Add to the wells a The following protocol is usually suitable: maintain the
series ofvolumes (20 ~lL, 60 !lL, lO0!lL and 140 !lL) ofthe test temperature in the manifold, except for the incubation coils,
solution or the reference solution and make up the volume at 15.0 oc. Pump into the manifold of the apparatus the red
in each well to 200 ~lL using dilution buffer (0.02-0.08 IU of blood ceH suspensiol1 at arate of 0.1 mL/min and a 3 giL
heparin per millilitre in the final reaction mixture). solution of methylcellulose 450 R at arate of 0.05 mL/min.
Introduce the dilutions oí the preparation to be examined and
End-point method. Transfer 40 !lL fr0111 each well to a second the reference preparation at arate of 0.1 mL/min fOl' 2 min,
series of wells, add 20 !lL of bovine factor Xa solution R and followed by the diluent solution at arate of 0.1 mL/min for
incubate at 37 oC for 30 S. Add 40 ~lL of a 1 mmol/L solution 4 min before the next dilution is introduced.
of factor Xa chromogenic substrate and incubate at 37 oC
Introduce air at arate of 0.6 mL/min. Incubate at 37 oC for
for 3 mino Terminate the reaction by lowering the pH by
18 min and then disperse the rouleaux by introducing at arate
the addition of a suitable reagent, such as a 20 per cent V/V
of 1.6 mL/min a 9 giL solution of sodium chloride R containing
solution of glacial acetic acid R and measure the absorbance
a suitable wetting agent (for example, polysorbate 20 R at a
at 405 nm (2.2.25). Appropriate reaction times are usually
final concentration of 0.2 giL) to prevent disruption of the
between 3 mil1 and 15 min, but deviations are permissible
bubble pattern. Allow the agglutinates to settle and decant
if better linearity of the dose-response relationship is thus
twice, first at 0.4 mL/min and then at 0.6 mL/min. Lyse the
obtained.
unagglutinated red bJood cells with a solution containing
5 giL of octoxinollO R, 0.2 giL of potassium ferricyanide R,
Kinetic method. Transfer 40 !lL from each well to a second
1 giL of sodium hydrogen carbonate R and 0.05 giL of
series of wells, add 20 !lL of bovine factor Xa solution R
potassium cyanide R at arate of 2.5 mL/min. A lO-minute
and incubate at 37 oC for 30 S. Add 40 !lL of a 2 mmol/L
delay coil is introduced to allow for conversion of the
solution offactor Xa chromogenic substrate, incubate at 37 oC
haemoglobin. Continuously record the absorbance (2.2.25) of
and measure the rate of substrate cleavage by continuous
the haemolysate at a wavelength between 540 nm and 550 nm.
measurement of the absorbance change at 405 11m (2.2.25),
Determine the range of antibody concentrations over which
thus allowing the initial rate of substrate cleavage to be
there is a linear relatiol1ship between the concentration and the
calculated. This rate must be linear with the concentration of
resultant change in absorbance (DA). From the results, prepare
residual factor Xa.
a standard curve and use the linear Dortion of the curve to
determine the activity of the prepar~tion to be examined.
Check the validity of the assay and calculate the heparin
activity of the test preparation by the usual statistical methods Calculate the potency of the preparation to be examined using
for a slope-ratio assay (for example, 5.3). the usual statistical methods (5.3).

General Notices (1) apply to al! monographs and other texts 249
2.7.13. Assay ofhuman anti-D immunoglobulin EUROPEAN PHARMACOPOEIA 8.0

METHOD B Centrifuge the plate at 350 g fol' 3 min, preferably at 4 oc.


The potency of human anti-D immunoglobulin is Without removing the supernatant, gently add 100 I1L of
determined by competitive enzyme-linked immunoassay on glutaraldehyde solution to each well and leave for 10 mino
erythrocyte-coated microtitre plates. The method is based Drain the wells by quicldy inverting the pIate and wash 3 times
011 the competitive binding between a polyclonal anti-D with 250-300 I1L of PBS. This may be done manually or using
immunoglobulin preparation and a biotinylated monoclonal a suitable automated plate washer. Either carry out the assay
anti-D antibody directed against a D-antigen-specific epitope. as described below, or store the plate at 4 oC after draining
The activity of the preparation to be examined is compared off the PBS and adding 100 I1L of cell-fixation buffer per well
with a reference preparation calibrated in International Units. and sealing with plastic film. Plates can be sto red at 4 oC for
The International Unit is the activity of a stated amount of up to 1 month.
International Reference Preparation. The equivalence in Test solutions. For freeze-dried preparations, reconstitute as
International Units of the International Reference Preparation stated on the labe!' Prepare 4 independent replicates of 5 serial
is stated by the World Health Organization. 2-fold dilutions starting with 30 IU/mL in PBS containing
Human anti-D immunoglobulin BRP is calibrated in 10 giL of bovine albumin R. If necessary, adjust the starting
International Units by comparison with the International dilution to obtain responses falling in the linear portion of
Standard and intended for use in the assay of human anti -D the dose-response curve.
immunoglobulin.
Referenee solutions. Reconstitute the reference preparation
MATERIALS according to instructions. Prepare 4 independent replicates
Reagents not specified are of analytical grade. of 5 serial 2-fold dilutions starting with 30 IU/mL in PBS
PBS (Phosphate-buffered saline). Dissolve 8.0 g of sodium
containing 10 giL of bovine albumin R.
ehloride R, 0.76 g of anhydrous disodium hydrogen phosphate R, Using U- or V-bottomed microtitre plates, add 35 flL of each
0.2 g of potassium ehloride R, 0.2 g of potassium dihydrogen of the dilutions of the test solution or reference solution to
phosphate R and 0.2 g of sodium azide R in water R and dilute each of a series ofwells. To each well add 35 flL ofbiotinylated
to 1000 mL with the same solvent. Brad-5 at 250 ng/m1.
TBS (Tris-buffered saline). Dissolve 8.0 g of sodium ehloride R Empty the wells of the red cell-coated plate by inverting and
and 0.6 g of tris(hydroxymethyl)aminomethane R in water R. draining on a paper towel. Add 250 flL of PBS containing
Adjust to pH 7.2 with 1 M hydroehlorie aeid and dilute to 20 giL of bovine albumin R and leave at room temperature
1000 mL with water R. for 30 mino
Papain solution. Prepare a solution by stirring 1 g of papain R
Empty the wells of the red cell-coated plate by inverting and
at 37 oC for 30 min in 10 mL of 0.067 M phosphate buffer draining on a paper towel and transfer 50 flL from each of the
solution pH 5.4 R, centrifuge at 10000 g for 5 min and filter dilutions of the test solution or reference solution containing
through a membrane filter (nominal pore size 0.22 flm). To biotinylated Brad-5 into the wells. Use 50 flL ofPBS containing
activate, combine 1 mL of the filtrate with 1 mL of a 48.44 giL 10 giL of bovine albumin R as negative control. Seal the plate
solution of L-cysteine R and 1 mL of a 3.72 giL solution of with plastic film and incubate at room temperature for 1 h.
sodium edetate R and dilute to 10 mL with 0.067 M phosphate
buffer solution pH 5.4 R. Freeze in aliquots at - 20 oC or below. Remove the liquid from the wells of the red cell-coated plate
Red blood cells. Use pooled D-positive red blood cells obtained
and wash 3 times with 250-300 I1L of TBS.
from not fewer than 3 group O R2 R2 donors. Wash the cells Dilute the alkaline phosphatase-conjugated avidin/streptavidin
4 times with PBS. Centrifuge the cells at 1800 g for 5 min, reagent in TBS containing 10 giL of bovine albumin R and add
mix a suitabIe volume of prewarmed packed cells with a 50 flL to each well. Incubate for 30 min at room temperature.
suitable volume of prewarmed papain solution (2 volumes to
1 volume has been found suitable) and incubate at 37 oC for Remove the liquid from the wells of the red cell-coated pI ate
10 mino Wash the cells 4 times with PBS. Store at 4 oC in an and wash 3 times with 250-300 flL of TBS.
appropriate stabiliser for up to 1 week. Add 100 I1L of substrate solution to each of the wells and
Biotinylated Erad-S. Use according to instructions. incubate at room temperature for 10 min in the darle To stop
the reaction, add 50 flL of 3 M sodium hydroxide to each of
Alkaline phosphatase-canjugated avidin/streptavidin reagent.
the wells.
Preferably modified to combine high specific activity with low
non-specific binding. Use according to instructions. Measure the absorbances at 405 nm and substract the negative
Substrate solution. Use para-nitrophenyl phosphate according control reading. Use the absorbance vaIues in the linear range
to instructions. of the titration curve to estimate the potency of the preparation
to be examined by the usual statistical methods (5.3).
Cell fixation buffer. Dissolve 18.02 g of g/ucase R, 4.09 g of
sodium eh/oride R, 1.24 g of borie acid R, 10.29 g of sodium
citrate R and 0.74 g of sodium edetate R in water R. Adjust to METHOD C
pH 7.2-7.3 using 1 M sodium hydroxide or 1 M hydroehloric The potency ofhuman anti-D immunoglobulin is determined
aeid, and dilute to 1000 mL with water R. Use directly from by flow cytometry in a microtitre pI ate format. The method is
storage at 4 oc. based on the specific binding between anti-D immunoglobulin
Glutaraldehyde solution. Immediately before use, add 750 I1L and D-positive red blood cells. The activity of the preparation
of a 250 giL solution of glutaraldehyde R to 50 mL of cold PBS. to be examined is compared with a reference preparation
calibrated in International Units.
Mierotitre plates. Plates to be coated with red blood cells
are flat-bottomed polystyrene plates with surface properties The International Unit is the activity of a stated amount of
optimised for enzyme immunoassay and high protein-binding International Reference Preparation. The equivalence in
capacity. Plates used to prepare immunoglobulin dilutions are International Units of the International Reference preparation
U- or V-bottomed polystyrene or poly(vinyl chloride) plates. is stated by the World Health Organization.
METHOD Human anti-D immunoglobulin ERP is calibrated in
Prepare a 0.1 per cent V/V suspension of papain-treated red International Units by comparison with the International
blood cells in cold cell-fixation buffer. Pipette 50 flL into each Standard and intended for use in the assay of human anti- D
well of the flat-bottomed microtitre plateo immunoglobulin.

250 See the informatian seetian on general monographs (ca ver pages)
EUROPEAN PHARMACOPOEIA 8.0 2.7.14. Assay of hepatitis A vaccine

MATERIALS 0112011:20714
Reagents not specified are of analytical grade.
PBS. Dissolve 8.0 g of sodium chloride R, 0.76 g of disodium
hydrogen phosphate R, 0.2 g of potassium ehloride R and 0.2 g
2.7.14. ASSAY OF HEPATITIS A
of potassium dihydrogen phosphate R in water R and dilute to VACCINE
1000 mL with the same solvent.
PBS-BSA solution. PBS containing 10.0 giL of bovine
The assay ofhepatitis A vaccine is carried out either in vivo, by
albumin R.
comparing in given conditions its capacity to induce specific
antibodies in mice with the same capacity of a reference
Red blood eells. Use D-positive red blood cells obtained from preparation, or in vitro, by an immunochemical determination
a group O R¡R¡ donor within 2 weeks of collection. Store if of antigen contento
necessary in an appropriate stabiliser at 4 oc. Wash the ceUs at
least twice with PBS-BSA solution and prepare a suspension
containing 1 x 10 4 cells per micro litre but not more than IN VIVO ASSAY
5 x 10 4 cells per microlitre in PBS-BSA solution. The test in mice shown below is given as an example of a
Use D-negative red blood cells obtained from a group O rr method that has been found suitable for a given vaccine; other
donor and prepared similarly. validated methods may also be used.
Secondary antibody. Use a suitable fluorescent dye-conjugated Selection and distribution of the test animals. Use in the
anti-IgG antibody fragment specific for human IgG or parts of test healthy mice from the same stock, about 5 weeks old and
it. Store and use according to the manufacturer's instructions. from a strain shown to be suitable. Use animals of the same
sexo Distribute the animals in at least 7 equal groups of a
Microtitres plates. Use flat-bottomed plates without surface
number suitable for the requirements of the assay.
treatment for enzyme immunoassays.
Determination of potency of the vacdne to be examined.
METHOD
Using a 9 giL solution of sadium chloride R containing the
Test solutions. For freeze-dried preparations, recol1stitute as aluminium adjuvant used for the vaccine, prepare at least
stated on the label. Prepare at least 3 independent replica tes 3 dillltions of the vaccine to be examined and matching
of at least 3 serial 1.5- or 2-fold dilutions starting with a dilutions of the reference preparation. Allocate the dilutions
concentration in the range of 1.2-0.15 IU/mL using PBS/BSA one to each of the groups of animals and inject subcutaneously
solution as diluent. lf necessary, adjust the starting dilution not more than 1.0 mL of each dilution into each animal in the
to obtain responses falling in the linear portion of the group to which that dilution is allocated. Maintain a group of
dose-response curve. unvaccinated controls, injected subcutaneously with the same
Reference solutions. Reconstitute the reference preparation volume of diluent. After 28 to 32 days, anaesthetise and bleed
according to instructions. Prepare at least 3 independent aH animals, keeping the individual sera separate. Assay the
replicates of at least 3 serial 1.5- or 2-fold dilutions starting individual sera for specific antibodies against hepatitis A virus
with a concentration in the range of 1.2-0.15 IU/mL using by a suitable immunochemical method (2.7.1).
PBS-BSA solution as diluent. lf necessary, adjust the starting Cakulations. Carry out the calculations by the usual
dilution to obtain responses falling in the linear portion of statistical methods for an assay with a quantal response (5.3).
the dose-response curve.
Distribute 50 IlL of the D-positive red blood cens into each well From the distribution of reaction levels measured on al! the
of a microtitre plateo Add 50 IlL of each of the dilutions of the sera in the unvaccinated group, determine the maximum
test solution or reference solution to each of a series of wells. reaction level that can be expected to occur in an llnvaccinated
Use 50 IlL of PBS-BSA solution as negative control. Distribute animal for that particular assay. Any response in vaccinated
50 IlL of the D-negative red blood ceUs into 4 wells of the animals that exceeds this level is by definition a seroconversion.
same microtitre plate and add 50 IlL of the lowest dilution of Make a suitable transformation of the percentage of animals
the test preparation. To monitor spurious reactions, distribute showing seroconversion in each group (for example, a
50 IlL of the D-positive red blood cells into 4 wells of the same pro bit transformation) and analyse the data according to a
microtitre plate and add 50 IlL of PBS-BSA solution. Seal with parallel-line log dose-response model. Determine the potency
plastic film and incubate at 37 oC for 40 mino of the test preparation relative to the reference preparation.
Centrifuge the plates at 50 g for 3 min, discard the supernatant Validity conditions. The test is not valid unless:
and wash the ceUs with 200-250 IlL ofPBS-ESA solution.
Repeat this at least once. - for both the test and the reference vaccine, the EDso lies
between the smallest and the largest doses given to the
Centrifuge the plates at 50 g for 3 min, discard the supernatant animals;
and add 50 IlL of the secondary antibody diluted with
PBS-BSA solution to a suitable protein concentration. Sea! - the statistical analysis shows no significant deviation from
with plastic film and incubate, protected from light, at room linearity or parallelism;
temperature for 20 mino
- the confidence limits (P = 0.95) are not les s than 33 per cent
Centrifuge the plates at 50 g for 3 min, discard the supernatant and not more than 300 per cent of the estimated potency.
and wash the cells with 200-250 IlL ofPES-ESA solution.
Potency requirement. The upper confidence limit (P = 0.95)
Repeat this at least once.
of the estimated relative potency is not less than 1.0.
Centrifuge the plates at 50 g for 3 min, resuspend the cells
into 200-250 IlL of PBS. Transfer the cen suspension into a
tube suitable for the f1ow-cytometry equipment available and IN VITRO ASSAY
further dilute by adding PBS to allow a suitable flow rateo Carry out an immunochemical determination (2.7.1) of
Proceed immediately with measurement of the median antigen content with acceptance criteria validated against the
fluorescence intensity in a flow cytometer. Record at least in vivo test. The acceptance criteria are approved for a given
10000 events without gating but excluding debris. reference preparation by the competent authority in the light
of the validation data.
Use the median fluorescence intensity in the linear range of the
dose-response curve to estimate the potency of the preparation Hepatitis A vaccine (inactivated, non-adsorbed) BRP is suitable
to be examined by the usual statistical methods (5.3). for use as a reference preparation.

General Notiees (1) apply ta all monagraphs and ather texts 251
2.7.15. Assay ofhepatitis B vaccine EUROPEAN PHARMACOPOEIA 8.0

0112012:20715 shown to be suitable. Suitable numbers of dilutions of the


vaccine to be examined and the reference preparation are used
and a parallel-line model is used to analyse the data, which
2.7.15. ASSAY OF HEPATITIS B may be suitably transformed. Kits for measming HBsAg in
VACCINE (rDNA) vitro are commercially available and it is possible to adapt
their test procedures for use as an in vitro potency assay.
The assay of hepatitis B vaccine (rDNA) is carried out either
in vivo, by comparing in given conditions its capacity to
The acceptance criteria are approved for a given reference
induce specific antibodies against hepatitis B surface antigen preparation by the competent authority in light of the
validatíon data.
(HBsAg) in mice 01" guinea-pigs with the same capacity of a
reference preparation, or in vitro, by an immunochemical
determination of the antigen content.

IN VIVO ASSAY 07/2012:20716


Selection and distribution of the test animals. Use in the
test hea1thy mice from the same stock, about 5 weeks old.
The strain of mice used for this test must give a significant
2.7.16. ASSAY OF PERTUSSIS VACCINE
slope for the dose-response curve to the antigen; mice with (ACELLULAR)
haplotype H-2 q or H-2 d are suitable. Healthy guinea-pigs
weighing 300 g to 350 g (about 7 weeks old) from the same The capacity of the vaccine to induce the formation of specific
stock are also suitable. Use animal s of the same sexo Distribute antibodies in mice or guinea-pigs is compared with the same
the animals in at ¡east 7 equal groups of a number appropriate capacity of a reference preparation examined in parallel;
to the requirements of the assay. antibodies are determined using a suitable immunochemical
method 1) such as enzyme-linked immunosorbent assay
Determination of potency of the vaccine to be examined. (ELISA).
Using a 9 giL solution of sodium chloride R containing
the aluminium adjuvant used for the vaccine or another For combinations containing pertussis components together
appropriate diluent, prepare at least 3 dilutions of the vaccine with diphtheria and tetanus components, the serological
to be examined and matching dilutions of the reference assay in guinea-pigs can be performed with the same group
preparation. Allocate the dilutions, 1 to each of the groups of of animals used for the serological assay of diphtheria
animals, and inject intraperitoneally not more than 1.0 mL vaccine (adsorbed) (2. and of tetanus vaccine (adsorbed)
of each dilution into each animal in the group to which (2.7.8) when the common immunisation conditions for
that dilution is allocated. One group of animals remains all components (for example, doses, duration) have been
unvaccinated and is injected intraperitoneally with the same demonstrated to be valid for the combined vaccine. The
volume of diluent. After an appropriate time interva1 (for guinea-pig model allows for a further reduction in the
example, 4-6 weeks), anaesthetise and bleed the animals, number of animal s required and must be considered by each
keeping the individual sera separate. the individual analyst in accordance with the provisions of the European
sera for specific antibodies against HBsAg a suitable Convention for the Protection of Vertebrate Animals Used for
immunochemical method (2.7.1). Experimental and Other Scientific Purposes.
Calculations. Calculations are carried out by the usual The design of the assays A and B described below uses
statistical methods for an assay with a quantal response (5.3). multiple dilutions for the test and reference preparations.
After validation for a given vaccine, it is possible to applya
From the distribution of reaction levels measured on al! the simplified model such as a single dilution for both test and
sera in the unvaccinated group, the maximum reaction level reference preparations. Such a model enables the analyst to
that can be expected to occur in an unvaccinated animal determine whether the immunogenicity of the test preparation
for that particular assay is determined. Any response in is comparable to the reference vaccine, but does not give
vaccinated animals that exceeds this leve! is by definition a information 011 linearity or parallelism of the dose-response
seroconversion. curves.
Make a suitable transformation of the percentage of animals
For serological assays, suitable indicators to monitor test
showing seroconversion in each group (for example, a
consistency are:
probit transformation) and analyse the data according to a
parallel-line log dose-response model. Determine the potency - the mean ancl standard deviation of relative antibody
of the test preparation reJative to the reference preparation. levels or scores of the serum samples obtained after
administration of a fixed dose of the vaccine reference
Validity conditions. The test is not valid unless: preparation;
- for both the test and the reference vaccine, the EDso lies
- the antibody levels or scores of run controls (reference
between the smallest and the largest doses given to the
antiserum and negative serum samples);
animals;
- the ratio of antibody levels or scores for the reference
the statistical analysis shows no significant deviation from
antiserum to the serum corresponding to the
linearity or parallelism;
reference vaccine.
- the confidence limits (P = 0.95) are not less than 33 per cent
and 110t more than 300 per cent of the estimated potency. Where a single-dilution assay is used, production and test
consistency over time are monitored via suitable indicators
Potency requil'ement. The upper confidence limit (P = 0.95) ane! by carrying out a full multiple-dillltion assay periodically,
of the estimated relative potency is not less than 1.0. for example every 2 years.
IN VITRO ASSAY METHOD A. SEROLOGY IN MI CE
Carry out an immunochemical determination (2.7.1) of Reference vaccine. A batch of vaccine shown to be effective
antigen content with acceptance criteria validated against the in clinical tri al s or a batch representative thereof is used as
in vivo test. a reference vaccine. For the preparation of a representative
Enzyme-linked immunosorbent assay (ELISA) and batch, strict adherence to the production pro ces s used for the
radio-immunoassay (RIA) using monoclonal antibodies batch tested in clinical trials is necessary The stability of the
specific for protection-inducing epitopes ofHBsAg have been reference vaccine shall be monitored and documented.

252 See the (cover pages)


EUROPEAN PHARMACOPOEIA 8.0 2.7.16. Assay oí pertussis vacdne (aceHular)

Reference antiserum. A reference antiserum of assigned Reference antiserum. An in-house guinea-pig reference
activity is used in the test and serves as the basis for calculation antíserum of assigned activity is used in the test and serves as
of the antibody levels in the test sera. Bordetella pertussis the basis for calculation of the antibody levels in the test sera.
mouse antiserum BRP is suitable for use as a reference Dilution of tile test and reference preparations. Using a
antiserum. 9 giL solutíon of sodium chloride R as diluent, prepare serial
The following test model is given as an example of a method dilutions of the vaccine to be examined and the reference
that has been found to be satísfactory. preparation; series differing by 2.5- to 5-fold steps have been
Selection and distribution oí the test animals. Use in the found to be suitable. Use not fewer than 3 dilutions within
test healthy mice (for example, CDl strain) of the same stock, the range found to be suitable for all the components in the
about 5 weeks old. Distribute the animals in 6 groups of a vaccine to be examined. Use the dilutions for immunisation
number appropriate to the requirements of the assay. Use preferably within 1 h of preparation. Allocate 1 dilution to
3 dilutions of the vaccine to be examined and 3 dilutions of a each group of guinea-pigs.
reference preparation and attribute each dilution to a group Immunisation. Inject subcutaneously into each guinea-pig
of mice. Inject intraperitoneally or subcutaneously into each 1.0 mL of the dilution allocated to its group.
mouse 0.5 mL of the dilution attributed to its group. During
Blood sampling. 35-42 days (5-6 weeks) after immunisation,
validation studies a further group of mice may be used as a
take a blood sample from each vaccinated and negative
negative control by injecting the animals with diluent alone.
control guinea-pig using a suitable method. Store the sera at
CoHecHon of serum samples. 4-5 weeks after vaccination, - 20 oC until used for antibody determination. Avoid frequent
bleed the mice individually under anaesthesia. Sto re the sera freezing and thawing of serum samples.
at - 20 oC until used for antibody determination.
Antibody determination. Assay the individual sera for
Antibody determination. Assay the individual sera for content of specific antibodies to each acellular pertussis
content of specific antibodies to each acellular pertussis antigen using a validated method such as the EUSA test
antigen using a validated method such as the EUSA test shown below.
shown below.
ELISA test. Suitable 96-well microtitre plates are coated with
ELISA test. Microtitre plates (poly(vinyl chloride) or the purified antigens (e.g. pertussis toxin (PT), pertactin
polystyrene as appropriate for the specific antigen) are coated (PRN), filamentous haemagglutinin (FHA) and/or fimbrial
with the purified antigen at a concentration of 100 ng per agglutinogens (Fim 2/3)) representing components in the
well. After washing, unreacted sites are blocked by incubating combined vaccine at a concentration of 200-400 ng per well.
the pi ates with a solution of bovine serum albumin and After washing, unreacted sites are blocked by incubating the
then washed. 2-fold dilutions of sera from individual mice plates with a suitable blocking buffer and then washed. 2-fold
immunised with test 01' reference vaccines are made on dilutions of sera from individual guinea-pigs immunised with
the plates. Reference antiserum is included on each plateo test or reference vaccines are made 011 the plates. Reference
After incubation at 22-25 oC for 1 h, the plates are washed. antiserum is included on each plateo After incubation at
A suitable solution of enzyme-conjugated anti-mouse IgG 37 oC for 1 h, the plates are washed. A suitable solution of
antibody is added to each well and incubated at 22-25 oC for enzyme-conjugate~d anti-guinea-pig IgG antibody is added
1 h. After washing, a chromogenic substrate is added from to each well and incubated at 37 oC for 1 h. After washing, a
which the bound enzyme conjugate liberates a chromophore chromogenic substrate is added from which the bound enzyme
that can be quantified by measurement of absorbance (2.2.25). conjugate liberates a chromophore that can be quantified by
Cakulations. The antibody titres in the sera of mice measurement of absorbance (2.2.25).
immunised with reference and test vaccines are calculated for Calculations. The antibody titres in the sera of guinea-pigs
each acellular pertussis antigen using the reference antiserum, immunised with reference and test vaccines are calculated for
and from the values obtained the relative potency of the test each acellular pertussis antigen using the reference antiserum,
vaccine in relation to the reference vaccine is calculated by the and from the values obtained the relative potency of the test
usual statistical methods (5.3). vaccine in relation to the reference vaccine is calculated by the
The assay is not valid unless: usual statistical methods (5.3).
- the confidence limits (P = 0.95) are not less than 50 per The assay is not valid unless:
cent and not more than 200 per cent of the relative potency - the confidence limits (P = 0.95) are not less than 50 per
estímate for each acellular pertussis antigen; cent and 110t more than 200 per cent of the relative potency
- the statistical anaIysis shows a significant slope and estímate for each acellular pertussis antigen;
no deviation from linearity and parallelism of the - the statistical analysis shows a significant slope and
dose-response curves (chapter 5.3 describes possible no deviation from linearity and parallelism of the
alternatives if significant deviations are observed). dose-response curves (chapter 5.3 describes possible
alternatives if significant deviations are observed).
METHOD B. SEROLOGY IN GUINEA-PIGS
Selection and distribution of the test ani.mals. Use in the The following section is published for information.
test healthy guinea-pigs from the same stock, each weighing
250-350 g. Use guinea-pigs of the same sex or with males and
females equally distributed between the groups. Distribute Assay of pertussis vaccine (acellular):
the guinea-pigs in not fewer than 6 equal groups; use groups guidelines
containing a number of animals sufficient to obtain results
that fulfil the requirements for a valid assay prescribed below. METHOD B. DETERMINATION OF ANTIBODIES IN
During validation studies a further group of guinea-pigs GUINEA-PIGS
is used as a negative control by injecting the animals with The EUSA shown below is given as an example of an
diluent alone. immunochemical method that has been found to be suitable.
ReÍerence vaccine. A batch of vaccine shown to be effective Determination of anHbody titre by ELISA method for
in clínica! tria!s or a batch representative thereof is used as pertussis toxin (PT), filamentous haemagglutinin (FHA),
a reference vaccine. For the preparation of a representative fimbrial agglutinogens (Fim 2/3) and pertactin (PRN).
batch, strict adherence to the production process used for the 2-fold dilutions of sera from test and reference vaccines are
batch tested in clinical tríaIs is necessary. The stability of the made on ELISA plates coated with acellular pertussis antigens
reference vaccine shall be monitored and documented. (PRN, PT, FHA or Fim 2/3). A guinea-pig reference antiserum

General Notices (1) apply to all monographs and other texts 253
2.7.17. of human anHthmmbin lB EUROPEAN PHARMACOPOEIA 8.0

and a negative guinea-pig serum are included on each plateo 01/2008:20717


Peroxidase-conjugated rabbit or directed against
guinea-pig IgG is addea, by a substrate.
Optical density is measured and the antibody titre is
calculated the usual statistiolmethods

to be
to inactivate

Peroxidase-conjugated rabbit or goat


guinea-pig IgG.
to be examinen are with
PT, FHA OI Fim 2/3). and the remaining thrombin
pH 9.6. Dissolve 1.59 g of a suitable chromogenic substrate.
sodium R and 2.93 g of sodium The International Unit is the of a stated amount
carbonate R in 1000 mL oí water R. Distribute
of the International Standard for human antithrombin nI
into mL bottles and sterilise by autoclaving at 121°C
concentrate. The in International Units oí
for 15 mino
the International 1S stated by the World Health
7.4 (PES). Dissolve with Organization.
of sodium R, 2.0 g
Method. Prepare 2 series of 3 01' 4 dilutions in the
R, 14.3 g of disodium
lrll.·ur.UlP R and 2.0 g oí R in range 1/75 to 1/200 from 1 IU/mL, for both the to
be examined and the reference using tris-EDTA
mL of water R. Sto fe at room temperature to prevent
ESA so/utíon 8.4 R J 5 IU of heparin per
OL""li,""lV!!. Dilute 10-fold with water R befare use.
millilitre.
- Citric acid solution. Dissolve 10.51 g of eitrie acid R in
1000 mL of water R and to pH 4.0 with a 400 giL Warm 200 of each dilutiol1 al 37 oC for 1-2 mino Add
solution of sodium R. to each dilution 200 flL of a solution of bovine thrombin R
containing 2 IU/mL in tris-EDTA ESA solution
PBS containing 0.5 of polysorbate 20 R. pH 8.4 R Mix and maintain at 37 oC for 1 lnin. Add
- Diluent block PBS containing 0.5 of 500 I1L of a suitable substrate
hnlllcrlrh,nfp 20 and 25 giL oí dried skimmed milk. D- phenylalanyl-L-pipecolyl-L-argi nine-4- nitroanilide,
- Peroxidase substrate. reconstituted in water R to a solution "-V"CO"'''''''i'o
of diammonium 2,2' 4 mmol/L and further to a concentration
R in 20 mL of the citrie acid solution. tris-EDTA~ BSA

before use add 5 of strong start measurement [he in


'-VHLllHHlJ14 the measurement
R.
of absorbance
below is given as an example of a
suitable others be used. Wells lA-H are
used for negative control serum. 2-12 A-H are used for
~eference antiserum in 2 positions) and
sera from guinea-pigs immunised with the test or The fate of change of absorbance is
reI'erence vaccine. to antitnrombin III
Coat each well oI' the ELISA with 100 I1L of the of [he assay and calculate the potency of the
solution FHA and Fim 2/3 at 2 [J.g/mL test the usual statistical methods (5.3).
in carbonate coating buffer pH 9.6).
overnight at 4 oC in a humid atmosphere. To
avoid temperature gradient effects, do not stack more than 4
high. On the following wash the plates thoroughly Ol!2008:20718
with the buffer. Block by addition of 150 ~lL
of the diluent buffer to each well. Incubate in a humid
at 37 oC for 1 h. Wash the plates thoroughly with 2.7.1 ASSAY OF HUMAN
buffer. Place 100 I1L of the diluent block buffer
of the except those of row A. Prepare
COAGULATION n
individual test and reference vaccine Human coagulation factor II i5
serum reference antiserum and negative control activation to form factor Ha, Factor
serum AlIocate the control serum to column comparing its in
L the reference antiserum to at 2 other columns and peptide substrate the same
individual test and reference vaccine sera to the remaining Standard or of a reference
columns and add 100 I1L of each serum to the first 2 wells of International Units.
the cO]l1mn to which it i5 allocated. Using a multichannel
micropipette, make 2-fold serial dilutions from row B down The International Unit is the factor Ir of a stated
the plate to row H, by transferring 100 flL from one well to the amount of the International Standard whieh consists of a
next. Discard 100 flL from the ¡ast row so that al! wells contain freeze-dried concentrate ofhuman blood coagulation factor II.
100 Incubate at 37 oC for 2 h. Wash thoroughly with the The equivalence in International Units of the International
buffer. Prepare a suitable dilution of the peroxidase Standard is stated by the World Health
in the diluent block buffer and add 100 I1L to each The assay method consists of 2
at 37 oC in a humid atmosphere for 1 h. Wash snake venom-dependent activation of factor II,
the pIates thoroughly with the washing buffer. Add 100 I1L of enzymatic of a factor Ha
the substrate to each well. Allow to stand at room to form a can be
protected fromlight, for 30 mino Read the spectrophotometrically.
nm in the same order as the addition of substrate was there is a linear relation between
made,

254 See the section 011 pages)


EUROPEAN PHARMACOPOEIA 8.0 2.7.20. In vivo assay of poliomyelitis vacdne (inactivated)

REAGENTS The chromogenic assay method consists of 2 steps:


Iliper venom specific factor JI activator (Ecarin). A protein snake venom-dependent activation of factor X, followed
derived from the venom of the saw-scaled viper (Echis by enzymatic cleavage of a chromogenic factor Xa
carinatus) which specifically activates factor n. Reconstitute substrate to form a chromophore that can be quantified
according to the manufacturer's instructions. Store the spectrophotometrically. Under appropriate assay conditio!1s,
reconstituted preparation at 4 oC and use within 1 m011th. there is a linear relation between factor Xa activity and the
Factor JIa ehromogenic substrate. SpeciJ1c chromogenic cleavage of the chromogenic substrate.
substrate for factor Ha such as: H-D-phenylalanyl-L-
pipecolyl-L-arginine-4-nitroanilide dihydrochloride, REAGENTS
4-toluenesulfonyl-glycyl-prolyl-L-arginine-4- nitroanilide, H- Russell's viper venam specifi e factor X activatar (RVV). A
D-cyclohexyiglycyl-a -aminobutyryl-L-arginine-4-nitroanilide, protein derived fro111 the venom of Russell's viper (Vipera
D-cyclohexylglycyl-L-alanyl-L-arginine-4-nitroanilide russelli) which speciJ1cally activates factor X. Reconstitute
diacetate. Reconstitute according to the manufacturer's according to the manufacturer's instructions. Store the
instructions. reconstituted preparation at 4 oC and use within 1 111onth.
Dilutian buffer. Solution containing 6.06 giL of Factor Xa chromogenic substrate. Specific chromogenic
tris(hydroxymethyl)aminomethane R, 17.53 giL of sodium substrate for factor Xa such as: N-a-benzyloxycarbonyl-D-
ehloride R, 2.79 giL of (ethylenedinitrilo)tetra-aeetie acid R arginyl- L-glycyl-L-arginine-4-nitroanilide dihydrochloride,
and 1 giL of bavine albumin R or human albumin R. Adjust to N- benzoyl-L-isoleucyl-L-glutamyl-glycyl-L-arginine-4-
pH 8.4 if necessary, using hydroehloric aeid R. nitroanilide hydrochloride, methanesulfonyl-D-leucyl-
glycyl-L-arginine-4-nitroanilide, methoxycarbonyl-D-
METHOD cyclohexylalanyl-glycyl- L-arginine-4-nitroanilide aceta te.
Test solutian. Dilute the preparation to be examined with Reconstitute according to the manufacturer's instructions.
dilution buffer to obtain a solution containing 0.015 IU of Dilution buffer. Solution containing 3.7 giL of
factor II per millilitre. Prepare at least 3 further dilutions in lris(hydroxymethyl)aminomethane R, 18.0 giL of sadium
dilution buffer. chloride R, 2.1 giL of imidazole R, 0.02 giL of hexadimethrine
Reference solution. Dilute the reference preparation to be bromide R and 1 giL of bovine albumin R or human albumin R.
examined with dilution butIer to obtain a solution containing Adjust to pH 8.4 if necessary using hydrochloric aeid R.
0.015 IU of factor II per millilitre. Prepare at least 3 further
dilutions in dilutiol1 buffer. METHOD
Warm all solutions to 37 oC in a water-bath shortly before the Test solutíon. Dilute the preparation to be examined with
test. dilution buffer to obtain a solution containing 0.18 IU of
factor X per millilitre. Prepare at least 3 further dilutions in
The following working conditions apply to microtitre pIates.
dilution buffer.
rf the assay is carried out in tubes, the volumes are adjusted
while maintaining the proportions in the mixture. Reference solution. Dilute the reference preparation to be
examined with dilution buffer to obtain a solution containing
Using a microtitre plate maintained at 37 oC, add 25 [lL of 0.18 IU of factor X per millilitre. Prepare at ¡east 3 further
each dilution of the test soJution or the reference solution to dilutions in dilution buffer.
each of a series of wells. To each well add 125 ~lL of dilution
buffer, then 25 [lL of ecarin and incubate for exactly 2 mino To Warm all solutions to 37 oC in a water-bath shortly before the
each well add 25 [lL of factor Ha chromogenic substrate. test.
The following working conditions apply to microtitre plates.
Read the rate of change of absorbance (2.2.25) at 405 nm
If the assay is carried out in tubes, the volumes are adjusted
continuously over a period of 3 mil1 and obtain the mean rate
while maintaining the proportiol1s in the mixture.
of change of absorbance (L'lAlmin). rf continuous monitoring
is not possible, read the absorbance at 405 nm at suitable Using a microtitre plate maintained at 37 oC, add 12.5 [lL of
consecutive intervals, for instance 40 s, plot the absorbances each dilution of the test solution or the reference solution to
against time 011 a linear graph and calculate L'lAlmin as the each of a series of wells. To each well add 25 ~lL of RVV and
510pe of the lineo From the L'lAlmin values of each individual incubate for exactly 90 s. To each well add 150 ~lL of factor Xa
dilution of standard and test preparations, calculate the chromogenic substrate, diIuted 1 in 6 in dilution buffer.
potency of the preparation to be examined and check the Read the rate of change of absorbance (2.2.25) (at 405 nm
validity of the assay by the usual statistical methods (5.3). continuously over a period of 3 min and obtain the mean rate
of change of absorbance (L'lAlmin). lf continuous monitoring
is 110t possible, read the absorbance at 405 nm at suitable
consecutive intervals, for instan ce 40 s, pIot the absorbances
against time on a linear graph and calcuIate L'lAlmin as the
0112008:20719 slope of the lineo From the L'lAlmin values of each individual
dilution of standard and test preparations, calculate the
potency of the preparation to be examined and check the
2.7.19. ASSAY OF HUMAN of the assay by the usual statistical methods (5.3).
COAGULATION FACTOR X
Human coagulation factor X is assayed following specific
activation to forl11 factor Xa. Factor Xa is estimated by 0112008:20720
comparing its activity in cleaving a speciJ1c chromogenic
peptide substrate with the same activity of the International
Standard or of a reference preparation calibrated in 2.7,20. IN VIVO ASSAY OF
International Units. POLIOMYELITIS VACCINE
The International Unit is the factor X activity of a stated (INACTIVATED)
amount of the International Standard which consists of a
freeze-dried concentrate of human coagulation factor X. The capacity of the vaccine to induce the formation of
The equivalence in International Units of the International neutralising antibodies is determined in vivo by one of the
Standard is stated by the World Health Organization. following methods.

General Notices (1) apply to all mOl1ographs and other texts 255
2.7.20. In vivo assay of poHornyditis vacdne (inactivated) EUROPEAN PHARMACOPOEIA 8.0

TEST IN CHICKS OR GUINEA-PIGS The following section is published for informatian.


Prepare a suitable series of not fewer than 3 dilutions of
the vaccine to be examined using a suitable buffered saline Guideline on waiving of the in vivo assay of
solution. Distribute either guinea-pigs weighing 250-350 g or poliomyelitis vaccine (inactivated) and its
3-week-old chicks into groups of 10, and allocate a group to
each dilution of the vaccine. Inject intramuscularly into each combinations
animal 0.5 mL of the dilution intended for its group. Bleed the This guideline applies to vaccines derived from wild strains of
animals after 5-6 days and separate the sera. Examine the sera poliavirus. The validation described should be carried out for
for the presence of neutralising antibodies, at a dilution of ea eh product befare waiving of the in vivo assay, and should
1 in 4, to each of the human poliovirus types 1, 2 and 3. Mix be repeated wherever there is a substantial ehange to the
100 CCID so of virus with the dilution of serum and incubate manufacturing process that may affect the in vitro or in vivo
at 37 oC for 4.5-6 h. Keep at 5 ± 3 oC for 12-18 h where assays.
necessary fo1' consistency of results. Inoculate the mixtures
The European convention on the protection of vertebrate
into ceH cultures for the detection of unneutralised virus and
animals used for experimental and other scientific purposes
read the results up to 7 days after inoculation. For each group
requires that tests in animals shall not be carried out if
of animals, note the numbe1' of sera that have neutralising
a scientifically satisfactory alternative is reasonably and
antibodies and calculate the dilution of the vaccine that gives
practically available. The aim of this guideline is therefore
an antibody response in 50 per cent of the animals. Carry out
to promote waiving of the in vivo assay whereve1' it can be
in parallel a control test using a suitable reference preparation.
shown for a given product that the in vitro assay (D-antigen
The vaccine complies with the test if a dilution of 1 to 100 or
determination) gives sufficient assurance of satisfactory
more produces an antibody response for each of the 3 types of
potency for routine batch control.
virus in 50 per cent of the animals.
For the in vivo assay, the test in rats is considered to be the
method of choice. For vaccines that are assayed using chicks or
TEST IN RATS guinea-pigs and that have an established record of production
A suitable in vivo assay method consists of intramuscular history, the in vivo assay may be waived if the rat assay is
injection into the hind limb(s) of not fewer than 3 dilutions also applied to the batches included in the validation study
of the vaccine to be examined and a reference vaccine, using described below. For vaccines not yet approved, the results of
for each dilution a group of 10 specific pathogen-free rats of a the rat assay on al! final bulks should be included in all data
suitable st1'ain. Use of 4 dilutions is often necessary to obtain generated for demonstration of consistency of production
valid results for all 3 serotypes. The number of animals per before waiving of the in vivo assay.
group must be sufficient to obtain results that meet the validity Once the in vivo assay has been waived, batches of vaccine will
criteria; groups of 10 rats are usually sufficient, although valid be released 011 the basis of the in vitro assay, and the in vivo
results may be obtained with fewer animals per group. If assay should not be used as an alternative for the release of a
animals of different sex are used, males and females are evenly batch that fails the in vitro assay. Repetition of the in vitro
distributed between al! groups. A weight range of 175-250 g assay may be performed according to an authorised procedure.
has been found to be suitable. An inoculum of 0.5 mL per rat
is used. The dose range is chosen such that a dose response PROCEDURE
to al! 3 poliovirus types is obtained. Bleed the animals after The following conditions should be met before performance
20-22 days. Neutralising titres against al! 3 poliovirus types of the validatiol1 study:
are measured separately using 100 CCID so of the Sabin - appropriate experience of the rat assay;
strains as challenge viruses, Yero or Hep2 as indicator cells,
- fuU validation of the D-antigen assay (linearity,
and neutralisation conditions of 3 h at 35-37 oC followed
repeatability, intermediate precision, accuracy and limits of
by 18 h at 2-8 oC where necessary for consistency of results.
quantification) ;
Results are read following fixation and staining after 7 days of
incubation at 35 oc. For a valid antibody assay, the titre of - establishment of acceptance criteria for the D-antigen assay
each challenge virus must be shown to be within the range based on a suitable number of consecutive finallots;
10 CCID so to 1000 CCID so and the neutralising antibody titre - establishment of production consistency on recent final
of a control serum must be within 2 twofold dilutions of the bulks using the currently approved in vivo assay; the final
geometric mean titre of the serum. The potency is calculated bulks should correspond to the finallots used to establish
by comparison of the proportion of responders for the vaccine the acceptance criteria for the D-antigen assay and should
to be examined and the reference vaccine by the probit method represent different inactivated harvests of each of the 3
or, after validation, using a parallel-line model. For the probit types of poliovirus.
method it is necessary to establish a cut -off neutralising The validation study should be performed on:
antibody titre for each poliovirus type to define a responder. - a final bulk/lot that is representative of the current
Due to interlaboratory variation, it is not possible to define production method;
cut-off values that could be applied by alllaboratories. Rather,
the cut -off values are determined for each laboratory based on - 2 sub-potent batches prepared, for example, by heating
a minimum series of 3 tests with the reference vaccine. The normal vaccine or mixing it with heat-treated vaccine; the
mid-point 011 a IOg2 scale of the minimum and maximum sub-potent batches should have expected titres of about
geometric mean titres of the series of 3 or more tests is used half that of the represel1tative final bulk/lot.
as the cut -off value. For each of the 3 poliovirus types, the These batches are assayed using as reference standard a
potency of the vaccine is 110t significantly less than that of the homologous production batch:
reference preparation. The test is not valid unless: - by the currently approved in vivo assay for the vaccine;
for both the vaccine to be examined and the refe1'ence - by the rat assay where this is not the currently approved
vaccine, the EDso lies between the smallest and the largest in vivo assay;
doses given to the animals; - by the D-antigen assay.
- the statistical analysis shows no significant deviation from Waiving of the in vivo assay is acceptable if the representative
linearity or parallelism; final bulk/lot camplíes with the in vivo and in vitro assays and
the sub-potent batches faíl to comply. If a sub-potent batch
- the confidence limits (P = 0.95) are not les s than 25 per cent fails to comply with the D-antigen assay but complies with the
and not more than 400 per cent of the estimated potency. in vivo assay, the latter may be repeated.

256 See the information section 011 general monographs (cover pages)
EUROPEAN PHARMACOPOEIA 8.0 2.7.21. Assay ofhuman von Willebrand factor

01/2008:20721 of von Willebrand factor to collagen fibrils and the


subsequent binding of polyclonal anti -von Willebrand factor
2.7.21. ASSAY OF HUMAN VON antibody conjugated to an e11zyme, which 011 addition of a
chromogenic substrate yields a product that can be quantitated
WILLEBRAND FACTOR spectrophotometrically. Under appropriate conditions,
The biological functions of human von Willebrand factor there is a linear relationship between von Willebrand factor
are numerous. At present, its ristocetin cofactor activity collagen -binding and absorbance.
and its collagen binding activity can be utilised for assays.
The potency of human von Willebrand factor is determined REAGENTS
by comparing, in given conditions, its activity with the Collagen. Use native equine or human fibrils of collagen type 1
same activity of a reference preparation calibrated against or lII. For ease of handling, collagen solutions may be used.
the International Standard, in International Units where Collagen diluent. Dissolve 50 g of glucase R in water R, adjust
applicable. to pH 2.7-2.9 with 1 M hydrochloric acid and dilute to 1000 mL
The International Unit is the activity of a stated amount of with water R.
the International Standard, which consists of a freeze-dried Phosphate-buffered saline (PBS). Dissolve 8.0 g of sodium
human von Willebrand factor concentrate. The equivalence in chloride R, 1.05 g of disodium hydrogen phosphate dihydrate R,
International Units of the International Standard is stated by 0.2 g of sodium dihydrogen phosphate R and 0.2 g of potassium
the World Health Organization (WHO). chloride R in water R. Adjust to pH 7.2 using 1 M sodium
RISTOCETIN COFACTOR ASSAY hydroxide or 1 M hydrochloric acid and dilute to 1000 mL
with water R.
The ristocetin cofactor activity of von Willebrand factor is
determined by measuring agglutination of a suspension of Washing buffer. PBS containing 1 giL of polysorbate 20 R.
platelets in the presence of ristocetin A. The assay can be Blocking reagent. PBS containing 1 giL of polysorbate 20 R and
carried out for quantitative determinations by using automated 10 giL of bovine albumin R.
instruments, or for semi-quantitative determinations by
visually assessing the endpoint of agglutination in a dilution Di/ution buffer. PBS containing 1 giL of polysorbate 20 R and
series. Quantitative assays are preferred. 50 giL of bovine albumin R.
REAGENTS Conjugate. Rabbit anti-human von Willebrand factor serum
Suspension of platelets. Use standardised and, for example, horseradish peroxidase conjugate. Use according to the
formaldehyde- or paraformaldehyde-fixed preparations of manufacturer's instructions.
freshly isolated and washed human platelets. The suspension Substrate solution. Immediately before use, dissolve a tablet
may also be freeze-dried. An appropriate amount of of o-phenylenediamine dihydrochloride and a tablet of urea
ristocetin A is added if necessary. Sorne platelet reagents may hydrogen peroxide in 20 mL of water R or use a suitable
already contain ristocetin A. volume of hydrogen peroxide. Protect from light.
Reference preparation. The reference preparation for von Micratitre plates. Flat-bottomed polystyrene plates with
Willebrand factor is the WHO International Standard for von surface properties optimised for enzyme immunoassay and
Willebrand factor concentrate. high protein -binding capacity.
METHOD METHOD
Semi-quantitative assay. Prepare suitable dilutions of the Test solutions. Reconstitute the preparation to be examined as
preparation to be examined and of the reference preparation, stated on the labe!' Dilute with dilution buffer to produce a
using as diluent a solution containing 9 giL of sodium solution containing approximately 1 IU of von Willebrand
chloride R and 10-50 giL of human albumin R. Add to each factor. Prepare 2 series of at least 3 further dilutions using
dilution an appropriate amount of the suspension of platelets dilution buffer.
and, if necessary, of ristocetin A. Mix on a glass slide by
moving it gently in circles for 1 mino Allow to stand for a Reference solutions. Reconstitute the reference preparation as
further 1 min and read the result against a dark background directed. Dilute with dilution buffer to produce a solution
with side lighting. The last dilution which clearly shows containing approximately 1 IU of von Willebrand factor.
visible agglutination indicates the ristocetin cofactor titre of Prepare 2 series of at least 3 further dilutions using dilution
the sample. Use diluent as a negative control. buffer.
Quantitative Assay. Reconstitute the entire contents of Allow the solution of collagen to warm to room temperature.
1 ampoule of the reference preparation and the preparation Dilute with collagen diluent to obtain a solution containing
to be examined by adding the appropriate quantity of the 30-75 flg/mL of collagen, mix gently to produce a uniform
recommended diluent (for example water R); use immediately. suspension of collagen fibrils. Pipette 100 flL into each well
Add sufficient prediluent to the reconstituted preparations to of the microtitre plateo Cover the plate with plastic film
produce solutions containing 0.5-2.0 IU/mL. The prediluent and incubate at 37 oC overnight. Empty the wells of the
consists of an isotonic non-chelating buffer containing, collagen-coated plate by inverting and draining on a paper
for example, 1-5 per cent of human or bovine albumin, toweL Add 250 flL of washing buffer. Empty the wells of the
and tris(hydroxymethyl)aminomethane or imidazole, plate by inverting and draining on a paper towel. Repeat this
appropriately buffered. operation 3 times. Add 250 flL of blocking reagent to each
The test is performed in accordance with the manufacturer's well, cover the plate with plastic film and incubate at 37 oC for
instructions with at least 2 dilution series with as many 1 h. Empty the wells of the plate by inverting and draining
dilutions as are needed to obtain a total of at least 3 different on a paper towel. Add 250 flL of washing buffer. Empty the
concentrations in the linear range of the assay. wells of the plate by inverting and draining on a paper towel.
Repeat this operation 3 times.
Check the validity of the assay and calculate the potency of
the test preparation using the usual statistical methods (for Add 100 flL each of the test solutions or reference solutions to
example, 5.3). the wells. Add 100 flL of dilution buffer to a series of wells
to serve as negative control. Cover the plate with plastic film
COLLAGEN-BINDING ASSAY and incubate at 37 oC for 2 h. Empty the wells of the plate
Collagen-binding is determined by an enzyme-linked by inverting and draining on a paper towel. Add 250 IlL of
immunosorbent assay on collagen-coated microtitre washing buffer. Empty the wells of the plate by inverting and
plates. The method is based on the specific binding draining on a paper towe!. Repeat this operation 3 times.

General Natices (1) apply to all monographs and other texts 257
2.7.22. Assay ofhuman coagulaHon factor XI EUROPEAN PHARMACOPOEIA 8.0

Prepare a suitable dilution of the conjugate (for example, a 0112008:20723


dilution factor of 1 to 4000) with PES containing 5 gIL of
bovine albumin R and add 100 flL to each well. Cover the 2.7,23, NUMERATION OF
plate with plastic film and incubate at 37 oC for 2 h. Empty
tÍ1e wells of the plate by inverting and draining on a paper CD34/CD45+ CELLS IN
towel. Add 250 ~lL of washing buffer. Empty the wells of the HAEMATOPOIETIC PRODUCTS
plate by invel'ting and draining 011 a paper towel. Repeat this
operatioll 3 times. This chapter describes immunolabelling and analysis
by flow cytometry (2.7.24) to determine the number of
Add 100 flL of substrate solution to each of the wells and CD34/CD45+ cells contained in haematopoietic products.
incubate at room temperature for 20 min in the darle Add The determination is carried out by a single platform method
100 ~lL of 1 M hydroch/oric acid to each of the wells. using calibrated fluorospheres, after Iysis of the sample red
Measure the absorbance at 492 nm. Use the absorbance values blood cells if necessary.
to estimate the potency of the preparation to be examined This method applies to all types of preparatiol1s and whole
using the usual statistical methods (5.3). blood. However, its level of precision makes it particularly
suitable for preparatiol1s containing very low percentages of
The assay is invalid if the absorbances measured fol' the
CD34/CD45+ cells.
negative controls are greater than 0.05.
Graft quality assessment by CD34/CD45+ (eH enumeration
A variety of studies have established that the 1-3 per cent
of cells in the bone marrow that express the CD34 cel!
surface antigen are capable of reconstituting long-term,
multilineage haematopoiesis after myeloablative therapy.
01/2008:20722 CD34/CD45+ cells are al50 Íound in the peripheral circulation
of normal individuals but are extremely rare (0,01-0.1 per
cent), However, CD34/CD45+ cells may also be mobilised
2.7.22, ASSAY OF HUMAN from marrow to the peripheral circulation in greater
COAGULATION FACTOR XI numbers by haematopoietic cytokines such as granulocyte
colony-stimulating factor and/or chemotherapy.
The principIe of the assay is to measure the ability of a The technique used for enumeration of CD34/CD45+ cells
factor XI preparation to reduce the prolonged coagulation must meet the following requirements:
time of factor XI -deficient plasma. The reaction is accelerated - high sensitivity, since haematopoietic stem cells are rare
by addition of a reagent containing phospholipid and a contact events;
activator, e.g. kaolin, silica 01' ellagic acid. The potency
clinically relevant results;
is assessed by comparing the dose-response curve of the
preparation to be examined to that of a reference preparation to
VUU'-,'UHH ,. clinically reliable l'esults;
cOl1sisting of human normal plasma. - speed, to provide real-time analysis.
1 unit of factor XI is equal to the of 1 mL of human Selection of parameters
normal plasma. Humañ normal plasma prepared by pooling The flow cytometry assay uses commercially available, directly
plasma units frol11 not fewer than 30 donors and stored at conjugated fluorochrome-labelled monoclonal antibodies,
- 30 oC 01' lower. routine staining and whole biood lysing procedures, and a
gating strategy using light scatter and immunofluorescence
Reconstitute separately the preparation to be exanIÍned
analysis using a pan-CD45/CD34 monoclonal antibody
and the reference preparation as stated on the labe! and
combinatiol1.
use immediately, Where applicable, determine the amount
of heparin present (2.7.12) and neutralise the heparin, 1t is possible to determine CD34/CD45+ cel! viability by
for example by addition of protarnine sulfate R (lO flg of appropriate l1ucleic acid staining with a stail1 that does
protamine sulfate neutralises 1 IU of heparin). Predilute the not cross the intact cell membrane (for example, with
preparation to be examined and the reference preparation in 7 -aminoactinomycin
factor XI-deficient plasma (for example plasma substrate R3) Selectlon of monodonal antibodies
to produce solutions containing 0.5-2,0 units/mL. Prepare CD34 antibodies. Use class III CD34 antibodies that detect al!
at least 3 appropriate dilutions for each material, preferably glycosylation variants of the molecule (for example, clone 8G 12
in duplicate, using a suitable buffer solution (for example or 581). To deteet rare events, use an antibody conjugated
imidazo/e buffer solution pH 7.3 R) containing 10 gIL ofbovine to the brightest fluorochrome excitable using an argon
or human albumino Use these dilutions immediately, laser-based flow cytometer, for example phycoerythrin (PE).
Use an apparatus suitable for measurement of coagulation CD4S antibodies. Pan-CD45 antibodies that detect al! isofonns
times or perform the assay with incubation tubes maintained and all glycoforms of this structure are required. A CD45
in a water bath at 37 oc. Place in each tube 0.1 mL of antibody conjugated to fluorescein isothiocyanate (FITC)
factor XI -deficient plasma (for example plasma substrate R3) fluorochrome is generally used example, J33, HLel, 2DI).
and 0.1 mL of one of the dilutions of the reference preparation Isotypic or isoclonic contro/s. A negative control is analysed to
or of the preparation to be examined. Add to each tube 0.1 mL detect any non-specific signal in the PE fluorescence region.
of a suitable Activated Partíal Thromboplastin Time (APTT) If using an isotypic control (a monoclonal antibody to an
reagent containing phospholipid and contact activator and irrelevant antigen of the same isotype as the CD34 antibody
incubate the mixture for a recommended time at 37 oc. employed), the PE-conjugated isotype is combined with
To each tube, add 0,1 mL of a 3.7 gIL solution of calcium CD45-FITC (or PerCP), Ifusing an isoclonic control, the
chloride R previously heated to 37 oc. Using a timer, measure unconjugated and PE-conjugated CD34 identical
the coagulation time, Le. the ¡nterval between the moment oí monoclonal antibody is combined with conjugated CD45.
the addition of the calcium chloride and the first indicatiol1 Alternative combinations may be used.
of the formation of fibrin. The volumes given aboye may be
adapted to the APTT reagent and apparatus used, Calculate Absolute count of CD34/CD45+
the potency using the usual statistical methods example, Depending on the technique used,
5.3). either consists of calibrated beads in

258 See the monographs (cover pages)


EUROPEAN PHARMACOPOEIA 8.0 2.7.24. Flow cytometry

suspension or is directly introduced into the associated tubes of moderate antigen density can be distinguished; PMT
by the manufacturero voltages are reviewed and adjusted periodically according
The absolute count of the CD34/CD45+ cells per microlitre is to standardised laboratory procedures.
calculated using the following expression: - Compensation: this must be acceptable fOl" the colour
spectra overlap (for example, FITC/PE) encountered in
.4
-x e cell-surface marker analysis; colour compensation is
B analysed and adjusted according to standardised laboratory
procedures.
A number of CD34/CD45+ cells counted;
- Flow rate: this must be consistent with routine cell-surface
B number of fluorosphere singlets counted; marker analysis.
C known fluorosphere concentration. - Gating regions: the gatil1g regions established for the
CD34/CD45 samples are maintained unaltered for the
Gating strategies analysis of the negative region.
The purpose of sequential gating is to select the population of Cakulation of absolute number of CD34/CD45+ cells
interest and simultaneously minimise interference from debris The absolute number of CD34/CD45+ cells is calculated using
and mature cells to which antibodies can bind non-specifically. the following express ion :
lf using a commercial kit, apply the gating recommended by
the manufacturero If using an in-house assay, it is preferable to nxDxV
applya currently recommended strategy. A gating strategy that
uses light scattering parameters and CD34/CD45 fluorescence 11 total number ofCD34/CD45+ ceUs per microlitre;
will aid in the accurate identification and enumeration of
D dilution factor;
CD34/CD45+ ceUs.
Nmnber of events analysed V volume of the product to be tested, in microlitres.
A sufficient number of events are analysed to maintain Results are reported as both the percentage of
acceptable precision, for example not fewer than CD34/CD45+ cells and the absolute number per
100 CD34+ events and not fewer than 60 000 CD45+ events; microlitre. They may also be reported as the absolute number
the total number of cells counted may be greater if the per kilogram of recipient body mass, where this is possible.
percentage of CD34 is 0.1 per cent or less.
Specimen coUection 01/2008:20724
Acid citrate dextrose (ACD) formula A is the anticoagulant
used in apheresis procedures. This anticoagulant allows both 2.7.24. FLOW CYTOMETRY
an automated leucocyte count and flow cytometry evaluation Flow cytometry consists of a multiparametric analysis of
to be performed on the same specimen. Edetic acid (EDTA) is optical properties of individual particles in a fluidic system.
the anticoagulant of choice for peripheral blood sampling.
Cells or particles in suspension are individually distributed
Spedmen transport into a linear array (stream), which flows through a detection
Transport conditions guarantee the physical and thermal device. Solid tissues have to be reduced to a single-cell
safety of samples. suspension to be analysed.
Specimen integrity and storage The spectrum of parameters measurable by flow cytometry
Fresh (less than 24 h old) apheresis products, whole blood includes volume and morphological complexity of cells
or cell-like structures, ceH pigments, DNA content, RNA
samples, umbilical cord blood specimens or bone marrow
content, proteins, cel! surface markers, intracellular markers,
samples can be processed. Old specimens (more than 24 h
old) and specimens that have been frozen and thawed are enzymatic activity, pH, membrane and fluidity.
stained with a viability dye. 011 receipt, the temperature It is possible to collect 2 morphological parameters plus 1 or
within the package is verified. more fluorescence signals per single cell. The multiparametric
analysis allows the definition of cell populations by their
TECHNIQUE phenotype.
Sample preparation APPARATUS
Ensure that the concentration of leucocytes is suitable prior to Focusing, magnifying, and choice of light source are
staining with monoclonal antibodies. lf necessary, dHute the optimised to allow the automatic detection and measurement
sample with medium that is compatible with the product to be of morphological differences and staining patterns. Flow
tested and the lysing system. Record the dilution factor. It is cytofluorimetric analysis meets the following criteria:
recommended to perform the test with a negative control.
- choice of light source depending on the parameters to be
Flow cytometry analysis analysed;
Autostandardisation - adjustment of instrument settings depending on the ceH
For analysis of cells labelled with a commercially available type to be analysed (for example, ceH cultures, leucocytes,
kit, manufacturers have developed some quality tools platelets, bacteria, spermatozoa, yeast) and the analysis
for setting the flow cytometer. These settings are then to be performed (for example, phenotyping, ceU cycle,
automatically transferred on protocol analysis of samples. apoptosis, cytokines, membrane fluidity, fluorescent
Specific fluorospheres are used to set the photomultiplier tube protein).
(PMT) on target values, compensation is set and the system Flow cytometry is characterised by the automated
is checked using a control preparation. quantification of set parameters for a high number of
System settings single cells during each analysis session. For example,
Discriminator/threshold: the forward angle light scatter 100000 particles or more (practically unlimited) are analysed
threshold is set to exclude debris (low forward scatter) but one after the other, typically in about 1 mino The detection
not smalllymphocytes from the light-scatter plot. limit is as low as 100 fluorescent molecules per cell.
- PMT high voltage settings: these must be consistent with A flow cytometer apparatus has 5 main components:
cell-surface marker analysis and established within each - a fluidic system and a flow ceH;
laboratory so that negative and positive cell populations - a light source;

General Notices (1) apply to all monographs and other texts 259
2.7.24. Flow cytometl'y EUROPEAN PHARMACOPOEIA 8.0

- a detection and Analogue to Digital Conversion (ADC) SIGNAL MANAGEMENT AND ANALOGUE TO DIGITAL
system; CONVERSION
Scatter and fluorescence signals emitted by ceUs when passing
- an amplification system;
across the laser beam are sorted and addressed to their
- a computer provided with software for analysis of the detectors using optical filters. The detectors are transducers
signals. (photomultiplier tubes (PMTs)) that convert light signals
radiated from the cells into voltage pulses.
FLUIDIC SYSTEM AND FLOW CELL
The single ceH is exposed to the light source and detected in The process of counting each pulse in the appropriate channel
the flow cell. The fluidic system carries the suspended cells is known as Analogue to Digital Conversion (ADC). The
individually from the sample tube to the laser intercept point. process is finally shown as a frequency histogram.
To achieve this, the sample stream is drawn out to a very thin Amplification. Voltage pulses need to be amplified for
fluid thread by a sheath fluid in the flow cen (hydrodynamic optimal visualisation. The amplification process accentuates
focusing). The light beam is focused in an elliptical shape, by the differences between ceH signals, and consequently
2 confocallenses, into the flow ceH channel through which the increases the resolution among ceH populations of different
cells pass. The flow rate must be constant during routine cel! characteristics (for example, the differentiation of viable
surface marker analysis and must ensure a suitable distance from non-viable ceUs, or non-specific fluorescence from
between the cells to allow counting. antigen-specific fluorescence after staining with a fluorescent
LIGHT SOURCES monoelonal antibody).
Commonly used light sources are: There are 2 methods of amplification: linear or logarithmic;
the choice between the 2 types is made for every single signal
- lamps (mercury, xenon);
according to the morphological characteristics of the cells
- high power water-cooled lasers (argon, krypton, dye laser); and the staining reagents used (for example, fluorescent
monoelonal antibodies, nueleic acid dyes).
- low power air-cooled lasers (argon (488 nm),
red helium-neon (633 nm), green helium-neon, Linear amplification, which enhances the differences among
helium-cadmium (UV)); strong pulses, is used with those parameters that generate high
intensity signals, for example:
- diode lasers (blue, green, red, violet).
- cell scatters;
SIGNAL DETECTION
When a particle passes across the light beam, it scatters sorne - fluorescence from nueleic acid dyes for ceU cyele studies.
of the light in all directions. Fluorescent dyes, when added to Logarithmic amplification, in contrast, is for weak pulses and
the particle, give off their own light (fluorescence), which is parameters or analysis conditions 'chat may generate both
also radiated in aU directions. 2 types of signals may thereby weak and strong pulses, for example:
be generated:
- ceH antigens;
- scatter of light;
- scatter from platelets, bacteria, yeast;
- fluorescence emission. fluorescence from nueleic acid dyes for apoptosis studies.
The instrument's light detectors collect sorne of this Compensation of fluorescence signals. Each fluorescent
scattered and fluorescent light and produce electronic signals dye has an absorption wavelength spectrum and a higher
proportional to the amount of Iight collected. emission wavelength spectrum. When using 2 or more
Scattel'. 2 parameters of light scattering are measured: fluorescent probes simultaneously for staining cells (for
example, 4-antigen immunophenotyping), the fluorochromes
- the amount scattered mainly forward (forward scatter (FS)) emission spectra may overlap. As a consequence, each
fluorescence detector will sense its own specific fluorescent
- the amount scattered at 90° from the direction of the light
light and a variable quantity of light emitted by the other
beam (side scatter (SS)).
fluorescent probes. This results in signal over-evaluation and
Forward scatter correlates with the ceH volume while side poor separation of the ceU populations.
scatter is influenced by parameters such as the shape of the The solution is in the use of an electronic matrix that allows
nueleus, the amount and type of cytoplasmic granules or the the selective subtraction of the interfering signals from
membrane roughness, and correlates with the morphological each fluorescence signal after detector sensing (fluorescence
complexity of the cell, so that the higher the SS intensity, the compensation).
higher the cel! complexity. As a function of the morphological
characteristics of ceUs, scatter signals will always be generated Fluorescence compensation requires the use of fluorescence
during a flow analysis; they are defined as intrinsic parameters. calibrators, preferably positive cell samples stained with the
fluorochromes of interest, combined in a manner equivalent
Fluorescence. Depending on the type and number of light to that for the antibody used for the analysis. -
sources, when a ceH passes through the sensing are a, it will
emit fluorescent light. Fluorescence signals are generated from SIGNAL PLOTTING AND DISPLAY
fluorescent dyes naturally present in the ceHs (for example, After amplification and compensation, the signals are plotted
co-enzymes, chlorophyll, seaweed pigments) and/or from in 2 or 3 dimensions. Histograms show the signal intensities
fluorescent probes taken up by the ceUs when stained for the versus the cel! counts for a given parameter. Cytograms, in
analysis of specific characteristics (for example, fluorescent which each dot represents a cell, result from the combination
antibodies, nueleic acid dyes, pH probes, calcium probes, of 2 signal intensities (dual-parameter dot plots). The type
fluorescent proteins). Nowadays, there is a large number and and number of plots and signal combinations are chosen on
a wide range of different types of fluorescent probes available. the basis of the specimens and dyes used. When analysing
The optical filters must be adapted to the fluorochromes acquired data, the flow cytometry software can also generate
used and changed if necessary. Each fluorescent probe is other kinds of graphs (such as overlays, surface plots,
characterised by its excitation spectrum and its emission tomograms, contour plots, density plots, overlay plots).
spectrum. They are chosen depending on the nature of the Statistical data such as mean fluorescent intensities (and their
excitation source and the detection system, and according to shifts in time or their dependence on ceU function) can also
the specific purpose of the analysis. be used.

260 See the information section on general monographs (ca ver pages)
EUROPEAN PHARMACOPOEIA 8.0 2.7.27. Flocculation value oftoxins and toxoids (Ramon assay)

DATA ANALYSIS plasmin inhibitor. Plasmin deavage of the chromogenic


Different kinds of cel! populations may be present in si de the substrate yields a chromophore that can be quantified
cel! suspensions to be analysed, sorne of which are unwanted spectrophotometrically.
(such as dead cells, debris or macro-aggregates), or simply The individual reagents for the assay may be obtained
not relevant for the analysis (for example, granulocytes when separately or in commercial kits. Both end-point and kinetic
studying lymphocytes). This depends on the cel! sample methods are available. Procedures and reagents may vary
type (whole blood, bone marrow, cel! cultures, biological between different kits and the manufacturer's instructions are
fluids, cel! suspensions from solid tissues) and on the followed. The essential features ofthe procedure are described
handling procedures (for example, staining methods, lysis, in the following example of a microtitre-plate kinetic method.
fixation, cryopreservation, thawing, paraffin-embedded tissue
preparation). REAGENTS
As a consequence, not all the signals generated during a flow Dilution buffer pH 7.5. According to the manufacturer's
cytometry analysis belong to the cells to be studied. 2 strategies instructions, a suitable buffer is used. Adjust the pH if
are adopted to exdude unwanted and irrelevant cell signals. necessary.
The 1st is used during data acquisition. It is a noise threshold, Plasmin. A preparation of human plasmin that does not
applied to 1 (or more) significant parameter(s), set to contain significant amounts of other proteases is preferably
acquire only the cells with signal intensities higher than the used. Reconstitute and stofe according to the manufacturer's
pre-defined discrimination value for that parameter. Due instructions.
to its characteristics of a strong signal with a low grade of Plasmin chromogenic substrate. A suitable specific
interference, forward scatter is the parameter most often used chromogenic substrate for plasmin is used: H-
as discriminator. D-cydohexylalanyl-norvalyl-Iysyl-p-nitroaniline
The 2nd , applied during data analysis, consists ofthe use of hydrochloride (H-D-CHA-Nva-Lys-pNA.HCl) or
gating regions to restrict the analysis only to signals from L-pyroglutamyl-L-phenylalanyl-L-lysyl-p-nitroaniline
those populations that satisfy given morphological and hydrochloride (Glp-Phe-Lys-pNA.HCl). Reconstitute in
expression profile characteristics. 2 types of logical gating are water R to give a suitable concentration according to the
commonly use d . T h e 1st is t1le morphologica1 gateo The cell manufacturer's instructions.
populations are identified using their morphological signals METHOD
(FS and SS). A region gate is drawn around the population
of interest (for example, Iymphocytes, viable cells) then the Varying quantities of the preparation to be examined are
fluorescence plots are gated into the selected region. The 2nd is mixed with a given quantity of plasmin and the remaining
the fluorescence-based gateo The cel! population of interest is plasmin activity is determined using a suitable chromogenic
identified on the basis of the expression intensity of an antigen substrate.
or a dye, then a gate region is drawn around it. Afterwards the Reconstitute or thaw the preparation to be examined according
fluorescence plots are gated into the selected region. to the manufacturer's instructions. Dilute with dilution buffer
The analysis software allows the creation of multiple gate pH 7.5 and prepare at least 2 independent series of 3 or
regions, using a sequentiallogic order. This feature is 4 dilutions for both the preparation to be examined and the
especially useful when studying rare ceH populations or for reference standard.
sorting purposes. Mix 0.020 mL of each dilutíon with 0.020 mL of dilution
buffer pH 7.5 and warm to 37 oc. Add 0.040 mL of a plasmin
CONTROLS
solution (test concentration in the range of 0.2 nkat/mL to
Internal control. The systenú optical alignment must be 1.6 nkat/mL) previously heated to 37 oC and leave at 37 oC for
validated before analysis using adapted fluorospheres and the 1 mino Add 0.020 mL of the chromogenic substrate solution,
optimum fluidic stability is checked. The data obtained are previously heated to 37 oC, to each mixture. Immediately start
reported and allow the periodical review of control values measurement of the change in absorbance at 405 nm (2.2.25)
against the mean performance value. A positive control is using a microtitre plate reader. Calculate the rate of change
highly desirable to prove that the test antibody is functional of absorbance (6A/min). Alternatively, an end-point assay
and to allow the proper setting of the flow cytometer. The might be used by stopping the reaction with acetic acid and
positive control must in dude samples known to be positive measuring the absorbance at 405 nm.
for the marker of interest. In both cases the duration of the deavage of the chromogenic
External control. To ensure reliability in the data obtained or substrate should be chosen to produce a linear increase in
to check inter-laboratory reproducibility, participation in a absorbance at 405 nm, before substrate depletion becomes
proficiency testing study is recommended. significant. If the assay is performed in test tubes or cuvettes
using a spectrophotometric method, the volumes of reagent
solutions are changed proportionally.
Substract the optical density of the blank (prepared with
07/2009:20725 dilution buffer pH 7.5) from the optical density of the
preparation to be examined. Check the validity of the assay
2.7.25. ASSAY OF HUMAN PLASMIN and calculate the potency of the preparation to be examined
by the usual statistical methods (5.3).
INHIBITOR
Human plasmin inhibitor, also called human u 2 -antiplasmin,
01/2008:20727
is a plasma protein that inhibits the plasmin (a serine
protease) pathway of fibrinolysis by rapidly forming a complex
with free plasmin. Furthermore, upon blood coagulation, 2.7.27. FLOCCULATION VALUE (Lf) OF
human plasmin inhibitor is cross-linked to fibrin strands by DIPHTHERIA AND TETANUS TOXINS
factor XIII, and interferes with binding of the proenzyme
plasminogen to fibrin. AND TOXOIDS (RAMON ASSAY)
The potency of human plasmin inhibitor is estimated by The content of toxin or toxoid in a sample can be expressed as
comparing the ability of the preparation to be examined to a flocculation value (Lf) using the Ramon assay. In this assay,
inhibit the deavage of a specific chromogenic substrate by antitoxin is added in increasing concentrations to a series of
plasmin with the same ability of a reference standard ofhuman tubes containing a constant amount of toxin or toxoid. At the

General Natices (1) apply ta all managraphs and ather texts 261
2.7.28. CFC assay for human haematopoietic progenitor cells EUROPEAN PHARMACOPOEIA 8.0

equivalence point of toxin/toxoid and antitoxin, flocculation If there is no indication of the expected Lf value of the sample
occurs in 1 or more tubes. The first tube in which flocculation available, it is advisable to obtain a rough estimate by use
occurs is used to determine the Lf value of the sample. of a wider range of antitoxin content in the tubes before
The Lf value of a toxin or toxoid is determined by the number proceeding to the final test.
oí units of antitoxin that, when mixed with the sample, Example
produces an optimally flocculating mixture (Ramon assay).
Tube A B e D E F
Practical experience has shown that the results of the
calibration of antitoxins in International Units (IU), for Antitoxin 20 30 45 70 100 150
content
example by comparison to international antitoxin standards, (Lf-eg.)
depends on the immunochemical method used. For this
reason, antitoxins used for the Ramon assay must be directly The level of toxin or toxoid and antitoxin concentration
calibrated against the in ternational biological reference in the test may be varied, but this will markedly affect the
reagents for diphtheria or tetanus toxoid for flocculation tests, flocculation time, so that at very low levels the test will take too
using the principies described below. The concentration thus long, whilst at a high concentration the onset of flocculation
determined may be indicated in Lf-equivalents per millilitre may be so rapid as to make it difficult to distinguish the first
(Lf-eq./mL). and second tubes to flocculate.
By definition, 1 Lf is the quantity of toxin or toxoid that Assay of low concenírations by blend flocculation
flocculates in the shortest time with 1 Lf-eq. of specific For very low concentrations, it is preferable to measure toxin
antitoxin. or toxoid by the method of blend flocculation. This involves
A range of volumes of the reference standard of antitoxin comparison of the Lf value of a known toxin or toxoid and
adjusted to a concentration of 100 Lf-eq./mL is dispensed into that of a mixture of the sample with that toxin or toxoid.
a series of, for example, 7 cm x 1 cm flocculation tubes. A When a toxin or toxoid with a known Lf value and a toxin
sufficient quantity of a 9 giL solution of sodium chloride R or toxoid with an unknown Lf value are flocculated together,
is added to each tube to give a constant total volume of, for the mixture will flocculate as the sum of their values if they
example, 1 mL. The test sample is diluted to give an expected are homogeneous. If non-homogenous toxins or toxoids
concentration of approximately 50 Lf/mL, and, for example, are mixed they will produce an aberrant pattern with
1 mL aliquots of this dilution are dispensed into each of the 2 flocculation maxima.
tubes containing antitoxin. The tubes are properly mixed by
shaking, then placed in a water-bath at a constant temperature
between 30 oC and 52 oC, and observed at regular intervals for
the first appearance of floccules. This may requi1'e the use of a 0112008:20728
magnifying lens and strong illumination.
The first and the second mixtures to flocculate are reco1'ded
as well as the time taken for the first flocculation to appear. 2,7.28. COLONY-FORMING
2 tubes may flocculate simultaneously. CELL ASSAY FOR HUMAN
The first tube to flocculate is the one that contains the amount HAEMATOPOIETIC PROGENITOR
of antitoxin closest in equivalence to the amount of antigen in
the sample. The antitoxin content of this tube can be used to
CELLS
calculate the Lf value of the sample. If 2 tubes flocculate at the The haematopoietic system represents a continuum of cells
same time, the mean from the tubes are given as the result. whose phenotype and properties change as they progress from
The time taken for the first tube to flocculate (Kf) is a stem cells to differentiated cells.
useful indicator of the quality of the antígeno If at a given Haematopoietic progenitor ceUs (HPCs) are capable of
temperature and concentration of toxoid and antitoxin the forming colonies or 'cen clusters' in cultures grown in
Kf value is increased compared with normal, this indicates semi-solid media and are said to be 'clonogenic'. The
that the antigen has been damaged. The Kf value may also determination of the number of colony-forming cells (CFCs)
change with the quality of the antitoxin used. in a cellular product is an indicator of the functional capacity
Example of the progenitor cells and is a predictor of haematopoietic
reconstitution. The measured number of CFCs correlates with
Tube A B e D E F
the minimum number of progenitor s present in the sample.
Antitoxin added 40 45 50 55 60 65
(Lf-eq.) CELL-SURFACE MARKERS
Antitoxin added 0.40 0.45 0.50 0.55 0.60 0.65 The capacity of colony-forming cells to give rise to
(mL) haematopoietic colonies in vitro and/or to reconstitute
Saline added 0.60 0.55 0.50 0.45 0.40 0.35 the haematopoietic system has been correlated with the
(mL) expression of specific cell-surface antigens. The expression of
Diluted sample 1.0 1.0 1.0 1.0 1.0 1.0 the membrane antigen CD34 is an accepted marker for most
added of the haematopoietic progenitors and stem cells.
If in this example the first tube to flocculate is tube C then the
Lf value of the diluted sample is 50 Lf/mL. However, if the first COLONY ASSAY SPECIFICITY
tube to flocculate is tube A or tube F this do es not indicate Colony-forming cells are identified with a nomenclature
equivalence at that leve!. It would be necessary to perform a based on the lineages of mature cells present in the colony
repeat test using either a different dilution of test sample or (for example, CFU-Mix, CFU-GEMM, CFU-GM, CFU-G,
selecting a different range of doses of reference antitoxin. CFU-M, BFU-E, CFU-E, CFU-Meg) and are a population of
More precision can be obtained by making allowance for progenitors able to give rise to colonies containing one or
the sequence of flocculation after the first tube. Thus, in the more lineages of haematopoietic cells. No or low capacity for
example quoted, if the second tube to flocculate had been self-renewal has been ascribed to this population of human
tube D, the final value fo1' the diluted sample would be 52, HPCs compared with the most immature stem cells.
whereas if the second tube to flocculate was tube B, the final The amount and type of growth factors supplied during the
value would be 48. The test may be performed in duplicate culture modulate the type and size of colonies that will be
with slightly different dilutions of the test sample. formed.

262 See the information section on general monographs (cover pages)


EUROPEAN PHARMACOPOEIA 8.0 2.7.29. Nudeated ceU count and viabiHty

Greater specificity 011 the general class of HPCs and on their Because of the of the medium, the solution cannot be
relative proliferative potential is provided by the time required plated with air displacement pipettes and the use of syringes
to differentiate in vitro into mature cells. The time required by with large bore (-s: 18-gauge) needles is required.
post-natal colony-forming cells to give rise to a colony formed The number of ceUs to be plated depends on the HPC
of mature cells in vitro is 10-14 days. concentration in the sample to be tested. So that no colony
is derived from 2 different HPCs, the number of cells plated
QUALITY ASSURANCE FOR A CFC ASSAY must allow between 40 and 80 colonies per plate (035 mm)
1t is paramount for the overall quality of the colo11Y-forming to be counted. The number of coJonies plate
cel! assay to apply a strictly standardised approach. of (or
It is therefore recommended to carry out i11tra- and concentration of CD34+ by flow
inter-laboratory validations. The source of the materials, (2.7.24) or from different dilutions of the cel!
including reagents, growth factors amI disposables, is suspension (usually 2 concenüations are tested).
identified. The plates are incubated in aerobic conditions with a carbon
The mail1 factors affecting variability in the CFC assay are the dioxide concentration of 5 per cent, at 37°C in a humid
number of ceUs plated and the identiflcation of colonies. Up (saturated) atmosphere for 10-14 days, and the number of
to 15 per cent intra-laboratory variability may be observed coJonies is then scored under an inverted microscope. Care
for the same test. If it is necessary to evaluate the number of must be taken when manipulating the dishes containing the
colony-forming cells in a purified cel! population, it is possible colonies as the methylcellulose-based medium is viscous
to use a limiting dilution approach where the number of wells but 110t jellified. An inclined plate wil! result in mixed and
positive for cel! proliferation i8 measured with an automated 'comet' -shaped colonies making the scoring likely to be
system. incorrect.
The other main source of variability stems from the use of IDENTIFICATION OF THE COLONIES
undefined materials (for example, foetal bovine serum or The size and structure oE the colonies on the type
bovine serum albumin) in the CFC assay. These products of mature cells that are their constituents. 50 cells per
derive from pools of source materials and provide a is usually considered a minimum. The presence of
non -specific stimulatiol1 of cellular proliferation. However, haemoglobinised cells identifies of the erythroid
it is not uncommon to have batches with particular lineage. As the amount of matm·e for each lineage
characteristics that selectively stirl1ulate the proliferation of largely OH the growth factors added to the cultures,
specific haematopoietic lineages. performing differentiated counts is not recommended unless
otherwise prescribed.
Finally, a low level of endotoxins (less than 0.01 IU/mL or less
thal1 0.01 IU/mg) in al! the materials used for the clonogenic EXPRESSION OF THE RESULTS
assay is advisable, as higher levels result first in a The results of CFe culture are expressed as the
skewil1g of the haematopoietic lineages expression in the arithmetic mean of the number of counted in at least
cultures, and afterwards in a more general inhibition of cel! 3 in the test. The mean number of colonies is then
proliferation and LlUllV",eJlleÓ to 10-1 or 105 viable nucleated ceUs in culture.

CFC CLONOGENIC ASSAY


The CFC assay is based on the cells to !H/2008:20729
form a colony when plated in a
in the presence of specific growth factors. Different types
semi-solid media may be used example, methylcellulose, NUCLEATED COUNT
collagen, agar and plasma-clot) depending 011 the desired AND VIABILITY
readout. Commercially available media usually give more
reproducible results. The determination of the requires
accurate measurements cel! cOl1centration and
MATERIALS percentage of viable cells. These data are essential to the
A validation is performed at least for the following critical decision-making pro ces s for celllllar products and
materials. for maintaining optimum The cel! count
Growth factors. Both multilineage (such as Kit-ligand or may be expressed as the number of cells per volume of ceH
stem cell factor (SCF), interleukin-3, granulocyte-macrophage suspension and the cel! viability as the l111mber of viable cells
colony-stimulating factor (GM-CSF)) and lineage-speciflc per volume of cell The cell-count procedure
(erythropoietin, granulocyte colony-stimulating factor may be performed manualJy (haemocytometer) or with an
(G-CSF)) growth factors are required to obtain the highest automated apparatus (for example, particle counter, flow
number of colonies from a cell suspension containing a mixed cytometer). Other methods than that described below may
population of HPCs. be used.

Other media components. Media may be supplemented by CELL NUMBER


serum (notably by foeta! bovine serum) and/or albumino MANUAL COUNTING
CELL CULTURE Descriptioll al the apparatus and The
Censo The sample placed in culture must be representative of materia!s are required:
the cellular product injected. Cell suspensions are required - a :a counting
for this assay. In the case ofbone marrow aspirates, such chamber available in desüms. It consists of a thick
suspensions can be obtained by forcing the bone marrow slide and a coverslip mounted to d~limit a chamber with
through a sieve or through progressively smaller calibre a specific volume for eaeh The thick slide of the
needles. Repeated passages through a 21-gauge needle are various of counting chambers
usually sufficient to disperse cell clusters into a ceH suspension. deep to avoid cross-fllling. The
counting chamber etched in the and cOlltains a grid
PLA TING AND SCORING
which is for each model;
The cells diluted in the culture medium are mixed in the
semi -solid medium. It is comm011 to pIate J mL of the mixture - a light -low power 10x to 40x magnification;
in an untreated sterile Petri dish (035 mm). of a suitable volume range.

General Notices (1) apply io all ,~r'Mr'Mn~J'"" and other texts 263
2.7.29. Nudeated (eH count and viability EUROPEAN PHARMACOPOEIA 8.0

The haemocytometer is used to quantify the number of cells membrane integrity but its results do 110t necessarily reflect
in a given solution by calculation of the cell concentration per cel! functionality. Recently trypsinised or thawed viable cells
millilitre (C) using the following expression; may have leaky membranes, causing them to absorb the dye.
Dye. Trypan bIue is the stain most commonly used to
a, x lOn x d
distinguish between viable al1d non-viable cells, but other
a number of cells counted; suitable dyes such as erythrosin B or nigrosin may also be
used. It is an acid dye (Mr 961), an anion with 4 sulfonate
d dilution factor (where applicable); groups that can easily bind to proteins; therefore the protein
n factor varying with the volume of the cOl1centration of the preparation to be tested must be as low
haemocytometer chamber. as possible.
Test conditio,ns. Dye fixation is strongly influenced by pH,
1t is possible to distinguish between mixed cel! populations within a range of 6.6 to 7.6. Fixation is optimal at pH 7.5.
provided they differ in size or pigmentation (for example, The other conditions, such as the dye concentration and the
leukocytes and erythrocytes). staining time are validated.
Preparatio,n o,f the co,unting chamber and analysis. Mount the Sto,rage conditions o,f the dye: Generallya 0.4 or 0.5 per cent
coverslip (slightly moistened on the edges) on the slide. Move trypan blue solution in sterile phosphate-buffered saline is
the coverslip back and forth over the slide, pressing slightly 011 used. Store protected from light and airo
the sides. Prepare a suitable dilution of the ceH suspension in
isotonic buffer or in haemolysis buffer. Test preparatio,n and analysis. Stain the ceH suspension at
the required dilution (usually in phosphate-buffered saline)
Add an appropriate volume of the dilution to the counting with, for example, a trypan bIue solution having a final
chamber. The liquid is added to the border of the coverslip and concentration of 0.1 to 0.2 per cent. Mix gentIy. Incubate for
is drained inside the chamber by capillarity. Carefully place not more than 2-4 min at room temperature. Mix gently and
the haemocytometer under the microscope and focus. Count place a suitable volume in a counting chamber. Count without
the ceUs in a zone of the grid. Calculate the ceH concentration delay.
in the diluted and original samples.
Determine the percentage of viable cells from the ratio of the
To increase the accuracy of the measurement, it is important number of unstained cells to the total number of cells under
to respect the foHowing basic precautions: a light microscope, considering al! stained cells as dead cells.
- use only suitably thickened coverslips; Viability (V) is calculated as a percentage using the following
- wherever possible, count more than 100 cells (if necessary, expression;
count more are as) ;
~ x 100
- where ceH clustering is detected (i.e. the ceH suspension N
is not monocellular), resuspend the ceUs before sampling
and count again; n number of unstained (viable) cells;
avoid underfilling or overflowing the chamber, otherwise N total number of cells (stained and unstained).
the volume will no longer be accurate.
AUTOMATED COUNTING METHODS It is essential that the incubation time be not more than 4 min
Particle co,unters based o,n conductivity variatio,n. Electronic as the number of stained cells may increase significantly
particle counting devices measure the size and number of afterwards. For a new determination, it may therefore be
particles in a solution. necessary to prepare a new test.
Particle counters are calibrated before use with a solution AUTOMATED METHODS
of particles of known concentration and size. To allow the Flow cytometry
counting of larger particles, tubes fitted with differently Test principie. The test is based on the ability of certain dyes to
calibrated orifices are available. These apparatuses do not cross damaged membranes and bind to DNA by intercalating
allow the discrimination between dead and live cells. As between bases so that dead cells may fluoresce and be detected
cell debris may also generate pulses that may cause errors, by flow cytometry (2.7.24). Non-viable cells are evaluated and
counters are also fitted with a threshold control allowing only discriminated by focusing 011 positive staining whereas viable
larger particles to be counted. cells remain unstained. This analysis is generally performed
The apparatus must be qualified for the counting of cellular with 7-aminoactinomycin D (7-AAD) or propidium
products (in terms of linearity, accuracy, etc.). iodide (PI) but other suitable dyes may also be used.
Particle counters based o,n flo,w cyto,metry (2.7.24). The flow Dye. 7-AAD and PI are given as examples of
cytometer is calibrated with reference particles of known membrane-impermeants that may be used as viability
concentration and size to give an absolute cell number per dyes.
volume. However, a calibrating solution is no longer necessary 7 -AAD is an analogue of actinomycin D that contains a
in instruments using 2 electro des inserted in the sampling substituted amino group at position 7 of the chromophore.
chamber where the fixed size of the sampling chamber and It intercalates between cytosine and guanine DNA bases.
distance between the 2 electrodes allow the measurement of The spectral properties of 7-AAD make this molecule
the content of a fixed volume. This type of instrument rarely particularly suitable for flow-cytometry analysis. The
needs to be calibrated after the initial setting. maximum absorption of the 7-AAD/DNA complex is situated
VIABILITY in the green spectral region and is thus suitable for an argon
laser-equipped cytometer (excitation wavelength of 488 nm).
This section applies to cell staining by viability dyes and The deep red fluorescence emission of the 7 -AAD viability dye
manual or automated analysis, under a light microscope or (635 nm to 675 nm) eases the use of the probe in combination
by flow cytometry, of a ceH suspension in order to determine with fluorescein isothiocyanate (PITC) and phycoerythrin
the percentage of viable cells. (PE)-conjugated antibodies, because in contrast to PI, the
Depending on the type of cells and the method used, the 7-AAD/DNA complex shows minimal overlap with FITC and
results may differ. PE.
MANUAL DYE-EXCLUSION METHOD PI binds to double-stranded DNA by intercalating between
Test principie. This test is based 011 the exclusion of the dye bases with little or no sequence preference and with a
from viable cells whereas dead or damaged cells absorb the dye stoichiometry of 1 dye molecule per 4-5 DNA base pairs. Once
and are coloured. 1t provides information on the cytoplasmic the dye is bound to nucleic acids, its fluorescence is enhal1ced

264 See the info,rmatio,n section o,n general mo,no,graphs (cover pages)
EUROPEAN PHARMACOPOEIA 8.0 2.7.30. Assay ofhuman protein e

20- to 30-fold, the fluorescence excitation maximum is shifted kits and the manufacturer's instructions are followed. The
around 30-40 nm towards the red and the fluorescence essential features of the procedure are described in the
emission maximum (615 nm) is shifted around 15 nm towards following example of a microtitre plate end-point method.
the blue. Although its absorptivity is quite low, PI exhibits a
sufficiently large Stokes shift to allow simultaneous detection REAGENTS
of nucleic acids and fluorescein-labelled antibodies, provided Dilution buffer pH 8.4. Dissolve 6.055 g of tris(hydroxy-
that the suitable optical filters are used. methyl)aminomethane R and 16.84 g of caesium chloride R in
Storage conditiol1S of nucleic acid dye solution: 5 ± 3 De. water R and adjust the pH if necessary. Dilute to 1000.0 mL
with water R.
Test preparatían and analysis. In the case of haematopoietic
ceUs, the dye may be added after CD45 labelling to obtain a Human protein e actívatar. Protein isolated from the venom
better separation of cells from debris and platelets with a side of the viper Agkistrodon contortrix contortrix that specifically
scatter (SS)/CD45+ gating region. The incubation conditions activates human protein e. Reconstitute and store according
of the cel! suspension with the dye are validated previously. to the manufacturer's instructions. Dilute to 0.25 U/mL with
water R before use in the assay.
Incubation is performed al room temperature protected from
light. Where necessary, lysis of red blood cells is performed Activated protein e chromogenic substrate. Specific
using, for example, ammonium chloride. If not, add buffer chromogenic substrate for APe, for example
alone. L- pyroglu tamyl-L-prolyl-L-arginyl-p-nitroaniline
Percentages of viable ceUs are directly given by the flow hydrochloride (pyroGlu-Pro-Arg-pNA.HCl). Reconstitute
cytometer and deduced from the analysis of positive cells with water R to give a concentration of 4.5 mmol/L. Further
(dead cells) in the SS/7-AAD or SS/PI cytogram (dot plots). dilute to 1.1 mmo1/L with dilution buffer pH 8.4 before use in
the assay.
Positive contro15 may consist of stabilised cells (dead cells)
mixed with fresh viable cells at a target value. METHOD
Digital i.maging of stained ceUs. Digital imaging allows the Reconstitute or thaw the preparation to be examined according
automation of dye-exclusion methods. The cel! suspension to the manufacturer's instructions. Dilute with water R to
and viability-dye solution are directly mixed by a machine. produce at least 3 separate dilutions for each preparation in
The system, which allows sample aspiration, reagent handling, the range 0.050-0.200 IV/mL, preferably in duplicate.
and subsequent instrument cleaning is fully automated. Once Step l. Mix 0.025 mL of each dilution with 0.050 mL of the
the cellular suspension has been aspirated and mixed with the human protein C activator, both previously heated to 37 oc,
dye solution, it is pumped to the flow cell for imaging. The and leave at 37 oC for exactly 10 mino For each dilution,
stained ceH suspension is aspirated through a chamber where prepare a blank in the same manner, using water R instead of
stroboscopic light allows a camera to photograph the flowing the human protein C activator.
ceUs. The images are digitalised and the number of dead or
live cells counted by the software. Step 2. Add 0.150 mL of diluted chromogenic substrate,
previously heated to 37 oC, to each mixture and leave at 37 oC
for exactly 10 mino The incubation time must be adjusted, if
07/2008:20730 necessary, to ensure a linear development of chromophore
corrected 7.0 with time. Terminate the reaction by adding 0.050 mL of a
50 per cent V/V solution of glacial acetic acid R.
2.7.30. ASSAY OF HUMAN PROTEIN e Cleavage of the chromogenic substrate by APC causes releas e
of the chromophore pNA, in proportion to the concentratiol1
1. CHROMOGENIC ASSAY of human protein e in the preparatiol1. Measure the optical
Human protein C is a vitamin K -dependent plasma protein density at a wavelength oí 405 nm. Subtract the optical density
that, upon activation to activated protein e (APC), can inhibit of the blank from the optical density of the test sample. Check
blood coagulation through the proteolytic cleavage of factors the validity of the assay and calculate the potency of the
Va and VlIIa. Human protein e activity is estimated using preparation to be examined using the usual statistical methods
a two-step method: in the 1st step, human protein e in the (5.3).
preparation is activated by a specific activator from snake
venom; in the 2 nd step, APC cleaves a specific chromogenic 2. CLOTTING ASSAY
substrate to form a chromophore that can be quantified Human protein e activity is estimated following cleavage
spectrophotometrically. to APC by a specific activator extracted from the venom of
the viper Agkistrodon contortrix contartrix. The resulting
Step 1
APC inactivates factors Va and VIlla, and thus prolongs the
human protein e activator APTT (Activated Partía! Thromboplastin Time) of a system
human protein e >- APe in which a11 the coagulation factors are present, constant and
in excess, except for human protein C, which is derived from
the preparation being tested. Prolongation of the clotting time
Step 2 is proportional to the concentratiol1 of human protein C in
the preparation.
APe 'd e + ehromop hore The potency of human protein C is estimated by comparing
chromogenic substrate ----i)oll!!>-peptl
the ability of the preparation to be examined to prolong the
The potency of human protein C is estimated by comparing clotting time with the same ability of a reference standard
the ability of the preparatiol1 to be examined to cleave a of human protein e calibrated in International Units. The
chromogenic substrate with the same ability of a reference International Unit is the activity of a stated amount of the
standard ofhuman protein C calibrated in International Units. International Standard for human protein C. The equivalence
The International Unit is the activity of a stated amount of the in International Units of the International Standard is stated
International Standard for human protein e. The equivalence by the World Health Organization.
in International Units of the International Standard is stated Individual reagents may be obtained separately or in
by the World Health Organization. commercial kits. Procedures and reagents may vary between
Individual reagents may be obtained separately or in different kits and the manufacturer's instructions are followed.
commercial bts. Both end-point and kinetic methods are The essential features of the procedure are described in the
available. Procedures and reagents may vary between different following example.

General Notices (1) apply to all monographs and other texts 265
2.7.31. Assay ofhuman protein S EUROPEAN PHARMACOPOEIA 8.0

REAGENTS sodium chloride R in water R and adjust the pH if necessary;


Dilution buffer pH 7.4. Isotonic non-chelating buffer. add 10 g of bovine albumin R or human albumin R and dilute
Human protein C-deficient plasma. Citrated human plasma to 1000.0 mL with water R.
with no measurable human protein C content. Reconstitute Human protein S-deficient plasma. Citrated human plasma
and 5tore according to the manufacturer's instructions. with no measurable human protein S content and, preferably,
Human protein C activator. Protein isolated from the venom also free of C4b-binding protein.
of the viper Agkistrodon contortrix contortrix that specifically Coagulation activator. This reagent is used to initiate
activates human protein e. Recol1stitute and store according coagulation in the human protein S-deficient plasma, and
to the manufacturer's instructions. thereby also provides a source of activated factor V. The
Coagulation activator. A suitable APTT reagent containing activator may consist of tissue factor, activated factor X, or
phospholipids and a contact activator may be used. Jt may be an agent capable of directly activating factor X that may be
combined with the human protein C activator. purified from the venom of Russell's viper (Vipera russelli).
The reagent also contains APC, phospholipids and calcium
METHOD chloride R, or, alternatively, calcium chloride may be added
Reconstitute or thaw the preparation to be examined according separately after a timed activation periodo
to the manufacturer's instructions. Dilute with dilution
METHOD
buffer pH 7.4 to produce at least 3 separate dilutions for each
preparation in the range 0.010-0.150 IU/mL, preferably in Reconstitute or thaw the preparation to be examined according
duplicate. to the manufacturer's instructions. Dilute with dilution
Mix 1 volume of each dilution with 1 volume of human buffer pH 7.4 to produce at least 3 separate dilutions for each
protein C-deficient plasma and 1 volume of the human preparation in the range 0.020-0.100 IU/mL, preferably in
protein C activator (combined with the APTT reagent where duplicate.
appropriate), all previously heated to 37 0e. Add 1 volume Mix 1 volume of each dilution with 1 volume of human
of 0.025 M calcium chloride solution R previously heated to protein S-deficient plasma, both previously heated to 37 oc.
37 oC, and record the clotting time. Add 2 volumes of the coagulation activator, previously heated
The clotting time is proportional to the concentration of to 37°C, and record the clotting time.
human protein C in each dilution. Check the validity of Alternative procedures may use a coagulation activator
the assay and calculate the potency of the preparation to be without calcium chloride, and require a precisely timed
examined using the usual statistical methods (5.3). activation period before the addition of calcium chloride and
the measurement of dotting time.
07/2008:20731 The clotting time is proportional to the concentration of
human protein S in each dilution. Check the validity of the
2.7.31. ASSAY OF HUMAN PROTEIN S assay and calculate the potency of the preparation to be
examined using the usual statistical methods (5.3).
Human pro te in S is a vitamin K-dependent plasma protein that
acts as a cofactor for the anticoagulant functions of activated
protein C (APC). Human protein S activity may be determined 07/2008:20732
by the clotting assay described below, which is sensitive to the
ability of human protein S to accelerate the inactivation of 2.7.32. ASSAY OF HUMAN
factor Va by APe. In practice, the assay involves the addition a-l- PROTEINASE INHIBITOR
of human protein S to a reagent mixture containing APC,
factor Va and human protein S-deficient plasma. Prolongation Human a-l-proteinase inhibitor (also known as
of the clotting time is proportional to the cOl1centration a-l-antitrypsin or Cl-l-antiproteinase) content is determined
of human protein S in the preparation. Methods in which by comparing the ability of the preparation to be examined
APC is added directly as a reagent are preferred to those in to inactivate the serine protease elastase (porcine pancreatic
which APC is generated during the assay by the addition of elastase OI human neutrophil elastase) with the same ability
a specific human protein C activator purifled from snake of a reference standard ofhuman a-l-proteinase inhibitor
venom. Activation of coagulation is initiated by the addition calibrated in milligrams of active (functional) Cl-l-proteinase
of an activating reagent such as thromboplastin or activated inhibitor. Varying quantities of the preparation to be examined
factor X, together with phospholipids and calcium chloride. are mixed with a given quantity of elastase and the remaining
During the assay, factor Va is generated from factor V in the elastase activity is determined using a sllitable chromogenic
human protein S-deficient plasma following the activation of substrate. The method described below is given as an example.
coagulation. The assay procedure must ensure that human
protein S is the only limiting factor. REAGENTS
The potency of human protein S is estimated by comparing Tris-albumin buffer solution. Dissolve 24.23 g of trometamol R
the ability of the preparation to be examined to prolong the in water R, adjust to pH 8.0 ± 0.3 using hydrochloric acid Rl
clotting time with the same ability of a reference standard and dilute to 1000 mL with water R. To 100 mL of this solution
of human protein S calibrated in International Units. The add 0.5 mL of a 20 per cent solution of human albumin R or
International Unit is the activity of a stated amount of the bovine albumin R.
International Standard for human protein S. The equivalence Buffer solution containing human or bovine albumin must
in International Units of the International Standard is stated be prepared fresh on the day of its use; otherwise, it can be
by the World Health Organization. conserved by sterile filtration (0.2 f-lm) and stored at 2-8 oC
Individual reagents may be obtained separately or in for up to 2 weeks.
commercial kits. Procedures and reagents may vary between METHOD
different kits and the manufacturer's instructions are followed.
The essential features of the procedure are described in the Prepare 2 series of 4 or 5 dilutions in an appropriate human
following example. a-l-proteinase inhibitor concentration range, for both the
preparation to be examined and the reference standard, using
REAGENTS the tris-albumin buffer solution.
Dilution buffer pH 7.4. Isotonic non-chelating Transfer 50 f-lL of the reference solution dilutions into the
buffer prepared as follows: dissolve 6.08 g of wells of a microtitre plate and to each well, add 150 f-lL of a
tris(hydroxymethyl)aminomethane R and 8.77 g of porcine pancreatic elastase solution diluted to an appropriate

266 See the information section on general monographs (caver pages)


EUROPEAN PHARMACOPOEIA 8.0 2.7.32. Assay ofhuman a-l-proteinase inhibitor

concentration with the tris-albumin buffer solution. Incubate 0.45 mg/mL. Immediately start measurement of the change in
for a defined period of time, 3 -1 O min, at room temperature. absorbance (2.2.25) at 405 nm using a microtitre plate reader,
Since the activity of the solutions of the different porcine continuing the measurement for at least 5 mino Calculate the
pancreatic elastases may vary, the concentration of elastase rate of change of absorbance (~A/min). Alternatively, an
can be adjusted by evaluation of blank values containing end-point assay may be used by stopping the reaction with
elastase but no human a-l-proteinase inhibitor, to exhibit a acetic acid and measuring the absorbance at 405 nm. lf the
suitable change of absorbance at 405 nm under the actual assay is performed in test tubes using spectrophotometers for
assay conditions. monitoring the change in absorbance at 405 nm, the volumes
Add to each well 100 I1L of a solution of chromogenic of reagent solutions are changed proportionally.
substrate N-succinyl- tri -L-alanyl 4-p-nitroanilide The fate of change of absorbance (~A/min) is inversely
(Suc-Ala-Ala-Ala-pNA), reconstituted in dimethyl sulfoxide R proportional to human a -1-proteinase inhibitor activity.
to give a solution containing 4.5 mg/mL, then further diluted Check the validity of the assay and calculate the potency of the
with the tris-albumin buffer solution to a concentration of test preparation by the usual statistical methods (5.3).

General Notices (1) apply to all monographs and other texts 267
EUROPEAN PHARMACOPOEIA 8.0

268 See the information section on general monographs (cover pages)


EUROPEAN PHARMACOPOEIA 8.0

ln
A

2.8. Methods in pharmacognosy............................................. 271 2.8.11. of L8-cincole in essential oils .......................... 272
2.8.1. Ash insoluble in hydrochloric add .............................. 271 2.8.12. Essential oils in herbal .................................... 273
2.8.2. Foreign matter.. ....................................................... " ..... 271 2.8.13. Pesticide residues ......................................................... 274
2.8.3. Stomata and stomatal index ...................... " .................. 271 2.8.14. Tannins in herbal ............................... " ........... 275
2.8.4. Swelling index ................................................................. 271 2.8.15. Bitterness value ............................................................. 276
2.8.5. Water in essential oils .................................................... 271 2.8.16. residue of extracts ................................................ 276
2.8.6. Foreign esters in essential oils ...................................... 271 2.8.17. Loss on drying of extracts ........................................... 276
2.8.7. Fatty oils and resinified essential oils in essential 2.8.18. Determination of aflatoxin Bl in herbal drugs ......... 276
oils ............................................................................................ 271 2.8.20. Herbal and sample preparation .... 278
2.8.8. Odour and taste of essential oils .................................. 272 2.8.21. Test Íor aristolochic in herbal drugs ................ 279
2.8.9. Residue on evaporation of essential oils ..................... 272 2.8.22. Determinatíon of ochratoxin A in herbal drugs ...... 281
2.8.10. Solubility in alcohol of essential oils .......................... 272 2.8.23. examination oÍherbal drugs., .............. 282

General Notices (1) apply lo all and olher texts 269


EUROPEAN PHARMACOPOEIA 8.0

270 See the information section on general monographs (cover pages)


EUROPEAN PHARMACOPOEIA 8.0 2.8.7. Fatty oHs and resinified essential oHs in essential oils

2.8. METHODS IN For each sample ofleaf, make not fewer than 10 determinatiol1s
and calculate the mean.
PHARMACOGNOSY
Ol/2008:20801

2.8. L ASH INSOLUBLE IN


HYDROCHLORIC ACID 2
Ash insoluble in hydrochloric acid is the residue obtained
after extracting the sulfated or total ash with hydrochloric
acid, calculated with reference to 100 g of drug.
To the crucible containing the residue from the determination
of sulfated or total ash, add 15 mL of water R and 10 mL of
hydrochloric acid R, cover with a watch-glass, boil the mixture
gently for 10 min and allow to cool. Filter through an ashless
filter, wash the residue with hot water R until the filtrate is 3 4
neutral, dry, ignite to dull redness, allow to cool in a desiccator
and weigh. Reheat until the difference between 2 consecutive
weighings is not more than 1 mg.

0112008:20802

2.8.2. FOREIGN MATTER Figure 2.8.3.-1


Herbal drugs should be free from moulds, insects and other
0112008:20804
animal contamination.
Foreign matter is material consisting of any or all of the
following:
2.8.4. SWELLING INDEX
1) Foreign organs: matter coming from the source plant but The swelling index is the volume in millilitres occupied by
not defined as the drug, 1 gram of a drug, including any adhering mucilage, after it has
2) Foreign elements: matter not coming from the source plant swollen in an aqueous liquid for 4 h.
and either of vegetable or mineral origino In a 25 mL ground-glass stoppered cylinder graduated over
a height of 125 ± 5 mm in 0.5 mL divisions, place 1.0 g of
DETERMINATION OF FOREIGN MATTER the drug, whole or of the degree of comminution prescribed
Weigh 100 g to 500 g of the substance to be examined, or the in the monograph. Unless otherwise prescribed, moisten the
minimum quantity prescribed in the monograph, and spread drug with 1.0 mL of alcohol R, add 25 rnL of water R and close
it out in a thin ¡ayer. Examine for foreign matter by inspection the cylinder. Shake vigorously every 10 mill for 1 h. AUow to
with the unaided eye or by use of a lens (6 x). Separate foreign stand for 3 h. At 90 min after the beginning of the test, release
matter and weigh it and calculate the percentage present. any large volumes of liquid retained in the layer of the drug
and any particles of the drug floating at the surface of the
0112008:20803 ¡¡quid by rotating the cylinder about a vertical axis. Measure
the volume occupied by the drug, including any adhering
2.8.3. STO MATA AND STOMATAL mucilage. Carry out 3 tests at the same time.
INDEX The swelling index is given by the mean of the 3 tests.

STOMATA 0112008:20805
There are several types of stomata (see Figure 2.8.3.-1),
distinguished by the form and arrangement of the surrounding 2.8.5. WATER IN ESSENTIAL OILS
cells:
(1) The anomocytic (irregular-celled) type: the stoma is Mix 10 drops of the essential oil with 1 mL of carbon
surrounded by a varying number of cells in no way differing disulfide R. The solution remains clear on standing.
from those of the epidermis generally,
(2) The anisocytic (unequal-celled) type: the stoma is usually 0112008:20806
surrounded by 3 subsidiary ceUs, of which one is markedly
smaller than the others, 2.8.6. FOREIGN ESTERS IN ESSENTIAL
(3) The diacytic (cross-celled) type: the stoma is accompanied OILS
by 2 subsidiary ceUs, whose common wall is at right angles
to the guard cells, Heat 1 mL of the essential oil for 2 min on a water-bath with
3.0 mL of a freshly prepared 100 giL solution of potassium
(4) The paracytic (parallel-celled) type: the stoma has on each
hydroxide R in alcohol R. No crystals are formen within
side one or more subsidiary ceUs parallel to the long axis of
30 min, even after cooling.
the pore and guard cells.
STOMATAL INDEX 0112008:20807
100 x S
Stomatal Index = ~~S­
E+ 2.8.7. FATTY OILS AND RESINIFIED
ESSENTIAL OILS IN ESSENTIAL OILS
S the number of stomata in a given area of leaf,
Allow 1 drop of the essential oil to fall onto filter paper. The
E the number of epidermal cells (including drop evaporates completely within 24 h without leaving any
trichomes) in the same area ofleaf. translucent or greasy spot.

General Notices (1) apply to al! monographs and other texts 271
2.8.8. Odour and taste of essential oils EUROPEAN PHARMACOPOEIA 8.0

0112008:20808 a burette of at least 20 mL capacity, add the alcohol of the


strength prescribed in the monograph by increments of
0.1 mL until solution is complete and then continue adding by
2.8.8. ODOUR AND TASTE OF increments of 0.5 mL to a total of 20 mL, shaking frequently
ESSENTIAL OILS and vigorously. Record the volume of alcohol added when a
clear solution has been obtained and, if the solution becomes
Mix 3 drops of the essential oil with 5 mL of alcohol (90 per doudy or opalescent before 20 mL of alcohol has been added,
cent V/V) R and stir in 10 g of powdered sucrose R. The odour record the volume added when the cloudiness or opalescence
and taste are similar to that of the plant or parts of the plant appears and, where applicable, the volume added when the
from which the essential oil has been obtained. cloudiness or opalescence disappears.

rf a clear solution has not been obtained when 20 mL of


alcohol of the prescribed strength has been added, repeat the
test using the next highest concentration of alcohol.
0112008:20809 An essential oi! is said to be "soluble in n volumes and more
of alcohol of given strength t" when the clear solution in n
volumes remains clear when compared with the undiluted oil
2.8,9. RESIDUE ON EVAPORATION OF after further addition of alcohol of the same strength up to a
ESSENTIAL OILS total of 20 volumes of alcohol.

The residue on evaporation of an essential oil is the percentage An essential oi! is said to be "soluble in n volumes of alcohol
by mass of the oil which remains after evaporation on a of given strength t, becoming doudy when diluted" when the
water-bath under the conditions specified below. dear solution in n volumes becomes cloudy in nI volumes
(nI less than 20) and stays so after further gradual addition
Apparatus. The apparatus (see Figure 2.8.9.-1) consists oE: of alcohol of the same strength up to a total of 20 volumes
- water-bath with a cover having holes of 70 mm d¡ameter; of alcohol.
- evaporating dish of heat -resistant glass which is inert to An essential oi! is said to be "soluble in n volumes of alcohol of
the contents; given strength t with cloudiness between nI and n2 volumes"
- desiccator. when the clear solution in n volumes becomes cloudy in
nI volumes (nI less than 20) and stays so after further gradual
addition of alcohol of the same strength up to a total of
n2 volumes of alcohol and then becomes clear (n 2 1ess than 20).

An essential oil is said to be "soluble with opalescence" when


the alcoholic solution shows a bluish tinge, similar to that of
a standard of opalescence freshly prepared as follows: mix
0.5 mL of silver nitrare solution R2 and 0.05 mL of nitric
acid R; add 50 mL of a 12 mg/L solution of sodium chloride R;
mix and allow tostand protected fram light for 5 mino
o
l{)

70

0112008:20811
Figure 2.8.9.-1
Dimensions in millimetres
2.8.11. ASSAY OF 1,8-CINEOLE IN
Method. Weigh the evaporating dish after having heated it on
the water-bath for 1 h and cooled it in the desiccator. Weigh
ESSENTIAL OILS
into the evaporating dish 5.00 g of the essential oil, unless
otherwise prescribed. Heat the oil on the vigorously boiling Weigh 3.00 g of the oil, recently dried with anhydrous sodium
water-bath in a draught-free atmosphere for the prescribed sulfate R, into a dry test-tube and add 2.10 g of melted cresol R.
time. Allow to cool in the desiccator and weigh. Place the tube in the apparatus for the determination of
During the test, the level of water in the bath is maintained freezing point (2.2.18) and allow to coo1, stirring continuously.
When crystallisation takes place there is a small rise in
about 50 mm beneath the level of the cover.
temperature. Note the highest temperature reached (tJ

Remelt the mixture on a water-bath at a temperature that


do es not exceed tI by more than 5 oC and place the tube in
the apparatus, maintained at a temperature 5 oC below tI'
01/2008:20810 When crystallisation takes place, or when the temperature of
the mixture has fallen 3 oC below tI' stir continuously. Note
the highest temperature at which the mixture crystallises (t 2 ).
2.8.10. SOLUBILITY IN ALCOHOL OF Repeat the operation until 2 highest values obtained for t 2
ESSENTIAL OILS do not differ by more than 0.2 oc. lf supercooling occurs,
induce crystallisation by adding a small crystal of the complex
Place 1.0 mL of the essential oil in a 25 mL or 30 mL consisting of 3.00 g of cineole R and 2.lO g of melted cresol R. rf
glass-stoppered cylinder. Place in a constant temperature t2 is below 27.4 oC, repeat the determination aÍter the addition
device, maintained at a temperature of 20 ± 0.2 oc. Using of 5.10 g of the complexo

272 See the information section on general monographs (cover pages)


EUROPEAN PHARMACOPOEIA 8.0 2.8.12. Essential oils in herbal drugs

The content of cineole corresponding to the highest


temperature observed (t2) is given in Table 2.8.11.-1. If
5.10 g of the complex has been added, calculate the cine ole
content per cent m/m from the expression:

2 (A - 50) D

where A is the value found in Table 2.8.11.-1.


The content of cine ole, corresponding to the highest
temperature observed (t2)' is obtained, where necessary, by
interpolation.

Table 2.8.11.-1
t2 cineole t2 cineole t, cineole t, cine ole
oc per oc per oc per oc per e
cent m/m cent m/m cent m/m cent m/m
24 45.5 32 56.0 40 67.0 48 82.0

25 47.0 33 57.0 41 68.5 49 84.0

26 48.5 34 58.5 42 70.0 50 86.0

27 49.5 35 60.0 43 72.5 51 88.5

28 50.5 36 61.0 44 74.0 52 9l.0

29 52.0 37 62.5 45 76.0 53 93.5

30 53.5 38 63.5 46 78.0 54 96.0

31 54.5 39 65.0 47 80.0 55 99.0

01/2008:20812
corrected 6.0
Figure 2.8.12.-1. - Apparatus for the determination of essential
oils in herbal drugs
2,8,12, ESSENTIAL OILS IN HERBAL Dimensions in millimetres
Method. Use a thoroughly cleaned apparatus. Carry out the
DRUGS assay according to the nature of the drug to be examined.
Place the prescribed volume of distillation liquid in the flask,
The determination of essential oils in herbal drugs is add a few pieces of porous porcelain and attach the condenser
carried out by steam distillation in a special apparatus in the assemblyo Introduce water R through the filling funDel N until
conditions described below. The distillate is collected in the it is at the leve! B. Remove the stopper K/ and introduce the
graduated tube, using xylene to take up the essential oil; the prescribed quantity of xylene R, using a pipette with its tip at
aqueous phase is automatically returned to the distillation the bottom of the tube K. Replace the stopper K / and ensure
flask. that the orifice coincides with the vent. Heat the liquid in the
Apparatus. The apparatus comprises the following parts: flask to boiling and adjust the distillation rate to 2-3 mL/min,
unless otherwise prescribed.
(a) a suitable round-bottomed flask with a short, ground-glass
To determine the rate of distillation, during distillation
neck having an internal diameter of about 29 mm at the wide
lower the leve! of the water by means of the three-way tap
end;
until the meniscus is at the leve! ofthe !ower mark (a) (see
(b) a condenser assembly (see Figure 2.8.12.-1) that closely fits Figure 2.8.12.-2). Close the tap and measure the time taken
the flask, the different parts being fused into one pie ce ; the for the liquid to reach the upper mark (b)o Open the tap and
glass used has a low coefficient of expansion: continue the distillation, modifying the heat to regulate the
distillation rateo Distil for 30 mino Stop the heating and after
- the stopper K/is vented and the tube K has an orifice of at least 10 min read off the volume of xylene in the graduated
diameter about 1 mm that coincides with the vent; the wide tube.
end of the tube K is of ground-glass and has an internal
diameter of 10 mm;
- a pear-shaped swelling, J, of 3 mL capacity;
the tube JL is graduated in 0.01 mL;
the bulb-shaped swelling L has a capacity of about 2 mL;
M is a three-way tap;
- the junction B is at a level20 mm higher than the uppermost
graduation;
Figure 2.8.12.-2
(c) a suitable heating device, allowing a fine control;
Introduce into the flask the prescribed quantity of the drug
(d) a vertical support with a horizontal ring covered with and continue the distillation as described aboye for the time
insulating material. and at the rate prescribed. Stop the heating and after 10 min

General No/ices (1) apply to all monographs and o/her texts 273
2.8.13. Pesticide residues EUROPEAN PHARMACOPOEIA 8.0

read the volume of liquid collected in the graduated tube and MRL HD maximum residue limit of the pesticide in the
subtract the volume of xylene previously noted. The difference herbal drug as given in Table 2.8.13.-1 or in
represents the quantity of essential oil in the mass of the drug EU texts or calculated using the expression
taken. Calculate the result as millilitres per kilogram of drug. mentioned above;
When the essential oil is to be used for other analytical DER drug/extract ratio, i.e. the ratio between the
purposes, the water-free mixture of xylene and essential oil quantity of herbal drug used in the manufacture
may be recovered as follows: remove the stopper K / and of a herbal drug preparation and the quantity of
introduce 0.1 mL of a 1 giL solution of sodium fluoresceinate R herbal drug preparation obtained;
and 0.5 mL of water R. Lower the mixture of xylene and MDD HP = daily dose of the herbal drug preparation, in
essential oil into the bulb-shaped swelling L by means of the kilograms.
three-way tap, allow to stand fOI 5 min and lower the mixture
slowly until it just reaches the level of the tap M. Open the tap The competent authority may grant total or partial exemption
anti-clockwise so that the water flows out of the connecting from the test when the complete history (nature and
tube BM. Wash the tube with acetone R and with a httle quantity of the pesticides used, date of eaeh treatment during
toluene R introduced through the filling funnel N. Turn the cultivation and after the harvest) of the treatment of the batch
tap anti-clockwise in order to recover the mixture of xylene is known and can be checked precisely according to good
and essential oil in an appropriate flask. agricultural and collection practice (GACP).
Table 2.8.13.-1
Substance Limit
(mg/kg)
Acephate 0.1
07/2008:20813 Alachlor 0.05

Aldrin and dieldrin (sum of) 0.05

2.8.13. PESTICIDE RESIDUES Azinphos-ethyl 0.1

Azinphos-methyl
Definition. Por the purposes of the Pharmacopoeia, a
pesticide is any substance or mixture of substances intended Bromide, inorganic (calculated as bromide ion) 50
for preventing, destroying or controlling any pest, unwanted Bromophos-ethyl 0.05
species of plants or animals causing harm during or otherwise
interfering with the production, processing, storage, transport Bromophos-methyl 0.05
or marketing of herbal drugs. The item includes substances Brompropylate 3
intended for use as growth-regulators, defoliants or desiccants
and any substance applied to crops, either before or after Chlordane (sum of cis-, trans - and oxychlordane) 0.05
harvest, to protect the commodity from deterioration during Chlorfenvinphos 0.5
storage and transport. Pesticide residues can be present and
are controlled in herbal drugs and herbal drug preparations. Chlorpyriphos-ethyl 0.2

Limits. Unless otherwise indicated in the monograph, the Chlorpyriphos-methyl 0.1


herbal drug to be examined at least complies with the limits 0.01
Chlorthal-dimethyl
indicated in Table 2.8.13.-1. The limits applying to pesticides
that are not listed in Table 2.8.13.-1 and whose presenee Cyfluthrin (sum of) 0.1
is suspected for any reason comply with the limits (levels)
i\-Cyhalothrin
eross referred to by Regulation (EC) No. 396/2005, including
annexes and suceessive updates. Limits for pesticides that are Cypermethrin and isomers (sum of)
not listed in Table 2.8.13.-1 l10r in European Union texts are
DDT (sum of o,p'-DDE, p,p'-DDE, o,p'-DDT, p,p'-DDT,
calculated using the following expression: o,p'-TDE and p,p'-TDE)
ADlxM Deltamethrin 0.5
MDD HD X 100 Diazinon 0.5

Dichlofluanid 0.1
ADI acceptable daily intake, as published by
FAO-WHO, in milligrams per kilogram of Dichlorvos
body mass; Dieofol 0.5
M body mass in kilograms (60 kg) ;
Dimethoate and omethoate (sum of) 0.1
lvJDD HD = daily dos e of the herbal drug, in kilograms.
Dithiocarbamates (expressed as CS2) 2

The limits for pesticides in herbal drug preparations are Endosulfan (sum of isomers and endosulfan sulfate)
calculated using the following expressions: 0.05
Endrin

Ethion 2

IfDER::; JO: !VIRLHO xDER Etrimphos 0.05

Fenchlorophos (sum of fenchlorophos and 0.1


fenchlorophos-oxon)
Fenitrothion 0.5

ADI x !VI Fenpropathrin 0.03


IfDER> JO:
!VIDD HP X 100 0.05
Fensulfothion (sum of fensulfothion, fenslllfothion-oxon,
fensulfothion-oxonsulfon and fensulfothion-sulfon)

274 See the information section on general monographs (cava pages)


EUROPEAN PHARMACOPOEIA 8.0 208.14. Tannins in herbal drngs

Sllbstance Limit - natural occurrence of sorne constituents is considered in the


(mg/kg) interpretation of results (e.g. disulfide from Cruciferaceae);
Fenthion (sum of fe11thio11, fenthion-oxon, 0.05 - tile concentration of test and reference solutions and the
fenthion ~oxon -sulfol1, fenthion -oxon -sulfoxid,
setting of the are such that the responses used
fenthion-sulfon and fenthion-Slllfoxid)
for of the residues are within the
Fenvalerate 1.5
dynamic of the detector; test solutions containing
Flucytrinate 0.05 at a level outside the dynamic range,
may be diluted within the calibration range, provided
T-Fluvalinate 0.05
that the concentration of the matrix in the solution is
Fonophos 0.05 adjusted in the case vvhere the calibration solutions must
be matrix-matched;
Heptachlor (sum ofheptachlor, cis-heptachlorepoxide and 0.05
tralls-heptachlorepoxide) - between 70 per cent to 110 per cent oI' eaeh pesticide is
Hexachlorbenzene 0.1
recovered;
- repeatability of the method: RSD is not greater than the
Hexachlorocyciohexane (sum 01' isomers a-, 0-, 0- and E) 0.3
values indicated in Table 2.8.13.-2;
Lindan ("y-hexachlorocyclohexane) 0.6 - reproducibility of the method: RSD is not greater than the
values indicated in Table 2.8.13.-2.
Malathion and malaoxon (sum of)

Mecarbam 0.05
Table 2.8.13.-2
Concentration range Repeatability (RSD) Reproducibility (RSD)
Methacriphos 0.05 (lf the pesticide (per ccut) (per cent)
Methamidophos 0.05 (mg/kg)
0.001 - 0.01 30 60
Methidalhion 0.2
> 0.01 - 0.1 20 40
Methoxychlor 0.05
> 0.1 - 1 15 30
lVIirex 0.01
> 1 10 20
Monocrotophos 0.1

Parathion-ethyl and Paraoxon-ethyl (sum of) 0.5

Parathion-methyl amI Paraoxon-methyl (surn of) 0.2 ¡H/2008:20814


Pendinlethalin 0.1
14. TANNINS IN HERBAL DRUGS
Pentachloranisol 0.01
out all the extraction alld dilution opemtions
Permethrin and isol11crs (5U111 00

Phosalone 0.1

Phosmet 0.05 or the extract in


a 250 mL flask add 150 mL of water R. Heat
Piperonyl butoxide on a water-bath for 30 mino Cool under
Pirimiphos-ethyl 0.05 to a 250 mL volumetric Rinse
and colleet the in the
Pirimiphos-methy! (sum 01' pirimiphos-methyl and 4 volumetric flask, then dilute to 250.0 mL with water R. Allow
N-desethyl-pirimiphos-methyl) the solids to settle and filter the liquid through a filter paper
Procymidone 0.1 125 mm in diameter. Discard the first 50 mL of the filtrate.
Profenophos 0.1 In the case of a liquid extract or a tincture, dHute the stated
amount of the liq~id extraet or tineture to 250.0 mL with
Prothiophos 0.05
water JI.. Filter the mixture through a filter paper 125 mm in
Pyrethrum (sum of cinerin 1, cinerin 11, jasmolin l, jasmolin diameter. Discard the first 50 mL of the filtrate.
Il, pyrethrin 1 and pyrethrin Il) Total polyphenols. Dilute 5.0 mL of the filtrate to 25.0 mL
Quinalphos 0.05 with water R. Mix 2.0 mL of this solution with 1.0 mL of
Quintozene (sum 01' quintozene, pentachloraniline amI
reagent R and 10.0 mL of water R
melhyl penthachlorpheny! sulfide) with a 290 giL solutiol1 of sodium
S-421 0.02 carbonate R. After 30 mil1 measure the absorbance (2.2.25) at
760 nm (Al)' using water Ras the compensation liquido
Tecnazene 0.05
Polyphenols not adsorbed by hide . To 10.0 mL of the
Tetradifon 0.3 filtrate, add 0.10 g of hide and shake
for 60 mino Filter and dilute 5.0 mL of the filtrate to
Vinclozolin 0.4
with water R. Mix 2.0 mL of this solution with 1.0 mL of
phosphomolybdotungstic R and 10.0 mL of water R
Sampling of herbal drugs. Sampling is done
and dilute to 25.0 mL with a giL solution of sodium
general chapter 2.8.20. Herbal drugs: sampling and
carbonate R. After 30 min measure the absorbance at
preparation.
760 n111 (Al)' using water Ras the liquido
Qualitative and quantitative analysis of pesticide residnes. Standard. Dissolve before use 50.0 mg oí
The analytical procedures used are validated (e.g. according pyrogallol R in water JI. and dilute to 100.0 mL with the
to Document N° SANCO/l023212006). In particular, they same solvent. Dilute 5.0 mL of the solution to 100.0 mL
satisfy the following criteria: with water R. Mix 2.0 mL of this solution with l.0 mL of
- the chosen method, especially the purification steps, is phosphomolybdotungstic reagent R and 10.0 mL of water JI.
suitable for the combinatiol1 pesticide residue/substance and dilute to 25.0 mL with a 290 solution of sodium
to be examined, and not susceptible to interferel1ce from carbonate R. After 30 min measure the absorbance (2.2.25) at
co-extractives; 76011111 (A}), water R as the liquido

General Notices (1) apply to all and other texts 275


2.8.15. Bittemess value EUROPEAN PHARMACOPOElA 8.0

Calculate the percentage content of tannins expressed as Starting with dilutíon C-4 each panel member determines
pyrogallol from the expression: the dilution which still has a bitter laste. This solution is
designated D. Note the DF of solution D is Y.
62.5 (Al ~ A 2 ) m2
Starting with solutiol1 D prepare the following sequence of
A3 x ml
dilutions:
m¡ mass of the sample to be examined, in grams; Solution D (111L) 1.2 l.5 2.0 3.0 6.0 8.0

m2 mass of pyrogallol, in grams. water R (111L) 8.8 8.5 8.0 7.0 4.0 2.0

Determine the number of millilitres of solution D which, when


01/2008:20815 diluted to 10.0 mL with water R, still has a bitter taste (X).
Calculate the bitterness value fo1' each panel member from
2.8.15. BITTERNESS VALUE the expression:

The bitterness value is the reciprocal of the dilution of a y x k \


(
compound, a liquid or an extnlct that still has a bitter taste. It X x 0.1)
is determined by comparison with quinine hydrochloride, the
Calculate the bitterness value of the sample to be examined as
bitterness value of which is set at 200 000.
the average value for all panel members.
Determination of the correction factor
A taste panel comprising at least 6 persons is recommended.
The mouth must be rinsed with water R before tasting. 0112008:20816
To correct for individual differences in tasting bitterness
amongst the panel members it is necessary to determine a 2,8.16. DRY RESIDUE OF EXTRACTS
carrection factor for each panel member. In a flat-bottomed dish about 50 mm in diameter and about
Stock solution. Dissolve 0.100 g of quinine hydrochloride R in 30 mm in height, introduce rapidly 2.00 g or 2.0 mL of the
water R and dilute to 100.0 mL with the same solvent. Dilute extract to be examined. Evaporate to dryness on a water-bath
l.0 mL of this solution to 100.0 mL with water R. and dry in an oven at 100-105 oC for 3 h. Allow to cool in a
Reference solutions. Prepare a series of dilutions by placing in desiccator over diphosphorus pentoxide R or anhydrous silica
a first tube 3.6 mL of the stock solution and increasing the gel R and weigh. Calculate the result as a mass percentage or
volume by 0.2 mL in each subsequent tube to a total of 5.8 mL; in grams per litre.
dilute the contents of each tube to 10.0 mL with water R.
Determine as follows the dilution with the lowest 01/2008:20817
concentration that still has a bitter taste. Take 10.0 mL of the
weakest solution into the mouth and pass it from side to side
over the back of the tongue for 30 s. If the solution is not 2.8.17. LOSS ON DRYING OF
found to be bitter, spit it out and wait for 1 mino Rinse the EXTRACTS
mouth with water R. After 10 min, use the next dilution in
arder of increasing concentration. In a flat-bottomed dish about 50 mm in diameter and about
30 mm in height, weigh rapidly 0.50 g of the extract to be
Calculate the correction factor k for each panel member from examined, finely powdered. Dry in an oven at 100-105 oC
the expression: far 3 h. Allow to cool in a desiccator over diphosphorus
n pentoxide R or anhydrous silica gel R and weigh. Calculate the
k=-
5.00 result as a mass percentage.

n number of millilitres of the stock solution in the


dilution of lowest concentration that is judged to 01/2008:20818
be bitter.
Persons who are unable to taste any bitterness when using the
2.8.18. DETERlVHNATION OF
reference solution prepared from 5.8 mL of stock solution AFLATOXIN Bl IN HERBAL DRUGS
have to be excluded from the panel.
CAUTION: aflatoxins are very toxic and carcinogenic. Perform
Sample preparation manipulations in a fume cupboard whenever possible. Talce
If necessary, reduce the sample to a powder (710) (2.9.12). particular precautions, such as use of a glove box, when
To 1.0 g of sample add 100 mL ofboiling water R. Heat on a toxins are in dry form because of their electrostatic properties
water-bath for 30 min, stirring continuously. AlIow to cool and the tendency to disperse through the working are as.
and dilute to 100 mL with water R. Shake vigorously and filter, Decontamination procedures for laboratory wastes of aflatoxins
discarding the first 2 mL of the filtrate. The filtrate is labelled were deve/oped by the International Agency for Research on
C-l and has a dilution factor (DF) of 100. Caneer (IARC).
If liquids have to be examined, 1 mL of the liquid is diluted Aflatoxins are naturally occurring mycotoxins produced
with a suitable solvent to 100 mL and designated C-l. mainly by Aspergillus flavus and Aspergillus parasiticus.
Determination of the bitterness value These fungi are common and widespread in nature and
Test solutions: are most often found when certain grains are grown under
conditions of stress such as drought. The mouId occurs in soil,
10.0 mL of C-l is diluted with water R to (DF = 1000) decaying vegetation, hay, and grains undergoing microbial
100 mL: C-2 spoilage, and invades al! types of organic substrates whenever
10.0 mL of C-2 is diluted with water R to (DF = 10000) and wherever the conditions are favourable for its growth.
100 mL: C-3 Favourable conditions in dude high moisture content and
high temperature. At least 13 different types of aflatoxin are
20.0 mL of C-3 is diluted with water R to (DF = 50 000) produced in nature and most of these are known to be highly
100 mL: C-3A toxic and carcinogenic. Aflatoxin B¡ is considered the mast
10.0 mL of C-3 is diluted with water R to (DF=100000) toxiCo Herbal drugs that are subject to contamination by
100 mL: C-4 aflatoxins are tested by a validated method.

276 See the information section on general monographs pages)


EUROPEAN PHARMACOPOElA 8.0 2.8.18. Determination of a:ílatoxin BI in herbal drugs

Unless otherwise indicated in the monograph, herbal drugs tightly in aluminium foil and stofe it below 4 oc. Befare use,
contain not more than 2 flg/kg of aflatoxin BI. The competent do not remove the aluminium foil until the contents have
authority may also require compliance with a limit of 4 ~lg/kg reached room temperature. lf the solution has to be stored
for the sum of aflatoxins Bl' B2, G¡ and G 2 • for a long period (for example, 1 month), weigh the flask and
The mechod described below is cited as an example of a record the mass before and after each use of the solution.
method that has been shown to be suitable for devil's claw root, Aflatoxin B 1 standard solutions. Place the volumes of aflatoxin
ginger and senna pods. Its suitability for other herbal drugs secondary stock solution indicated in Table 2.8.18.-1 in
must be demonstrated or another validated method used. separate 250 mL volumetric flasks. Pass a stream of nitrogen
through at room temperature until the solvent has just
METHOD evaporated. To each flask, add 75 mL of methanol R, allow the
Liquid chromatography (2.2.29). aflatoxin Bl to dissolve and dilute to 250 mL with water R.
Aflatoxins are subject to light degradation. Carry out the Table 2.8.18.-1. - Aflatoxin Bl standard solutions
determination protected from daylight by using UV-absorbing
foil on windows in combination with subdued light, or curtains Final concentration
Volume of secoudary
Standard solution of standard solution
or blinds in combination with artificiallight (jluorescent tubes stock solution (rlL)
(ng/rnL)
are acceptable). Protect aflatoxin-containing solutions from
125 0.05
daylight.
Rinse glassware before use with a 10 per cent V/V solution of 2 250 0.1
sulfuric acid R and then rinse carefully with distilled water R 3 500 0.2
until no more acid is present.
4 750 0.3
Test solution. Use an immunoaffinity column containing
antibodies against aflatoxin B¡ with a capacity of not less than 5 1000 0.4
100 ng of aflatoxin B¡ and which gives a recovery of not less
than 80 per cent when a solution of 5 ng of aflatoxin B¡ in a Calibration curve. Prepare the calibration curve using
mixture of 12.5 mL of methanol R and 87.5 mL of water R is aflatoxin B¡ standard solutions 1 to 5, which cover a range
passed through. Allow the immunoaffinity column to reach equivalent to 1-8 flg/kg of aflatoxin B¡ in the herbal drug.
room temperature. To 5.00 g of the powdered drug (500) Check the plot for linearity. lf the content of aflatoxin B¡ in
(2.9.12) add 100 mL of a mixture of 30 volumes of water R and the sample to be examined is outsicle of the calibration range,
70 volumes of methanol R and extract by sonication for 30 mino the test solution must be diluted to an aflatoxin content that is
Filter through folded filter papero Pipette 10.0 mL of the clear appropriate for the established calibration curve.
filtrate into a 150 mL conical flask. Add 70 mL of water R. Column:
Pass 40 mL through the immunoaffinity column at a flow rate - size: 1= 0.25 m, 0 = 4.6 mm;
of 3 mLlmin (not exceeding 5 mLlmin). Wash the column
- stationary phase: octadecylsilyl si/ica gel for
with 2 volumes, each of 10 mL, of water R at a flow rate not
chromatography R (5 flm).
exceeding 5 mLlmin and dry by applying a slight vacuum for
5-10 s or by passing air through the immul10affinity column Mobile phase:
by means of a syringe for 10 s. Apply 0.5 mL of methanol R - mobile phase A (for post-column derivatisation with
to the column and allow to pass through by gravity. Collect photochemical reactor or pyridinium bromide):
the eluate in a 5 mL volumetric flask. After 1 min, apply a acetonitrile R, methanol R, water R (2:3:6 V/V/V);
2 nd portion of 0.5 mL of methanol R. After a further 1 min, - mobile phase B (for post-column derivatisation with
apply a 3,d portion of 0.5 mL of methanol R. Collect most of electrochemically derived bromine): add 0.12 g of
the applied elution solvent by pressing air through or applying potassium bromide R and 350 of dilute nitric acid Rl
vacuum to the column. Dilute to 5 mL with water R and shake per litre of mobile phase A.
well. rf the solution is clear it can be used directly for analysis.
Otherwise, pass it through a disposable filter unít prior to Flow rate: 1 mL/min.
injection. Use a disposable filter unit (e.g. 0.45 I.un pore size Detection: f1uorescence detector with a 360 nm excitation
polytetrafluoroethylene filter) that has been shown not to filter and a 420 nm cut-off emission filter, or equivalent.
cause loss of aflatoxin by retention. Recommended settings for adjustable detectors are 365 11m
Aflatoxin B1 primary stock solution. Dissolve aflatoxin B1 R in
(excitation wavelength) and 435 nm (emission wavelength).
a mixture of 2 volumes of acetonitrile R and 98 volumes of Injection: 500 1lL.
toluene R to give a 10 flg/mL solution. To determine the exact Post-column derivatisation with pyridinium hydrobromide
concentration of aflatoxin B¡ in the stock solution, record perbromide (PBPB):
the absorption curve (2.2.25) between 330 nm and 370 nm - pulseless pump;
in quartz cells.
- T-piece with zero dead volume;
Calculate the aflatoxin B ¡ mass concentration, in micrograms
per millilitre, using the following expression: - polytetrafluoroethylene reaction tube, 1= 0.45 m,
0= 0.5 mm;
A x NI x 100
- mobile phase A;
ex1
- post -column derivatisation reagent: dissolve 50 mg of
pyridinium hydrobromide perbromide R in 1000 mL of
A absorbance determined at the maximum of the water R (store protected from light and use within 4 clays);
absorption curve;
- flow rate ofthe derivatisation reagent: 0.4 mL/min.
M molar mass of aflatoxin Bl (312 g/mol);
Post-column derivatisation with photochemical reactor
E molar absorptivity of aflatoxin B¡ in the (PHRED)
toluene-acetonitrile mixture (1930 m 2/mol); - reactor unit with one 254 nm low pressure mercury UV
optical path length of the cell (l cm). bulb (minimum 8 W) ;
Aflatoxin B 1 secondary stock solution. Prepare a secondary - polished support plate;
stock solution containing 100 ng/mL aflatoxin B¡ by diluting - knitted reactor coil: polytetrafluoroethylene tubing knitted
aflatoxin B¡ primary stock solution with a mixture of 2 volumes tightly around the UV bulb, 1= 25 m, 0 = 0.25 mm,
of acetonitrile R and 98 volumes of toluene R. Wrap the flask nominal void volume 1.25 mL;

General Notices (1) apply to all monographs and other texts 277
2.8.20. Herbal drugs: sampling alld sample pl'eparatioll EUROPEAN PHARMACOPOEIA 8.0

- exposure time: 2 min; Take one sample from eaeh container to be sampled. The
- mobile phase A. sample is taken from the upper, middle or lower section of the
container, su eh that the samples taken are representative of
Post-column derivatisation with electrochemically generated
different parts of the containers. In the case oflarge bales or
bromine (KOBRA):
bags, samples must be taken from a depth of at least 10 cm.
- KOBRA-cell: electrochemical ceH that generates a reactive The mas s of the material taken from each container is such
form ofbromine for derivatisation of aflatoxins, resulting in that the total mass of the bulk sample complies with the
enhanced f1uorescence; available from various commercial following values.
suppliers;
- Derivation direct-current suppIy in series with the Minimum mass of samples as
Mass of herbal drug in the
KOBRA-cell, providing a constant current of about 100 flA; a percentage of the mas s of the
batch (kg)
batch of herbal drug
polytetrafluoroethylene reaction tube, 1 = 0.12 m,
< 50 1.00*
0= 0.25 mm;
- mobile phase B. 50 - 100 0.50

Elution order: aflatoxin G 2, aflatoxin Gl' aflatoxin B2, aflatoxin > 100 - 250 0.25
Bl'
> 250 - 500 0.20
Calculation: calculate the calibration curve y = ax + b, with
aflatoxin Bl concentl'ation (ng/mL) on the x-axis and the > 500 - 1000 0.18
signal (S) on the y-axis. The aflatoxin Bl concentration (C) in
> 1000 - 2500 0.15
a measured solution is equal to S;;b.
Calculate the aflatoxin Bj content of the herbal drug, in > 2500 - 5000 0.10
nanograms pel' gram, using the following expression: > 5000 - 10 000 0.08
V1 xV2xC > 10 000 - 25 000 0.05
mxV;
NOTE: if the mas s of the batch is greater than 25 000 kg, it is divided
into sub-batches, and the procedure is applied to each sub-batch as
m mass of the herbal drug taken for analysis, in though it were a homogeneous batch.
grams; * subject to a minimum total mas s of 125 g for the bulk sample; ifthis
voIume of the solvent used for extraction, in minimum requirement represents more than 10.0 per cent of the mass
of herbal drug in the batch, the whole batch may be used as the sample.
millilitres;
aliquot taken fol' immunoaffinity clean-up, in Prepare the bulk sample by combining and thoroughly
millilitres; mixing the samples taken Írom each of the randomly selected
final volume of solutiol1 after elution from the containers (see Table 2.8.20.-1).
immunoaffinity column and dilution, in millilitres;
C measured aflatoxin Bl concentration of the test TEST SAMPLE
solution, in nanograms per millilitre. Unless otherwise prescribed in the monograph, prepare the
The presence of aflatoxin Bl may be confirmed by recording test sample as follows.
the chromatogram without post-column derivatisation, which Reduce the size of the bulk sample by quartering (see Note
leads to a large decrease (greater than lO-fold) in the response below) or by any other method that produces a homogeneous
due to aflatoxin Bl' sample, making sure that each retained portion remains
representative of the whole, until the minimum retained
0112008:20820 quantity complies with the following conditions.

Type oí herbal drug Minimum weight of test sample


2.8.20. HERBAL DRUGS: SAMPLING
Roots, rhizomes. bark, herbs 500 g or mas s of whole sample if
AND SAMPLE PREPARATION bulk sample is less than 500 g

In order to reduce the effect of sampling in qualitative Leaves, flowers, seeds, fruits 250 g or mass of whole sample if
bulk sample is less than 250 g
and quantitative analysis, it is necessary to ensure that the
composition of the sample used is representative of the batch Broken or fragmented drugs (where 125 g
average mass of the pie ces is less
of material being examined. The following procedures are than 0.5 g)
the minimum considered applicable for herbal drugs. NOTE:
other procedures may be used if they can be demonstrated to NOTE: quartering consists of placing the bulk sample,
produce representative batch samples. thoroughly mixed, as a level and square-shaped heap and
dividing it diagonally into 4 equal parts. 2 opposite quarters
BULK SAMPLE are retained and carefully remixed. The process is repeated as
Where externa! examination of containers, markings and necessary until the required minimum mass is obtained for the
Iabels of a batch indicate that it can be considered to be test sample.
homogeneous, sample the number of randomly selected
Mili the test sample in a single pass through a 1 mm screen
containers indicated below. Where a batch cannot be
01' the screen size specified in the monograph. The use of a
considered to be homogeneous, divide it into sub-batches that
milling machine is recommended.
are as homogeneous as possible, then sample each sub-batch
as a homogeneous batch using, as a minimum, the number of Pass the milled sample through a 1 mm standard sieve or the
randomly selected containers indicated below. sieve specified in the monograph. The residue retained on
the sieve must not be more than 10 per cent of the total mass
Number of containers
Number of containers in batch (N) of the milled sample, of which not more than 2 per cent oí
to be sampled (n)
the total mass of the milled sample may be of a particle size
1- 3 all greater than 1.5 mm or 1.5 times the specified particle size
> 3 n* = VN + 1 in the monograph. If these eonditions are met, the sample
and residue are to be well mixed to form the test sample for
* round 11 up to the next integer analysis.

278 See the information section on general monographs (cover pages)


EUROPEAN PHARMACOPOEIA 8.0 2.8.21. Test for aristolochic acids in herbal drngs

TabIe 2.8.20.-1. - Operation of the sampling procedure in order to obtain the prescribed bulk sample
Mass of herba!
drug in container 0.5 1 5
(kg)
Total mass of No.of No.of Total mass No.of No. oí Total mass No.of No.of Total mass
herbal drug in containers containers to ofsamples containers containers to of samples containers containers to oí samples
the batch (kg) in batch be sampled (g) in batch be sampled (g) in batch be sampled (g)
0.5 1 1 125 - - - - - -

1 2 2 125 1 1 125 - - -

5 10 5 125 5 4 125 1 1 125

10 20 6 125 10 5 125 2 2 125

25 - - - 25 6 250 5 4 250
100 - - - 100 11 500 20 6 500
250 - - - - - - 50 9 625

500 - - - - - -
100 11 1000

Mass of herbal
drug in container 25 125 500
(kg)
Total mass of No.of No.of Total mass No.of No.of Total mass No.of No.of Total mas s
herbal drug in containers containers to of samples containers containers to of samples containers containers to ofsamples
the batch (kg) in batch be sampled (g) in batch be sampled (g) in batch be sampled (g)
25 1 1 250 - - - - - -

100 4 3 500 - - - - - -

250 10 5 625 2 2 625 - - -

500 20 6 1000 4 3 1000 1 1 1000

1000 40 8 1800 8 4 1800 2 2 1800

2000 80 10 3000 16 5 3000 4 3 3000

3000 120 12 3000 24 6 3000 6 4 3000


5000 200 16 5000 40 8 5000 10 5 5000

10000 400 21 8000 80 10 8000 20 6 8000


25000 800 30 12500 160 14 12500 40 8 12500

In those cases where these requirements are not met, the These methods are not designed for inclusion as assay methods
test sample for analysis is composed of the 2 parts measured in monographs on those drugs that produce aristolochic acids
separateIy. Therefore, the quantity required for each analysis as secondary metabolites; for these, a more sensitive, validated
is derived by weighing proportional quantities of the powder method is required.
and the residue.
NOTE: for determination of microscopic characters, a portion METHOD A: SCREENING TEST FOR ARISTOLOCHIC
of the milled test sample is re-milled through a 0.355 mm screen. ACIDS
Thin-Iayer chromatography (2.2.27).
0112011:20821
Solvent mixture: anhydrous formic acid R, water R, methanol R
(1:9:40 V/V/V).
2.8.21. TEST POR ARISTOLOCHIC
Test solution. To 1.0 g of the powdered herbal drug (710)
ACIDS IN HERBAL DRUGS (2.9.12) add 10.0 mL ofthe solvent mixture, sonicate for
CAUTION: aristolochic acids are very toxic and carcinogenic. 10 min and centrifuge.
Perform manipulations in a fume cupboard whenever possible. Reference solution (a). Disperse an amount of aristolochia HRS
Take particular precautions, such as use of a glove box, when corresponding to 0.10 mg of aristolochic acid I in 20.0 mL of
the substance is in dry form beca use of its electrostatic properties the solvent mixture, sonicate for 10 min and centrifuge.
and the tendency to disperse through the working areas.
Reference solution (b). Dilute 1.0 mL of reference solution (a)
Methods A and B are intended to be cross-reÍerenced to 25.0 mL with methanol R.
in monographs on herbal drugs that, according to
chemotaxonomic knowledge, are expected to be free from Plate: TLC silica gel F254 plate R (2-10 flm).
aristolochic acids, but that may be subject to adulteration or Mobile phase: anhydrous formic acid R, water R, ethyl
substitution with plant material containing aristolochic acids. acetate R, toluene R (3:3:30:60 V/V/V/V); use the upper layer.
Methods A and B are intended to be used in the screening of Application: 20 flL as bands of 8 mm.
herbaI drugs for aristolochic acids at the stated limits and will
usually be complemented by macroscopic and/or microscopic Development: over a path of 6 cm.
tests to exclude plant material containing aristolochic acids. Drying: in a current of cold air for 5 mino
Method C will not be used in specific monographs but is Detection: spray with a 100 giL solution of stannous chloride R
provided as a method to confirm the presence of aristolochic in dilute hydrochloric acid R untiI the pI ate is slightly wet, heat
acid I at Ievels equal to or greater than 2 ppm. It may be at 100 oC for 1 min and examine in ultraviolet light at 365 nm.
applied if chromatographic data suggests the presence of
aristolochic acid 1.

General Notices (1) apply to all monographs and other texts 279
2.8.21. Test for aristolochic adds in herbal drugs EUROPEAN PHARMACOPOEIA 8.0

System suitability: METHOD C: CONFIRMATORY TEST FOR


- the chromatogram obtained with reference solution (a) ARISTOLOCHIC ACm 1
shows 2 greenish-blue zones due to aristolochic acids I Liquid chromatography (2.2.29) coupled with mass
and II between RF = 0.35 and RF = 0.55, which may not be spectrometry (2.2.43).
completely separated; Solvent mixture: acetonitrile R, water R (50:50 V/V).
- the chromatogram obtained with reference solution (b) Test solution. Weigh 2.0 g of the powdered herbal drug (710)
shows at least 1 of these zones (corresponding to 2 ppm (2.9.12) into a 250 mL, brown, screw-cap bottle and add
of aristolochic acid 1). 100.0 mL of the solvent mixture. Sonicate for 30 min and filter
Results: in the chromatogram obtained with the test solution through a membrane filter (nominal pore size 0.45 flm).
no zone is similar in position and fluorescence to any of the Reference so/ution (a). Dissolve the contents of a vial of
zones due to aristolochic acids in the chromatogram obtained aristoloehie acid 1 CRS in the solvent mixture to obtain a
with reference solution (a). concentration of 0.04 ¡.tg/mL of aristolochic acid 1.
If the chromatogram obtained with the test solution shows Reference solution (b). Prepare a solution according to the
any zones similar in position and fluorescence to any of the instructions supplied with aristolochic acid 1 CRS to obtain a
zones due to aristolochic acids 1 and II in the chromatogram concentration of 0.45 flg of aristolochic acid 1 in 10.0 mL of
obtained with reference solution (a), apply method B. the test solution.
Column:
METHOD B: LIMIT TEST FOR ARISTOLOCHIC ACm 1
- size: 1 = 0.15 m, 0 = 2.1 mm;
Liquid chromatography (2.2.29). - stationary phase: octadecylsilyl si/ica gel for
Solvent mixture: acetonitrile R, water R (50:50 V/V). chromatography R (3.5 flm);
Test solution. Weigh 2.0 g of the powdered herbal drug (710) - temperature: 40 oc.
(2.9.12) into a 250 mL, brown, screw-cap bottle and add Mobile phase:
100.0 mL ofthe solvent mixture. Stir for 30 min at about
- mobi/e phase A: anhydrous formic acid R, 1 giL solution of
300 r/min and filter through a membrane filter (nominal pore
ammonium acetate R in water R (0.1:99.9 V/V);
size 0.45 ¡.tm).
- mobile phase B: anhydrous formic acid R, 1 giL solution of
Reference solution (a). Dissolve the contents of a vial of ammonium acetate R in methanol R (0.1:99.9 V/V);
aristolochic acid 1 CRS in the solvent mixture to obtain a
concentration of 0.04 ¡.tg/mL of aristolochic acid 1. Time Mobile phase A Mobile phase B
(min) (per cent V/V) (per cent V/V)
Reference solution (b). Dissolve the contents of a vial of
O - 15 70 ~ O 30 ~ 100
aristolochic acid for system suitability CRS (containing
aristolochic acids 1 and II) in the solvent mixture and dilute to 15 - 16 O 100
20.0 mL with the solvent mixture.
16 - 17 O ~ 70 100 ~ 30
Column:
- size: 1 = 0.15 m, 0 = 2.1
mm; Flow rate: 0.4 mL/min.
- stationary phase: octadecylsilyl silica gel for Injection: 20 ¡.tL; inject reference solution (a) twice, the test
chromatography R (3.5 ~un); solution twice, reference solution (a) twice, then reference
solution (b) twice.
- temperature: 40 oc.
Detection: mass detector as described below under A or B.
Mobile phase: Adjust the flow rate, the temperature and the detector settings
- mobile phase A: trifluoroacetic acid R, water R so as to comply with the system suitability criterion.
(0.1:99.9 V/V); A. Ion-trap mass spectrometer equipped with an electro spray
- mobile phase B: trifluoroacetic acid R, acetonitri/e R ionisation (ESI) interface and MSll analyser.
(0.1:99.9 V/V); Set the mass spectrometer parameters for the MS 3 mode
Time Mobile phase A Mobile phase B
as follows:
(min) (pe,. cent V/V) (per cent V/V) Paren! Isolation width Relative collision
0-25 85 ~ 35 15 ~ 65 Mode (miz) (miz) energy (per cenl)

25 - 30 35 ~ O 65 ~ 100 359
MS 2 2.0 30
[M + NH,l+
30 - 31 O ~ 85 100 ~ 15
MS 3 298 2.0 35
Flow rate: 0.3 mL/min.
Deteetion: spectrophotometer at 390 nm. - ful! sean of product ions: from miz 80 to miz 370;
Injection: 25 ¡.tL. - product ions to be monitored: miz 252, miz 268 and
miz 281.
System suitability:
System suitabi/ity:
- resolution: minimum 3.0 between the peaks due to
- signal-to-noise ratio: minimum 100 for the monitored
aristolochic acids 1 and JI in the chromatogram obtained
product ions in the chromatogram obtained with
with reference solution (b);
reference solution (a);
- signal-to-noise ratio: minimum 10 for the peak due to matrix interference test: the average of the 2 ratios of
aristolochic acid 1 in the chromatogram obtained with reference solution (b) is inside the ± 40 per cent interval
reference solution (a). of the average of the 2 ratio s of reference solution (a);
Limit: otherwise it is necessary to adjust the detector settings.
- the sample complíes with the test if the chromatogram Results: evaluate the average ratios (252/268 and 2811268)
obtained with the test solution shows no peak with the of the relative intensity of the 3 product ion s of aristolochic
same retention time as the peak due to aristolochic acid 1 acid 1 in the test solution; evaluate the average of the
in the chromatogram obtained with reference solution (a) 2 ratios of the signals at the retention time of aristolochic
(2 ppm). acid 1 in reference solution (a); if the average of the 2 ratios

280 See the information sec/ion on general monographs (ca ver pages)
EUROPEAN PHARMACOPOEIA 8.0 2.8.22. Determinatlon of ochratoxin A in herbal drugs

of the test solution is within the ± 40 per cent interval of the thoroughly 5.0 mL (Vi) of the elear supernatant with 30 mL
average of the 2 ratios of reference solution (a), aristolochic solution 7.4 R and pass the whole solution through
acid 1 is present in the test solution. column at a ílow rate oí 3 mLlmin (do not
B. Triple-quadrupole mass spectrometer equipped with an the columl1 first with 10 mL bufler
ESI interÜ1Ce and Msn analyser. solution pH 7.4 R then with 2 each of 10 mL, of
water R ~t a flow rate not 5 mUmin and dry by
Set the mass spectrometer parameters for the MS 2 mode applying a slight vacuum for 5-10 s or by passing air through
as follows: the immunoaffinity coJumE means oí a for 10 S.
precursor ion: miz 359 [M + 0.5 mL of rnethanol R to the column and to pass
- product ions to be monitored: miz 265, miz 281 and by
miz 296. Collect the eluate in a 4 mL vial. AÍter 30 s, apply a 2 nd
System suitability: quantity of 0.5 mL of methmlOl R and allow to pass through
the column into the same glass vial. After a further
- signal-to-noise ratio: minimum 100 for the monitored 30 s, repeat portion of 0.5 mL of methanol R. Collect
product ions in the chromatogram obtained with any solvent retained 011 the column by pressing air through
reference solution (a); or applying vacuum to the column. Evaporate the combined
- matrix inteljerence test: the average of the 2 ratios of eluates completely to using a thermal block with a
reference solution (b) is inside the ± 40 per cent interval nitrogen b!anket (40 Dissolve the residue in 0.5 mL (V2 )
of the average of the 2 ratios of reference solution (a); oí solution A. rE the solution is elear it can be used directly Íor
otherwise it is necessary to adjust the detector settings. Otherwise, pass it a clisposable fiIter unit
Results: evaluate the average ratios (265/281 and 2961281) prior to injection. Use a filter unít (e.g. 0.45 flm
of the relative intensity of the 3 product ions oí aristolochic filter) that has been shown
acid 1 in the test solution; evaluate the average of the A by retention.
2 ratio s of the signals at the retention time oí aristolochic Ochratoxin A stock solution. Dilute 1.0 mL of
acid 1 in reÍerence solution (a); ifthe average oÍtbe 2 ratios to 100.0 mL with methanol R and
of the test soJution is within the ± 40 per cent interval of the
average of the 2 ratios oí reÍerence solution (a), aristolochic Ochratoxin A stock solution. Dilute 10.0 mL of
acid 1 is present in the test solution. ochratoxin A stock solution to 100.0 mL with
methanol R shake thoroughly.
Ochratoxin A standard solutions. Place the volumes of
stock solution or ochratoxin A
01/2010:20822
indicated in Table 2.8.22.-1 inta
and make up to 50.0 111L with solution A.
2,8.22, DETERMINATION OF Table 2.8.22.-1. - Ochratoxin A standard solutio175
OCHRATOXIN IN HERBAL DRUGS
Standard solution Volmne off Fin.a! concentratlon
ocurato"i" A prlmary of ochratoxin A in
CAUTION: ochratoxin A is and stock soiutiol1 ("L) standard sohüion
nephrocarcinogenic. Perform manipulation5 in a (ng/mi)
cupboard. Take particular precaution5, such as use g/ove sooo 50
box, when toxins are in dry form beca use of their electrostatic
properties and the tendency to through the 2 2500 25
areas. Decontamination l(lOO lO
containing ochratoxin A are necessary (see
500 s
Herbal drugs that are subject to contamination by ochratoxin A
are tested by a validated method. 5 250 2.5

The method described below is cited as an example of a Standard solution Vo!ume of Final concentratlon
method that has been shown to be suitable Íor liquorice oelIrato"i" A of ocnratoxin A in
extraet and Iiquorice root. Its suitability for otber herbal drugs secondary stock standard solution
solution (flL) (ng/mL)
must be demonstrated or another validated method used.
6 500 0.5
METHOD 7 100 0.1
Liquid chromatography (2.2.29).
Calibration curve. the calibration curve using
Use brown glassware that is free Í1"Om detergent residues. If
ochratoxin A solutions 1 to 7, which cover a range
necessary rinse glassware before use with a 10 per cent V/V
to 0.5- 250 flg/kg oE ochratoxin A in the herbal drug.
solution of sulfuric acid R and then rinse carefully with
the for If the content of ochratoxin A in
distilled water R until no more acid is present.
the sample to be outside of tbe calibration range,
Solution A. Mix 80 volumes of water R, previously adjusted the test solution must be diluted to a11 oehratoxin A content
to pH 2.3 with anhydrous formic acid R, and 20 volumes of that is appropriate for the established calibration curve.
acetonitrile R.
Column:
Test solution. Use an immunoaffinity column
antibodies against ochratoxin A with a capacity of not les s - size: 1 = 0.15 m, 0 = 4.6 mm;
than 100 ng oí ochratoxin A and which gives a recovery of silica gel for
not less than 70 per cent. Allow the immunoaffinity column
to reach room temperature. - tempcrature: 45 oc.
To 2.00 g of the powdered drug (250) (2.9.12) add 80 mL Mobile
of a 30 giL solution of sodium hydrogen carbonate R and
extract by sonieation for 30 min (change water of ultrasonic - mobile A: water R to pH 2.3 with phosphoric
bath after 15 min). Coo! to room temperature and dilute acid R;
to 100.0 mL (V¡) with tIle same solution. Mix - mobile B: (lee/onUrilc R;

General Notices (1) and other texts 281


2.8.23. Microscopic examination of herbal dmgs EUROPEAN PHARMACOPOEIA 8.0

Time Mobile phase A Mobile phase B tissues, ruthenium red solution R, which is used to show the
(min) (per cent V/V) (per cent V/V) presence of mucilage in cells or glyeerol R used to show the
0-30 80 -¿ 40 20 -¿ 60 presence of starch and inulin.
30 - 35 40 -¿ 20 60 -¿ 80 Examination under polarised light (between crossed nicol
prisms) is used to identify starch granules (black cross
35 - 37 20 80 phenomenon), calcium oxalate crystals (refringence) or
37 - 40 20 -¿ 80 80 -¿ 20 lignified structures.
MOUNTING IN CHLORAL HYDRATE SOLUTION
Flow rate: 0.8 mUmin.
Place 2-3 drops of ehloral hydrate so/ution R on a glass
Deteetion: fluorescence detector; recommended settings fol'
microscope slide. Disperse a very small quantity of the
adjustable detector s are 330 nm (excitation wavelength) and
powdered drug in the liquid and cover the preparation with
460 nm (emission wavelength).
a cover slip. Heat the preparation very gently to boiling on a
Injection: 20 flL. hot pi ate or a micro gas bumer. Maintain gentle boiling for
ealeulation: calculate the calibration curve y = ax + b, with a short time. Make sure that the quantity of mounting fluid
ochratoxin A concentration (in nanograms per millilitre) on is sufficient. If necessary, add more fluid using a tapered
the x-axis and the signal (S) on the y-axis. The ochratoxin A glass pipette. Allow to cool and then examine under a
concentration (e) in a measured solution is equal to S~b. microscope. Repeat the heating until the starch granules and
Calculate the ochratoxin A content of the herbal drug, in the water-soluble contents of the ceUs are no longer visible.
nanograms per gram, using the following expression: Examine under a microscope.
Chloral hydrate tends to crystallise as long needles. To avoid
V1 XV2 XC this, proceed as follows: after heating, remove the cover slip;
mxVi to the preparation add 1 drop of a 10 per cent V/V mixture of
ehloral hydrate solution R in glyeerol R; place a clean cover slip
m mass of the herbal drug used to prepare the test on the preparation; examine under a microscope.
solution, in grams;
MOUNTING IN A 50 PER CENT V/V SOLUTION OF
V¡ volume of dilution, in millilitres; GLYCEROL
aliquot taken for immunoaffinity clean-up, in Place 2 drops of a 50 per cent V/V solution of glyeerol R on a
millilitres; glass microscope slide. Disperse a very small quantity of the
volume in which the residue is taken up, in powdered drug in the liquid and cover the preparation with a
millilitres; cover slip. Examine under a microscope.
e measured ochratoxin A concentration of the test MOUNTING IN A 10 PER CENT V/V ALCOHOLIC
solution, in nanograms per millilitre. SOLUTION OF PHLOROGLUCINOL AND
HYDROCHLORIC ACm
Appendix: Decontamination procedures for Place a very smal! quantity of the powdered drug on a glass
microscope slide. Add 1-2 drops of a 10 per cent V/Valcoholic
laboratory glassware solution of phloroglucinol R. Mix and allow the solvent to
Rinse glassware with methano/ R and decontaminate by evaporate almost completely. Add 1-2 drops of hydrochloric
immersion in strong sodium hypochlorite so/ution R fol' at least acid R and cover the preparation with a cover slip. Examine
2 h, then wash thoroughly with water. immediately under a microscope. The red colour indicates
the presence of lignin.
MOUNTING IN LACTIC REAGENT
04/2010:20823 Place 2-3 drops of lactic reagent Ron a glass microscope slide.
Disperse a very small quantity of the powdered drug in the
liquid and cover the preparation with a cover slip. Heat the
2,8.23. MICROSCOPIC EXAMINATION preparation very gently to boiling. Maintain gentle boiling for
OF HERBAL DRUGS a short time. Make sure that the quantity of mounting fluid is
sufficient. If necessary, add more fluid using a tapered glass
The microscopic examination ofherbal drugs is carried out on pipette. Allow to cool and then examine under a microscope.
the powdered drug (355) (2.9.12) unless otherwise prescribed Lignified structures stain bright yellow; structures containing
in the monograph. cellulose remain colourless. Starch granules stain more or less
ehloral hydrate solution R is the most commonly prescribed violet; certain secretions (e.g., essential oils, resins, oleoresins)
reagent. However, certain features are not visible or 110t stail1 orange and cork stains red.
easily se en after mounting in this reagent. In this case, other
reagents are used, for example, a 50 per cent V/V solution of MOUNTING IN RUTHENIUM RED SOLUTION
glycerol R, which makes it possible to visualise starch granules. Place 2 drops of ruthenium red solution R on a glass
It may also be necessary to prescribe specific reagents in microscope slide. Disperse a very small quantity of the
a monograph, for example: laetic reagent R which is used powdered drug in the liquid and cover the preparation with
to show the presence of various features, 10 per cent V/V a cover slip. After about 1 minute, allow a drop of distilled
alcoholic solution of phloroglucinol R and hydrochlorie acid R, water R to be taken up between the slide and the cover slip.
which are used to identify the presence of lignin in cells or Examine under a microscope. The mucilage stains violet red.

282 See the information section on general


EUROPEAN PHARMACOPOEIA 8.0

Pharmaceutical technical
rocedures
2.9. Pharmaceutical technical procedures ............................. 285 2.9.26. Specific surface are a by gas adsorption ..................... 329
2.9.1. Disintegration of tablets and capsules ......................... 285 2.9.27. Uniformity of mass of delivered doses from multidose
2.9.2. Disintegration of suppositories and pessaries ............ 287 containers ................................................................................ 331
2.9.3. Dissolution test for solid dosage forms ....................... 288 2.9.29. Intrinsic dissolution ..................................................... 331
2.9.4. Dissolution test for transdermal patches .................... 295 2.9.31. Partiele size analysis by laser light diffraction .......... 333
2.9.5. Uniformity of mass of single-dose preparations ........ 297 2.9.32. Porosity and pore-size distribution of solids by
2.9.6. Uniformity of content of single-dose preparations .... 298 mercury porosimetry............................................................. 336
2.9.7. Friability of uncoated tablets ........................................ 298 2.9.33. Characterisation of crystalline and partially crystalline
2.9.8. Resistance to crushing of tablets .................................. 299 solids by X-ray powder diffraction (XRPD) ....................... 339
2.9.9. Measurement of consistency by penetrometry........... 299 2.9.34. Bulk density and tapped density of powders ............ 343
2.9.10. Ethanol content.. .......................................................... 301 2.9.35. Powder nneness ............................................................ 346
2.9.11. Test for methanol and 2-propanol.. ........................... 304 2.9.36. Powder flow................................................................... 346
2.9.12. Sieve test.. ...................................................................... 305 2.9.37. Optical microscopy...................................................... 349
2.9.14. Specific surface are a by air permeability................... 305 2.9.38. Particle-size distribution estimation by analytical
2.9.16. Flowability..................................................................... 307 sieving ...................................................................................... 351
2.9.17. Test for extractable volume ofparenteral 2.9.39. Water-solid interactions: determination of
preparations ............................................................................ 308 sorption-desorption isotherms and of water activity......... 353
2.9.18. Preparations for inhalation: aerodynamic assessment 2.9.40. Uniformity of dosage units ......................................... 357
of fine partieles ........................................................................ 309 2.9.4l. Friability of granules and spheroids .......................... 359
2.9.19. Particulate contamination: sub-visible partieles ...... 321 2.9.42. Dissolution test for lipophilic solid dosage forms ... 361
2.9.20. Particulate contamination: visible partieles ............. 323 2.9.43. Apparent dissolution ................................................... 361
2.9.22. Softening time determination of iipophilic 2.9.44. Preparations for nebulisation: characterisation ....... 363
suppositories ........................................................................... 323 2.9.45. Wettability of porous solids ineluding powders ....... 365
2.9.23. Gas pycnometric density of solids ............................. 324 2.9.47. Demonstration of uniformity of dosage units using
2.9.25. Dissolution test for medicated chewing gums ......... 325 large sample sizes .................................................................... 368

General Notices (1) apply to all monographs and other texts 283
EUROPEAN PHARMACOPOEIA 8.0

284 See the informatíon section on general monographs (cover pages)


EUROPEAN PHARMACOPOEIA 8.0 2.9.1. Disintegration of tablets and capsules

2.9. PHARMACEUTICAL holes extend between the ends of the cylinder. One of the
holes is centered on the cylindrical axis. The other holes
TECHNICAL PROCEDURES are centered 6 ± 0.2 mm from the axis on imaginary lines
perpendicular to the axis and parallel to each other. 4 identical
trapezoidal-shaped planes are cut into the v,all of the cylinder,
nearly perpendicular to the ends of the cylinder. The
04/2011:20901 trapezoidal shape is symmetrical; its parallel sides coincide
with the ends of the cylinder and are parallel to an imaginary
line connecting the centres of 2 adjacent holes 6 mm Írom
2.9.1. DISINTEGRATION OF TABLETS the cylindrical axis. The parallel side of the trapezoid on the
AND CAPSULES(l) bottom of the cylinder has a length of 1.6 ± 0.1 mm and its
bottom edges lie at a depth of 1.5 mm to 1.8 mm from the
This test is provided to determine whether tablets or capsules cylinder's circumference. The parallel side of the trapezoid
disintegrate within the prescribed time when placed in a liquid on the top of the cylinder has a length of 9.4 ± 0.2 mm and
medium under the experimental conditions presented below. its centre hes at a depth of 206 ± 0.1 mm from the cylinder's
For the purposes of this test, disintegration does 110t circumference. AH surfaces of the disc are smooth.
imply complete dissolution of the ul1it or even of its active rf the use of discs is specifled, add a disc to each tube and
constituent. Complete disintegration is deflned as that state in operate the apparatus as directed under Procedure. The discs
which any residue of the unit, except fragments of insoluble eonform to the dimensions shown in Figure 2.9.1.-1.
coating or capsule shell, remaining on the screen of the test
apparatus or adhering to the lower surface of the discs, if used, The use of automatic detection employing modifled discs
is a 50ft mass having no palpably flrm coreo is permitted where the use of dises is specifled or allowed.
Such discs must comply with the requirements of density and
·Use apparatus A for tablets and capsules that are not dimension given in this chapter.
greater than 18 mm long. For larger tablets or capsules use
apparatus B. + Procedure. Place 1 dosage unit in each of the 6 tubes of the
basket and, if prescribed, add a dise. Operate the apparatus
TEST A - TABLETS AND CAPSULES OF NORMAL SIZE using the specifled medium, maintained at 37 ± 2 oC, as
the immersion fluid. At the end of the specifled time, lift
Apparatus. The apparatus consists of a basket-rack assembly, the basket from the fluid and observe th-e dosage units:
a 1 L, low-form beaker, 149 ± 11 mm in height and having all of the dosage units have disintegrated completely. lf
an inside diameter of 106 ± 9 mm for the immersion fluid, a 1 or 2 dosage units fail to disintegrate, repeat the test on
thermostatic arrangement for heating the fluid between 35 oC 12 additional dosage units. The requirements of the test are
and 39 oC, and a device for raising and lowering the basket in met if not less than 16 of the 18 dosage units tested have
the immersion fluid at a constal1t frequency rate between 29 disintegrated.
and 32 cycles per minute, through a distance of 55 ± 2 mm.
The volume of the fluid in the vessel is such that at the highest
point of the upward stroke the wire mesh remains at least tTEST B - LARGE TABLETS AND LARGE CAPSULES
15 mm below the surface of the fluid, and descends to not less Apparatus. The main part of the apparatus (Figure 2.9.1.-2.)
than 25 mm from the bottom of the vessel on the downward is a figid basket-rack assembly supporting 3 cylindrical
stroke. At no time should the top ofthe basket-rack assembly transparent tubes 77.5 ± 2.5 mm long, 33.0 mm ± 0.5 mm in
become submerged. The time required for the upward stroke internal diameter, and with a wall thickness of 2.5 ± 0.5 mm.
is equal to the time required for the downward stroke, and the Each tube is provided with a cylindrical dise 31.4 ± 0.13 mm
change in stroke direction is a smooth transition, rather than in diameter and 15.3 ± 0.15 mm thick, made oftransparent
an abrupt reversal of motion. The basket-rack assembly moves plastic with a relative density of 1.18-1.20. Each d¡sc is pierced
vertically along its axis. There is no appreciable horizontal by 7 holes, each 3.15 ± 0.1 mm in diameter, 1 in the centre
motion or movement of the axis from the vertical. and the other 6 spaced equally on a circle of radius 4.2 mm
Basket-rack assembly. The basket-rack assembly consists of from the centre of the dise. The tubes are held vertically
6 open-ended transparent tubes, each 77.5 ± 2.5 mm long by 2 separate and superimposed rigid plastic plates 97 mm
and having an inside diameter of 21.85 ± 1.15 mm and a wall in diameter and 9 mm thick, with 3 holes. The holes are
1.9 ± 0.9 mm thick; the tubes are held in a vertical position equidistant from the centre of the pI ate and equally spaced.
by 2 plates, each 90 ± 2 mm in diameter and 6.75 ± 1.75 mm Attached to the under side of the lower plate is a piece of
in thickness, with 6 holes, each 24 ± 2 mm in diameter, woven gauze made from stainless steel wire 0.63 ± 0.03 mm
equidistant from the centre of the plate and equally spaced in diameter and having mesh apertures of 2.0 ± 0.2 mm.
from one another. Attached to the under surface of the lower The plates are held rigidly in position and 77.5 mm apart
plate is a woven stainless steel wire c1oth, which has a plain by vertical metal rods at the periphery. A metal rod is also
square weave with 2.0 ± 0.2 mm mesh apertures and with a flxed to the centre of the upper plate to enable the assembly
wire diameter of 0.615 ± 0.045 mm. The parts of the apparatus lo be attached to a meehanical device capable of raising and
are assembled and rigidly held by means of 3 bolts passing lowering it smoothly at a COl1stant frequency of between
through the 2 plates. A suitable means is provided to suspend 29 and 32 cycles per minute, through a distance of 55 ± 2 mm.
the basket-rack assembly from the raising and lowering device The assembIy is suspended in the specifled liquid medium
using a point on its axis. in a suitable vessel, preferably a 1 L beaker. The volume of
The design of the basket-rack assembly may be varied the liquid is such that when the assembly is in the highest
somewhat provided the specifications for the glass tubes and position the wire mesh is at least 15 mm below the surface of
the screen mesh size are maintained. The basket-rack assembly the liquid, and when the assembly is in the lowest position the
conforms to the dimensions shown in Figure 2.9.1.-1. wire mesh is at least 25 mm aboye the bottom of the beaker
and the upper open ends of the tubes remain aboye the surÍace
Discs. The use of discs is permitted only where specified
of the liquido A suitable device maintains the temperature of
or allowed. Each tube is provided with a cylindrical disc
the liquid at 35-39 oc.
9.5 ± 0.15 mm thick and 20.7 ± 0.15 mm in diameter.
The disc is made of a suitable, transparent plastic material The design of the basket-rack assembly may be varied provided
having a speciflc gravity of 1.18-1.20. 5 parallel 2 ± 0.1 mm the speciflcations for the tubes and wire mesh are maintained.

(1) Ihis chapter has llndergone pbarmacopoeial harmonisatiol1. See (hapter 5.8. Phannacopociallwrmonisatioll.

General Notices (1) apply to all monographs and other texts 285
2.9.1. Disintegration oftablets and capsules EUROPEAN PHARMACOPOEIA 8.0

Method. Test 6 tablets or capsules either by using 2 basket-rack the specified liquido Operate the apparatus for the prescribed
assemblies in parallel or by repeating the procedure. In each period, withdraw the assembly and examine the state of the
of the 3 tubes, place 1 tablet or capsule and, if prescribed, tablets or capsules. To pass the test, al! 6 of the tablets or
add a disc; suspend the assembly in the beaker containing capsules must have disintegrated. +

Basket-rack
assembly

1.9 ± 0.9 Disc

~
6'
2.6 + 0.1 " ,x ()~

l!)
1'-:
" Top
vlew
l!)
N
'"
+1
l!)
9.4 ± 0.2
r--:
t-

iN gj \\ _~:I
Side
view
1/
+1
l!)
(J)

ci
N
+1
'<:j'
N
j Boltom
view
I

Figure 2.9.1.-1. - Disintegration apparatus A

Dimensions in millimetres

286 See the information section on general monographs (ca ver pages)
EUROPEAN PHARMACOPOEIA 8.0 2.9.2. Disintegration of suppositories and pessaries

l!)
o
+1
l!)
3.15 ± 0.1
Nt¡==~~
l!)
o
+1
o
c0
(")

l!)

o
+1
(")
l!)

Figure 2.9.1.-2. - Disintegration apparatus B


Dimensions in millimetres
0112008:20902 d) rupture of the gelatin sheH of rectal or vaginal capsules
occurs allowing release of the contents,
2.9.2. DISINTEGRATION OF e) no residue remains on the perforated dise or if a residue
SUPPOSITORIES AND PESSARIES remains, it eonsists only of a soft or frothy mass having no
solid core offering resistance to pressure of a glass rod (vaginal
The disintegration test determines whether the suppositories tablets).
or pessaries soften or disintegrate within the prescribed
Apparatus. The apparatus (Figure 2.9.2.-1) consists of a
time when placed in a liquid medium in the experimental
sleeve of glass or suitable transparent plastic, of appropriate
conditions described below.
thickness, to the interior of which is attached by means of three
Disintegration is considered to be achieved when: hooks a metal device consisting of two perforated stainless
a) dissolution is complete, metal discs each eontaining 39 holes 4 mm in diameter; the
b) the components of the suppository or pessary have diameter of the discs is similar to that of the interior of the
separated: melted fatty substances colleet on the surface of sleeve; the discs are about 30 mm apart. The test is carried out
the liquid, insoluble powders fal! to the bottom and soluble using three such apparatuses each containing a single sample.
components dissolve, depending on the type of preparation, Each apparatus is placed in a beaker with a capacity of at least
the components may be distributed in one or more of these 4 L filled with water maintained at 36-37 oC, unless otherwise
ways, prescribed. The apparatuses may also be placed together in a
c) there is softening of the sample that may be accompanied vessel with a capacity of at least 12 L. The beaker is fitted with a
by appreciable change of shape without complete separation slow stirrer and a device that will hold the cylinders vertically
of the components, the softening is such that the suppository not less than 90 mm below the surface of the water and allow
or pessary no longer has a solid core offering resistance to them to be inverted without emerging from the water.
pressure of a glass rod,

General Notices (1) apply to all monographs and other texts 287
2.9.3. Dissolution test for solid dosage forms EUROPEAN PHARMACOPOEIA 8.0

Method. Use three suppositories or pessaries. Place each one


on the lower disc of a device, place the latter in the sleeve
and secure. Invert the apparatuses every 10 mino Examine lA
the samples after the period prescribed in the monograph. To
pass the test aH the samples must have disintegrated.

o lE
ID
I

A. glass plate D. water

B. vaginal tablet E. dish, beaker

C. water surface

Figure 2.9.2.-2.

0112012:20903

2.9.3. DISSOLUTION TEST POR SOLID


DOSAGE FORMS
This test is provided to determine compliance with the
dissolution requirements for solid dosage forms administered
orally. In this chapter, a dosage unit is deflned as 1 tablet or
1 capsule or the amount specifled.

APPARATUS
Apparatus 1 (Basket apparatus). The assembly consists
of the following; a vessel, which may be covered, made of
glass or other inert, transparent material (2) ; a motor; a drive
shaft; and a cylindrical basket (stirring element). The vessel is
partially immersed in a suitable water-bath of any convenient
size or heated by a suitable device su eh as a heating jacket.
The water-bath or heating device permits maintaining the
temperature inside the vessel at 37 ± 0.5 oC during the test
Figure 2.9.2.-1. - Apparatus for disintegration of suppositories and keeping the dissolution medium in constant, smooth
and pessaries motion. No part of the assembly, including the environment in
which the assembly is placed, contributes signiflcant motion,
Dimensions in millimetres agitation, or vibration beyond that due to the smoothly
rotating stirring elemento Apparatus that permits observation
of the preparation and stirring element during the test is
preferable. The vessel is cylindrical, with a hemispherical
METHOD OF OPERATION FOR VAGINAL TABLETS
bottom and a capacity of 1 L. Its height is 160-210 mm and its
Use the apparatus described aboye, arranged so as to rest on inside d¡ameter is 98-106 mm. Its sides are flanged at the topo
A fltted cover may be used to retard evaporation(3). The shaft
the hooks (see Figure 2.9.2.-2). Place it in a beaker of suitable
diameter containing water maintained at 36-37 oC with the is positioned so that its axis is not more than 2 mm at any
level just below the upper perforated dise. Using a pipette, point from the vertical axis of the vessel and rota tes smoothly
adjust the level with water at 36-37 oC until a uniform film and without signiflcant wobble that could affect the results. A
covers the perforations of the dise. Use three vaginal tablets. speed-regulating device is used that allows the shaft rotation
Place each one on the upper plate of an apparatus and cover speed to be selected and maintained at a specifled rate, within
the latter with a glass plate to maintain appropriate conditions ± 4 per cent.
of humidity. Examine the state of the samples after the period Shaft and basket components of the stirring element are
prescribed in the monograph. To pass the test al! the samples fabricated of stainless steel, type 316 or equivalent, to the
must have disintegrated. speciflcations shown in Figure 2.9.3.-1.

(2) The materials must not 5orb, reaet, or interfere with the preparatian to be tested.
(3) If a caver is used, it provides ~ufficient openings to allow ready insertion of the lhermometer and withdrawal of samples.

288 See the information section on general monographs (cover pages)


EUROPEAN PHARMACOPOEIA 8.0 2.9.3. Dissolution test for solid dosage forms

A basket having a gold coating of about 2.5 flm (0.0001 inch) the test. The metallic or suitably inert, rigid blade and shaft
thick may be used. The dosage unit is p!aced in a dry basket comprise a single entity. A suitable two-part de tachable design
at the beginning of each test. The distan ce between the may be used provided the assembly remains firmly engaged
inside bottom of the vessel and the bottom of the basket is during the test. The paddle blade and shaft may be coated with
maintained at 25 ± 2 mm during the test. a suitable coating so as to make them inert. The dosage unít
is allowed to sink to the bottom of the vessel before rotation
of the blade is started. A small, loose piece of non-reactive
material, such as not more than a few turns of wire helix, may
be attached to dosage units that would otherwise floal. An
alternative sinker device is shown in Figure 2.9.3.-3. Other
validated sinker devices may be used.
Apparatus 3 (Redprocating eyUnder). The assembly consists
of a set of cylindrical, flat -bottomed glass vessels; a set of
6.3 lo 6.5 glass reciprocating cylinders; inert fittings (stainless steel
or 9.4 lo 10.1 type 316 or other suitable material) and screens that are made
of suitable nonsorbing and nonreactive material, and that
are designed to tit the tops and bottoms of the reciprocating
l!)
cylinders; a motor and drive assembly to reciprocate the
ci
Vent hole +1 cylinders vertically inside the vessels, and if desired, index
2.0 ± 0.5 the reciprocating cylinders horizontally to a different row
oí vessels. The vessels are partially immersed in a suitable
water-bath of any convenient size that permits holding the
Retenlion spring temperature at 37 ± 0.5 oC during the test. No part of the
with 3 tangs assembly, induding the environment in which the assembly is
on 120' centers
I• •I placed, contributes significant motion, agitation, or vibration
Clear openíng beyond that due to the smooth, vertically reciprocating
20.2 ± 1.0 cylinder. A device is used that allows the reciprocation
rate to be selected and maintained at the specified dip rate,
within ± 5 per cent. An apparatus that permits observation of
the preparations and reciprocating cylinders is preferable. The
Screen 0.0
vessels are provided with an evaporation cap that remains in
22.2 ± 1.0
. place for the duration of the test. The components conform
to the dimensions shown in Figure 2.9.3.-4 unless otherwise
+1 specified.
o
r--.
C'\I
Apparatu.s 4 (Flow-through ceH). The assembly consists
of a reservoir and a pump for the dissolution medium; a
flow-through ceH; a water-bath that maintains the dissolution
medium at 37 ± 0.5 oc. Use the specified cel! size.
A The pump force s the dissolution medium upwards through
the flow-through cell. The pump has a delivery range
between 240 mLlh and 960 mLlh, with standard flow rates
of 4 mLlmin, 8 mLlmin, and 16 mLlmin. It must deliver a
constant flow (± 5 per cent of the nominal t10w rate); the Ílow
profile is sinusoidal with a pulsation of 120 ± 10 pulses/mino
o A pump without pulsation may also be used. Dissolution test
C'"i procedures using the flow-through ceH must be characterised
+1
o with respect to rate and any pulsation.
l!)
C'\I
The flow-through ceH (see Figures 2.9.3.-5 and 2.9.3.-6) of
transparent and inert material is mounted vertically, with a
filter system that prevents escape of undissolved partides
from the top of the cell; standard cel! diameters are 12 mm
and 22.6 mm; the bottom cone is usually filled with small
1) Screen with welded seam: 0.22-0.31 mm wire
diameter with wire opening of 0.36-0.44 mm. glass beads of about 1 mm diameter, with 1 bead of about
After welding the screen may be slighty altered. 5 mm positioned at the apex to protect the fluid entry tube; a
2) Maximum allowable mnoul al "/( is 1.0 mm tablet holder (see Figures 2.9.3.-5 and 2.9.3.-6) is available for
when the part is rotated on eenler line axis with positioning of specia! dosage forms. The cel! is immersed in a
basket mounted. water-bath, and the temperature is maintained at 37 ± 0.5 oc.
Figure 2.9.3.-1. - Apparatus 1, Basket stirring element The apparatus uses a clamp mechanism and 2 O-rings for the
Dimensions in millimetres fixation of the ceH assembly. The pump is separated from
the dissolution unit in order to shield the latter against any
Apparatus 2 (Paddle apparatus). Use the assembly from
vibrations originating from the pump. The position of the
Apparatus 1, except that a paddle formed from a blade and a
pump must not be on a level higher than the reservoir flasks.
shaft is used as the stirring element. The shaft is positioned so
Tube connections are as short as possible. Use suitably inert
that its axis is not more than 2 mm from the vertical axis of the
tubing, such as polytetrafluoroethylene, with a 1.6 mm inner
vessel, at any point, and rotates smoothly without significant
diameter and inert flanged-end connections.
wobble that could affect the results. The vertical center line
of the blade passes through the axis of the shaft so that the Apparatus suitability. The determination of suitability of
bottom of the blade is flush with the bottom of the shaft. The the apparatus to perform dissolution testing must indude
paddle conforms to the specifications shown in Figure 2.9.3.-2. conformance to the dimensions and toleran ces of the
The distan ce of 25 ± 2 mm between the bottom of the blade apparatus as given aboye. In addition, critica! test parameters
and the inside bottom of the vessel is maintained during that have to be monitored periodically during use indude

General Natices (1) apply ta all monographs and other texts 289
2.9.3. Dissolution test for solid dosage forms EUROPEAN PHARMACOPOEIA 8.0

9.41010.1

ce
LD
('") LD
o
+1
o
ai

42.0

o
+1
74.5 ± 0.5
o
-of

A and B dimensions do not vary more than 0.5 mm when part is rotated on center Une axis. Tolerances
are ± l.0 mm unless otherwise stated.
Figure 2.9.3.-2. - Apparatus 2, Paddle stirring element
Dimensions in millimetres
volume and temperature of the dissolution medium, rotation PROCEDURE
speed (Apparatus 1 and 2), dip rate (Apparatus 3), and flow APPARATUS 1 AND 2
rate of medium (Apparatus 4).
Conventional-release solid dosage forms
Determine the acceptable performance of the dissolution test
assembly periodically. Procedure. Place the stated volume of the dissolution medium
(± 1 per cent) in the vessel of the specified apparatus.
Assemble the apparatus, equilibrate the dissolution medium
to 37 ± 0.5 oC, and remove the thermometer. The test may
also be carried out with the thermometer in place, provided
it is shown that results equivalent to those obtained without
the thermometer are obtained.

290 See the information section on general monographs (cover pages)


EUROPEAN PHARMACOPOEIA 8.0 2.93. Dissolution test for solid dosage rorms

3.5-4.0 3.0-3.5 3.5-4.0


B
_____1---+_1---

t
3.5-4.0

11--"'IlI---1 25-26 12.0 ± 0.2

A: acid-resistant wire clasp B: acid-resistant 'ivire support


Figure 2.9.3.-3. - Altemative sinker
Dimensions in millimetres

Place 1 dosage unit in the apparatus, taking care to exclude 1 dosage unit in the apparatus, cover the vessel and operate
air bubbles from the surface of the dosage unit. Operate the apparatus at the specifled rateo After 2 h of operation
the apparatus at the specifled rateo Within the time interval in 0.1 M hydrochloric acid, withdraw an aliquot of the fluid
specifled, or at each of the times stated, withdraw a specimen and proceed immediately as directed under Buffer stage.
from a zone midway between the surface of the dissolution Perform an analysis of the aliquot using a suitable assay
medium and the top of the rotating basket or blade, not les s method.
than 1 cm from the vessel wall. Where multiple sampling
- Buffer stage. Complete the operations of adding the buffer
times are specifled, replace the aliquots withdrawn for analysis
and adjusting the pH within 5 mino With the apparatus
with equal volumes of fresh dissolution medium at 37 oC Of,
operating at the rate specifled, add to the fluid in the
where it can be shown that replacement of the medium is not
vessel 250 mL of a 0.20 M solution of trisodium phosphate
necessary, correct for the volume change in the calculation.
dodecahydrate R that has been equilibrated to 37 ± 0.5 oc.
Keep the vessel covered for the duration of the test and verify
Adjust, if necessary, with 2 M hydrochloric acid R or 2 M
the temperature of the medium at suitable times. Perform the
sodium hydroxide R to a pH of 6.8 ± 0.05. Continue to
analysis using a suitable assay methodW Repeat the test with
operate the apparatus for 45 min, or for the specified time.
additional dosage units.
At the end of the time period, withdraw an aliquot of
If automated equipment is used for sampling or the apparatus the fluid and perform the analysis using a suitable assay
is otherwise modifled, verification that the modified apparatus method.
will produce results equivalent to those obtained with the
apparatus described in this chapter, is necessary. Method B
Dissolution medium. A suitable dissolution medium is used. - Acid Stage. Place 1000 mL of 0.1 M hydrochloric acid in
The volume specifled refers to measurements made between the vessel and assemble the apparatus. Allow the medium
20 oC and 25 oc. If the dissolution medium is a buffered to equilibrate to a temperature of 37 ± 0.5 oc. Place
solution, adjust the solution so that its pH is within 0.05 units 1 dosage unit in the apparatus, cover the vessel, and operate
of the specifled pH. Dissolved gases can cause bubbles to the apparatus at the specifled rateo After 2 h of operation in
form, which may change the results of the test. In such cases, 0.1 M hydrochloric acid, withdraw an aliquot of the fluid,
dissolved gases must be removed prior to testing(sJ. and proceed immediately as directed under Buffer stage.
Time. Where a single time speciflcation is given, the test Perform an analysis of the aliquot using a suitable assay
may be concluded in a shorter period if the requirement method.
for minimum amount dissolved is met. Samples are to be - Buffer stage. For this stage of the procedure use buffer
withdrawn only at the stated times, within a tolerance of that has previously been equilibrated to a temperature of
± 2 per cent. 37 ± 0.5 oc. Drain the acid from the vessel and add 1000 mL
Prolonged-release solid dosage forms of pH 6.8 phosphate buffer, prepared by mixing 3 volumes
of 0.1 M hydrochloric acid with 1 volume of a 0.20 M
Procedure. Proceed as described for conventional-release
solution of trisodium phosphate dodecahydrate R and
dosage forms.
adjusting, if necessary, with 2 M hydrochloric acid R or 2 M
Dissolution medium. Proceed as described for sodium hydroxide R to a pH of 6.8 ± 0.05. This may also be
conventional-release dosage forms. accomplished by removing from the apparatus the vessel
Time. The test-time points, generally 3, are expressed in hours. containing the acid and replacing it with another vessel,
containing the buffer and transferring the dosage unit to
Delayed-release solid dosage forms the vessel containing the buffer. Continue to operate the
Procedure. Use Method A or Method B. apparatus for 45 min, or for the specifled time. At the end
Method A of the time period, withdraw an aliquot of the fluid and
perform the analysis using a suitable assay method.
- Acid stage. Place 750 mL of 0.1 M hydrochloric acid in the
vessel, and assemble the apparatus. Allow the medium Time. AH test times stated are to be observed within a
to equilibrate to a temperature of 37 ± 0.5 oc. Place tolerance of ± 2 per cent, unless otherwise specifled.

(4) Test are filtcred sampling llnless filtratlon is demonstrated to be unnecessary. Use an inert filter that does not cause adsorption of the active substance or
contain substallce~ that would with tbe
(5) A method of dcaeration is <l'i follmvs: hc;:lt the mediulD, slirring gently, to abolLt 41°C, immediatdy filter under vacuum using ;} filter having a porosity 01' OA5 ~lIn or less, with
vigorous stirring, and continue stirring undcr VaCllUl11 for about 5 mino Otber validated deaeration lechniques fO!" removal 01' dissolved gases lJlay be used.

General Notices (1) apply to allmonographs and other texts 291


2.9.3. Dissolution test foJ:' solid dosage fonns EUROPEAN PHARMACOPOEIA 8.0

50.8 ± 1 APPARATUS 3
.. 38.1 ± 1 .- 1"1 Air holes Conventional-release solid dosage forrns
~03.9±0.1 Procedure. Place the stated volume of the dissolution medium
(± 1 per cent) in each vessel of the apparatus. Assemble the
---+,,L,+-I'.-!-t'-T---,
r - -4
apparatus, equilibrate the dissolution medium to 37 ± 0.5 oc,
r l and remove the thermometer. Place 1 dosage unit in each of
I I the reciprocating cylinders, taking care to exclude air bubbles
+1 I I ---- Evaporation cap from the surface of each dosage unit, and immediately operate
ro the apparatus as specified. During the upward and downward

CD
I I stroke, the reciprocating cylinder moves through a total
I I distan ce of9.9-1O.1 cm. Within the time interval specified, or
at each of the times stated, raise the reciprocating cylinders
and withdraw a portion of the medium from a zone midway
between the surface of the dissolution medium and the bottom
of each vessel. Perform the analysis as directed. If necessary,
repeat the test with additional dosage units.

1IIIr---- Mesh sereen


Replace the aliquot withdrawn for analysis with equal volumes
of fresh dissolution medium at 37 oC or, where it can be shown
that replacement of the medium is not necessary, correct for
the volume change in the calculation. Keep the vessel covered
23 L26 I with the evaporation cap for the duration of the test and verify
the temperature of the medium at suitable times.
Dissolution medium. Proceed as described for
I conventional-release dosage forms under Apparatus 1 and 2.
Time. Proceed as described for conventional-release dosage
forms under Apparatus 1 and 2.
I
+1
o
o I .. Glass reciproeating
eylinder
Prolonged-release dosage forms
Procedure. Proceed as described for conventional-release
dosage forms under Apparatus 3.
I Dissolution medium. Proceed as described for
I prolonged-release dosage forms under Apparatus 1 and 2.
I Time. Proceed as described for prolonged-release dosage

JLJ ¡ l. Mesh sereen


forms under Apparatus 1 and 2.
Delayed-release dosage forms
Procedure. Proceed as described for delayed-release dosage
I forms, Method B, under Apparatus 1 and 2, using one row
of vessels for the acid stage media and the following row of

'"
47 ± 1.4
.. vessels for the buffer stage media, and using the volume of
medium specified (usually 300 mL).
Time. Proceed as directed for delayed-release dosage forms
under Apparatus 1 and 2.
I
I
APPARATUS4

---- Conventional-release dosage forms


I
I
Glass vessel
I Procedure. Place the glass beads into the cen specified. Place
I I I
I 1 dosage unit on top of the beads or, if specified, on a wire
I I carrier. Assemble the filter head and fix the parts together
+1
o I
ro by means of a suitable clamping device. Introduce by the
pump the dissolution medium warmed to 37 ± 0.5 oC through
I the bottom of the cell to obtain the flow rate specified and
I measured with an accuracy of 5 per cent. Collect the eluate by
fractions at each of the times stated. Perform the analysis as
directed. Repeat the test with additional dosage units.
I Dissolution medium. Proceed as described for
I
conventional-release dosage forms under Apparatus 1 and 2.
Time. Proceed as described for conventional-release dosage
forms under Apparatus 1 and 2.
I
I I
Prolonged-release dosage forms
I
L ___ + ___ ..J
I I
Procedure. Proceed as described for conventional-release
dosage forms under Apparatus 4.
I Dissolution medium. Proceed as described for
conventional-release dosage forms under Apparatus 4.
Figure 2.9.3.-4. - Apparatus 3, glass vessel and reciprocating Time. Proceed as described for conventional-release dosage
cylinder forms under Apparatus 4.
Dimensions in millimetres unless otherwise specified Delayed-release dosage forms
Procedure. Proceed as described for delayed-release dosage
forms under Apparatus 1 and 2, using the specified media.

292 See the information section on general monographs (ca ver pages)
EUROPEAN PHARMACOPOEIA 8.0 2.9.3. Dissolution test for solid dosage forffis

f*B*Bf*B*H ____ Filler chamber


Sieve
- - - - - d = 0.2 w = 0.45

020 l 0.2

l()
o 022.6 ± 0.2
+1
l()

______ Score lor the holder

U
03

<:0
¿
'E
I 00.8 ± 0.05

-!~

l()
c:i

l()
N
c:i
+1
l()
24.0 + 8. 5
N

Figure 2.9.3.-5. - Apparatus 4, large ceU for tablets and capsules (top), tablet holder for the large ceU (bottom)
Dimensions in millimetres unless otherwise specified

Time. Proceed as described for delayed-release dosage forms tested conform to Table 2.9.3.-1. Continue testing through
under Apparatus 1 and 2. the 3 levels unless the results conform at either SI or 52'
The quantity Q, is the specifled amount of dissolved active
INTERPRETATION substance, expressed as a percentage of the labelled content;
Conventional-release solid dosage forms the 5 per cent, 15 per cent, and 25 per cent values in the Table
are percentages of the labelled content so that these values
Unless otherwise specifled, the requirements are met if the and Q are in the same terms.
quantities of active substance dissolved from the dosage units

General No/ices (1) apply to all monographs and other texts 293
2.9.3. Dissolution test for solid dosage forms EUROPEAN PHARMACOPOEIA 8.0

Hf-+Hf-***-lH-**-l<I _________ Filter chamber

~~~~iP;ttí;:t;:tj~ ---Sieve d = 0.2 w = 0.45


020 ± 0.2

LO 012 ± 0.2
c:i
+1
o
LO

_.,...--_ _ _ _ Score for the holder

¿+--------\,
·E¡----=:=:¡¡;tItl-l..-_ _ _ _ 00.8 ± 0.05
- ___+-+...------ 03

~
+ 0.5
13.5 O

Figure 2.9.3.-6. - Apparatus 4, small cell fo1' tablets and capsules (top), tablet holder for the small cell (bottom)
Dimensions in millimetres unless otherwise specifíed

Table 2.9.3.-1 Prolonged-release dosage forms


Leve! Number Acceptance criteria Unless otherwise specified, the requirements are met if the
tested
quantities of active substance dissolved from the dosage units
S, 6 Each unit is not less than Q + 5 per cen!. tested conform to Table 2.9.3.-2. Continue testing through
S2 6 Average of 12 units (S, + S,) is equa! to or greater than Q, the 3 levels unless the results conform at either L¡ or L2 •
and no unit is less than Q - 15 per cen!. Limits on the amounts of active substance dissolved are
S3 12 Average of 24 units (S, + S, + 53) is egual to or greater expressed in terms of the percentage of labelled content. The
than Q, not more than 2 units are les s than Q - 15 per limits embrace each value of Qi' the amount dissolved at each
cent, and no is less than Q - 25 per cen!. specified fractional dosing interval. Where more than one
range is specified, the acceptance criteria apply individually
to each range.

294 See the information section on general monographs (caver pages)


EUROPEAN PHARMACOPOEIA 8.0 2.9.4. Dissolution test for transdermal patches

Table 2.9.3.-2 l. DISK ASSEMBLY METHOD


Leve! N umber Acceptance crHeria Equipment. Use the paddle and vessel assembly from the
testea paddle apparatus described in the dissolution test for solid
Ll 6 No individual value lies outside each of the stated ranges oral dosage forms (2.9.3) with the addition of a stainless steel
and no individual value is less lhan the statea amoun! at disk assembly (SSDA) in the form of a net with an aperture of
the final test time.
125 flm (see Figure 2.9.4.-1).
L, 6 The average value of the 12 units (L l + L,) lies within each
of the stated ranges and is not les s than the stated amount 125 jJm mesh
at the final test time; none is more than 10 per cent of stainless steel net 4.5mm
labelled content outside each of the stated ranges; and
none is more than 10 per cent oflabelled content below
1-1
the stated amount al the final test time.
L, 12 The average value of the 24 unils (L l + L, + L 3 ) lies within
each of the stated ranges, and is not less than the stated \j D i 3.3mm
amount at the final test time; not more than 2 of the
24 units are more than 10 per cent oflabelled cantent
outside each of the stated ranges; not more than 2 of the
24 nnits are more than 10 per cent oflabelled cantent
below the stated amount at the final test time; and none
of the units is more than 20 per cent oflabelled cantent
outside each of the stated ranges or more than 20 per cent
oflabelled content below the stated amount at lhe final
test time.

Delayed-release dosage forms


Acid stage. Unless otherwise specified, the requirements of
this portion of the test are met if the quantities, based on the
percentage of the labelled content of active substance dissolved
from the units tested conform to Table 2.9.3.-3. Continue
testing through the 3 levels unless the results of both acid and
buffer stages conform at an earlier leve!.
Table 2.9.3.-3
Level Number Acceptance (riteria ~ 41.2mm ~
tested
Al 6 No individual value exceeds 10 per cent dissolved. Figure 2.9.4.-1. - Disk assembly
A, 6 The average value of the 12 units (Al + A 2 ) is not more The SSDA holds the system at the bottom of the vessel and is
than 10 per cent dissolved, and no individual unit is designed to minimise any dead volume between the SSDA and
greater than 25 per cent dissolved. the bottom of the vesse!. The SSDA holds the patch flat, with
A3 12 The average value of the 24 units (Al + A 2 + A 3 ) is not the release surface uppermost and parallel to the bottom of the
more than 10 per cent dissolved, and no individual unit ís
greater than 25 per cent díssolved.
paddle blade. A distance of 25 ± 2 mm between the bottom of
the paddle blade and the surface of the SSDA is maintained
Buffer stage. Unless otherwise specified, the requirements during the test (see Figure 2.9.4.-2). The temperature is
are met if the quantities of active substance dissolved from maintained at 32 ± 0.5 oc. The vessel may be covered during
the units tested conform to Table 2.9.3.-4. Continue testing the test to minimise evaporation.
through the 3 levels unless the results of both stages conform
at an earlier level. The value of Q in Table 2.9.3.-4 is 75 per
cent dissolved unless otherwise specified. The quantity, Q,
is the specified total amount of active substance dissolved in
both the acid and buffer stages, expressed as a percentage of
the labelled content. The 5 per cent, 15 per cent and 25 per Dissolution vessel
cent values in the Table are percentages of the labelled content
so that these values and Q are in the same terms.
Table 2.9.3.-4 +-----r- Paddle

Leve! Number Acceptance criteria


tested
Bl 6 No unít ís less than Q + 5 per cent.

B, 6 The average value of the 12 units (B l + B, ) is equal to or


greater than Q, and no unít is less than Q - 15 per cent.
! 1f mm
25 2

\133m~
B3 12 The average value of the 24 units (B l + B, + BJ is equal
to or greater than Q, not more than 2 units are less than
Q - 15 per cent, and no unit ís less than Q - 25 per cent.
A
Disk assembly
Recommendations on dissolution testing are given in general
chapter 5.17.1. Figure 2.9.4.-2. - Paddle and disk
Procedure. Place the prescribed volume of the dissolution
medium in the vessel and equilibrate the medium to the
01/2008:20904 prescribed temperature. Apply the patch to the SSDA,
ensuring that the release surface of the patch is as flat as
possible. The patch may be attached to the SSDA by a
2.9.4. DISSOLUTION TEST FOR prescribed adhesive or by a strip of a double-sided adhesive
TRANSDERMAL PATCHES tape. The adhesive or tape are previously tested for the absence
of interference with the assay and of adsorption of the active
This test is used to determine the dissolution rate of the active ingredient(s). Press the patch, releas e surface facing up, onto
ingredients of transdermal patches. the side of the SSDA made adhesive. The applied patch must

General Natices (1) apply to a/l monographs and other texts 295
2.904. Dissolution test for transdermal patches EUROPEAN PHARMACOPOEIA 8.0

not overlap the borders of the SSDA. For this purpose and Cover. The cover has a central opening with a diameter
provided that the preparation is homogeneous and uniformly selected according to the size of the patch to be examined.
spread on the outer covering, an appropriate and exactly The patch can thus be precisely centred, and its releasing
measured piece of the patch may be cut and used for testing sur fa ce limited. The following diameters may be used: 20 mm,
the dissolution rateo This procedure may also be necessary 32 mm, 40 mm, 50 mm corresponding to are as of 3.14 cm',
to achieve appropriate sink conditiol1S. This procedure must 8.03 cm 2 , 12.56 cm 2 , 19.63 cm 2 , respectively. The cover is
not be applied to membrane-type patches. Place the patch held in place by nuts screwed onto bolts projecting from the
mounted on the SSDA flat at the bottom of the vessel with support. The cover is sealed to the support by a rubber ring
the release surface facing upwards. Immediately rotate the set on the reservo ir.
paddle at 100 r/min, for example. At predetermined intervals,
withdraw a sample from the zone midway between the surface
Extraction cell. The cell holds the patch flat, with the release
surface uppermost and parallel to the bottom of the paddle
of the dissolution medium and the top of the blade, not less
blade. A distance of 25 ± 2 mm is maintained between the
than 1 cm from the vessel wall.
paddle blade and the surface of the patch (se e Figure 2.9.4.-4).
Perform the assay on each sample, correcting for any volume The temperature is maintained at 32 ± 0.5 oc. The vessel may
losses, as necessary. Repeat the test with additional patches. be covered during the test to minimise evaporation.
Procedure. Place the prescribed volume of the dissolution
2. CELL METHOD
medium in the vessel and equilibrate the medium to the
Equipment. Use the paddle and vessel assembly from the prescribed temperature. Precisely centre the patch in the
paddle apparatus described in the dissolution test for solid cel! with the releasing surface uppermost. Close the cell, if
oral dosage forms (2.9.3) with the addition of the extraction necessary applying a hydrophobic substance (far example,
cell (cell). petrolatum) to the flat surfaces to ensure the seal, and ensure
The ceU is made of chemically inert materials and consists of that the patch stays in place. Introduce the cell flat into
a support, a cover and, if necessary, a membrane placed on the bottom of the vessel with the cover facing upwards.
the patch to iso late it from the medium that may modify or Immediately rotate the paddle, at 100 r/min for example. At
adversely affect the physico-chemical properties of the patch predetermined intervals, withdraw a sample from the zone
(see Figure 2.9.4.-3). midway between the surface of the dissolution medium and
the top of the paddle blade, not less than 1 cm from the vessel
wall.
Perform the assay on each sample, correcting for any volume
losses, as necessary. Repeat the test with additional patches.

o
50 mm
19.63 cm'
--->Cover
o
'--::=====::::::==--_____-_':.1 8.5 mm --------t----+ Paddle
o
o o -----+ Membrane
o ---+----+ Dissolution vessel

R-----+Bolis
2.6 mm -"----------

i -> Rubber ring \'-----J------+-. A

74------>0. Reservoir 125 ± 2 mm

~ ___=-=--+_+--__L¡ 9.6 mm
{!:!:=-~. ~. -~. ~-:·!;·;..;..:..~.. ~:E:íLt=:Extraction cell
~ /
52 mm
21.23 cm'
I
I
Figure 2.9.4.-4. - Paddle over extraction ceU
I 70.5 mm
3. ROTATING CYLINDER METHOD
L27mm
. . i Equipment. Use the assembly of the paddle apparatus
I , 38 mm • I
1~-----+45 mm described in the dissolution test for solid oral dosage forms
(2.9.3). Replace the paddle and shaft with a stainless steel
~--- 52 mm cylinder stirring element (cylinder) (see Figure 2.9.4.-5). The
patch is placed on the cylinder at the beginning of each test.
The distance between the inside bottom of the vessel and the
cylinder is maintained at 25 ± 2 mm during the test. The
Figure 2.9.4.-3. - Extraction cell temperature is maintained at 32 ± 0.5 oc. The vessel is covered
Support. The central part of the support forms a cavity during the test to minimise evaporation.
intended to hold the patch. The cavity has a depth of 2.6 mm Procedure. Place the prescribed volume of the dissolution
and a diameter that is appropriate to the size of the patch to medium in the vessel and equilibrate the medium to the
be examined. The following diameters can be used: 27 mm, prescribed temperature. Remove the protective liner from
38 mm, 45 mm, 52 mm, corresponding to volumes of 1.48 mL, the patch and place the adhesive side on a piece of suitable
2.94 mL, 4.13 mL, 5.52 mL, respectively. inert porous membrane that is at least 1 cm larger on all

296 See the information section on general monographs (caver pages)


EUROPEAN PHARMACOPOEIA 8.0 2.9.5. Uniformity of mass af single-clase preparatians

1.270
Four holes at 1.111 dia.
equally spaced on 2.540
Inlerference fit
dia. b.c. at 63.4 ± OS
angle to surface

Maximu m rad ius 0.300 ----iE:-.-J---=:::::.

This adaptar
sectian is to
I be used for
TOLERANCES : ± 0.0127 k-2.045 larger systems
FINISH : Degrease before
final assembly or rod
and cylinder
MATERIAL: Stainless steel

Figure 2.9.4.-5. - Cylinder stirring element

Dimensions in centimetres

sides than the patch. Place the patch on a clean surface with 0112008:20905
the membrane in contact with this surface. Two systems for
adhesion to the cylinder may be used:
- applya suitable adhesive to the exposed membrane borders
and, if necessary, to the back of the patch, 2.9,5. UNIFORMITY OF MASS OF
- apply a double-sided adhesive tape to the external wall of SINGLE-DOSE PREPARATIONS
the cylinder.
Using gentle pressure, carefully apply the non-adhesive side Weigh individually 20 units taken at random or, for single-dose
of the patch to the cylinder, so that the release surface is in preparations presented in individual containers, the contents
contact with the dissolution medium and the long axis of the of 20 units, and determine the average mass. Not more than
patch fits around the circumference of the cylinder. 2 of the individual masses deviate from the average mass by
The system for adhesion used is previously tested for absence more than the percentage deviation shown in Table 2.9.5.-1
of interference with the assay and of adsorption of the active and none deviates by more than twice that percentage.
ingredient( s).
Place the cylinder in the apparatus, and immediately rotate the For capsules and powders fol' parenteral administration,
cylinder at 100 r/min, for example. At determined intervals, proceed as described below.
withdraw a sample of dissolution medium from a zone
midway between the surface of the dissolution medium and
the top of the rotating cylinder, and not less than 1 cm from
the vessel wall. CAPSULES
Perform the assay on each sample as directed in the individual Weigh an intact capsule. Open the capsule without losing any
monograph, correcting for any volume withdrawn, as part of the shell and remove the contents as completely as
necessary. Repeat the test with additional patches. possible. For soft shell capsules, wash the shell with a suitable
Interpretation. The requirements are met if the quantity of solvent and allow to stand until the odour of the solvent is no
active ingredient(s) released from the patch, expressed as the longer perceptible. Weigh the shell. The mass of the contents
amount per surface area per time unit, is within the prescribed is the difference between the weighings. Repeat the procedure
limits at the defined sampling times. with another 19

General Nolices (1) apply lo all monographs and other texts 297
2.9.6. Uniformity of content of single-dose preparations EUROPEAN PHARMACOPOEIA 8.0

Table 2.9.5.-1 TEST B


Pharmaceutical Form Average Mass Percentage Capsules, powders other than for parenteral administration,
deviation granules, suppositories, pessaries. The preparation complíes
Tablets (uncaated and 80 mg ar less 10 with the test if not more than one individual content is outside
film-mated) the limits of 85 per cent to 115 per cent of the average content
More than 80 mg and
less than 250 mg
7.5 and none is outside the limits of 75 per cent to 125 per cent of
the average content. The preparation fails to comply with the
250 mg or more 5 test if more than 3 individual contents are outside the limits of
Capsules, granules Less than 300 mg 10 85 per cent to 115 per cent of the average content or if one or
(uncoated, single-dosel more individual contents are outside the limits of 75 per cent
and powders (single-dose) 300 mg or more 7.5 to 125 per cent of the average content.
Powders for parenteral More than 40 mg 10 If 2 or 3 individual contents are outside the limits of 85 per
administration * cent to 115 per cent but within the limits of 75 per cent to
(single-dose) 125 per cent, determine the individual contents of another
Suppositories and pessaries Al! masses 5 20 dosage units taken at random. The preparation complies
10
with the test if not more than 3 individual contents of the
Powders for eye-drops and Less than 300 mg
powders for eye lotions 7.5
30 units are outside the limits of 85 per cent to 115 per cent
300 mg or more
(single-dose) of the average content and none is outside the limits of 75 per
* When the average mass is equal to or below 40 mg, the preparation cent to 125 per cent of the average contento
is not submitted to the test for uniformity of mas s but to the test for
uniformity of content of single-dose preparations (2.9.6). TEST C
Transdermal patches. The preparation complies with the test
POWDERS FOR PARENTERAL ADMINISTRATION if the average content of the 10 dosage units is between 90 per
Remove any paper labels from a container and wash and dry cent and 110 per cent of the content stated on the label and if
the outside. Open the container and without delay weigh the the individual content of each dosage unít is between 75 per
container and its contents. Empty the container as completely cent and 125 per cent of the average content.
as possible by gentle tapping, rinse it if necessary with water R
and then with alcohol R and dry at 100-105 oC for 1 h, or, if the 0112010:20907
nature of the container precludes heating at this temperature,
dry at a lower temperature to constant mass. A110w to cool 2.9.7. FRIABILITY OF UNCOATED
in a desiccator and weigh. The mass of the contents is the TABLETS(6)
difference between the weighings. Repeat the procedure with
another 19 containers. This chapter provides guídelines for the friability
determination of compressed, uncoated tablets. The test
procedure presented in this chapter ís generally applicable to
0112008:20906 most compressed tablets. Measurement of tablet friability
supplements other physical strength measurements, such as
2.9.6. UNIFORMITY OF CONTENT OF tablet breaking force.
SINGLE-DOSE PREPARATIONS Use a drum, with an internal diameter between 283-291 mm
and a depth between 36-40 mm, of transparent synthetic
The test for uniformity of content of single-dose preparations polymer with polished internal surfaces, and subject to
is based on the assay of the individual contents of active minimum static build-up (see Figure 2.9.7.-1.). One side of
substance(s) of a number of single-dose units to determine the drum is removable. The tablets are tumbled at each turn
whether the individual contents are within limits set with of the drum by a curved projection with an inside radius
reference to the average content of the sample. between 75.5-85.5 mm that extends from the middle of the
The test is not required for multivitamin and trace-element drum to the outer waH. The outer diameter of the central
preparations and in other justified and authorised ring is between 24.5-25.5 mm. The drum is attached to the
circumstances. horizontal axis of a device that rotates at 25 ± 1 r/min. Thus,
Method. Using a suitable analytical method, determine the at each turn the tablets ro11 or slide and faH onto the drum
individual contents of active substance(s) of 10 dosage units wa11 or onto each other.
taken at random. For tablets with a unit mass equal to or less than 650 mg, take
Apply the criteria of test A, test B or test C as specified in the a sample of whole tablets corresponding as near as possible to
monograph for the dosage form in question. 6.5 g. For tablets with a unit mass of more than 650 mg, take a
sample of 10 whole tablets. The tablets are carefully dedusted
TEST A prior to testing. Accurately weigh the tablet sample, and place
Tablets, powders for parenteral administration, ophthalmic the tablets in the drum. Rotate the drum 100 times, and
inserts, suspensions for injection. The preparation complies remove the tablets. Remove any loose dust from the tablets as
with the test if each individual content is between 85 per cent before, and accurately weigh.
and 115 per cent of the average content. The preparation fails Generally, the test is run once. rf obviously cracked, cleaved, or
to comply with the test if more than one individual content is broken tablets are present in the tablet sample after tumbling,
outside these limits or if one individual content is outside the the sample fails the test. If the results are difficult to interpret
limits of 75 per cent to 125 per cent of the average content. or if the weight loss is greater than the targeted value, the test
rf one individual content is outside the limits of 85 per is repeated twice and the mean of the 3 tests determined. A
cent to 115 per cent but within the limits of 75 per cent to maximum 105s of mas s (obtained from a single test or from
125 per cent, determine the individual contents of another 20 the mean of 3 tests) not greater than 1.0 per cent is considered
dosage units taken at random. The preparation complies with acceptable for most products.
the test if not more than one of the individual contents of the rf tablet size or shape causes irregular tumbling, adjust the
30 units is outside 85 per cent to 115 per cent of the average drum base so that the base forms an angle of about 10° with the
content and none is outside the limits of 75 per cent to 125 per horizontal and the tablets no longer bind together when lying
cent of the average content. next to each other, which prevents them from falling freely.

(6) This chapter has undergone pharmacopoeial hannonisation. See chapter 5.8. Pharrnacopoeial harmonisation.

298 See the information section on general monographs (caver pages)


EUROPEAN PHARMACOPOEIA 8.0 2.9.9. Measurement of consistency by penetrometry

~ ~
010.0 ± 0.1 mm
~ ~
156.0 ± 2.0 mm ~ ~
Drop 0287.0 ± 4.0 mm ~ ~
height (in!.)
~ ~
~ ~
~ ~
~
~~
J

~~ ~
~ ~
~ ~
~ ~
~ ~
~ ~

, o 302.5 ± 4.0 mm
o 25.0 ± 0.5 mm
~
.-".~
~
~

38.0 ± 2.0 mm

Figure 2.9.7.-l. - Tablet friability apparatus

Effervescent tablets and chewable tablets may have different respect to the direction of application of the force. Carry out
specifications as far as friabílity is concerned. In the case of the measurement on 10 tablets, taking care that al! fragments
hygroscopic tablets, a humidity-controlled environment ís of tablets have been removed before each determination.
required for testing.
This procedure does not apply when fully automated equipment
A drum with dual scooping projections, or apparatus with is used.
more than one drum, for the running of multiple samples at
one time, are also permitted. EXPRESSION OF RESULTS
Express the results as the mean, minimum and maximum
values of the forces measured, all expressed in newtons.
Indicate the type of apparatus and, where applicable, the
0112008:20908 orientation of the tablets.

2.9.8. RESISTANCE TO CRUSHING OF


TABLETS
07/2008:20909
This test is intended to determine, under defined conditions,
the resistance to crushing of tablets, measured by the force
needed to disrupt them by crushing. 2.9.9. MEASUREMENT OF
CONSISTENCY BY PENETROMETRY
APPARATUS
This test is intended to measure, under determined and
The apparatus consists of 2 jaws facing each other, one of validated conditions, the penetration of an object into the
which moves towards the other. The flat surfaces of the jaws product to be examined in a container with a specified shape
are perpendicular to the direction of movement. The crushing and size.
surfaces of the j aws are flat and larger than the zone of contact
with the tablet. The apparatus is calibrated using a system
APPARATUS
with a precision of 1 newton.
The apparatus consists of a penetrometer made up of a stand
OPERATING PROCEDURE and a penetrating object. A suitable apparatus is shown in
Figure 2.9.9.-l.
Place the tablet between the jaws, taking into account, where
applicable, the shape, the break-mark and the inscription;
for each measurement orient the tablet in the same way with

General Notices (1) apply to all monographs and other texts 299
2.9.9. Measurement of consistency by penetrometry EUROPEAN PHARMACOPOEIA 8.0

The stand is made up of:


- a vertical shaft to maintain and guide the penetrating
object;
- a horizontal base;
- a device to ensure that the penetrating object is vertical;

__~~____________ A - a device to check that the base is horizontal;


- a device to retain and releas e the penetrating object;
- a scale showing the depth of penetration, graduated in
tenths of a millimetre.
----_B The penetrating object, made of a suitable material, has a
smooth surface, and is characterised by its shape, size and
++-___._ _ _ ~. ~ ___ e mass (m).
Suitable penetrating objects are shown in Figures 2.9.9.-2 and
------------D 2.9.9.-3.

+-~----------- E PROCEDURE
Prepare the test samples according to one of the following
procedures.
---+----~---------F
A. Carefully and completely fill 3 containers, without forming
air bubbles. Level if necessary to obtain a flat surface.
~--G Store the samples at 25 ± 0.5 oC for 24 h, unless otherwise
prescribed.
B. Store 3 samples at 25 ± 0.5 oC for 24 h, unless otherwise
prescribed. Apply a suitable shear to the samples for 5 mino
Carefully and completely fill 3 containers, without forming
Figure 2.9.9.-1. - Penetrometer air bubbles, and level if necessary to obtain a flat surface.
A. Scale showing the depth of penetration, graduated in C. Melt 3 samples and carefully and completely fill
tenths of millimetres. 3 containers, without forming air bubbles. Sto re the
B. Vertical shaft to maintain and guide the penetrating object. samples at 25 ± 0.5 oC for 24 h, unless otherwise prescribed.
C. Device to retain and to release the penetrating object Determination of penetration. Place the test sample on
automatically and for a constant time. the base of the penetrometer. Verify that its surface is
D. Device to ensure that the penetrating object is vertical and perpendicular to the vertical axis of the penetrating object.
that the base is horizontal. Bring the temperature of the penetrating object to 25 ± 0.5 oC
E. Penetrating object (see Figures 2.9.9.-2 and 3). and then adjust its position such that its tip just touches the
surface of the sample. Release the penetrating object and hold
F. Container. it free for 5 s. Clamp the penetrating object and measure the
G. Horizontal base. depth of penetration. Repeat the test with the 2 remaining
H. Control for the horizontal base. containers.
55 ± 0.25 0

3.14 ± 0.04 0

r;1...
o(----- A
co
90' ±1,"5!-'_t77I__
+1
CJ)

en
5.4±1.50
"
1
o(

od

N h
'"
+1

j
CJ)

en

+1 f»'''------~ B

~~+-l--~::~-=--=--c ----L

------
8.38 ± 0.02 0

Figure 2.9.9.-2. - Cone (m = 102.5 ± 0.05 g), suitable container (d = 102 mm or 75 mm; h 2: 62 mm)
and shaft (l = 162 mm; m = 47.5 ± 0.05 g).
Dimensions in millimetres

300 See the information section on general monographs (cover pages)


EUROPEAN PHARMACOPOEIA 8.0 2.9.10. Ethanol content

121 12.65
121 9.5

r
;:
121 3.2

121 8.2 ~,-J7+bJ...,oC-~~~r~

1 ~l~ 1

Figure 2.9.9.-3 - Micro-cone (m = 7.0 g), suitable container and shaft (l = 116 mm; m = 16.8 g)
Dimensions in millimetres
EXPRESSION OF THE RESULTS B

The penetration is expressed in tenths of a millimetre


as the arithmetic mean of the 3 measurements. lf any of
the individual results differ from the mean by more than
3 per cent, repeat the test and express the results of the 6 180
measurements as the mean and the relative standard deviatioll.

A 500 ml

o
o e
04/2013:20910 '""

2.9.10. ETHANOL CONTENT


These methods are intended for the examination of liquid
pharmaceutical preparations and their ingredients that contain I-"----D
ethanol. The ethanol content of a liquid is expressed as the
number of volumes of ethanol contained in 100 volumes of
the liquid, the volumes being measured at 20 ± 0.1 oc. This is
known as the 'percentage of ethanol by volume' (per cent V/V).
The content may also be expressed in grams of ethanol per
100 g of the liquido This is known as the 'percentage of ethanol
by mass' (per cent m/m).
E

METHODA

Where preparations contain dissolved substances, the


dissolved substances must be separated from the ethanol that Figure 2.9.10.-1. - Apparatus for the determination of ethanol
is to be determined by distillation. Where distillation would content
distil volatile substances other than ethanol and water, the Dimensions in millimetres
appropriate precautions are stated in the monograph. Apparatus. The apparatus (see Figure 2.9.10.-1) consists of a
round-bottomed flask (A) fittea with a distillation head (B)
The relation between the density at 20 ± 0.1 oC, the relative
with a steam trap and attached to a vertical condenser (e). The
density (corrected to vacuum) and the ethanol content of
latter is fitted at its lower part with a tube (D), which carries the
a mixture of water and ethanol is given in the tables of the
distillate into the lower part of a 100 mL or 250 mL volumetric
International Organisation for Legal Metrology (1972),
flask. The volumetric flask is immersed in a mixture of ice
International Recommendation No. 22.

General Notices (1) apply to all monographs and other texts 301
2.9.10. Ethanol content EUROPEAN PHARMACOPOEIA 8.0

and water (E) during the distillation. A disc having a circular P20 Relative density of the Ethanol content in
aperture 6 cm in diameter is placed under the flask (A) to (kg.m- 3 ) distillate measured in air per cent V/V
reduce the risk of charring of any dissolved substances. 20
d 20 at 20 oC

Method 983.0 0.9848 11.44

983.5 0.9853 ! 1.02


Pycnometer method/oscillating transducer density meter
method. Transfer 25.0 mL of the preparation to be examined, 984.0 0.9858 10.60
measured at 20 ± 0.1 oC, to the distillation flask. Dilute with
984.5 0.9863 10.18
100-150 mL of distilled water R and add a few pieces of
pumice. Attach the distillation head and condenser. Distil and 985.0 0.9868 9.76
coUeet not less than 90 mL of distillate in a 100 mL volumetric
985.5 0,9873 9.35
flask. Adjust the temperature to 20 ± 0.1 oC and dilute to
100.0 mL with distilled water R at 20 ± 0.1 oc. Determine 986.0 0.9878 8.94
the relative density at 20 ± 0.1 oC using a pycnometer or an
986.5 0.9883 8.53
oscillating transducer density meter.
987.0 0.9888 8.13
The values indicated in Table 2.9.10.-1, column 3, are
multiplied by 4 to obtain the percentage of ethanol by volume 987.5 0.9893 7.73
(V/V) contained in the preparation. After calculation of the 988.0 0.9898 7.34
ethanol content using the table, round off the result to 1
decimal place. 988.5 0.9903 6.95

989.0 0,9908 6.56


Table 2.9.10.-1. - Relationship between density, relative density
and ethanol content 989.5 0.9913 6.17

990.0 0.9918 5.79


P,0 Relative density of the Ethanolcontentin
(kg.m- 3 ) distillate measured in air per cent V/V 990.5 0.9923 5.42
20
d 20 al 20 oC
991.0 0.9928 5.04
968.0 0.9697 25.09
991.5 0.9933 4.67
968.5 0.9702 24.64
992.0 0.9938 4.30
969.0 0.9707 24.19
992.5 0.9943 3.94
969.5 0.9712 23.74
993.0 0.9948 3.58
970.0 0.9717 23.29
993.5 0.9953 3.22
970.5 0.9722 22.83
994.0 0.9958 2,86
97l.0 0.9727 22.37
994.5 0.9963 2.51
971.5 0.9733 21.91
995.0 0.9968 2.16
972.0 0.9738 21.45
995.5 0.9973 1.82
972.5 0.9743 20.98
996.0 0.9978 1.47
973.0 0.9748 20.52
996.5 0.9983 1.13
973.5 0.9753 20.05
997.0 0.9988 0.80
974.0 0.9758 19.59
997,5 0.9993 0.46
974.5 0.9763 19.12
998.0 0.9998 0.13
975.0 0.9768 18.66

975.5 0.9773 18.19 Hydrometer method. Transfer 50.0 mL of the preparation to


976.0 0.9778 17.73
be examined, measured at 20 ± 0.1 oC, to the distillation flask,
add 200-300 mL of distilled water R and distil, as described
976.5 0.9783 17.25 aboye, into a volumetric flask until at least 180 mL has been
977.0 0.9788 16.80
collected. Adjust the temperature to 20 ± 0.1 oC and dilute to
250.0 mL with distilled water R at 20 ± 0.1 oc.
977.5 0.9793 16.34
Transfer the distillate to a cylinder whose diameter is at least
978.0 0.9798 15.88 6 mm wider than the bulb of the hydrometer. If the volume is
978.5 0.9803 15.43 insufficient, double the quantity of the sample and dilute the
distillate to 500.0 mL with distilled water R at 20 ± 0.1 oc.
979.0 0.9808 14.97
Multiply the strength by 5 to allow for the dilution during the
979.5 0.9813 14.52
determination. After calculation of the ethanol content using
980.0 0.9818 14.07 Table 2.9.10.-1, round off the result to 1 decimal place.
980.5 0.9823 13.63

981.0 0.9828 13.18 METHOD B


981.5 0.9833 12.74 Head-space gas chromatography (2.2.28).
982.0 0.9838 12.31 Internal standard solution. Dilute 1.0 mL of propanol Rl
982.5 0.9843 11.87
to 100.0 mL with water R. Dilute 1.0 mL of the solution to
20,0 mL with water R.

302 See the information section on general monographs (cover pages)


EUROPEAN PHARMACOPOEIA 8.0 2.9.10. Ethanol content

Test solution. Dilute a volume of the preparation to be METHODC


examined eorresponding to 1 g of ethanol to 50.0 mL with Gas ehromatography (2.2.28).
water R. Dilute 1.0 mL of the solution to 20.0 mL with water R.
To 2.0 mL of this solution add l.0 mL of the internal standard Internal standard solution. Dilute l.0 111L of propano/ Rl to
solution and dilute to 20.0 mL with water R. 100.0 111L with water R.
Referenee solu/ion (a). Dilute 5.0 mL of anhydrous ethanol R Test solution. Dilute a volume of the preparation to be
to 100.0 mL with water R. Dilute 25.0 mL of the solution exa111ined corresponding to 1 g of ethanol to 50.0 mL with
to 100.0 mL with water R. Dilute 1.0 mL of this solution to water R. To 1.0 mL ofthis solution add l.0 mL of the internal
20.0 mL with water R. standard solution and dilute to 20.0 I11L with water R.
Reference so/ution (b). Mix 0.5 mL of referenee solution (a) Reference solution (a). Dilute 1.0 111L of anhydrous ethanol R
and 1.0 mL of the internal standard solution and dilute to to 50.0 mL with water R.
20.0 mL with water R. Reference solution (b). Dilute 1.0 111L of methanol R2 to
Reference so/ution (e). Mix 1.0 mL of referenee solution (a) 100.0 111L with water R. Dilute 1.0 mL of the solution to
and 1.0 mL of the internal standard solution and dilute to 20.0 mL with water R.
20.0 mL with water R. Reference solution (e). Mix 1.0 mL of the internal standard
Referenee so/ution (d). Mix l.5 mL ofreferenee solution (a) solution, 1.0 mL of referenee solution (a) and 2.0 mL of
and 1.0 mL of the internal standard solution and dilute to reference solution (b) and dilute to 20.0 I11L with water R.
20.0 mL with water R. Co/umn:
Referenee so/u/ion (e). Dilute 1.0 mL of methano/ R2 to - material: fused silica;
100.0 mL with water R. Dilute 1.0 mL of the solution to - size: 1 =o 30 m, 0 =o 0.53 mm;
20.0 mL with water R.
- stationary phase: po/y[(cyanopropyl)(phenyl)] [dimeth-
Reference so/ution (j). Mix l.0 mL of the internal standard yl]siloxane R (film thickness 3 !-uu).
solution, 2.0 mL of referenee solution (a) and 2.0 mL of
Carrier gas: helium for chromatography R.
referenee solution (e) and dilute to 20.0 mL with water R.
Flow rate: 3 mLlmin.
Co/umn:
Split ratio: 1:50.
- material: fused silica;
Temperature:
- size: 1 =o 30 lTI, 0 =o 0.53 mm;
Time Temperature
- stationary phase: po/y[(cyanopropy/)(pheny/)] [dimeth-
(min) (oC)
yl]siloxane R (film thiekness 3 ~lm).
Column O - 1.6 40
Carrier gas: helium for chromatography R.
F/ow rate: 3 mL/min. 1.6 - 9.9 40 -7 65

Split ratio: 1:50. 9.9 - 13.6 65 -7175

Static head-space conditions that may be used: 13.6 - 20 175


- equilibration temperature: 85 oC; Injection port 200
- equilibration time: 20 mino Detector 200
Temperature:
DeteeUon: flame ionisation.
Time Temperature
(min) (oC) Injection: 1.0 ¡.tL of the test solutiol1 and referenee solution (e),
O - 1.6 40
at least 3 times.
Column
E/ution order: methanol, ethanol, l-propanol.
1.6 - 9.9 40 -7 65
Relative retention with referenee to ethanol (retention
9.9 - 13.6 65 -7175 time =o about 5.3 min): methanol =o about 0.8;
13.6 - 20 175 l-propanol =o about 1.6.
System suitability: referenee solution (e):
Injection port 200
- resolution: mínimum 5 between the peaks due to methanol
Detector 200 and ethanol.
Detection: flame ionisation. Caiculate the ethanol eontent in per cent V/V using the
following expression:
Injection: 1.0 mL of the gaseous phase of the test solution and
reference solutions (b), (e), (d) and (0, at least 3 times. Al x h x 100
Elution order: methanol, ethanol, l-propanol. A 2 X h X Vl
Relative retention with referenee to ethanol (retention area of the peak due to ethanol in the chromatogram
time =o about 5.3 min): methanol =o about 0.8;
obtained with the test solution;
l-propanol =o about 1.6.
area of the peak due to ethanol in the ehromatogram
System suitability: referenee solution (f): obtained with reference solution (e);
- resolution: minimum 5 between the peaks due to metlianol area of the peak due to the internal standard in the
and ethanol. ehromatogram obtained with the test solution;
Establish a ealibration curve with the coneentration of ethanol are a of the peak dne to the internal standard
in referenee solutions (b), (e), (d) and (f) as the abscissa and 12 in the chromatogram obtained with reference
the mean ratio of the peak area of ethanol to the peak area of solution (e);
the internal standard in the eorresponding ehromatograms as
the ordinate. volume of the preparation to be examined in the
test solution, in millilitres.
Calculate the percentage eontent of ethanol in the preparation
to be examined.

General Notices (1) apply to all monographs and other texts 303
2.9.11. Test far methanol and 2-propanol EUROPEAN PHARMACOPOEIA 8.0

04/2013:20911 Calculate the methanol eontent in per eent V/V using the
following expression:

2.9.11. TEST FOR METHANOL AND


2-PROPANOL
area of the peak due to methanol in the
METHODA ehromatogram obtained with the test solution;
Head-spaee gas ehromatography (2.2.28). area of the peak due to methanol in the
ehromatogram obtained with reference
Internal standard solution. DiIute 1.0 mL of propanol Rl solution (e);
to 100.0 mL with water R. DiIute 1.0 mL of the solution to
area of the peak due to the internal standard in the
20.0 mL with water R.
ehromatogram obtained with the test solution;
Test salution. Mix 1.0 mL of the internal standard solution are a of the peak due to the internal standard
and 4.0 mL of the preparation to be examined and dilute to in the ehromatogram obtained with referenee
20.0 mL with water R. solution (e).
Reference solution (a). Mix 1.0 mL of methanol R2 and 1.0 mL
of 2-propanol R2 and dilute to 100.0 mL with water R. Dilute
1.0 mL of the solution to 20.0 mL with water R. Calculate the 2-propanol eontent in per cent V/V using the
Reference solution (b). DiIute 5.0 mL of anhydrous ethanol R following expression:
to 100.0 mL with water R. Dilute 25.0 mL of the solution
to 100.0 mL with water R. DiIute 1.0 mL of this solution to
20.0 mL with water R.
Referenee solution (e). Mix 1.0 mL of the internal standard area of the peak due to 2-propanol in the
solution, 2.0 mL of referenee solution (a) and 2.0 mL of ehromatogram obtained with the test solution;
referenee solution (b) and dilute to 20.0 mL with water R.
area of the peak due to 2-propanol in the
Column: ehromatogram obtained with referenee
solution (e);
- material: fused silica;
area of the peak due to the internal standard in the
- size: 1 = 30 m, 0 = 0.53 mm; chromatogram obtained with the test solution;
stationary phase: poly[(eyanopropyl)(phenyl)] [dimeth- are a of the peak due to the internal standard
yl]siloxane R (film thiekness 3 [lm). in the ehromatogram obtained with referenee
solution (e).
Carrier gas: helium for chromatography R.
Flow rate: 3 mL/min.
Split ratia: 1:50.
METHODB
Static head-space canditions that may be used:
Gas chromatography (2.2.28).
- equilibratíon tempera tu re: 85 oC;
Internal standard solution. Dilute 1.0 mL of propanol Rl to
- equilibration time: 20 mino 100.0 mL with water R.
Temperature:
Test solutian. Mix 1.0 mL of the internal standard solution
Time Temperature
and 4.0 mL of the preparation to be examined and dilute to
(min) (oC) 20.0 mL with water R.
Column O - 1.6 40 Reference solution (a). Mix 1.0 mL of methanol R2 and 1.0 mL
1.6 - 9.9 40 -7 65 of 2-propanol R2 and dilute to 100.0 mL with water R. Dilute
1.0 mL of the solution to 20.0 mL with water R.
9.9 - 13.6 65 -7 175
Reference solution (b). Dilute 1.0 mL of anhydrous ethanol R
13.6 - 20 175
to 50.0 mL with water R.
Injection port 200
Referenee solution (e). Mix 1.0 mL of the internal standard
Detector 200 solution, 1.0 mL ofreferenee solution (b) and 2.0 mL of
reference solution (a) and dilute to 20.0 mL with water R.
Deteetion: flame ionisation.
Column:
Injection: 1.0 mL of the gaseous phase of the test solution and
referenee solution (e), at least 3 times. - material: fused silica;

Elution order: methanol, ethanol, 2-propanol, 1- propano!. - size: 1 = 30 m, 0 = 0.53 mm;


Relative retention with referenee to ethanol (retention - stationary phase: poly[(cyanopropyl)(phenyl)][dimeth-
time = about 5.3 min): methanol = about 0.8; yl]siloxane R (film thickness 3 [lm).
2-propanol = about 1.2; l-propanol = about 1.6.
Carrier gas: helium far chromatography R.
System suitability: re fe rene e solution (e):
Flow rate: 3 mUmin.
- resolution: minimum 5 between the peaks due to methanol
and ethano!. Split ratio: 1:50.

304 See the information section on general monographs (caver pages)


EUROPEAN PHARMACOPOEIA 8.0 2.9.14. Spedfie surfaee area by air permeability

Temperature: 0112008:20912

Time Temperature
(min) ('C)
2.9.12. SIEVE TEST
Column 0-1.6 40
The degree of flneness of a powder may be expressed by
1.6 - 9.9 40 -¿ 65
reference to sieves that comply with the specifleations for
9.9 - 13.6 65 -¿ 175 non-analytical sieves (2.1.4).
13.6 - 20 175 Where the degree of flneness of powders is determined by
sieving, it is deflned in relation to the sieve number(s) used
Injection port 200
either by means of the following terms or, where such terms
Detector 200 cannot be used, by expressing the finen es s of the powder as a
percentage m/m passing the sieve(s) used.

Detectian: flame ionisation. The following terms are used in the description of powders :
Coarse powder. Not les s than 95 per eent by mass pass es
Injectian: 1.0 I1L ofthe test solution and referenee solution (c), through a number 1400 sieve and not more than 40 per cent
at least 3 times. by mass passes through a number 355 sieve.
Moderately fine powder. Not less than 95 per cent by mas s
Elutian arder: methanol, ethanol, 2-propanol, l-propanol. passes through a number 355 sieve and not more than 40 per
cent by mass pass es through a number 180 sieve.
Relative retentian with referenee to ethanol (retention Fine powder. Not les s than 95 per cent by mass passes
time = about 5.3 min): methanol = about 0.8; through a number 180 sieve and not more than 40 per cent by
2-propanol = about 1.2; l-propanol = about 1.6. mass passes through a number 125 sieve.
Very fine powder. Not less than 95 per cent by mass passes
System suitability: referenee solution (e): through a number 125 sieve and not more than 40 per cent by
mass passes through a number 90 sieve.
- resalutian: minimum 5 between the peaks due to methanol
and ethanol. lf a single sieve number is given, not les s than 97 per cent of
the powder passes through the sieve of that number, unless
otherwise preseribed.
Calculate the methanol eontent in per eent V/V using the
following expression: Assemble the sieves and operate in a suitable manner until
sifting is practically complete. Weigh the separated fractions
of the powder.

area of the peak due to methanol in the


ehromatogram obtained with the test solution;
are a of the peak due to methanol in the
chromatogram obtained with referenee 0112008:20914
solution (e);
II are a of the peak due to the internal standard in the
ehromatogram obtained with the test solution; 2.9.14. SPECIFIC SURFACE AREA BY
I2 area of the peak due to the internal standard AIR PERMEABILITY
in the ehromatogram obtained with referenee
solution (e). The test is intended for the determination of the speeific
surface area of dry powders expressed in square metres per
gram in the sub-sieve region. The effect of molecular flow
("slip flow") which may be important when testing powders
Calculate the 2-propanol eontent in per eent V/V using the eonsisting of partides less than a few mierometres is not taken
following expression: into aceount in the equation used to calculate the specifie
surface area.
A3 X h

APPARATUS

area of the peak due to 2-propanol in the The apparatus eonsists of the following parts:
ehromatogram obtained with the test solution;
(a) a permeability cell (see Figure 2.9.14.-1), which consists
are a of the peak due to 2-propanol in the of a cylinder with an inner di ame ter of 12.6 ± 0.1 mm (A),
ehromatogram obtained with referenee constructed of glass or non-corroding metal. The bottom
solution (e); of the cel! forms an airtight connection (for example, via an
are a of the peak due to the internal standard in the adapter) with the manometer (Figure 2.9.14.-2). A ledge
ehromatogram obtained with the test solution; 0.5 mm to 1 mm in width is located 50 ± 15 mm from the
area of the peak due to the internal standard top of the celI. It is an integral part of the eel! or firmly fixed
in the ehromatogram obtained with reference so as to be airtight. It supports a perforated metal disk (B),
solution (e). construeted of non-corroding metal. The disk has a thickness
of 0.9 ± 0.1 mm and is perforated with thirty to forty holes
1 mm in diameter evenly distributed over this area.

General Notices (1) apply ta all manographs and other texts 305
2.9.14. Specific surface area by air permeability EUROPEAN PHARMACOPOEIA 8.0

016 v bulk volume of the compacted bed of powder,


P density of the substance to be examined in grams
per millilitre,
f porosity of the compacted bed of powder.

(e)

I
012.6±O.1
-~ I
-
A)
I
I
¡
t!)
CD
I
l[)

(B) I '"
. ::.:!:.:. .
1
~ ~::I:::
~

¡
(D)

¡~
Ol
¡\ c::i
I (B)

I o
(!)

I
t!)
0J

Figure 2.9.14.-1. - Permeability cell


Dimensions in millimetres
The plunger (e) is made ofnon-corroding metal and fits into
the ceH with a clearance of not more than 0.1 mm. The bottom
of the plunger has sharp square edges at right angles to the
principal axis. There is an air vent 3 mm long al1d 0.3 mm Figure 2.9.14.-2. - Manometer
deep on one side of the plunger. The top of the plunger has
a collar such that when the plunger is placed in the cell and Dimensions in millimetres
the collar is brought into contact with the top of the ceH, the
Assume first a porosity of 0.5 and introduce this value in Eq. 1
distance between the bottom of the plunger al1d the top of the
to calculate the mass (M) of the powder to be examined.
perforated disk (E) is 15 ± 1 mm.
Place a filter paper disk on top of the perforated metal disk (E).
The filter paper disks (D) have smooth edges and the same Weigh the calculated mass (M) of the powder to be examined
diameter as the inside of the cell. to the nearest 1 mg. Carefully transfer the powder into the
(b) a U-tube manometer (E) (Figure 2.9.14.-2) is made of cleaned, tared permeability cell and careful!y tap the cell so
nominal 9 mm outer diameter and 7 mm inner diameter that the surface of the powder bed is level and (over it with
glass tubing with standard walls. The top of one arm of the a second filter paper disk. Slowly compact the powder by
manometer forms an airtight connection with the permeability means of the plunger, avoiding rotary movement. Mail1tain
cell (F). The manometer arm connected to the permeability the pressure until the plunger is completely inserted into the
cel! has a line etched around the tube at 125 mm to 145 mm permeability cel!o rf this is not possible, decrease the quantity
below the top of the side outlet and three other lines at of the powder used. lf, on the contrary, there is not enough
distances of 15 mm, 70 mm and 110 mm aboye that line (G). resistance, increase the quantity of the powder. In this case
The side outlet 250 mm to 305 mm aboye the bottom of the calculate the porosity again. After at least 10 s, remove the
manometer is used to evacuate the manometer arm connected plunger.
to the permeability cell. A tap is provided on the side outlet Attach the permeability cell to the tube of the manometer by
not more than 50 mm from the manometer armo means of an airtight connection. Evacuate the air from the
The manometer is mounted firmly in such a manner that the manometer by means of a rubber bulb until the level of the
arms are vertical. It is filled to the lowest mark with dibutyl coloured liquid is at the highest mark. Close the tap and check
phthalate R containing a lipophilic dye. that the apparatus is airtight by closing the upper el1d of the
cel!, for example with a rubber stopper. Remove the stopper
and, using a timer, measure the time taken for the liquid to
METHOD fal! from the second to the third mark.
lf prescribed, dry the powder to be examined and sift through Using the measured flow time, calculate the specific surface
a suitable sieve (for example no. 125) to disperse agglomerates. area (5), expressed in square metres per gram, from the
Calculate the mass (M) of the powder to be used from the following expression:
fol!owing express ion :
s= KxJE3xVt (2)
M=Vxpx(l-E) (1) px(l-E)xylri

306 See the information section on general rnonographs (ca ver pages)
EUROPEAN PHARMACOPOEIA 8.0 2.9.16. Flowability

flow time in seconds, The density of mercury and the viscosity of air over a range of
temperatures are shown in Table 2.9.14.-1.
11 dynamic viscosity of air in millipascal seconds (see
Table 2.9.14.-1), Table 2.9.14.-1.
K apparatus constant determined according to
Equation (4), Temperature Density of mercury Viscosity of air ('1) vfTi
(oC) (g/mL) (mPa·s)
P density of the substance to be examined in grams
16 13.56 0.01800 0.1342
per millilitre,
E porosity oí the compacted bed of powder. 17 13.56 0.01805 0.1344

18 13.55 0.01810 0.1345

19 13.55 0.01815 0.1347


CALIBRATION OF THE APPARATUS
20 13.55 0.01819 0.1349
The bulk volume of the compacted bed oí powder is
21 13.54 0.01824 0.1351
determined by the mercury displacement method as follows:
22 13.54 0.01829 0.1353
Place two filter paper disks in the permeability cel!, pressing
down the edges with a rod slightly smaller than the ceH 23 13.54 0.01834 0.1354
diameter until the filter disks lie flat on the perforated metal 24 13.54 0.01839 0.1356
disk; fill the ceH with mercury, removing any air bubbles
adhering to the wall of the ceH and wipe away the excess to
create aplane surface of mercury at the top of the celI. IÍ the
ceH is made of material that will amalgamate, grease the ceH
and the metal disk first with a thin layer of liquid paraffin.
Pour out the mercury into a tared beaker and determine the
mass (MA ) and the temperature ofthe mercury. Ol/2008:20916
Make a compacted bed using the reference powder and
again fill the ceH with mercury with aplanar surface at the
top of the cell. Pour out the mercury in a tared beaker and
2.9.16. FLOWABILITY
again determine the mass of the mercury (MB). Calculate the
bulk volume (V) of the compacted bed of powder from the The test for flowability is intended to determine the ability of
following expression: divided solids (for example, powders and granules) to flow
vertically under defined conditions.

(3)
APPARATUS

difference between the determined masses According to the flow properties of the material to be tested,
of mercury in grams, funnels with or without stem, with different angles and
orifice diameters are used. Typical apparatuses are shown
density of mercury at the determined in Figures 2.9.16.-1 and 2.9.16.-2. The funnel is maintained
temperature in grams per millilitre. upright by a suitable device. The assembly must be protected
from vibrations.
Repeat the procedure twice, changing the powder each time;
the range ofvalues for the calculated volume (V) is not greater
than 0.01 mL. Use the mean value ofthe three determined METHOD
volumes for the calculations.
lnto a dry funnel, whose bottom opening has be en blocked by
The apparatus constant K is determined using a reÍerence suitable means, introduce without compacting a test sample
powder with known specific surface area and density as weighed with 0.5 per cent accuracy. The amount of the sample
follows: depends on the apparent volume and the apparatus used.
Unblock the bottom opening of the funnel and measure the
Calculate the required quantity of the reference powder to time needed for the entire sample to flow out of the funnel.
be used (Eq. 1) using the stated density and the determined Carry out three determinations.
volume of the compacted powder bed (Eq. 3).

Homogenise and loosen up the powder by shaking it for 2 min EXPRESSION OF RESULTS
in a 100 mL bottle. Prepare a compacted powder bed and
measure the flow time of air as previously described. Calculate The flowability is expressed in seconds and tenths of seconds,
the apparatus constant (K) from the following expression: related to 100 g of sample.
The results depend on the storage conditions of the material
K = _Ss_p_x_P-;oX=(c--l_-_E~)_X---,-VTi_1) to be tested.
(4)
Fc3 x Vt The results can be expressed as the following:
stated specific surface are a of the reference powder, a) the mean of the determinations, if none of the individual
values deviates from the mean value by more than 10 per
density of the substance to be examined in grams
cent;
per millilitre,
E porosity of the compacted bed of powder, b) as a range, if the individual values deviate from the mean
value by more than 10 per cent;
flow time in seconds,
c) as a plot of the mass against the flow time;
dynamic viscosity of air in millipascal seconds (see
Table 2.9.14.-1). d) as an infinite time, if the entire sample fails to flow through.

General Notices (1) apply to all monographs and other texts 307
2.9.17. Test for extractable volume ofparenteral preparations EUROPEAN PHARMACOPOEIA 8.0

0110 0412010:20917

2.9.17. TEST FOR EXTRACTABLE


VOLUME OF PARENTERAL
PREPARATIONS(7)
Suspensions and emulsions are shaken before withdrawal of
the contents and before the determination of the density. Oily
and viscous preparations may be warmed according to the
instructions on the labe!, if necessary, and thoroughly shaken
immediately before removing the contents. The contents are
Connecting nut then cooled to 20-25 oC before measuring the volume.

SINGLE-DOSE CONTAINERS
Select 1 container if the nominal volume is 10 mL or more,
3 container s if the nominal volume is more than 3 mL and les s
030.2.g , than 10 mL, or 5 containers if the nominal volume is 3 mL or
less. Take up individually the total contents of each container
Nozzle 1, 2 or 3 selected into a dry syringe of a capacity not exceeding 3 times
enlarged the volume to be measured, and fitted with a 21-gauge needle
not less than 2.5 cm in length. Expel any air bubbles from the
syringe and needle, then discharge the contents of the syringe
without emptying the needle into a standardised dry cylinder
(graduated to contain rather than to deliver the designated
-d volumes) of such size that the volume to be measured occupies
at least 40 per cent of its graduated volume. Alternatively, the
volume of the contents in millilitres may be calculated as the
Nozzle Diameter (d) of the outtlow mass in grams divided by the density.
opening (millimetres)
For container s with a nominal volume of 2 mL or less, the
1 10 ± 0.01 contents of a sufficient number of containers may be pooled to
2 15 ± 0.01 obtain the volume required for the measurement provided that
a separate, dry syringe assembly is used fOI each container.
3 25 ± 0.01 The contents of containers holding 10 mL or more may be
Figure 2.9.16.-1. - Flow funnel and nozzle. Nozzle is made of determined by opening them and emptying the contents
stainless, acid-resistant steel (V4A, CrNi) directly into the graduated cylinder or tared beaker.
The volume is not less than the nominal volume in case of
Dimensions in millimetres containers examined individually, or, in case of containers
with a nominal volume of 2 mL or less, is not less than the sum
of the nominal volumes of the containers taken collectively.

MULTIDOSE CONTAINERS
For injections in multidose containers labelled to yield
a specific number of doses of a stated volume, select one
container and proceed as directed for single-dose containers
using the same number of separate syringe assemblies as the
number of doses specified.
The volume is such that each syringe delivers not les s than
the stated dose.

CARTRIDGES AND PREFILLED SYRINGES


Select 1 container if the nominal volume is 10 mL or more,
3 containers if the nominal volume is more than 3 mL and less
than 10 mL, or 5 containers if the nominal volume is 3 mL
or less. lf necessary, fit the containers with the accessories
required for their use (needle, piston, syringe) and transfer the
entire contents of each container without emptying the needle
into a dry tared beaker by slowly and constantly depressing
the piston. Determine the volume in millilitres calculated as
the mass in grams divided by the density.
1.25_......1- The volume measured for each of the containers is not les s
than the nominal volume.

PARENTERAL INFUSIONS
Select one container. Transfer the contents into a dry
measuring cylinder of such a capacity that the volume to
Figure 2.9.16.-2 be determined occupies at least 40 per cent of the nominal
volume of the cylinder. Measure the volume transferred.
Dimensions in millimetres The volume is not less than the nominal volume.

(7) This chapter has undergone pharmacopoeial hannonisation. See chapter S.S. Pharmacopoeial harmonisation.

308 See the information section on general monographs (caver pages)


EUROPEAN PHARMACOPOEIA 8.0 2.9.18. Preparations for inhalation

0112008:20918 Table 2.9.18.-1. - Component specification for apparatus A


in Figure 2.9.18.-1
Code Item Description Dimen-
2.9.18. PREPARATIONS FOR sions*
INHALATION: AERODYNAMIC A Mouthpiece Moulded ruboer adapter for
adaptor actuator mouthpiece.
ASSESSMENT OF FINE PARTICLES B Throat Modified round-bottomed flask: 50mL

This test is used to determine the fine particle characteristics - ground-glass inlet socket 29/32
of the aerosol douds generated by preparations for inhalation.
- ground-glass outlet cone 24/29
Unless otherwise justified and authorised, one of the following C Neck Modified glass adapter:
apparatus and test procedures is used.
- ground-glass inlet socket 24/29
Stage mensuration is performed periodically together with
confirmation of other dimensions critical to the effective - ground-glass outlet cone 24/29
operation of the impactor. Lower outlet section of
precision-bore glass tubing:
Re-entrainment (for apparatus D and E). To ensure efficient
partide capture, coat each plate with glycerol, silicone oil or - bore diameter 14
similar high viscosity liquid, typically deposited from a volatile Se1ected bore light-wall glass
solvento Plate coating must be part of method validation and tubing:
may be omitted where justified and authorised. - external diameter 17
Mass balance. The total mass of the active substance is not les s D Upper Modified round-bottomed flask 100mL
than 75 per cent and not more than 125 per cent of the average
delivered dose determined during testing for uniformity of impingement - ground-glass inlet socket 24/29
delivered dose. This is not a test of the inhaler but it serves to chamber - ground-glass outlet cone 24/29
ensure that the results are valido
E Couplingtube Medium-wall glass tubing:

APPARATUS A - GLASS IMPINGER - ground-glass cone 14/23

The apparatus is shown in Figure 2.9.18.-1 (see also Bent section and upper vertical
section:
Table 2.9.18.-1).
- external diameter 13

Lower vertical section :


95 - external diameter 8

F Screwthread, Plastic screw cap 28/13

side-arm Silicone rubber ring 28/11

adaptor PTFE washer 28/11


707 Glass screwthread:

- thread size 28

Side-arm outlet to vacuum pump :

- minimum bore diameter 5

G Lower jet Modified polypropylene filler see


assembly holder connected to lower vertical Figure
section of coupling tube by PTFE 2.9.18.-1
tubing.
63
Acetal circular disc with the centres
of four jets arranged on a projected
75 cirele of diameter 5.3 mm with an
integral jet spacer peg: 10
- peg diameter 2

- peg protrusion 2

~G H Lower

impingement
Conical flask

- ground-glass inlet socket


250mL

24/29
chamber
* Dimensions in millimetres, unless otherwise stated.

Procedure for nebulisers


Introduce 7 mL and 30 mL of a suitable solvent into the upper
and lower impingement chambers, respectively.
Connect all the component parts. Ensure that the assembly is
07.85 ± 0.125 vertical and adequately supported and that the jet spacer peg
of the lower jet assembly just touches the bottom of the lower
impingement chamber. Connect a suitable pump fitted with
Figure 2.9.18.-1. - Apparatus A: glass impinger
a filter (of suitable pore size) to the outlet of the apparatus.
Dimensions in millimetres (tolerances ± 1 mm unless otherwise Adjust the air flow through the apparatus, as measured at the
prescribed) inlet to the throat, to 60 ± 5 L/min.

General Notices (1) apply to all monographs and other texts 309
2.9.18. Preparations for inhalation EUROPEAN PHARMACOPOElA 8.0

Introduce the liquid preparation for inhalation into the the discharge sequence. The number of discharges should
reservoir of the nebuliser. Fit the mouthpiece and connect it be minimised and typically would not be greater than 10.
by means of an adapter to the device. Dismantle the apparatus.
Switch on the pump of the apparatus and after 10 s switch Wash the inner surface of the inIet tube to the lower
on the nebuliser. impingement chamber and its outer surface that projects into
the chamber with a suitable solvent, collecting the washings
After 60 s, unless otherwise justified, switch off the nebuliser,
in the lower impingement chamber. Determine the content
wait for about 5 s and then switch off the pump of the
of active substance in this solution. Calculate the amount of
apparatus. Dismantle the apparatus and wash the inner
active substance collected in the lower impingement chamber
surface of the upper impingement chamber collecting the
per discharge and express the results as a percentage of the
washings in a volumetric flask. Wash the inner surface of the
dose stated on the label.
lower impingement chamber collecting the washings in a
second volumetric flask. Finally, wash the filter preceding the
pump and its connections to the lower impingement chamber Fine partide close ancl partide size
and combine the washings with those obtained from the
lower impingement chamber. Determine the amount of active distribution
substance collected in each of the 2 flasks. Express the results
for each of the 2 parts of the apparatus as a percentage of the APPARATUS C - MULTI-STAGE LIQUID IMPINGER
total amount of active substance. The multi-stage liquid impinger consists of impaction stages 1
Procedure for pressurised inhalers (pre-separator), 2, 3 and 4 and an integral filter stage (stage 5),
see Figures 2.9.18.-4/6. An impaction stage comprises an
Place the actuator adapter in position at the end of the throat upper horizontal metal partition wall (B) through which
so that the mouthpiece end of the actuator, when inserted to a a metal inlet jet tube (A) with its impaction plate (D) is
depth of about 10 mm, lines up along the horizontal axis of protruding. A glass cylinder (E) with sampling port (F) forms
the throat and the open end of the actuator, which accepts the the vertical wall of the stage, and a lower horizontal metal
pressurised container, is uppermost and in the same vertical partition wall (G) through which the tube (H) connects to the
plane as the rest of the apparatus. next lower stage. The tube into stage 4 (U) ends in a multi-jet
Introduce 7 mL and 30 mL of a suitable solvent into the upper arrangement. The impaction plate (D) is secured in a metal
and lower impingement chambers, respectively. frame (J) which is fastened by 2 wires (K) to a sleeve (L)
secured on the jet tube. The horizontal face of the coHection
Connect al! the component parts. Ensure that the assembly plate is perpendicular to the axis of the jet tube and centrally
is vertical and adequately supported and that the lower aligned. The upper surface of the impaction plate is slightly
jet -spacer peg of the lower jet assembly just touches the raised above the edge of the metal frame. A recess around the
bottom of the lower impingement chamber. Connect a perimeter of the horizontal partition wall guides the position
suitable pump to the outlet of the apparatus. Adjust the air of the glass cylinder. The glass cylinders are sealed against
flow through the apparatus, as measured at the inlet to the the horizontal partition walls with gaskets (M) and clamped
throat, to 60 ± 5 L/min. together by 6 bolts (N). The sampling ports are sealed by
Prime the metering valve by shaking for 5 s and discharging stoppers. The bottom-side of the lower partition wall of
once to waste; after not less than 5 s, shake and discharge stage 4 has a concentrical protrusion fitted with a rubber
again to waste. Repeat a further 3 times. O-ring (P) which seals against the edge of a filter placed in
the filter holder. The fiIter holder (R) is constructed as a
Shake for about 5 s, switch on the pump to the apparatus basin with a concentrical recess in which a perforated filter
and locate the mouthpiece end of the actuator in the adapter, support (S) is flush-fitted. The filter holder is dimensioned for
discharge once immediately. Remove the assembled inhaler 76 mm diameter filters. The assembly of impaction stages is
from the adapter, shake for not less than 5 s, relocate the clamped onto the filter holder by 2 snap-locks (T). Connect
mouthpiece end of the actuator in the adapter and discharge an induction port (see Figure 2.9.18.-7) onto the stage 1 inlet
again. Repeat the discharge sequence. The number of jet tube of the impinger. A rubber O-ring on the jet tube
discharges should be minimised and typically would not be provides an airtight connection to the induction port. A
greater than 10. After the final discharge wait for not les s than suitable mouthpiece adapter is used to provide an airtight seal
5 s and then switch off the pump. Dismantle the apparatus. between the inhaler and the induction porto The front face
Wash the inner surface of the inlet tube to the lower of the inhaler mouthpiece must be flush with the front face
impingement chamber and its outer surface that projects into of the induction port.
the chamber with a suitable solvent, collecting the washings
in the lower impingement chamber. Determine the content Table 2.9.18.-2. - Component specification for apparatus e in
of active substance in this solution. Calculate the amount of Figures 2.9.18.-4/6
active substance collected in the lower impingement chamber Code* Item Description Dimen-
per discharge and express the results as a percentage of the sions**
dos e stated on the label.
A,H Jet tube Metal tube screwed onto partition see Figure
Procedure for powder inhalers wall sealed by gasket (C), polished 2.9.18.-5
inner surface
Introduce 7 mL and 30 mL of a suitable solvent into the upper
and lower impingement chambers, respectively. B,G Partition Circular metal plate
wall
Connect al! the component parts. Ensure that the assembly is
vertical and adequately supported and that the jet-spacer peg - diameter 120
of the lower jet assembly just touches the bottom of the lower - thickl1ess see Figure
impingement chamber. Without the inhaler in place, connect 2.9.18.-5
a suitable pump to the outlet of the apparatus. Adjust the air
C Gasket e.g. PTFE to fi.t jet
flow through the apparatus, as measured at the inlet to the tube
throat, to 60 ± 5 L/min.
D Impaction Porosity O sintered-glass disk
Prepare the inhaler for use and locate the mouthpiece in the
apparatus by means of a suitable adapter. Switch on the pump plate - diameter see Figure
2.9.18.-5
for 5 s. Switch off the pump and remove the inhaler. Repeat

310 See the information section on general monographs (caver pages)


EUROPEAN PHARMACOPOEIA 8.0 2.9.18. Preparations for inhalation

Q
Code* Item Description Dimen-
sions**

E Glass Plane polished cut glass tube


eylinder

- height, including gasleets 46


Stage 1
- outer diameter 100
(pre-separalor)
- wall thieleness 3.5

- sampling port (F) diameter 18

- stapper in sampling port ISO 24/25


Stage 2
j Metal frame L-profiled circular frame with slit

- inner diameter to fit


impaction
plate

- height 4

- thieleness af horizontal sectian 0.5 Slage 3

- thieleness of vertical sectian 2

K Wire Steel wire interconneeting metal


frame and sleeve (2 for each frame)

- diameler 1 Slage 4

L Sleeve Metal sleeve seeured on jet tube


by serew
Slage 5 (filler)
- inner diameter to fit jet
tube
- height 6 Oulle!
- thieleness 5
-R
M Gasket e.g. silicone to fit glass
eylinder

N Boll Metal bolt with nut (6 pairs)


- length 205 Figure 2.9.18.-4. - Apparatus C: multi-stage liquid impinger
- diameter 4 Procedllre for pressurised inhalers
Dispense 20 mL of a solvent, capable of dissolving the
P O-ring Rubber O-ring
active substance into each of stages 1 to 4 and replace the
- diameter x thic1cness 66.34x 2.62 stoppers. Tilt the apparatus to wet the stoppers, thereby
neutralising electrostatic charge. Place a suitable filter capable
Q O-ring Rubber O-ring
of quantitatively collecting the active substance in stage 5
- diameter x thieleness 29.1 x 1.6 and assemble the apparatus. Place a suitable mouthpiece
adapter in position at the end of the induction port so that
R Filter holder Metal housing with stand and see Figure
outle! 2.9.18.-6 the mouthpiece end of the actuator, when inserted, lines up
along the horizontal axis of the induction port and the inhaler
S Filter Perforated sheet metal is positioned in the same orientation as intended for use.
support Connect a suitable vacuum pump to the outlet of the apparatus
- diameter 65 and adjust the air flow through the apparatus, as measured at
the inlet to the induction port, to 30 L/min (± 5 per cent).
- hale diameter 3 Switch off the pump.
- distance bctween hales 4 Unless otherwise prescribed in the patient instructions, shake
(centre-paints) the inhaler for 5 s and discharge 1 delivery to waste. Switch
T Snap-Ioeks on the pump to the apparatus, locate the mouthpiece end of
the actuator in the adapter and discharge the inhaler into
U Mullí-jet jet tube (H) ending in mulli-jet see inserts the apparatus, depressing the valve for a sufficient time to
tube arrangement. Figure
2.9.18.-5
ensure complete discharge. Wait for 5 s before removing the
assembled inhaler from the adapter. Repeat the procedure.
* Refers to Figure 2.9.18.-4. The number of discharges should be minimised and typically
** Measures in millimetres with toleranees according to iso 2768-m would not be greater than 10. The number of discharges is
unless otherwise stated. sufficient to ensure an accurate and precise determination of
the fine particle dose. After the final discharge, wait for 5 s
and then switch off the pump.
Dismantle the filter stage of the apparatus. Carefully remove
the filter and extract the active substance into an aliquot of the
solvent. Remove the induction port and mouthpiece adapter
from the apparatus and extract the active substance into an
aliquot of the solvent. If necessary, rinse the inside of the inlet
jet tube to stage 1 with solvent, allowing the solvent to flow
into the stage. Extract the active substance from the inner

General Notices (1) apply to all monographs and other texts 311
2.9.18. Preparations for inhalation EUROPEAN PHARMACOPOEIA 8.0

stage center

~I
s

6x)
v H

4 L
K

multí-jet U only

Figure 2.9.18.-5. - Apparatus C: details ofjet tube and impaction plate. Inserts show end of multi-jet tube U leading to stage 4.
(Numbers and lowercase letters refer to Table 2.9.18.-3 and uppercase letters refer to Figure 2.9.18.-4).

walls and the coHection plate of each of the 4 upper stages Type Code (2 ) Stage 1 Stage 2 Stage 3 Stage 4 Filter
of the apparatus into the solution in the respective stage by (stage 5)
carefully tilting and rotating the apparatus, observing that no Radius l41 r 16 22 27 28.5 O
liquid transfer occurs between the stages. s n.a.
Radius 46 46 46 46
Using a suitable method of analysis, determine the quantity of n.a.
Radius t 50 50 50 50
active substance contained in each of the aliquots of solvento
Angle w 10" 53" 53" 53° 53°
Calculate the fine particle dose (see Calculations).
Angle u n.a. n.a. n.a. 45° n.a.
Table 2.9.18.-3. - Dimensions(J) ofjet tube with impaction plate
Angle v n.a. n.a. n.a. 60° n.a.
of apparatus C
Type Code (2) Stage 1 Stage 2 Stage 3 Stage 4 Filter (1) Measures in millimetres with toleran ces according to ISO 2768-m
unless otherwise stated
(stage 5)
(2) Refer to Figure 2.9.18.-5
Distance 1 9.5 5.5 4.0 6.0 n.a.
(3) Including gasket
(-.0+.5) (-.0+.5) (-.0+.5) (-.0+.5)
(4) Relative centreline of stage compartment
Distance 2 26 31 33 30.5 o
n.a. = not applicable
Distance 3 8 5 5 5 5
<________ • ___ l'!_ªL ______ _
Distance 4 3 3 3 3 n.a.
060
Distance 5 o 3 3 3 3

Distance 6 (31 20 25 25 25 25

Distance 7 n.a. n.a. n.a. 8.5 n.a.

Diameter e 25 14 8.0 21 14
(± .1)
Diameter d 50 30 20 30 n.a.

Diameter e 27.9 16.5 10.5 23.9 n.a.

Diameter f 31.75 22 14 31 22
(-.0+.5)
Diameter g 25.4 21 l3 30 21

Diameter h n.a. n.a. n.a. 2.70 n.a.


(± .5) Figure 2.9.18.-6. - Apparatus C: details of the fi/ter stage
Diameter j n.a. n.a. n.a. 6.3 n.a. (stage 5). Numbers refer to dimensions (0 = diameter).
Uppercase letters refer to Table 2.9.18.-2.
Diameter k n.a. n.a. n.a. 12.6 n.a.
Dimensions in millimetres unless otherwise stated

312 See the information section on general monographs (ca ver pages)
EUROPEAN PHARMACOPOEIA 8.0 2.9.18. Preparations fal" inhalatian

Drill, counter-bore and tap


for an M-4 cap screw.
19.0
Note: use minimum clearance
for screw in the lower part to aid
in precise alignment.

(;J
oo
1+
->.
o

Do not break edge

Joint must be leak tight

~
M-4 socket /
head cap screw

Isometric view of induction part

1. Material may be aluminium, stainless steel or other suitable material.


2. Machine from 38 mm bar stock.
3. Bore 19 mm hole through bar.
4. Cut tube lo exact 45° as shown.
5. The inner bores and lapers should be smooth - surface roughness Ra approx. 0.4 fim.
6. Mili joining cads of stock to provide a liquid tight leak-free sea!.
7. Set up a holding fixture for aligning the inner 19 mm bore and for drilling and tapping M4 x 0.7 threads. There must be virtually no mismatch of
the inner bores in the miter joint.

Figure 2.9.18.-7. - Induction port


Dimensions in millimefres unless otherwise stated

Procedure for powdel" inhalers Adjust the flow control valve to achieve steady flow through
Place a suitable low resistance filter capable of quantitatively the system at the required rate, Qout (± 5 per cent). Switch off
collecting the active substance in stage 5 and assemble the the pump. Ensure that critical flow occurs in the flow control
apparatus. Connect the apparatus to a flow system according valve by the following procedure.
lo the scheme specified in Figure 2.9.18.-8 and Table 2.9.18.-4. With the inhaler in place and the test flow rate established,
Unless otherwise defined, conduct the test at the flow rate, measure the absolute pressure on both sides of the control
Qout' used in the test for uniformity of delivered close, drawing valve (pressure reading points P2 and P3 in Figure 2.9.18.-8).
4 L of air from the mouthpiece of the inhaler and through A ratio P3/P2 ofless than or equal to 0.5 indicates critical flow.
the apparatus. Switch to a more powerful pump and re-measure the test flow
Connect a flowmeter to the induction port. Use a flowmeter rate if critical flow is not indicated.
calibrated for the volumetric flow leaving the meter, or Dispense 20 mL of a solvent, capable of dissolving the active
calculate the volumetric flow leaving the meter (Qout) using substance into each of the 4 upper stages of the apparatus and
the ideal gas law. For a meter calibrated for the entering replace the stoppers. Tilt the apparatus to wet the stoppers,
volumetric flow (Qin)' use the following expression: thereby neutralising electrostatic charge. Place a suitable
_ Qin X Po mouthpiece adapter in position at the end of the induction
Q port.
out - Po _ /::"P

Po atmospheric pressure,
!1P pressure drop over the meter.

General Notices (1) apply to all monographs and other texts 313
2.9.18. Preparations for inhalation EUROPEAN PHARMACOPOElA 8.0

APPARATUS D - ANDERSEN CASCAD E IMPACTOR


E The Andersen 1 ACFM non-viable cascade impactor
consists of 8 stages together with a final filter. Material of
construction may be aluminium, stainless steel or other
D P3 P2 Impactar
suitable material. The stages are clamped together and sealed
with 0- rings. Critical dimensions applied by the manufacturer
of apparatus D are provided in Table 2.9.18.-5. In use, sorne
Vacuum
pump occlusion and wear of holes will occur. In-use mensuration

I
Flow
toleran ces need to be justified. In the configuration used
Control for pressurised inhalers (Figure 2.9.18.-9) the entry cone
Valve of the impactor is connected to an induction port (see
Figure 2.9.18.-7). A suitable mouthpiece adapter is used to
F VacuumTubing provide an airtight seal between the inhaler and the induction
Connector port. The front face of the inhaler mouthpiece must be flush
A B with the front face of the induction port.
In the configuration for powder inhalers, a pre-separator
Figure 2.9.18.-8. - Experimental set-up for testing powder is placed aboye the top stage to collect large masses of
inhalers non-respirable powder. It is connected to the induction port
as shown in Figure 2.9.18.-10. To accommodate high flow
rates through the impactor, the outlet nipple, used to connect
Table 2.9.18.-4. - Component specificationfor Figure 2.9.18.-8
the impactor to the vacuum system is enlarged to have an
Code Item Description internal di ame ter of greater than or equal to 8 mm.
A Connector ID 2: 8 mm. e.g .. short metal coupling, with Table 2.9.18.-5. - Critical dimensions for apparatus D
low-diameter branch to P3. Description Number Dimension (mm)
B Vacuum tubing A length of suitable tubing having an ID 2: 8 mm
and an internal volume of 25 + 5 mL. Stage O nozzle diameter 96 2.55 ± 0.025
C 2-way solenoid A 2-way, 2-port solenoid valve having a Stage 1 nozzle diameter 96 1.89 ± 0.025
valve minimum airflow resistance orifice with
ID 2: 8 mm and an opening time $ 100 ms. Stage 2 nozzle diameter 400 0.914 ± 0.0127
(e.g. type 256-A08, Bürkert GmbH,
D-74653 Ingelfingen), or equivalent. Stage 3 nozzle diameter 400 0.711 ± 0.0127
D Vacuumpump Pump must be capable of drawing the required
Stage 4 nozzle diameter 400 0.533 ± 0.0127
flow rate through the assembled apparatus
with the powder inhaler in the mouthpiece Stage 5 nozzle diameter 400 0.343 ± 0.0127
adapter (e.g. product type 1023, 1423 or 2565,
Gast Manufacturing Ine., Benton Harbor, MI Stage 6 nozzle diameter 400 0.254 ± 0.0127
49022), or equivalent. Connect the pump to the
2-way solenoid valve using short and/or wide Stage 7 nozzle diameter 201 0.254 ± 0.0127
(ID 2: 10 mm) vacuum tubing and connectors
to minimise pump capacity requirements.
Procedure for pressurised inhalers
E Timer Timer capable to drive the 2-way solenoid
valve for the required duration (e.g. type Assemble the Andersen impactor with a suitable filter in
G814, RS Components International, Corby, place. Ensure that the system is airtight. In that respect, follow
NN17 9RS, UK), or equivalent. the manufacturer's instructions. Place a suitable mouthpiece
P2 P3 Pressure Determine under steady-state flow condition adapter in position at the end of the induction port so that the
measurements with an absolute pressure transducer. mouthpiece end of the actuator, when inserted, lines up along
F Flow control Adjustable regulating valve with maximum the horizontal axis of the induction port and the inhaler unit
valve C,2: 1, (e.g. type 8FV12LNSS, Parker Hannifin is positioned in the same orientation as the intended use.
plc., Barnstaple, EX311NP, UK), or equivalent.
Connect a suitable pump to the oudet of the apparatus and
adjust the air flow through the apparatus, as measured at
Prepare the powder inhaler for use according to patient
the inlet to the induction port, to 28.3 L/min (± 5 per cent).
instructions. With the pump running and the 2-way solenoid
Switch off the pump.
valve closed, locate the mouthpiece of the inhaler in the
mouthpiece adapter. Discharge the powder into the apparatus Unless otherwise prescribed in the patient instructions, shake
by opening the valve for the required time, T (± 5 per cent). the inhaler for 5 s and discharge one delivery to waste. Switch
Repeat the procedure. The number of discharges should be on the pump to the apparatus, locate the mouthpiece end of
minimised and typically would not be greater than 10. The the actuator in the adapter and discharge the inverted inhaler
number of discharges is sufficient to ensure an accurate and into the apparatus, depressing the valve for a sufficient time to
precise determination of fine particle do se. ensure complete discharge. Wait for 5 s before removing the
assembled inhaler from the adapter. Repeat the procedure.
Dismantle the filter stage of the apparatus. Carefully remove The number of discharges should be minimised and typically
the filter and extract the active substance into an aliquot of the would not be greater than 10. The number of discharges is
solvent. Remove the induction port and mouthpiece adapter sufficient to ensure an accurate and precise determination of
from the apparatus and extract the active substance into an the fine particle dose. After the final discharge, wait for 5 s
aliquot of the solvent. rf necessary, rinse the inside of the inlet and then switch off the pump.
jet tube to stage 1 with solvent, allowing the solvent to flow Dismantle the apparatus. Carefully remove the filter and
into the stage. Extract the active substance from the inner extract the active substance into an aliquot of the solvent.
walls and the collection plate of each of the 4 upper stages Remove the induction port and mouthpiece adapter from the
of the apparatus into the solution in the respective stage by apparatus and extract the active substance into an aliquot of
carefully tilting and rotating the apparatus, observing that no the solvent. Extract the active substance from the inner walls
liquid transfer occurs between the stages. and the coHection plate of each of the stages of the apparatus
into aliquots of solvent.
Using a suitable method of analysis, determine the amount of
Using a suitable method of analysis, determine the quantity of
active substance contained in each of the aliquots of solvent.
active substance contained in each of the aliquots of solvent.
Calculate the fine particle dose (see Calculations). Ca1culate the fine partic!e dose (see Calculations).

314 See the information section on general monographs (cover pages)


EUROPEAN PHARMACOPOEIA 8.0 2.9.18. Preparations for inhalation

Figure 2.9.18.-9. - Apparatus D: Andersen cascade impactor used for pressurised inhalers

Procedure for powder inhalers may be omitted, where justified and authorised. Stages 6
and 7 may also be omitted at high flow rates, if justified. The
The aerodynamic cut-off diameters of the individual stages of pre-separator may be coated in the same way as the pIates
this apparatus are currently not well-established at flow rates or may contain 10 mL of a suitable soIvent. Connect the
other than 28.3 L/min. Users must justify and validate the use apparatus to a flow system according to the scheme specified
of the impactor in the chosen conditions, when flow rates in Figure 2.9.18.-8 and TabIe 2.9.18.-4.
different from 28.3 L/min are selected.
UnIess otherwise defined, conduct the test at the flow rate,
Assemble the Andersen impactor with the pre-separator and a Qout' used in the test for uniformity of delivered dose drawing
suitable filter in place and ensure that the system is airtight. 4 L of air fram the mouthpiece of the inhaler and through
Depending on the product characteristics, the pre-separator the apparatus.

General Notices (1) apply to all monographs and other texts 315
2.9.18. Preparations for i.nhalation EUROPEAN PHARMACOPOEIA 8.0

44 :n01
38.3+ 0.2 ,
19 -O~
Cross-section
15 ~I
14.4~ R 12. 7 -----..!
13 .
S -----!
-------, IJIr<--- R15.87=8?05
Ir/1+1--- R 14.4
Top view Groove for O-ring
R12.7 R19
R44

Ri5.S7 ~g?05

R38.3 ~~.2
RS
R6.70±0.03 107 R6.70 ± 0.03
Do not break edge /106
45°± 3 ° V ---102
~100
~ 94
Material: aluminium, stainless steel
or other suitable material.
Except where noted, all edges to be broken
and burrs removed.

27 Surface to be clean machine - tooled finish.


1','.', ¡..,..,..",.....",.-,,-, /_22 Inferior bore surface roughness Ra
_14 approx. 0.4 fJm.
o O-ring: nominal dimensions: ID 29 mm,
'\----- OD 32 mm, width 1.8 mm.
~ Do not break edge
Figure 2.9.18.-10. - Cannectian af the inductian part ta the preseparatar af the Andersen cascade impactar
Dimensians in millimetres unless atherwise stated

Connect a flowmeter to the induction port. Use a flowmeter into an aliquot of the solvent. Extract the active substance
calibrated for the volumetric flow leaving the meter, or from the inner walls and the collection plate of each of the
calculate the volumetric flow leaving the meter (Qout) using stages of the apparatus into aliquots of solvent.
the ideal gas law. For a meter calibrated for the entering Using a suitable method of analysis, determine the quantity of
volumetric flow (Qin)' use the following expression: active substance contained in each of the aliquots of solvent.
Qin X Po Calculate the fine particle dose (see Calculations).
Q011.t = Po - !:1P
APPARATUS E
Po atmospheric pressure,
Apparatus E is a cascade impactor with 7 stages and a
l,.P pressure drop over the meter. micro-orifice collector (MOC). Over the flow rate range of
30 L/min to 100 L/min the 50 per cent-efficiency cut-off
Adjust the flow control valve to achieve steady flow through diameters (D so values) range between 0.24 11m to 11.7 11m,
the system at the required rate, Qout (± 5 per cent). Ensure that evenly spaced on a logarithmic scale. In this flow range, there
critical flow occurs in the flow control valve by the procedure are always at least 5 stages with Dso values between 0.5 11m
described for Apparatus C. Switch off the pump. and 6.5 flm. The collection efficiency curves for each stage are
Prepare the powder inhaler for use according to the patient sharp and minimise overlap between stages.
instructions. With the pump running and the 2-way solenoid
Material of construction may be aluminium, stainless steel or
valve closed, locate the mouthpiece of the inhaler in the
other suitable material.
mouthpiece adapter. Discharge the powder into the apparatus
by opening the valve for the required time, T (± 5 per cent). The impactor configuration has removable impaction cups
Repeat the discharge sequence. The number of discharges with al! the cups in one plane (Figures 2.9.18.-11/14). There
should be minimised and typically would not be greater are 3 main sections to the impactor; the bottom frame
than 10. The number of discharges is sufficient to ensure an that holds the impaction cups, the seal body that holds the
accurate and precise determination of fine particle dose. jets and the lid that contains the interstage passageways
Dismantle the apparatus. Carefully remove the filter and (Figures 2.9.18.-11/12). Multiple nozzles are used at all but
extract the active substance into an aliquot of the solvento the first stage (Figure 2.9.18.-l3). The flow passes through the
Remove the pre-separator, induction port and mouthpiece impactor in a saw-tooth pattern.
adapter from the apparatus and extract the active substance Critical dimensions are provided in Table 2.9.18.-6.

316 See the informatian sectian on general managraphs (ca ver pages)
EUROPEAN PHARMACOPOEIA 8.0 2.9.18. Preparatiolls for inhalation

Induction por!

Pre-separator

/ Impactor body

Airflow outlet

Clamping mechanism

Figure 2.9.18.-11. - Apparatus E (shown with the pre-separator in place)


Table 2.9.18.-6. - Critical dimensions for apparatus E In routine operation, the seal body and lid are held together as
Description Dhnension
a single assembly. The impaction cups are accessible when this
(mm)
assembly is opened at the end of an inhaler test. The cups are
held in a support tray, so that all cups can be removed froID
Pre-separator (dimension a - see Figure 2.9.18.-15) 12.8 ± 0.05
the impactor simultaneously by Iifting out the tray.
Stage 1* Nozzle diameter 14.3 ± 0.05 An induction port with internal dimensions (relevant to
Stage 2* Nozzle diameter 4.88 ± 0.04 the airflow path) defined in Figure 2.9.18.-7 connects to the
impactor inlet. A pre-separator can be added when required,
Stage 3* Nozzle diameter 2.185 ± 0.02 typically with powder inhalers, and connects between the
Stage 4* Nozzle diameter 1.207 ± 0.01 induction port and the impactor. A suitable mouthpiece
adapter is used to provide an airtight seal between the inhaler
Stage 5* Nozzle diameter 0.608 ± 0.01 and the induction porto
Stage 6* Nozzle diameter 0.323 ± 0.01 Apparatus E contains a terminal Micro-Orifice
Collector (MOC) that for most formulations will eliminate
Stage 7* Nozzle diameter 0.206 ± 0.01 the need for a final filter as determined by method validation.
MOC* approx. 0.070 The MOC is an impactor plate with nominally 4032 holes,
each approximately 70 11m in diameter. Most partides not
Cup depth (dimension b see Figure 2.9.18.-14) 14.625 ± 0.10 captured on stage 7 of the impactor will be captured on the cup
Collection cup surface roughness (Ra) 0.5 - 2 flm surface below the MOC. For impactors operated at 60 L/min,
the MOC is capable of collecting 80 per cent of 0.14 11m
Stage 1 nozzle to seal body distance** - dimension c O ± !.l8 partides. For formulations with a significant fraction of
Stage 2 nozzle to seal body distance" - dimension e 5.236 ± 0.736 partides not captured by the MOC, there is an optional filter
holder that can replace the MOC or be placed downstream of
Stage 3 nozzle to seal body distance** - dimension e 8.445 ± 0.410 the MOC (a glass fibre filter is suitable).
Stage 4 nozzle to se al body distance** - dimension c 11.379 ± 0.237 Procedure fol' pressul'ised inhalel's
Stage 5 nozzle to seal body distance** - dimension e 13.176 ± 0.341 Place cups into the apertures in the cup tray. Insert the cup
tray into the bottom frame, and lower into place. Close the
Stage 6 nozzle to seal body distance** - dimension c 13.999 ± 0.071 impactor lid with the seal body attached and operate the
Stage 7 nozzle to seal body distance** - dimension e 14.000 ± 0.071 han dIe to lock the impactor together so that the system is
airtight.
MOC nozzle to seal body distance" - dimension c 14.429 to 14.571
Connect an induction port with internal dimensions defined
* See Figure 2.9.18.-13 in Figure 2.9.18.-7 to the impactor inlet. Place a suitable
** See Figure 2.9.18.-14 mouthpiece adapter in position at the end of the induction
port so that the mouthpiece end of the actuator, when
inserted, lines up along the horizontal axis of the induction
port. The front face of the inhaler mouthpiece must be flush
with the front face of the induction port. When attached to
the mouthpiece adapter, the inhaler is positioned in the same

General Notices (1) apply to all monographs and other texts 317
2.9.18. Preparations for inhalation EUROPEAN PHARMACOPOEIA 8.0

Stage 1 nozzle Interstage passageway

Micro-orifice
collector (MOC)

Removable
impaction cups

+---- Lid with seal


body attached

Location pin

- - - - - Location pin recess

Boltom frame with


cup tray in place

Figure 2.9.18.-12. - Apparatus E showing component parts


orientation as intended for use. Connect a suitable pump to Calculate the fine particle dose (see Calculations).
the outlet of the apparatus and adjust the air flow through the Procedure for powder inhalers
apparatus, as measured at the inlet to the induction port, to
30 L/min (± 5 per cent). Switch off the pump. Assemble the apparatus with the pre-separator
Unless otherwise prescribed in the patient instructions, shake (Figure 2.9.18.-15). Depending on the product characteristics,
the inhaler for 5 s and discharge 1 delivery to waste. Switch the pre-separator may be omitted, where justified.
on the pump to the apparatus. Prepare the inhaler for use Place cups into the apertures in the cup tray. Insert the cup
according to the patient instructions, 10cate the mouthpiece tray into the bottom frame, and lower into place. Close the
end of the actuator in the adapter and discharge the inhaler impactor lid with the seal body attached and operate the
¡nto the apparatus, depressing the valve for a sufficient time handle to lock the impactor together so that the system is
to ensure a complete discharge. Wait for 5 s before removing airtight.
the assemb1ed inhaler from the adapter. Repeat the procedure.
The number of discharges should be minimised, and typically When used, the pre-separator should be assembled as follows:
would not be greater than 10. The number of discharges is assemble the pre-separator insert into the pre-separator base.
sufficient to ensure an accurate and precise determination of Fit the pre-separator base to the impactor inlet. Add 15 mL
the fine particle dose. After the final discharge, wait for 5 s of the solvent used for sample recovery to the central cup of
and then switch off the pump. the pre-separator inserto Place the pre-separator body on top
Dismantle the apparatus and recover the active substance as of this assembly and close the 2 catches.
follows: remove the induction port and mouthpiece adapter Connect an induction port with internal dimensions defined
from the apparatus and recover the deposíted active substance in Figure 2.9.18.-7 to the impactor inlet or pre-separator
into an aliquot of solvent. Open the impactor by releasing inlet. Place a suitable mouthpiece adapter in position at the
the handle and lifting the lid. Remove the cup tray, wíth the end of the induction port so that the mouthpiece end of the
collection cups, and recover the active substance in each cup inhaler, when inserted, lines up along the horizontal axis of
into an aliquot of solvent. the induction port. The front face of the inhaler mouthpiece
Using a suitable method of analysis, determine the quantity of must be flush with the front face of the induction port. When
active substance contained in each of the aliquots of solvent. attached to the mouthpiece adapter, the inhaler is positioned
Stage 2 Stage 4 Stage 6 MOC
6 hales 52 hales 396 holes 4032 holes

Stage 1 Stage 3 Stage 5 Stage 7


1 hole 24 hales 152 hales 630 holes
Figure 2.9.18.-13. - Apparatus E: nozzle conÍiguration

318 See the inÍormation section on general monographs (cover pages)


EUROPEAN PHARMACOPOEIA 8.0 2.9.18. Pl'epal'ations fol' inhalation

Interstage passage Interstage passage


( to next stage ( from previous stage

I / ,r- Lid
t t
1 , - Seal body
J

J~ iDI~~I~~pl~
,-JL-.::
L
------.. \ .,.- Cup tray
'( J>
~
Cup tray /"Bottom
, frame
),

Collection cup . - / Multi-nozzle stage./

Figure 2.9.18.-14. - Apparatus E: configuration of interstage passageways

Pre-separator body

/
/
Catch
- _ Nozzle diameter,
dimension a

Central cup

t
o = o

t
Pre-separator insert

Figure 2.9.18.-15. - Apparatus E: pre-separator configuration

in the same orientation as intended for use. Connect the Prepare the powder inhaler for use according to the patient
apparatus to a flow system according to the scheme specified instructions. With the pump running and the 2-way solenoid
in Figure 2.9.18.-8 and TabIe 2.9.18.-4. valve closed, locate the mouthpiece of the inhaler in the
mouthpiece adapter. Discharge the powder into the apparatus
Unless otherwise prescribed, conduct the test at the flow rate, by opening the valve for the required time, T (± 5 per cent).
Qout' used in the test for uniformity of delivered dose drawing Repeat the discharge sequence. The number of discharges
4 L of air from the mouthpiece of the inhaler and through should be minimised and typically would not be greater
the apparatus. Connect a flowmeter to the induction port. than 10. The number of discharges is sufficient to ensure an
Use a flowmeter calibrated for the voIumetric flow Ieaving the accurate and precise determination of fine particle dose.
meter, or calculate the volumetric flow leaving the meter (Qout)
using the ideal gas law. For a meter calibrated for the entering Dismantle the apparatus and recover the active substance as
volumetric flow (Q¡n)' use the following expression: follows: remove the induction port and mouthpiece adapter
from the pre-separator, when used, and recover the deposited
Q _ Qin X Po active substance into an aliquot of solvent. When used,
out - Po - 6.P remove the pre-separator from the impactor, being careful to
avoid spilling the cup liquid into the impactor. Recover the
Po atmospheric pressure, active substance from the pre-separator.
Open the impactor by releasing the han dIe and lifting the lid .
.6.P pressure drop over the meter. Remove the cup tray, with the collection cups, and recover the
active substance in each cup into an aliquot of solvent.
Adjust the flow control valve to achieve steady flow through
the system at the required rate, Qout (± 5 per cent). Ensure that Using a suitable method of analysis, determine the quantity of
critical flow occurs in the flow control valve by the pro ce dure active substance contained in each of the aliquots of solvent.
described for Apparatus C. Switch off the pump. Calculate the fine particle dose (see Calculations).

General Notices (1) apply to all monographs and other texts 319
2.9.18. Preparations for inhalation EUROPEAN PHARMACOPOEIA 8.0

CALCULATIONS for Apparatus D, 2.9.18.-9 for Apparatus E). Calculate by


interpolation the mass of the active substance less than 5 fIm.
From the analysis of the solutions, calculate the mass of active This is the Fine Particle Dose (FPD).
substance deposited on each stage per discharge and the mass lf necessary, and where appropriate (e.g., where there is a
of active substance per discharge deposited in the induction log-normal distribution), plot the cumulative fraction of active
port, mouthpiece adapter and when used, the pre-separator. substance versus cut-off diameter (see Tables 2.9.18.-7/9) on
log probability paper, and use this plot to determine values
Starting at the final coHection site (filter or MOC), derive for the Mass Median Aerodynamic Diameter (MMAD)
atable of cumulative mass versus cut-off diameter of the and Geometric Standard Deviation (GSD) as appropriate.
respective stage (see Tables 2.9.18.-7 for Apparatus C, 2.9.18.-8 Appropriate computational methods may also be used.

vi
Table 2.9.18.-7. - Calculations for Apparatus C. Use q = (60/Q), where Q is the test flow rate in litres per minute (Qou,!or
powder inhalers)

Cut-off diameter Mass of active substance deposited Cumulative mass oí active substance Cumulative fraction of active substance
(flm) per discharge deposited per discharge (per cent)
d 4 = 1.7 x q mass from stage 5, m 5 * c4 ::: lTIs
f l = (e,/e) x 100

d 3 = 3.1 x q mass from stage 4, m 4 c) ::::: c 4 + ll14 fl = (e¡le) x 100

d, = 6.8 x q mass from stage 3, m 3 e2 ::::: c3 + ID) f, = (c/c) x 100

mass from stage 2, m, e::: e2 + In] 100

* Stage 5 is the filter stage

Table 2.9.18.-8. - Calculations for Apparatus D when used at a flow rate of 28.3 L/min

Cut-off diameter Mass of active substance deposited Cl1m111ative mass of active sl1bstance Cumulative fraetion of active
(flm) per diseharge deposited per discharge sl1bstance (per cent)
d7 = 0.4 mass from stage 8, ms f7 = (c/c) x 100

d6 = 0.7 mass from stage 7, m 7 f6 = (e 6 /c) x 100

d 5 = l.l mass from stage 6, m 6 f5 = (c/c) x 100


d4 = 2.1 mass from stage 5, ms f4 = (c,le) x 100
d 3 = 3.3 mass from stage 4, m, [3 = (e 3 /e) x 100

d, = 4.7 mass from stage 3, m l f, = (e,le) x 100

di = 5.8 mass from stage 2, m, fl = (e¡le) x 100

do = 9.0 mass from stage 1, mi fc, = (eo/e) x 100

mass from stage O, mo 100

Table 2.9.18.-9. - Calculations for Apparatus E. Use q = (60IQY, where Q is the test flow rate in litres per minute, and x
is listed in the table

Cut -off diameter x Mass of active substance Cumulative mass of active substance Cumulative fraction of active
(flm) deposited per discharge deposited per discharge substance (per cent)
d, = 0.34 x q 0.67 mass from MOC or terminal e7 :::: ln s F7 = (e 7 /e) x 100
filter, m,
d 6 = 0.55 x q 0.60 mass from stage 7, m, c6 :::: e7 + ID7 F6 = (e 6 /e) x 100

d, = 0.94 x q 0.53 mass from stage 6, m 6 cS=cfi+m ó F, = (es/e) x 100

d, = 1.66 x q 0.47 mass from stage 5, m 5 c 4 = es + ffis F, = (e)e) x 100

d 3 = 2.82 x q 0.50 mass from stage 4, m, c3 :::: c4 + ln~ Fl = (c/c) x 100

d, = 4.46 x q 0.52 mass from stage 3, m l e2 ::::: c] + ID3 F, = (e,le) x 100

di = 8.06 x q 0.54 mass from stage 2, m, el = e 2 + ffi 2 FI = (e¡le) x 100

mas s from stage 1, mi e:::: el + In, 100

320 See the information section on general monographs (cover pages)


EUROPEAN PHARMACOPOEIA 8.0 2.9.19. Partkulate contamination: sub-visible partídes

04/2011:20919 for the test are not sufficient. The preparatory steps must
be repeated until the environment, glassware and water are
suitable for the test.
2.9.19. PARTICULATE
Method
CONTAMINATION: SUB-VISIBLE
Mix the contents of the sample by slowly inverting the
PARTICLES(8) container 20 times successively. If necessary, cautiously
Particulate contamination of injections and infusions consists remove the sealing closure. Clean the outer surfaces of the
of extraneous, mobile undissolved partides, other than gas container opening using a jet of particle-free water R and
bubbles, unintentionally present in the solutions. remove the dosure, avoiding any contamination of the
contents. Eliminate gas bubbles by appropriate measures such
For the determination of particulate contamination as allowing to stand for 2 min or sonicating.
2 procedures, Method 1 (Light Obscuration Partide Count
Test) and Method 2 (Microscopic Particle Count Test), For large-volume parenterals, single units are tested. For
are specified hereinafter. When examining injections and small-volume parenterals less than 25 mL in volume, the
infusions for sub-visible particles, Method 1 is preferably contents of lOor more units are combined in a cleaned
applied. However, it may be necessary to test sorne container to obtain a volume of not les s than 25 mL; where
preparations by the light obscuration particle count test justified and authorised, the test solution may be prepared by
followed by the microscopic partide count test to reach a mixing the contents of a suitable number of vials and diluting
conclusion on conformance to the requirements. to 25 mL with particle-free water R or with an appropriate
solvent without contamination of particles when particle-free
Not all parenteral preparations can be examined for sub-visible water R is not suitable. Small-volume parenterals having a
particles by one or both of these methods. When Method 1 volume of 25 mL or more may be tested individually.
is not applicable, e.g. in case of preparations having reduced
clarity or increased viscosity, the test is carried out according Powders for parenteral administration are reconstituted with
to Method 2. Emulsions, colloids, and liposomal preparations particle-free water R or with an appropriate solvent without
are examples. Similarly, products that produce air or gas contamination of particles when particle-free water R is not
bubbles when drawn into the sensor may also require suitable.
microscopic particle count testing. If the viscosity of the The number of test specimens must be adequate to provide a
preparation to be tested is sufficiently high so as to preclude statistically sound assessment. For large-volume parenterals
its examination by either test method, a quantitative dilution or for small-volume parenterals having a volume of 25 mL
with an appropriate diluent may be made to deerease viscosity, or more, fewer than 10 units may be tested, based on an
as necessary, to allow the analysis to be performed. appropriate sampling plan.
The results obtained in examining a discrete unit or group Remove 4 portions, each of not less than 5 mL, and count
of units for particulate contamination cannot be extrapolated the number of particles equal to or greater than 10 [lm and
with certainty to other units that remain untested. Thus, 25 [lm. Disregard the result obtained for the first portion, and
statisticalIy sound sampling plans must be developed if valid calculate the mean number of particles for the preparation to
inferences are to be drawn from observed data to characterise be examined.
the level of particulate contamination in a large group of units. Evaluation

METHOD 1. LIGHT OBSCURATION PARTICLE COUNT For preparations supplied in container s with a nominal
TEST volume of more than 100 mL, apply the criteria of test l.A.
Use a suitable apparatus based on the principIe of light Por preparations supplied in containers with a nominal
blockage which allows an automatic determination of the size volume of less than 100 mL, apply the criteria of test 1.B.
of particles and the number of particles according to size. • Por preparations supplied in containers with a nominal
The apparatus is calibrated using suitable certified reference volume of 100 mL, apply the criteria of test 1.B .•
materials consisting of dispersions of spherical particles of If the average number of partides exceeds the limits, test the
known sizes between 10 [lm and 25 [lm. These standard preparation by the microscopic partide count test.
partides are dispersed in particle-free water R. Care must be Test 1.A - Solutions for infusion or solutions for injection
taken to avoid aggregation of particles during dispersion. supplied in containers with a nominal content of more than
General precautions 100 mL
The test is carried out under conditions limiting particulate The preparation complies with the test if the average number
contamination, preferably in a laminar-flow cabinet. of particles present in the units tested does not exceed 25 per
Very carefully wash the glassware and filtration equipment millilitre equal to or greater than 10 [lm and does not exceed
used, except for the membrane filters, with a warm detergent 3 per millilitre equal to or greater than 25 [lm.
solution and rinse with abundant amounts of water to remove Test l.B - Solutions for infusion or solutions for injection
all traces of detergent. Immediately before use, rinse the supplied in containers with a nominal content of less than
equipment from top to bottom, outside and then inside, with 100 mL
particle-free water R. The preparation complies with the test if the average number
Take care not to introduce air bubbles into the preparation to of particles present in the units tested do es not exceed 6000 per
be examined, especialIy when fractions of the preparation are container equal to or greater than 10 [lm and do es not exceed
being transferred to the container in which the determination 600 per container equal to or greater than 25 [lm.
is to be carried out.
METHOD 2. MICROSCOPIC PARTICLE COUNT TEST
In order to check that the environment is suitable for the
test, that the glassware is properly cleaned and that the water Use a suitable binocular microscope, filter assembly for
to be used is particle-free, the folIowing test is carried out: retaining particulate contamination and membrane filter for
determine the particulate contamination of 5 samples of examination.
particle-free water R, each of 5 mL, according to the method The microscope is equipped with an ocular micro meter
described below. If the number of particles of 10 [lm or greater calibrated with an objective micrometer, a mechanical stage
size exceeds 25 for the combined 25 mL, the precautions taken capable of holding and traversing the entire filtration area

(8) This chapter has undergone pharmacopoeial harmonisatiol1. See chapter 5.8. Pharmacopoeial hannonisation.

General Notices (1) apply to all monographs and other texts 321
2,9,19, Particulate contamination: sub-visible partides EUROPEAN PHARMACOPOEIA 8.0

of the membrane filter, 2 suitable illuminators to provide For large-volume parenterals, single units are tested. For
episcopic illumination in addition to oblique illumination, small-volume parenterals les s than 25 mL in volume, the
and is adjusted to 100 ± 10 magnifications. contents of lOor more units are combined in a cleaned
The ocular micrometer is a circular diameter graticule (see container; where justified and authorised, the test solution
Figure 2.9.19.-1.) and consists of a large circle divided by may be prepared by mixing the contents of a suitable number
crosshairs into quadrants, transparent and black reference of vials and diluting to 25 mL with particle-free water R or
circles 10 flm and 25 flm in diameter at 100 magnifications, with an appropriate solvent without contamination of particles
and a linear scale graduated in 10 flm increments. It is when particle-free water R is not suitable. Small-volume
calibrated using a stage micro meter that is certified by either parenterals having a volume of 25 mL or more may be tested
a domestic or international standard institution. A relative individually.
error of the linear scale of the graticule within ± 2 per cent is Powders for parenteral administration are constituted with
acceptable. The lárge circle is designated the graticule field particle-free water R or with an appropriate solvent without
ofview (GFOV). contamination of particles when particle-free water R is 110t
2 illuminators are required. One is an episcopic brightfield suitable.
illuminator internal to the microscope, the other is an externa!, The number of test specimens must be adequate to provide a
focusable auxiliary illuminator adjustable to give reflected statistically sound assessment. For large-volume parenterals
oblique illumination at an angle of 10-20°. or for small-volume parenterals having a volume of 25 mL
The filter assembly for retaining particulate contamination or more, fewer than 10 units may be tested, based on an
consists of a filter holder made of glass or other suitable appropriate sampling plan.
material, and is equipped with a vacuum source and a suitable Wet the inside of the filter holder fitted with the membrane
membrane filter. filter with several millilitres of particle-free water R. Transfer
The membrane filter is of suitable size, black or dark grey to the filtration funnel the total volume of a solution pool or
in colour, non-gridded or gridded, and 1.0 flm or finer in of a single unit, and apply vacuum. If needed, add stepwise
nominal pore size. a portion of the solution until the entire volume is filtered.
After the last addition of solution, begin rinsing the inner
walls of the filter holder by using a jet of particle-free water R.
Maintain the vacuum until the surface of the membrane filter
GFOV circle
is free from liquido Place the filter in a Petri dish and allow the
filter to air-dry with the cover slightly ajar. After the filter has
been dried, place the Petri dish on the stage of the microscope,
scan the entire membrane filter under the reflected light from
00 00
the illuminating device, and count the number of particles
that are equal to or greater than 10 flm and the number of
Reference
circle particles that are equal to or greater than 25 flm. Alternatively,
partial filter count and determination of the total filter count
by calculation is allowed. Calculate the mean number of
particles for the preparation to be examined.
The particle sizing process with the use of the circular diameter
graticule is carried out by transforming mentally the image of
Cross
hairs each particle into a circle and then comparing it to the 10 flm
and 25 flm graticule reference circles. Thereby the particles
11111111111111111111111111111111111111111111111111111111111111111111111111111111111111111111111111111 ~ Linear scale are not moved from their initiallocations within the graticule
field of view and are not superimposed on the reference
Figure 2.9.19.-1. - Circular diameter graticule circles for comparison. The inner diameter of the transparent
graticule reference circles is used to size white and transparent
General precautions
particles, while dark particles are sized by using the outer
The test is carried out under conditions limiting particulate diameter of the black opaque graticule reference circles.
contamination, preferably in a laminar-flow cabinet.
In performing the microscopic particle count test do not
Very carefully wash the glassware and filter assembly used, attempt to size or enumerate amorphous, semi-liquid, or
except for the membrane filter, with a warm detergent solution otherwise morphologically indistinct material s that have
and rinse with abundant amounts of water to remove all traces the appearance of a stain or discoloration on the membrane
of detergent. Immediately before use, rinse both sides of filter. These materials show little or no surface relief and
the membrane filter and the equipment from top to bottom, present a gelatinous or film-like appearance. In such cases
outside and then inside, with particle-free water R. the interpretation of enumeration may be aided by testing a
In order to check that the environment is suitable for the sample of the solution by the light obscuration particle count
test, that the glassware and the membrane filter are properly test.
cleaned and that the water to be used is particle-free, the Evaluation
following test is carried out: determine the particulate
contamination of a 50 mL volume of particle-free water R For preparations supplied in containers with a nominal
according to the method described below. If more than volume of more than 100 mL, apply the criteria of test 2.A.
20 particles 10 ~lm or larger in size or if more than 5 particles For preparations supplied in container s with a nominal
25 [.1m or larger in size are present within the filtration area, volume of less than 100 mL, apply the criteria of test 2.B.
the precautions taken for the test are not sufficient. The • For preparations supplied in containers with a nominal
preparatory steps must be repeated until the environment, volume of 100 mL, apply the criteria of test 2.B .•
glassware, membrane filter and water are suitable for the test. Test 2.A - Solutions for infusion or solutions for injection
Method supplied in containers with a nominal content of more than
Mix the contents of the samples by slowly inverting the 100 mL
container 20 times successively. lf necessary, cautiously remove The preparation complíes with the test if the average number
the sealing closure. CIean the outer surfaces of the container of particles present in the units tested does not exceed 12 per
opening using a jet of particle-free water R and remove the millilitre equal to or greater than 10 flm and does not exceed
closure, avoiding any contamination of the contents. 2 per millilitre equal to or greater than 25 flm.

322 See the information section on general monographs (cover pages)


EUROPEAN PHARMAeOPOEIA 8.0 2.9.22. Softening time determination oí lipophilic suppositories

Test 2.E - Solutions for infusion or solutions for injection APPARATUS A


supplied in containers with a nominal content of less than The apparatus (see Figure 2.9.22.-1) consists of a glass tube
100 mI 15.5 mm in internal diameter with a flat bottom and a length
The preparation complies with the test if the average number of about 140 mm. The tube is closed by a removable pi as tic
of particles present in the units tested does not exceed 3000 per cover having an opening 5.2 mm in diameter. The apparatus
container equal to or greater than 10 [lm and does not exceed comprises a rod 5.0 mm in diameter which becomes wider
300 per container equal to or greater than 25 [lm. towards the lower end, reaching a diameter of 12 mm. A
metal needle 2 mm in length and 1 mm in diameter is fixed
on the flat underside.
0112008:20920 The rod consists of 2 parts, a lower part made of pi as tic
material and an upper part made of plastic material or metal
2.9,20. PARTICULATE with a weight disk. The upper and lower parts are either fitted
CONTAMINATION: VISIBLE together (manual version) or separate (automated version).
The weight of the entire rod is 30 ± 0.4 g. The upper part of the
PARTICLES rod carríes a sliding mark ringo When the rod is introduced
into the glass tube so that it touches the bottom, the mark ring
Particulate contamination of injections and infusions consists
is adjusted to coincide with the upper level of the plastic cover.
of extraneous, mobile undissolved particles, other than gas
bubbles, unintentionally present in the solutions.
~ii~
The test is intended to provide a simple procedure for the
visual assessment of the quality of parenteral solutions as
-~I 1I I<~

I,~I
regards visible particles. Other validated methods may be
used.
APPARATUS
The apparatus (see Figure 2.9.20.-1) consists of a viewing
station comprising:
- a matt black panel of appropriate size held in a vertical
position,
a non-glare white panel of appropriate size held in a vertical
position next to the black panel,
an adjustable lampholder fitted with a suitable, shaded,
white-light source and with a suitable light diffuser (a
viewing illuminator containing two 13 W fluorescent
tubes, each 525 mm in length, is suitable). The intensity of
illumination at the viewing point is maintained between
2000 lux and 3750 lux, although higher values are preferable
for coloured glass and plastic containers.
Adjustable lampholder

/
~II I<.~
, 015.5
·~i :~

Figure 2.9.22.-1. - Apparatus A for measuring the softening


Matl
black panel time of lipophilic suppositories
panel Dimensions in millimetres
Method. Place the glass tube containing 10 mL ofwater in a
water-bath and equilibrate at 36.5 ± 0.5 oc. Fix the glass tube
vertically and immerse to a depth of at least 7 cm below the
surface but without touching the bottom of the water-bath.
Figure 2.9.20.-1. - Apparatus for visible particles Introduce a suppository, tip first, into the tube followed by
METHOD the rod with the free gliding plastic cover into the glass tube
until the metal needle touches the flat end of the suppository.
Remove any adherent labels from the container and wash and Put the cover on the tube (beginning of time measurement).
dry the outside. Gently swirl or invert the container, ensuring Note the time which elapses until the rod sinks down to the
that air bubbles are not introduced, and observe for about 5 s bottom of the glass tube and the marIz ring reaches the upper
in front of the white panel. Repeat the procedure in front of level of the plastic cover.
the black paneL Record the presence of any particles.
APPARATUS B
The apparatus (see Figure 2.9.22.-2) consists of a water-bath (B)
!H/2008:20922 into which an inner tube (A) is inserted and fixed with a
stopper. The inner tube is closed by a stopper at the bottom.
2.9.22. SOFTENING TIME The apparatus is fitted with a thermometer. 2 insets are
DETERMINATION OF LIPOPHILIC available:
- a glass rod (el) in the form of a tube sealed at both ends,
SUPPOSITORIES carrying a rim at its lower end weighed with lead shot,
The test is intended to determine, under defined conditions, which has a weight of 30 ± 0.4 g,
the time which elapses until a suppository maintained in - a penetration inset (e2) consisting of a rod (7.5 ± 0.1 g) in
water softens to the extent that it no longer offers resistance a tube which has an enlargement for the suppository, both
when a defined weight is applied. made of stainless steel.

General Notices (1) apply to all monographs and other texts 323
2.9.23. Gas pycnometric density of soHds EUROPEAN PHARMACOPOEIA 8.0

Method. Pour 5 mL of water at 36.5 ± 0.5 oC into the inner The gas pycnometrie density measurement is performed at a
tube (A), introduce a suppository with the tip downwards and temperature between 15 oC and 30 oC that does not vary by
onto that, place the inset (Cl or C2). Note the time which more than 2 oC during the course of measurement.
elapses between this moment and the moment when the The apparatus is calibrated, whieh means that the volumes Ve
lower, rimmed end of the glass rod (C 1) or the steel rod (C2) and Ve are determined using a suitable calibration standard
reaches the narrowed part of the inner glass tube. Melting or whose volume is known to the nearest 0.001 cm). The
dissolution is then considered as complete. procedure described below is followed in 2 runs, firstly with
an empty test eel!, and secondly with the calibration standard
placed in the test cell. The volumes Ve and V, are ealculated
using the equation for the sample volume (V,), taking into
account that Vs is zero in the first runo

,,
,
A C1 C2
o
o
N

o B
m

©
1.9.11
1..1'L.
~
37'C
A valve;
expansion volume, in cubie centimetres;
ceH volume, in cubie centimetres;
Figure 2.9.22.-2. - Apparatus B for measuring the softening
time of lipophilic suppositories sample volume, in cubie centimetres;
Dimensions in millimetres M manometer.
Figure 2.9.23.-1. - Schematic diagram of a gas pycnometer
07/2008:20923 METHOD
Volatile contaminants in the powder are removed by degassing
2.9.23. GAS PYCNOMETRIC DENSITY the powder under a constant purge of helium prior to the
OF SOLIDS measurement. Occasionally, powders may have to be degassed
under vacuum. Because volatiles may be evolved during the
Gas pycnometric density is determined by measuring the measurement, weighing of the sample is carried out after the
volume occupied by a known mass of powder, which is pycnometric measurement of volume.
equivalent to the volume of gas displaced by the powder using
Weigh the test ceH of the pycnometer and record the mass. Fill
a gas displacement pycnometer. In gas pycnometric density
the test cell with a given mass of powder of the substance to
measurements, the volume determined excludes the volume
be examined. Seal the test cell in the pycnometer. Record the
occupied by open pores; however, it includes the volume
system reference pressure (P r) as indicated by the manometer
oceupied by sealed pores or pores inaccessible to the gas.
while the valve that connects the expansion cell with the test
Usual!y, helium is used as a test gas due to its high diffusivity cel! is open. Close the valve to separate the expansion cel! from
into smal! open pores. If gases other than helium are used, the test cell. Pressurise the test cell with the gas to an initial
different values would be obtained, since the penetration of pressure (P) and record the value obtained. Open the valve to
the gas is dependent on the size of the pore as well as the connect the expansion cel! with the test cell. Record the final
cross-sectional area of the gas molecules. pressure (PJ)' Repeat the measurement sequence for the same
The measured density is a volume-weighted average of the powder sample until consecutive measurements of the sample
densities of individual powder particles. It is cal!ed the particle volume (V) agree to within 0.2 per cent. Unload the test cell
density, distinct from the true density of a solid or the bulk and measure the final powder mass (m), expressed in grams.
density of a powder. The density of solids is expressed in grams If the pycnometer differs in operation or construction from
per cubie centimetre (g/cm)), although the International Unit the one shown in Figure 2.9.23.-1, follow the instructions of
is the kilogram per cubie metre (l g/cm 3 = 1000 kg/m 3 ). the manufacturer of the pycnometer.
APPARATUS EXPRESSION OF THE RESULTS
The apparatus (see Figure 2.9.23.-1) consists ofthe following: The sample volume (V,) is given by the equation:
- a sealed test cell, with empty cel! volume Ve' connected Ve
through a valve to an expansion cel!, with volume Vr; Vs=Vc - p - p
, T _ 1
- a system capable of pressurising the test cel! with the p¡ - Pe
measurement gas until a defined pressure (P) indieated by
a manometer; The density (p) is given by the equation:
- the system is eonnected to a source of measurement gas, m
preferably helium, unless another gas is specified. p= -
Vs

324 See the information section on general monographs (cover pages)


EUROPEAN PHARMACOPOEIA 8.0 2.9.25. Dis§olution test for medicated chewing gums

The sample conditioning is indicated with the results. For The funnel and guides are mounted on the central chamber.
example, indicate whether the sample was tested as is or dried The O-rings are incorporated in the pisto n recess with the
under specific conditions such as those described for 10ss on sealing ring around it; the sealing rings ensure that the
drying. chamber is watertight. The horizontal pistons are placed in
the chewing chamber through the guides.
The gum is artificially chewed by the horizontal pistons, and
04/2012:20925 the vertical piston ensures thatthe gum stays in the right
place between chews.
Machine speed is controlled to ensure a constant cyele. One
2.9.25. DISSOLUTION TEST FOR cyele (chew) is defined as follows: the horizontal pistons
MEDICATED CHEWING GUMS start from their outermost position, move to their innermost
position then return to their outermost position. Within one
PRINCIPLE cyele, the vertical piston moves from its lowest position to its
The test is used to determine the dissolution rate of active uppermost position and back to its lowest position.
substances in medicated chewing gums. This is done by Each horizontal piston has a stroke of 25.0 mm. The
applying a mechanical kneading procedure to a piece of gum maximum distance between these 2 pistons is 50 mm. The
placed in a small chamber designed to simulate the process minimum distan ce between the 2 horizontal pistons is 0.1 mm
of chewing. to 1.0 mm. The vertical pisto n has a stroke of 22.0 mm.
APPARATUSA Horizontal piston movement is controlled so that the 2 pistons
are at their innermost position at the same time. Vertical
Chewing apparatus A (Figure 2.9.25.-1) consists of: piston movement is controlled so that it does not conflict with
- 1 chewing chamber; the movement of the horizontal pistons.
- 1 vertical piston; lf necessary, the machine can be constructed so that the
- 2 horizontal pistons with O-rings and sealing rings. horizontal pistons rotate around their own axes in opposite
direction to each other by the end of the chew in order to
The chewing chamber consists of 4 individual parts:
obtain maximum chewing.
- 1 central chamber;
AH parts of the apparatus that may come into contact with the
- 1 funnel (Figure 2.9.25.-2); preparation or the dissolution medium are chemically inert
- 2 guides with bushes (Figure 2.9.25.-3). and do not adsorb, react with or interfere with the sample.

016 M6x16

o
<D
E
N

070

SECTION G-G

SECTION F-F

112
~ F

-fff-[3J F
1---
1- - r - , ~~J F~!--&-~+'--! Q

1----------11 ~ :-¡1---_ _12c-1_.4_ _----1


A B e
A. horizontal piston B. guide C. chewing chamber D. funnel E. vertical piston

Figure 2.9.25.-1 - Apparatus A - Chewing chamber and pistons


Dimensions in millimetres

General Notices (1) apply fo al! monographs and other texts 325
2.9.25. Dissolution test for medicated chewing gums EUROPEAN PHARMACOPOEIA 8.0

I I
Ir
~
.~'\
..... /
11 LT--T...J 11 ,
. . . . ) H: : nI
,,"
,,"
I1 1 I
",'
,"
I 11
'.----~./'
I11 I I 11 I
,,"
111
",'
11 1 /t
./' f

I:::~":"~~
Figure 2.9.25.-2 - Funnel
11 - ~ I --
( "(
Dimensions in millimetres ~. "
~- '"

Itl. ~ L_$

036

024.95

A. revolving device for the upper E. upper chewing surface


chewing surface
B. stand F. lower chewing surface

C. test ceH G. base chamber

D. axle H. device for up-and-down


chewing motion

Figure 2.9.25.-4 - Apparatus B

The test cells may also be equipped with 1 or 2 glass sampling


63 tubes, coming through the thermostatic double wall. These
tubes also make it possible to have an external sink, which
Figure 2.9.25.-3 - Guide (section G-G) may be necessary to achieve sink conditions for sparingly
soluble substances.
Dimensions in millimetres The gum is usually sandwiched between 2 circular plastic nets
to prevent disintegration.
APPARATUS B Nets made from nylon (PA6) with an aperture of lA mm and
a wire diameter of 00405 mm may be used.
Chewing apparatus B (Figure 2.9.25.-4) consists of:
Al! parts of the apparatus that may come into contact with the
- 1 test ceH (Figure 2.9.25.-5 or 2.9.25.-6); preparation or the dissolution medium are chemically inert
- 1 vertical axle with upper chewing surface (Figures 2.9.25.-7 and do not adsorb, react with or interfere with the sample.
and 2.9.25.-8); PROCEDURE
- 1 base chamber with lower chewing surface (Figures For each determination, the following information is needed:
2.9.25.-9 and 2.9.25.-10); - apparatus used (type A or type B);
- 1 device for up-and-down chewing motion; composition, volume and temperature of the dissolution
medium;
- 1 revolving device for the vertical axle.
- number of chews per minute;
The gum is artificially chewed by the lower and upper chewing
- time and sampling method;
surfaces. Machine speed is controHed to ensure a constant
cyele. The distan ce between the lower and upper chewing - whether the analysis is performed on the gum residue or
surfaces may be set up to 5 mm. The turning angle of the on the dissolution medium;
revolving device is about 20°. - method of analysis.

326 See the information section on general monographs (caver pages)


EUROPEAN PHARMACOPOEIA 8.0 2.9.25. Dissolution test fOl" medicated chewing gums

059 ±2

/
"'
/1\
I
1,. .
/

I I \
I I I 1
I I I I
I I I I
I I I I
I I I I
I I I
I II
I
I
I
I
I
I
I
I
I
N I
Ol

Figure 2.9.25.-5 - Test cell


Dimensions in millimetres
o 59±2

/
/ , "'\
I
1,. .
/

I \
I I I
I I I
I I I
I I I
I I I
I I I
I I
I
I
I
I
6$0 I
I
I
I
I
I
N
Ol I
I
I
I
I
I
I
I
\
"'
~-
I
/ -
I
I
I
L.J J1 tL...J

l. 037.2 ± 0.1
.1 1d'

Figure 2.9.25.-6 - Test cell (straight)


Dimensions in millimetres
Place the prescribed volume of dissolution medium in the or with a thermostat (apparatus B). Set the machine speed at
chewing chamber, usually 20 mL of phosphate buffer solution the prescribed number of chews per minute (typically up to
pH 6.0 R2. Maintain the medium temperature at 37 ± 0.5 oC 60). Accurately weigh a portion of gum or the whole gum, put
using an electrical device with external control (apparatus A) it into the chewing chamber and start the machine.

General Notices (1) apply to all monographs and other texts 327
2.9.25. Dissolution test for medicated chewing gums EUROPEAN PHARMACOPOEIA 8.0

1~
61.6
"1 M16x1
1 014

o
N
I
I I ~~
I

I
I

;
o
al

I
I

1
I

I
I

¡
N
N
1
fTl 037 -SS
"'1
/ ~-+_-!
" 8.8 (2x)
-1 1" 08.5
N
N M~
~1
j I
1 -'-
M8
¡ ---t-
~==i-:lL._--,
11
11
111
, 11 ,..--_..L.f=== t
j
ro -t---
~ I 1I
111 11111 I
~
08.5 ~===l ti! ,11 i ~===
I
l!)
027 r-..:

Figure 2.9.25.-9 - Base chamber


Figure 2.9.25.-7 - Axle
Dimensions in millimetres
Dimensions in millimetres M8

029

037

Blasted surface RA 1.5-2.1

028

Blasted suliace RA 1.5-2.1

Figure 2.9.25.-10 - Lower chewing surface


Dimensions in millimetres
I !
SAMPLING AND EVALUATION
Stop the apparatus at the prescribed time. Remove the
gum residue and take a sample of the dissolution medium.
Figure 2.9.25.-8 - Upper chewing surface Determine the content of active substance(s) by a suitable
method. Medium replacement may be made after each
Dimensions in millimetres sampling procedure; compensation by calculation of medium

328 See the information section on general monographs (cover pages)


EUROPEAN PHARMACOPOEIA 8.0 2.9.26. Spedfic surface area gas adsorption

volume change or sample dilution is needed. Alternatively,


determine the content of active substance(s) remaining in the s= VrniVa
m x 22400 (2)
gum residue. Carry out the test successively on 6 medicated
chewing gums.
N Avogadro constant (6.022 x 10 23 mol- 1),
The quantity oí active substance(s) dissolved in a specified
time is expressed as a percentage of the content stated on the a eÍfective cross-sectional are a of one adsorbate
label. molecule, in square metres (0.162 nm 2 Íor
nitro gen and 0.195 nm 2 Íor krypton),
m mass of test powder, in grams,
0112010:20926
22400 volume occupied by 1 mole of the adsorbate
gas at STP allowing for minor departures from
2.9.26. SPECIFIC SURFACE AREA BY the ideal, in millilitres.
GAS ADSORPTION(9) A minimum of 3 data points is required. Additional
INTRODUCTION measurements may be carried out, especially when
The specific surface are a oí a powder is determined by non-linearity is obtained at a PIPo value c10se to 0.3. Because
physical adsorption oí a gas on the surface of the solid and non-Iinearity is often obtained at a PIPo value below 0.05,
by calculating the amount of adsorbate gas corresponding to values in this region are not recommended. The test Íor
a monomolecular layer on the surface. Physical adsorption linearity, the treatment of the data, and the calculation of the
results from relatively weak force s (van der Waals forces) specific surface area of the sample are described aboye.
between the adsorbate gas mo1ecules and the adsorbent surface SINGLE-POINT MEASUREMENT
of the test powder. The determination is usually carried out Normally, at least 3 measurements oí Va each at difÍerent
at the temperature of liquid nitro gen. The amount of gas values of PIPo are required for the determination of specific
adsorbed can be measured by a volumetric or continuous flow surface are a by the dynamic flow gas adsorption technique
procedure. (Method 1) or by volumetric gas adsorption (Method JI).
However, under certain circumstances described below, it
BRUNAUER, EMMETT AND TELLER (BET) THEORY AND may be acceptable to determine the specific surface area of a
SPECIFIC SURFACE AREA DETERMINATION powder from a single yalue of Va measured at a single value of
MULTI-POINT MEASUREMENT PIPo such as 0.300 (corresponding to 0.300 mole of nitrogen
The data are treated according to the Brunauer, Emmett and or 0.001038 mole Íraction of krypton), using the following
Teller (BET) adsorption isotherm equation: equation for calculating

(3)
(1)

The specific surface area is then calculated from the value of


V", by equation (2) given aboye.
P partial vapour pressure of adsorbate gas in The single-point method may be employed directly for a series
equilibrium with the surface at 77.4 K (b.p. of of powder samples of a given material for which the material
liquid nitrogen), in pascals, constant C is l11uch greater than unity. These circllmstances
Po saturated pressure of adsorbate gas, in pascals, may be verified by comparing values of specific surface area
Va volume of gas adsorbed at standard temperature determined by the single-point method with that determined
and pressure (STP) [273.15 K and atmospheric by the multiple-point method Íor the series of powder
pressure (1.013 x 10 5 Pa)], in millilitres, samples. Close similarity between the single-point values and
V", volume of gas adsorbed at STP to produce an multiple-point vallles suggests that l/C approaches zero.
apparent monolayer on the sample surface, in The single-point method may be employed indirectly for
millilitres, a series of very similar powder samples of a given material
C dimensionless constant that is related to the for which the material constant C is not infinite but may be
enthalpy of adsorption of the adsorbate gas on the assumed to be invariant. Under these circumstances, the error
powder sample. associated with the single-point method can be reduced or
A value of Va is measured at each of not less than 3 values of eliminated by using the multiple-point method to evaluate C
for one of the samples of the series from the BET plot, from
PIPo' which C is calculated as (l + slope/intercept). Then V m is
Then the BET value calculated from the single value of Va measured at a single
1 value of PI Po by the equation:

is plotted against PIPo according to equation (1). This plot Vrn = Va ( pPo - 1
) [1e + -c:
e - 1x ( P )]
Po (4)
should yield a straight line usually in the approximate relatiye
pressure range 0.05 to 0.3. The data are considered acceptable The specific surface area is calcu1ated from byequation (2)
if the correlation coefficient, r, of the linear regression is not given aboye.
less than 0.9975; that is, ¡2 is not less than 0.995. From the
resulting linear plot, the slope, which is equal to (C - l)IV",C,
EXPERIMENTAL TECHNIQUES
and the intercept, which is equal to llVmC, are evaluated
by linear regression ana1ysis. From these values, V m is This section describes the methods to be used for the sample
calcu1ated as l/(slope + intercept), while C is calculated as preparation, the dynamic flow gas adsorption technique
(slope/intercept) + 1. From the value of V m so determined, the (Method 1) and the volumetric gas adsorption technique
specific surface area, S, in m 2 'g- 1 , is calculated by the equation: (Method II).

(9) This chapter has llndergone pharmacopoeial harmonisation. See chapter 5.8. Pharmacopoeiaf lwrmonisalion.

General Notices (1) apply ta all monagraphs and other texts 329
2.9.26. Spedfic surface area by gas adsorption EUROPEAN PHARMACOPOEIA 8.0

SAMPLE PREPARATION pressure. For this purpose, a thermal conductivity detector


Outgassing with an electronic integrator is one among various suitable
types. A minimum of 3 data points within the recommended
Before the speciflc surface area of the sample can be range of 0.05 to 0.30 for PIPo is to be determined.
determined, it is necessary to remove gases and vapours that
may have become physically adsorbed onto the surface after Procedure
manufacture and during treatment, handling and storage. If A known mixture of the gases, usually nitro gen and helium,
outgassing is not achieved, the speciflc surface area may be is passed through a thermal conductivity cel!, through the
reduced or may be variable because an intermediate area of the sample, again through the thermal conductivity ceH and then
surface is covered with molecules of the previously adsorbed to a recording potentiometer.
gases or vapours. The outgassing conditions are critical for Immerse the sample ceH in liquid nitro gen, then the sample
obtaining the required precision and accuracy of specific adsorbs nitro gen from the mobile phase. This unbalances
surface are a measurements on pharmaceuticals because of the the thermal conductivity cel!, and a pulse is generated on a
sensitivity of the surface of the materials. recorder chart.
Conditions. The outgassing conditions must be demonstrated Remove from the coolant; this gives a desorption peak equal
to yield reproducible BET plots, a constant weight of test in are a and in the opposite direction to the adsorption peak.
powder, and no detectable physical or chemical changes in Since this is better deflned than the adsorption peak, it is the
the test powder. one used for the determination.
The outgassing conditions defined by the temperature, To effect the calibration, inject a known quantity of adsorbate
pressure and time should be chosen so that the original surface into the system, sufficient to give a peak of similar magnitude
of the solid is reproduced as elosely as possible. Outgassing of to the desorption peak and obtain the proportion of gas
many substances is often achieved by applying a vacuum, by volume per unit peak area.
purging the sample in a flowing stream of a non-reactive, dry Use a nitrogen/helium mixture for a single-point
gas, or by applying a desorption-adsorption cyeling method. determination and several such mixtures or premixing
In either case, elevated temperatures are sometimes applied to 2 streams of gas for a multiple-point determination.
increase the rate at which the contaminants leave the surface. Calculation is essentially the same as for the volumetric
Caution should be exercised when outgassing powder samples method.
using elevated temperatures to avoid affecting the nature of
the surface and the integrity of the sample. Method n: the volumeíric method
If heating is employed, the recommended temperature Principie
and time of outgassing are as low as possible to achieve In the volumetric method (se e Figure 2.9.26.-2), the
reproducible measurement of specific surface area in an recommended adsorbate gas is nitro gen which is admitted
acceptable time. For outgassing sensitive samples, other into the evacuated space aboye the previously outgassed
outgassing methods such as the desorption-adsorption cyeling powder sample to give a defined equilibrium pressure, P, of
method may be employed. the gas. The use of a diluent gas, such as helium, is therefore
unnecessary, although helium may be employed for other
Adsorbate
purposes, such as to measure the dead volume.
The standard technique is the adsorption of nitrogen of Since only pure adsorbate gas, instead of a gas mixture, is
analytical quality at liquid nitro gen temperature.
employed, interfering effects of thermal diffusion are avoided
For powders of low specific surface are a « 0.2 m 2K 1) the in this method.
proportion adsorbed is low. In such cases the use of krypton Procedure
at liquid nitro gen temperature is preferred because the low
vapour pressure exerted by this gas greatly reduces error. The Admit a small amount of dry nitro gen into the sample tube
use oflarger sample quantities where fe asible (equivalent to prevent contamination of the elean surface, remove the
to 1 m 2 or greater total surface area using nitro gen) may sample tube, insert the stopper, and weigh it. Calculate the
compensate for the errors in determining low surface areas. weight of the sample. Attach the sample tube to the volumetric
apparatus. Cautiously evacuate the sample down to the
Al! gases used must be free from moisture. specified pressure (e.g. between 2 Pa and 10 Pa). Alternatively,
Quantity of sample sorne instruments operate by evacuating to a defined rate of
Accurately weigh a quantity of the test powder such that the pressure change (e.g. less than 13 Pa/30 s) and holding for a
total surface of the sample is at least 1 m 2 when the adsorbate defined period of time before commencing the next step.
is nitro gen and 0.5 m 2 when the adsorbate is krypton. If the principIe of operation of the instrument requires the
Lower quantities of sample may be used after appropriate determination of the dead volume in the sample tube, for
validation. example, by the admission of a non-adsorbed gas, such as
helium, this procedure is carried out at this point, followed
MEASUREMENTS by evacuation of the sample. The determination of dead
Because the amount of gas adsorbed under a given pressure volume may be avoided using difference measurements, that
tends to increase 011 decreasing the temperature, adsorption is, by means of reference and sample tubes connected by a
measurements are usually made at a low temperature. differential transducer. The adsorption of nitrogen gas is then
Measurement is performed at 77.4 K, the boiling point of measured as described below.
liquid nitro gen.
Raise a Dewar vessel containing liquid nitro gen at 77.4 K
Method 1: the dynamk How method up to a defined point on the sample cell. Admit a sufficient
Principie volume of adsorbate gas to give the lowest desired relative
In the dynamic flow method (see Figure 2.9.26.-1), the pressure. Measure the volume adsorbed, Va' For multipoint
recommended adsorbate gas is dry nitrogen or krypton, while measurements, repeat the measurement of Va at successively
helium is employed as a diluent gas, which is not adsorbed higher PIPo values. When nitro gen is used as the adsorbate
under the recommended conditions. gas, PIPo values of 0.10,0.20, and 0.30 are often suitable.
A minimum of 3 mixtures of the appropriate adsorbate gas REFERENCE MATERIALS
with helium are required within the PIPo range 0.05 to 0.30. Periodically verify the functioning of the apparatus using
The gas detector-integrator should provide a signal that is appropriate reference materials of known surface area, such as
approximately proportional to the volume of the gas passing a-alumina, which should have a specific surface area similar
through it under defined conditions of temperature and to that of the sample to be examined.

330 See the information section on general monographs (caver pages)


EUROPEAN PHARMACOPOEIA 8.0 2.9.29. Intrinsic dissolution

Self seals quick connection

o ring seals
Calibrating
Septum Flow I t
.- r@- Path
selection
meter
l Diffusion

Flow
1 valve baffle

-;
control :y
ro
valve
3
~ "'-
ro

• ......
.o
Cold
§:
cr
"-
Sample Outgassing
station
trap
ª-=>

cell

Differential [) eo-
flow ro
controller Short Long
path path
ballast ballast
,...--
On-off ~
valve l Digital
A
~ B
display
,.. Vent
~

G as inlet
Detector Detector

Figure 2.9.26.-1. - Schematic diagram of the dynamic flow method apparatus

9
To cold traps and
4 3 vacuum pumps

7 10 11
8

Helium
reservoir

Vapour 1~=::¡;21c~vacuum
j, Air
pressure
manometer

Figure 2.9.26.-2. - Schematic diagram of the volumetric method apparatus

0112008:20927 0112008:20929

2.9.27. UNIFORMITY OF MAS S 2.9.29. INTRINSIC DISSOLUTION


OF DELIVERED DOSES FROM The test is intended to determine the intrinsic dissolution rate
of pure solid substances following compaction. It is carried
MULTIDOSE CONTAINERS out under specifled experimental conditions such that a
The following test is intended for oral dosage forms such as practical measure of the intrinsic dissolution rate is obtained.
granules, powders for oral use and liquids for oral use, which The intrinsic dissolution rate is a theoretical value referring
are supplied in multidose containers provided at manufacture to pure solid substances having null porosity, but, practically,
with a measuring device. intrinsic dissolution rate is determined on substances having a
Weigh individually 20 doses taken at rarrdom from one or minimal porosity.
more containers with the measuring device provided and PRINCIPLE
determine the individual and average masses. Not more than
The intrinsic dissolution rate is defined as the dissolution
2 of the individual masses deviate from the average mass by
rate of pure substances following compaction under the
more than 10 per cent and norre deviates by more than 20 per
condition of constant surface area. Its assessment is useful in
cent.
the characterisation of active substances and excipients.
The dissolution rate of pure substances can be affected by all
the solid state properties such as crystal habit, crystallinity,
amorphism, polymorphism, pseudo-polymorphism, particle
size and specific surface area. In addition, it can also be

General Notices (1) apply to all monographs and other texts 331
2.9.29. Intrinsic dissolution EUROPEAN PHARMACOPOEIA 8.0

influenced by extrinsie factors (test conditions), such as the powder to be tested is placed. The punch is then inserted
hydrodynamics, temperature, viscosity, pH, buffer strength in the die cavity and the material is compressed, generally
and ionie strength of the dissolution medium. using a benchtop hydraulic press. A hole through the head of
The assessment of intrinsie dissolution rate of a solid the punch allows insertion of a metal rod to facilitate removal
substance involves the preparation of a compact. Assurance of fram the die after the test. A compact is formed in the cavity
appropriate compaction properties of the powder to be tested with a single face of defined are a exposed on the bottom of
is needed prior to performing the test. the die (Figure 2.9.29.-1). The bottom of the die cavity is
threaded so that at ¡east 50-75 per cent of the compact can
The intrinsie dissolution rate is determined by exposing a
dissolve without falling out of the die. The top of the die has
constant are a of the compacted substance to an appropriate
a threaded shoulder that allows it to be attached to a holder.
dissolution medium, while maintaining constant stirring rate,
The holder is mounted on a laboratory stirring deviee, and the
temperature, ionic strength and pH.
entire die, with the compact still in place, is immersed in the
The intrinsic dissolution rate is expressed in terms of dissolved dissolution medium and rotated by the stirring device.
mass of substance per time per exposed area, typieally in
milligrams per minute per square centimetre (mg.min- 1·cm- 2 ).PROCEDURE
Weigh the material onto a piece of weighing papero Attach the
APPARATUS surface plate to the underside of the die, and secure it with the
A typical apparatus consists of a punch and die fabricated out 3 provided screws. Transfer the sample of powder tested into
of hardened steel. The base of the die has 3 threaded holes the die cavity. Place the punch into the chamber, and secure
for the attachment of a surface plate made of polished steel, the metal plate on the top of the assembly. Compress the
providing a mirror-smooth base for the compacto The die has powder using a hydraulic press by applying a suitable pressure
a 0.1-l.0 cm d¡ameter cavity into which a measured amount of for a sufficient dwell time to ensure a stable compact with

09.75 ± 0.35 - - - 1 F

041.1 ± 0.4

o 8.0 --il----_>{
E

A. Surface plate B. Die C. Neoprene gasket D. Punch E. Holder and shaft assembly F. Die underside

Figure 2.9.29.-1. - Typical apparatus used to obtain the compact for the determination of the intrinsic dissolution
Dimensions in millimetres

332 See the information section on general monographs (ca ver pages)
EUROPEAN PHARMACOPOEIA 8.0 2.9.31. Partide size analysis by laser Hght diffraction

minimal porosity; the disintegration of the compact has to be The technique cannot distinguish between scattering by single
prevented as far as possible, since it would cause an increase partieles and scattering by elusters of primary particles, i.e.
in surface area and hence in dissolution rateo Detach the by agglomerates or aggregates. As most particulate samples
surface plate, and screw the die with punch still in place into contain agglomerates or aggregates and as the focus of interest
the holder. Tighten securely. Remove allloose powder from is generally on the size distribution of primary particles, the
the surface of the die by blowing compressed air or nitrogen elusters are usually dispersed into primary partieles before
across the surface of the compact. measurement.
Slide the die-holder assembly into the dissolution test chuck Por non-spherical particles, an equivalent sphere-size
and tighten. Position the shaft in the spindle so that when the distribution is obtained because the technique assumes
test head is lowered, the exposed surface of the compact will spherical partieles in its optical model. The resulting
be 3.8 cm from the bottom of the vessel. The disc assembly is partiele-size distribution may differ from those obtained
aligned to minimise wobble and air bubbles are not allowed by methods based on other physical principies (e.g.
to form as this could decrease the compact surface in contact sedimentation, sieving).
with the dissolution medium. If possible, sink conditions are This chapter provides guidance for the measurement
maintained throughout the test. However, in order to obtain of size distributions of particles in different dispersed
detectable concentrations of solute, the use of a relatively small systems, for example, powders, sprays, aerosols, suspensions,
volume of medium may be necessary as a consequence of the emulsions, and gas bubbles in liquids, through analysis of
limited surface available for dissolution. their angular Iight -scattering patterns. It does not address
Warm the dissolution medium to the temperature chosen for specific requirements of partiele size measurement of specific
the test. Lower the test head into position before rotation. products.
Care should be taken to ensure that air bubbles are exeluded
from the surface of the compact as this could decrease the PRINCIPLE
compact surface in contact with the dissolution medium. A representative sample, dispersed at an adequate
Operate the apparatus immediately at the speed of rotation concentration in a suitable liquid or gas, is passed through
chosen for the test. a beam of monochromatic light, usually a laser. The light
Collect samples at fixed time intervals and assay them by scattered by the particles at various angles is measured by a
means of an analytical method of suitable sensitivity and multi-element detector. Numerical values representing the
accuracy. scattering pattern are then recorded for subsequent analysis.
These scattering pattern values are then transformed, using an
ASSESSMENT OF THE RESULTS appropriate optical model and mathematical procedure, to
The data for the cumulative amount dissolved at each time yield the proportion of total volume to a discrete number of
point are corrected for sampling losses. To calculate the size elasses, forming a volumetric partiele-size distribution.
intrinsic dissolution rate, plot the cumulative amount of
sample dissolved per unit area of the compact against time. INSTRUMENT
The cumulative amount dissolved per unit area is given by the The instrument is located in an environment where it is
cumulative amount dissolved at each time point divided by the not affected by electrical noise, mechanical vibrations,
surface area exposed. Linear regression is then performed on temperature fluctuations, humidity or direct bright light.
the normalised experimental data relevant to an appropriate An example of a set-up of a laser light diffraction instrument
time interva! preceding the possible disintegration of the is given in Figure 2.9.31.-1. Other equipment may be used.
compacto The intrinsic dissolution rate of the substance tested, The instrument comprises a laser light source, beam
expressed in milligrams per minute per square centimetre, processing optics, a sample measurement region (or ceH), a
is determined from the slope of the regression lineo The Fourier lens, and a multi-element detector for measuring the
result for intrinsic dissolution rate must be accompanied by scattered light pattern. A data system is also required for
a statement of the precise conditions of compact preparation decol1volution of the scattering data into a volumetric size
and test method (dissolution medium, volume of medium distribution and associated data analysis and reporting.
used, stirring rate, temperature etc.).
The partieles can enter the laser beam in 2 positions. In
NOTE: when necessary and justified, an apparatus with a the conventional case the particles enter the parallel beam
different configuration may be used, such as a die holder that before the collecting lens and within its working distance. In
holds the compact in a fixed vertical position, with agitation so-called reversed Fourier optics the particles enter behind the
provided by a paddle positioned at a defined distance from the collecting lens and thus, in a converging beam. The advantage
surface of the compact. of the conventional set-up is that a reasonable path length for
the sample is allowed within the working distan ce of the lens.
The second set-up allows only small path lengths but enables
01/2010:20931 measurement of scattered light at larger angles, which is useful
when submicron partieles are present.
2.9.31. PARTIeLE SIZE ANALYSIS The interaction of the incident light beam and the ensemble of
dispersed partieles results in a scattering pattern with different
BY LASER LIGHT DIFFRACTION light intensities at various angles. The total angular intensity
The method is based on the ISO standards 13320-1(1999) and distribution, consisting of both direct and scattered light,
9276-1(1998). is then focused onto a multi-element detector by a lens or a
series of lenses. These lenses create a scattering pattern that,
INTRODUCTION within limits, does not depend on the location of the partieles
The laser light diffraction technique used for the determination in the light beam. Hence, the continuous angular intensity
of partiele-size distribution is based on the analysis of the distribution is converted into a discrete spatial intensity
diffraction pattern produced when partieles are exposed distribution on a set of detector elements.
to a beam of monochromatic light. Historically, the early It is assumed that the measured scattering pattern of the
laser diffraction instruments only used scattering at small partiele ensemble is identical to the sum of the patterns from
angles. However, the technique has sin ce been broadened to al! individual single scattering partieles presented in random
inelude laser light scattering in a wider angular range and relative positions. Note that only a Iimited angular range of
application of the Mie theory, in addition to the Fraunhofer scattered light is collected by the lens( es) and, therefore, by
approximation and anomalous diffraction. the detector.

General Notices (1) apply to all monographs and other texts 333
2.9.31. Partide size analysis by laser Hght diffraction EUROPEAN PHARMACOPOEIA 8.0

9 11

8 10
7 8
8 '--o

- -~-~-~--+~-t===t
(1)
3
I
2
4
5
l. Obscuration detector 5. Scattered light not collected by lens (4) 9. Working distance oflens (4)

2. Scattered beam 6. Particle ensemble 10. Multi~element detector

3. Direct beam 7. Light source laser 1l. Focal distance oflens (4)

4. Fourier lens 8. Beam processing unit

Figure 2.9.31.~ 1. ~ Example of a set~up of a laser light diffraction instrument


DEVELOPMENT OF THE METHOD stirrer and ultrasonic elements, a pump, and tubing.
The measurement of particle size by laser diffraction can give Non~recirculating, stirred ceUs are useful when only small
reproducible data, even in the sub~micron region, provided the amounts of a sample are available or when special dispersion
instrument used and the sample tested are carefully controlled liquids are used.
to limit variability of the test conditions (e.g. dispersion Dry powders can also be converted into aerosols through the
medium, method of preparation of the sample dispersion). use of suitable dry powder dispersers, which apply mechanical
Traditionally, the measurement of particle size using laser force for deagglomeration or deaggregation. Generally, the
diffraction has been limited to particles in the range of dispersers use the energy of compressed gas or the differential
approximately 0.1 f.lm to 3 mm. Because of recent advances pressure of a vacuum to disperse the particles to an aerosol,
in len s and equipment design, newer instruments are capable which is blown through the measuring zone, usually into the
of exceeding this range routinely. With the validation report inlet of a vacuum unit that collects the particles. However, for
the user demonstrates the applicability of the method for its free flowing, coarser particles or granules the effect of gravity
intended use. may be sufficient to disperse the particles adequately.
Sampling. The sampling technique must be adequate to lf the maximum particle size of the sample exceeds the
obtain a representative sample of a suitable volume for the measuring range of the instrument, the material that is too
particle~size measurement. Sample splitting techniques such coarse can be removed by sieving and the mass and percentage
as rotating riffler or the con e and quartering method may be of removed material are reported. However, after pre-sieving,
applied. note that the sample is no longer representative, unless
otherwise proven.
Evaluation of the dispersion procedure. Inspect the
sample to be analysed, visualIy or with the aid of a Optimisation of the liquid dispersiol1. Liquids, surfactants,
microscope, to estimate its size range and particle shape. The and dispersing aids used to disperse powders must:
dispersion procedure must be adjusted to the purpose of the - be transparent at the laser wavelength and practically free
measurement. The purpose may be such that it is preferable from air bubbles or particles;
to deagglomerate clusters into primary particles as far as
- have a refractive index that differs from that of the test
possible, or it may be desirable to retain clusters as intact as
material;
possible. In this sense, the particles of interest may be either
primary particles or clusters. - be non ~solvent of the test material (pure liquid or
pre~filtered, saturated solution);
For the development of a method it is highly advisable to
check that comminution of the particles does not occur, - not alter the size of the test materials (e.g. by solubility,
and conversely, that dispersion of particles or clusters solubility enhancement, or recrystallisation effects);
is satisfactory. This can usually be done by changing - favour easy formation and stability of the dispersion;
the dispersing energy and monitoring the change of the
- be compatible with the materials used in the instrument
particle~size distribution. The measured size distribution must
(such as O~rings, gaskets, tubing, etc.);
not change significantly when the sample is well dispersed and
the particles are neither fragile nor soluble. Moreover, if the - possess a suitable viscosity to facilitate recirculation,
manufacturing process (e.g. crystallisation, milling) of the stirring and filtration.
material has changed, the applicability of the method must be Surfactants and/or dispersing aids are often used to wet the
verified (e.g. by microscopic comparison). particles and to stabilise the dispersion. For weak acids and
Sprays, aerosols and gas bubbles in a liquid should be weak bases, buffering of the dispersing medium at low or high
measured directly, provided that their concentration is pH respectively can assist in identifying a suitable dispersant.
adequate, because sampling or dilution generally alters the A preliminary check of the dispersion quality can be
particle~size distribution. performed by visual or microscopic inspection. It is also
In other cases (such as emulsions, pastes and powders), possible to take fractional samples out of a well~mixed stock
representative samples may be dispersed in suitable liquids. dispersion. Such stock dispersions are formed by adding a
Dispersing aids (wetting agents, stabilisers) and/or mechanical liquid to the sample while mixing it with, for example, a glass
forces (e.g. agitation, sonication) are often applied for rod, a spatula or a vortex mixer. Care must be taken to ensure
deagglomeration or deaggregation of clusters and stabilisation the transfer of a representative sample and that settling of
of the dispersion. For these liquid dispersions, a recirculating larger particles does not occur. Therefore a sample paste is
system is most commonly used, consisting of an optical prepared or sampling is carried out quickly from a suspension
measuring cel!, a dispersion bath usually equipped with maintained under agitation.

334 See the information section on general monographs (ca ver pages)
EUROPEAN PHARMACOPOEIA 8.0 2.9.31. Partide size analysis by laser light diffraction

Optimisation of the gas dispersion. For sprays and dry depends mostly on the instrument hardware. Accuracy should
powder dispersions, a compressed gas free from oil, water be confirmed through an appropriate instrument qualification
and particles may be used. To remove such materials from and comparison with microscopy, while precision may be
the compressed gas, a dryer with a filter can be used. Any assessed by means of a repeatability determination.
vacuum unit should be located away from the measurement
The attainable repeatability of the method mainly depends
zone, so that its output do es not disturb the measurement.
on the characteristics of the material (milled/not milled,
Determination of the concentration mnge. In order to robust/fragile, width of its size distribution, etc.), whereas
produce an acceptable signal-to-noise ratio in the detector, the required repeatability depends on the purpose of the
the particle concentration in the dispersion must exceed a measurement. Mandatory limits cannot be specified in this
minimum leve!. Likewise, it must be below a maximum level chapter, as repeatabilities (different sample preparations) may
in order to avoid multiple scattering. The concentration range vary appreciably from one substance to another. However, it
is influenced by the width of the laser beam, the path length of is good practice to aim at acceptance criteria for repeatability
the measurement zone, the optical properties of the particles, such as srel :;:; 10 per cent [n == 6] for any central value of the
and the sensitivity of the detector elements. distribution (e.g. for x so ). Values at the sides of the distribution
In view of the aboye, measurements must be performed at (e.g. x¡O and x 90 ) are oriented towards less stringent acceptance
different particle concentrations to determine the appropriate criteria such as S,el :;:; 15 per cent [n == 6]. Below 10 ~lm, these
concentration range for any typical sample of material. (Note: values must be doubled. Robustness may be tested during the
in different instruments, particle concentrations are usually selection and optimisation of the dispersion media and forces.
represented by differently scaled and differently named The change of the dispersing energy may be monitored by the
numbers, e.g. obscuration, optical concentration, proportional change in the particle-size distribution.
number of total mass).
MEASUREMENT
Determination of the measuring time. The time of
measurement, the reading time of the detector and the Precautions. The instructions given in the instrument manual
acquisition frequency are determined experimentally in are followed:
accordance with the required precision. Generally, the time - never look into the direct path of the laser beam or its
for measurement permits a large number of detector scans or reflections;
sweeps at short time intervals.
- earth al! instrument components to prevent ignition of
Selection of an appropriate optical model. Most instruments solvents or dust explosions;
use either the Fraunhofer or the Mie theory, though other
approximation theories are sometimes applied for calculation - check the instrument set-up (e.g. warm-up, required
of the scattering matrix. The choice of the theoretical model measuring range and lens, appropriate working distan ce,
depends on the intended application and the different position of the detector, no direct bright daylight);
assumptions (size, absorbance, refractive index, roughness, - in the case oí wet dispersions, avoid air bubbles,
crystal orientation, mixture, etc.) made for the test material. evaporation of liquid, schlieren or other inhomogeneities
lf the refractive index values (real and imaginary parts in the dispersion; similarly, avoid improper mass-flow
for the used wavelength) are not exactly known, then the from the disperser or turbulent air-flow in the case of dry
Fraunhofer approximation or the Mie theory with a realistic dispersions; such effects can cause erroneous particle-size
estimate of the refractive index can be used. The former distributions.
has the advantages that it is simple and it does not need
Measurement of the light scattering of dispersed sample( s).
refractive index values; the latter usually provides less-biased
particle-size distributions for smal! particles. For instance, After proper alignment of the optical part of the instrument,
if the Fraunhofer model is used for samples containing a blank measurement of the partide-free dispersion medium
must be performed using the same method as that used for the
an appreciable amount of small, transparent particles,
a significantly larger amount of small particles may be measurement of the sample. The background signal must be
calculated. In order to obtain traceable results, it is essential below an appropriate threshold. The detector data are saved
to document the refractive index values used, since small in order to substract them later from the data obtained with
differences in the values assumed for the real and imaginary the sample. The sample dispersion is measured according to
part of the complex refractive index may cause significant the developed method.
differences in the resulting particle-size distributions. Small For each detector element, an average signal is calculated,
values of the imaginary part of the refractive index (about sometimes together with its standard deviation. The
0.01-0.1 i) are often applied to allow the correction of the magnitude of the signal from each detector element depends
absorbance for the surface roughness of the particles. It upon the detection area, the light intensity and the quantum
should be noted, in general, that the optical properties of the efficiency. The co-ordinates (size and position) of the
substance to be tested, as well as the structure (e.g. shape, detector elements together with the focal distance of the lens
surface roughness and porosity), bear upon the final resulto determine the range of scattering angles for each element.
Validation. Typically, the validity of a procedure may be Most instruments also measure the intensity of the central
assessed by the evaluation of its specificity, linearity, range, (unscattered) laser beam. The ratio of the intensity of a
accuracy, precision and robustness. In particle-size analysis dispersed sample to that in its absence (a blank measurement)
by laser light diffraction, specificity as defined by ICH is indicates the proportion of scattered light and hence the
not applicable as it is not possible to discriminate between partide concentration.
different components in a sample, nor is it possible to Conversion of scattering pattern inlo partide-size
discriminate agglomerates from dispersed particles unless distribution. This deconvolution step is the inverse of the
properly complemented by microscopic techniques. Exploring calculation of a scattering pattern for a given partide-size
a linear relationship between concentration and response, or distribution. The assumption of spherical particle shape
a mathematical model for interpolation, is not applicable to is particularly important as most algorithms use the
this procedure. Rather than evaluating linearity, this method mathematical solution for scattering from spherical partides.
requires the definition of a concentration range within which Furthermore, the measured data always contain sorne random
the result of the measurements does not vary significantly. and systematic errors, which may vitiate the size distributions.
Concentrations below that range produce an error due to a Several mathematical procedures have been developed
poor signal-to-noise ratio, while concentrations aboye that for use in the available instruments. They contain sorne
range produce an error due to multiple scattering. The range weighting of deviations between measured and calculated

General Natices (1) apply ta all managraphs and ather texts 335
2.9.32. Porosay and pore-size distribution by mercury porosimetry EUROPEAN PHARMACOPOElA 8.0

scattering patterns (e.g. least squares), some constraints agreed, detailed operating procedure. The use of reference
(e.g. non-negativity for amounts of particles), and/or some values from methods other than laser diffraction may
smoothing of the size distribution curve. cause a significant bias. The reason for this bias is that the
The algorithms used are specific to each make and model different principIes inherent in the various methods may
of equipment, and are proprietary. The differences in the lead to different sphere-equivalent diameters for the same
algorithms between different instruments may give rise to non-spherical particle.
differences in the calculated particle-size distributions. Although the use of certified reference materials is preferred,
other well-defined reference materials may also be employed.
Replicates. The number of replicate measurements (with
They consist of substances of typical composition and
individual sample preparations) to be performed depends on
the required measurement precision. It is recommended to set particle-size distribution for a specified class of substances.
this number in a substance-specific method. Their particle-size distribution has proven to be stable over
time. The results must comply with previously determined
REPORTING OF RESULTS data, with the same precision and bias as fOI the certified
reference material.
The particle-size distribution data are usually reported
as cumulative undersize distribution and/or as density Qualification of fue system. In addition to the calibration,
distribution by volume. The symbol x is used to denote the the performance of the instrument must be qualified at regular
particle size, which in turn is defined as the diameter of a time intervals or as frequently as appropriate. This can be
volume-equivalent sphere. Q3(x) denotes the volume fraction undertaken using any suitable reference material as mentioned
undersize at the particle size x. In a graphical representation, in the previous paragraph.
x is plotted on the abscissa and the dependent variable Q3 The qualification of the system is based on the concept that
on the ordinate. Most common characteristic values are the equipment, e1ectronics, software and analytical operations
calculated from the particle-size distribution by interpolation. constitute an integral system, which can be evaluated as an
The particle sizes at the undersize values of 10 per cent, 50 per entity. Thus the entire measurement procedure is examined,
cent, and 90 per cent (denoted as xlO' x so' and x go respectively) including sample collection, sample dispersion, sample
are frequently used. x so is also known as the median particle transport through the measuring zone, and the measurement
size. It is recognised that the symbol d is also widely used to and deconvolution procedure. It is essential that the total
designate the particle size, thus the symbol x may be replaced operational procedure is fully described.
byd. In general, unless otherwise specified in the individual
Moreover, sufficient information must be documented about monograph, the response of a laser diffraction instrument
the sample, the sample preparation, the dispersion conditions, is considered to meet the requirements if the x so value does
and the ceH type. As the results depend on the particular not deviate by more than 10 per cent from the range of values
instrument, data analysis program, and optical model used, of the reference materiaL If optionally the values at the sides
these details must also be documented. of the distribution are evaluated (e.g. x lO and x 90 ), then these
values must not deviate by more than 15 per cent from the
CONTROL OF THE INSTRUMENT PERFORMANCE certified range of values. Below 10 flm, these values must be
Use the instrument according to the manufacturer's doubled.
instructions and carry out the prescribed qualifications at an NOTE: for calibration of the instrument, stricter requirements
appropriate frequency, according to the use of the instrument are laid down in the paragraph Calibration.
and substances to be tested.
Calibration. Laser diffraction systems, although assuming 0712008:20932
idealised properties of the particles, are based on first
principIes of laser light scattering. Thus, calibration in the
strict sense is not required. However, it is still necessary to 2.9.32. POROSITY AND PORE-SIZE
confirm that the instrument is operating correctly. This can DISTRIBUTION OF SOLIDS BY
be undertaken using any certified reference material that is MERCURY POROSIMETRY
acceptable in industrial practice. The entire measurement
procedure is examined, including sample collection, sample INTRODUCTION
dispersion, sample transport through the measuring zone, In general, different types of pores may be pictured as
measurement, and the deconvolution procedure. It is essential apertures, channels OI cavities within a solid body, or as space
that the total operational procedure is fully described. (i.e. interstices or voids) between solid particles in a bed,
The preferred certified reference materials consist of spherical compact or aggregate. Porosity is a term that is often used
particles of a known distribution. They must be certified to indicate the porous nature of solid material, and is more
as to the mass-percentage size distribution by an absolute precisely defined as the ratio of the volume of accessible pores
technique, if available, and used in conjunction with an agreed, and voids to the total volume occupied by a given amount
detailed operation procedure. It is essential that the real and of the solido In addition to the accessible pores, a solid may
imaginary parts of the complex refractive index of the material contain closed pores, which are isolated from the external
are indicated if the Mie theory is applied in data analysis. The surface and into which fluids are not able to penetrate. The
representation of the particle-size distribution by volume characterisatioll oí closed pores, i.e. cavities with no access to
will equal that of the distribution by mass, provided that the an external surface, is not covered in this chapter.
density of the particles is the same for all size fractions. Porous materials may take the form of fine or coarse
The response of a laser diffraction instrument is considered to powders, compacts, extrudates, sheets or monoliths. Their
meet the requirements if the mean value of x so from at least characterisation usually involves the determination of the total
3 independent measurements do es not deviate by more than pore volume or porosity as well as the pore-size distribution.
3 per cent from the certified range of values of the certified It is well established that the performance of a porous solid
reference material. The mean values for x¡o and X gO must not (e.g. its strength, reactivity, permeability or adsorbent power)
deviate by more than 5 per cent from the certified range of is dependent upon its pore structure. Many different methods
values. Below 10 flm, these values must be doubled. have been developed for the characterisation of pore structure.
Although the use of materials consisting of spherical particles In view of the complexity of most porous solids, it is not
is preferable, non -spherical particles may also be employed. surprising to find that the results obtained are not always in
Preferably, these particles have certified or typical values agreement and that no single technique can be relied upon to
from laser diffraction analyses performed according to an provide a complete picture of the pore structure. The choice

336 See the information section on general rnonographs (cover pages)


EUROPEAN PHARMACOPOEIA 8.0 2.9.32. Porosity and pore-size distribution by mercury porosimetry

of the most appropriate method depends on the application between the mercury column in the capillary tube and a metal
of the porous solid, its chemical and physical nature and the sleeve around the outside of the capillary tube. lf precise
range of pore-size. measurements are required the expected total void and pore
This chapter provides guidance for measurement of porosity volume of the sample should be between 20 per cent and
90 per cent of the internal volume of the capillary tube. Since
and pore-size distribution by mercury porosimetry. It is a
different materials exhibit a wide range of open poro sities,
comparative test, usually destructive, in which the volume of
mercury penetrating a pore or void is determined as a function a number of penetrometers with different capillary tube
of an applied hydrostatic pressure, which can be related to diameters and sample volumes may be required. A typical
a pore diameter. Other information such as pore shape and set-up for a mercury porosimeter instrument is given in
inter-connectivity, the internal and external surface area, Figure 2.9.32.-1. The porosimeter may have separate ports
for high- and low-pressure operation, or the low-pressure
powder granulometry, bulk and tapped density could also be
inferred from volume-pressure curves; however, these aspects measurement may be carried out on a separate unit.
of the technique do not fall under the scope of this chapter. The pressure range is typically 4-300 kPa for low-pressure
Practical considerations presently limit the maximum applied operation and aboye 300 kPa for high-pressure operation,
absolute pressure reached by sorne equipment to about depending on the design of the particular apparatus and on
400 MPa, corresponding to a minimum equivalent pore the intended use.
diameter of approximately 0.003 [lm. The maximum diameter
will be limited for samples having a significant depth due to METHOD
the difference in hydrostatic head of mercury from the top to Sample preparation
the bottom of the sample. For most purposes this limit may
be regarded as 400 [lm. The sample is pre-treated to remove adsorbed material that
can obscure its accessible porosity, for example by heating
Inter-particle and intra-particle porosity can be determined, and/or evacuation, or by flowing inert gas. It may be possible
but the method does not distinguish between these porosities to passivate the surface of wettable or amalgam-forming
where they co-exist. solids, for example by producing a thin layer of oxide, or by
The method is suitable for the study of most porous materials. coating with stearate.
Samples that amalgamate with mercury, such as certain The sample of the pre-treated solid is weighed and transferred
metals, may be unsuitable for this technique or may require to the penetrometer. The pore system of the sample is then
a preliminary passivation. Other materials may deform or degassed in a vacuum to a maximum residual pressure of 7 Pa.
compact under the applied pressure. In sorne cases it may
be possible to apply sample-compressibility corrections and Filling the penetrometer with mercury
useful comparative data may still be obtained. The mercury used is of analytical quality. Overlay the sample
Mercury porosimetry is considered to be comparative, as with mercury under vacuum. The vacuum is required to
for most porous media a theory is not available to allow ensure the transfer of mercury from the reservoir to the
an absolute calculation of results of pore-size distribution. penetrometer. In a filled penetrometer the filling pressure
Therefore this technique is mainly recommended for comprises the applied pressure plus the pressure contribution
development studies. created by the head of mercury contacting the sample. A
typical filling pressure would be about 4 kPa. The hydrostatic
Mercury is toxico Appropriate precautions must be observed pressure of the mercury over the sample may be minimised by
to safeguard the health of the operator and others working in filling the penetrometer in the horizontal position.
the area. Waste material must also be disposed of in a suitable
manner, according to local regulations. Low-pressure measurement
Admit air or nitro gen in a controlled manner to increase
PRINCIPLE the pressure either in stages corresponding to the particular
The technique is based on the measurement of the mercury pore sizes of interest, or continuously at a slow rateo The
volume intruded into a porous solid as a function of the concomitant change in the length of the mercury column in
applied pressure. The measurement includes only those pores the capillary tube is recorded. When the maximum required
into which mercury can penetrate at the pressure applied. pressure has been reached, return to atmospheric pressure.

A non-wetting liquid penetrates into a porous system only High-pressure measurement


under pressure. The pressure to be applied is in inverse After measurement at low pressure, the penetrometer filled
proportion to the inner diameter of the pore aperture. In with mercury is transferred to the high-pressure port or unit
the case of cylindrical pores, the correlation between pore of the instrument and overlaid with hydraulic fluid. Mercury
diameter and pressure is given by the Washburn equation: is intruded into the pore system via the hydraulic fluid.
lncrease the pressure in the system to the maximum pressure
dp= - 4jf cos e reached in the low-pressure measurement and record the
intrusion volume at this pressure, since subsequent intrusion
volumes are calculated from this initial volume. Increase the
dp pore diameter, in me tres ;
pressure either in stages corresponding to the particular pore
()' surface tension, in newtons per metre; sizes of interest, or continuously at a slow rateo The faH in the
mercury column is measured up to the maximum required
e contact angle of mercury on the sample, in degrees; pressure. lf required the pressure may be decreased either in
p applied pressure, in pascals. stages or continuously at a slow rate to determine the mercury
extrusion curve.
APPARATUS Corrections are made to take account of changes in the volume
of the mercury, the penetrometer and other components of
The sample holder, referred to as penetrometer or dilatometer,
the volume detector system under elevated pressure. The
has a calibrated capillary tube, through which the sample
extent of the corrections may be determined by means of
can be evacuated and through which mercury can entero
blank measurements under the same conditions.
The capillary tube is attached to a wider tube in which the
test sample is placed. The change in the volume of mercury An experimentally determined volume-pressure curve is
intruded is usually measured by the change in capacitance shown in Figure 2.9.32.-2.

General Natíces (1) apply ta all managraphs and ather texts 337
2.9.32. Porosity and pore-size distribution by mercury porosimetry EUROPEAN PHARMACOPOEIA 8.0

c;=J I
1
I
I
I
I
I
I
I
I
I

-8
A. Low-pressure hydraulic fluid E. High-pressure hydraulic fluid J. Penetration volume N. Sample
reservoir reservoir indicator
B. Hydraulic pump F. Vacuum pump with gauge K. Capillary tube

C. Pressure muItiplier G. Mercury reservoir 1. High -pressure chamber

D. Pressure transducer H.Oil M. Mercury

Figure 2.9.32.-1. - Example of the set-up of a mercury porosimeter instrument

Oí 300 :2i 300


1 '"E
$ 240 E
(j) ~240
E
:J E
o 180 ::J
> (5
> 180
""'ü"8. 120 ü

"'
(j)
t <;:::

:!i 120
> U)
~ 60 (J)
'3 >
E
:J ~ 60
U o 10' 10° 10' 10'
::J
E
::J
Pressure (MPa) Ü O
10° 10' 10' 10 3
Figure 2.9.32.-2. - Volume-pressure curve as semilogarithmic Pore diameter (nm)
plot

REPORTING OF RESULTS Figure 2.9.32.-3. - Pore-size distribution as semilogarithmic


plots of the cumulative and the normalised density distribution
The pressure readings can be converted to pore diameters by
means of the Washburn equation or by another model. Extrusion curves may not be used for calculating the pore-size
The surface tension of mercury (a) depends not only on the distribution (for hysteresis, see Figure 2.9.32.-2), because
temperature, but also, in the case of markedly curved surfaces an intruded part of the mercury always remains in the pore
areas, on the radius of curvature. In general, values between system. The retention ratio may however be useful for the
0.41 N·m- 1 and 0.52 N·m- 1 are measured at room temperature. qualitative characterisation of pores that are only accessible
lf the value is not known, (J = 0.48 N-m- 1 can be used. via narrow openings ('ink-bottle pores').
The contact angle of mercury (8) in most cases is more Most common characteristic values, such as the total intruded
than 90°. lt may be determined using a contact angle specific volume and the mean and median pore diameters,
instrument. lf the value is not known, 8 = 130° can be used. are calculated fram the pore-size distribution. Moreover,
The values of contact angle and surface tension and the model sufficient information must be documented about the sample,
used in the calculation are reported. the sample preparation, the evacuation conditions and the
Visualisation of the data can be done with several types instrument used.
of graphs. Frequently, in a graphical representation the
pore diameter is plotted on the abscissa and the intruded
volume per sample mass on the ordinate to give the pore-size CONTROL OF INSTRUMENT PERFORMANCE
distribution. It is apprapriate here to choose a logarithmic
scale for the abscissa (see Figure 2.9.32.-3). The spaces As mercury porosimetry is considered to be used as a
between the partides of the solid sample are induded as pores comparative test, no details are given in this chapter. However,
in the calculation. lf the pores differ in size from the voids, the it is recommended that a stable comparison material is tested
latter can be separated by choosing the appropriate pore-size on a regular basis to monitor instrument calibration and
range. performance.

338 See the information section on general monographs (ca ver pages)
EUROPEAN PHARMACOPOEIA 800 2.9.33. Characterisation of crystalline solids by XRPD

0112009:20933 These interferences are constructive when the path difference


between 2 diffracted X -ray waves differs by an integral number
of wavelengthso This selective condition is described by the
2.9.33. CHARACTERISATION OF Bragg equation, also called Bragg's law (see Figure 2090330-1):
CRYSTALLINE AND PARTIALLY
CRYSTALLINE SOLIDS BY X-RAY
POWDER DIFFRACTION (XRPD) The wavelength A of the X -rays is of the same order of
magnitude as the distan ce between successive crystallattice
Every crystalline phase of a given substance produces a planes, or dhkl (also called 'd-spacings')o 8hkl is the angle
characteristic X -ray diffraction patterno between the incident ray and the family of lattice planes,
and sin8 hk1 is inversely proportional to the distance between
Diffraction patterns can be obtained from a randomly successive crystal planes or d-spacingso
oriented crystalline powder composed of crystallites or crystal
fragments of finite sizeo Essentially 3 types of information can The direction and spacing of the planes with reference to
be derived from a powder diffraction pattern: angular position the unit ceH axes are defined by the Miller índices {hkl}o These
of diffraction lines (depending on geometry and size of indices are the reciprocals, reduced to the next-Iower integer,
the unit ceH); intensities of diffraction hnes (depending mainly of the intercepts that aplane makes with the unit cel! axes.
on atom type and arrangement, and particle orientation The unit cel! dimensions are given by the spacings a, b and e
within the sample) ; and diffraction line profiles (depending on and the angles between them, G, ~, and yo
instrumental resolution, crystallite size, strain and specimen The interplanar spacing for a specified set of parallel hkl
thickness)o planes is denoted by dhk10 Each such family of planes may show
higher orders of diffraction where the d values for the related
Experiments giving angular positions and intensities of families of planes nh, nk, ni are diminished by the factor 1/n
hnes can be used for applications such as qualitative phase (n being an integer: 2, 3, 4, etc)
analysis (for example, identification of crystalline phases)
and quantitative phase analysis of crystalline materialso An Every set of planes throughout a crystal has a corresponding
estimate of the amorphous and crystalline fractions(10) can Bragg diffraction angle, 8hkl , associated with it (for a specific
also be madeo wavelength Al.
A powder specimen is assumed to be polycrystalline so that
The X-ray powder diffraction (XRPD) method provides an
at any angle 8hkl there are always crystallites in an oríentation
advantage over other means of analysis in that it is usually
allowing diffraction according to Bragg's law(J 1). For a given
non-destructive in nature (specimen preparation is usually
X-ray wavelength, the positions of the diffraction peaks
limited to grinding to ensure a randomly oriented sample)o
(also referred to as 'lines; 'reflections' or 'Bragg reflections')
XRPD investigations can also be carried out under in situ
are characteristic of the crystallattice (d-spacings), their
conditions on specimens exposed to non-ambient conditions,
theoretical intensities depend on the crystallographic unit ceH
such as low or high temperature and humidity
content (nature and positions of atoms), and the line profiles
on the perfection and extmt of the crystallatticeo Under these
PRINCIPLE conditions the diffraction peak has a finite il1tensity arising
X-ray diffraction results from the interaction between X-rays from atomic arrangement, type of atoms, thermal motion
and electron clouds of atomso Depending on the atomic and structural imperfections, as well as from instrument
arrangement, interferences arise from the scattered X-rayso characteristicso

/~ /
/
/
/

Figure 2090330-L - Diffraction of X-rays by a crystal according to Bragg's law

(lO) There are many other applications of the X-ray powder diffraction technique that can be applied to crystalline pharmaceutical substances such as: determination of cryslal structl1res,
refinement of crystal structures, determination of crystallographic purity of erystalline phases, characterisation of crystallographic texture, etc. These applications are not described in
this ehapter.
(11) An 'ideal' powder for diffraetion experiments eonsists of a large number uf small, randomly oriented spherical crystallites (coherently diffracting crystalline domains). If this numbcr is
sufficiently large, lhere are always enough crystallites in any diffracting orientation to give reproducible diffraction patterns.

General Notiees (1) apply to all monographs and other texts 339
2.9.33. Characterisation of crystalline solids by XRPD EUROPEAN PHARMACOPOEIA 8.0

The intensity is dependent upon many factors such as Depending on the type of analysis to be performed
structure factor, temperature factor, crystallinity, polarisation (phase identification, quantitative analysis, lattice
factor, multiplicity and Lorentz factor. parameters determination, ete.), different XRPD instrument
The main characteristics of diffraction line pro files configurations and performance levels are required. The
are 28 position, peak height, peak area and shape simplest instruments used to measure XRPD patterns are
(characterised by, for example, peak width or asymmetry, powder cameras. The replacement of photographic film as
analytical function, empirical representation). An example the detection method by photon detectors has led to the
of the type of powder patterns obtained for 5 different solid design of diffractometers in which the geometric arrangement
phases of a substance are shown in Figure 2.9.33.-2. of the optics is not truly focusing but parafocusing, such
as in the Bragg-Brentano geometry. The Bragg-Brentano
In addition to the diffraction peaks, an X-ray diffraction parafocusing configuration is currently the most widely used
experiment also generates a more-or-less uniform background, and is therefore briefly described here.
upon which the peaks are superimposed. Besides specimen
A given instrument may provide a horizontal or vertical
preparation, other factors contribute to the background,
for instan ce the sample holder, diffuse scattering from air 8/28 geometry or a vertical 8/8 geometry. For both geometries,
and equipment, other instrumental parameters such as the incident X-ray beam forms an angle 8 with the specimen
surface plane and the diffracted X -ray beam forms an angle 28
detector noise, general radiation from the X-ray tube, ete. The
with the direction of the incident X-ray beam (an angle
peak-to-background ratio can be increased by minimising
8 with the specimen surface plane). The basic geometric
background and by choosing prolonged exposure times.
arrangement is represented in Figure 2.9.33.-3. The divergent
INSTRUMENT beam of radiation from the X-ray tube (the so-called 'primary
beam') passes through the parallel plate collimators and a
Instrument set-up. X-ray diffraction experiments are usually divergence slit assembly and illuminates the flat surface of
performed using powder diffractometers 01' powder cameras. the specimen. AH the rays diffracted by suitably oriented
A powder diffractometer gene rally comprises 5 main parts: crystallites in the specimen at an angle 28 converge to a line
an X-ray source; incident beam optics, which may perform at the receiving slit. A second set of parallel plate collimators
monochromatisation, filtering, collimation and/or focusing and a scatter slit may be placed either behind or befo re the
of the beam; a goniometer; diffraction beam optics, which receiving slit. The axes of the line focus and of the receiving
may perform monochromatisation, filtering, collimation slit are at equal distan ces from the axis of the goniometer. The
and focusing or parallelising of the beam; and a detector. X-ray quanta are counted by a radiation detector, usually a
Data-coHection and data-processing systems are also required scintillation counter, a sealed-gas proportional counter, or a
and are generally included in current diffraction measurement position -sensitive solid -state detector such as imaging plate or
equipment. CCD detector. The receiving slit assembly and the detector are

Form C

Form A

amorphous

I I I I i I 1 I I T~I----¡---ITI--' I 1 I i I I '-:--r-r
ii-'-n-l-T-r--¡--I i I I
7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29' 30 31

2eo" Cu) - Scale


Figure 2.9.33.-2. - X-ray powder diffraction patterns collected for 5 different solid phases of a substance
(the intensities are normalised)

340 See the information section on general monographs (caver pages)


EUROPEAN PHARMACOPOEIA 8.0 2.9.33. Characterisation of crystaUine solids by XRPD

/
/

/
/ H

\
/
F

\ /

/
A. X-ray tube C. sample E. receiving slit G. detector receiving slit J. diffractometer cirde
B. divergence slit D. anti-diffusion slit F. monochromator H. detector K. focusing cirde

Figure 2.9.33.-3. - Geometric arrangement of the Bragg-Brentano parafocusing geometry

coupled together and move tangentially to the focusing circle. obtained using K~ filters, i.e. metal filters selected as having an
For e/2e scans the goniometer rotates the specimen about the absorption edge between the Ka and K~ wavelengths emitted
same axis as that of the detector, but at half the rotational by the tube.
speed, in a e/2e motion. The surface of the specimen thus Such a filter is usually inserted between the X-ray tube
remains tangential to the focusing circle. The parallel plate and the specimen. Another, more-and-more-commonly
collimator limits the axial divergence of the beam and hence used way to obtain a monochromatic X-ray beam is via
partially controls the shape of the diffracted line pro file. a large monochromator crystal (usually referred to as a
A diffractometer may also be used in transmission mode. The 'monochromator'). This crystal is placed before or behind
advantage with this technology is to lessen the effects due to the specimen and diffracts the different characteristic peaks
preferred orientation. A capillary of about 0.5-2 mm thickness of the X-ray beam (i.e. Ka and K~) at different angles, so
can also be used for small sample amounts. that only one of them may be seIected to enter into the
X-ray radiation. In the laboratory, X-rays are obtained by detector. It is even possible to separate Ka! and Ka2 radiations
bombarding a metal anode with electrons emitted by the by using a specialised monochromator. Unfortunately, the
thermionic effect and accelerated in a strong electric field gain in getting a monochromatic beam by using a filter or a
(using a high -voltage generator). Most of the kinetic energy of monochromator is counteracted by a loss in intensity. Another
the electrons is converted to heat, which limits the power of way of separating Ka and K~ wavelengths is by using curved
the tubes and requires efficient anode cooling. A 20- to 30-fold X-rays mirrors that can simultaneously monochromate and
increase in brilliance can be obtained using rotating ano des focus or parallelise the X-ray beam.
and by using X-ray optics. Alternatively, X-ray photons may RADIATION PROTECTION. Exposure of any part ofthe
be produced in a large-scale facility (synchrotron). human body to X-rays can be injurious to health. It is therefore
The spectrum emitted by an X-ray tube operating at sufficient essential that whenever X-ray equipment is used, adequate
voltage consists of a continuous background of polychromatic precautions are taken to protect the operator and any other
radiation and additional characteristic radiation that depends person in the vicinity. Recommended practice for radiation
on the type of anode. Only this characteristic radiation is protection as well as limits for the levels of X-radiation exposure
used in X-ray diffraction experiments. The principal radiation are those established by nationallegislation in each country.
sources utilised for X -ray diffraction are vacuum tubes utilising If there are no official regulations or recommendations in
copper, molybdenum, iron, cobalt or chromium as anodes; a country, the la test recommendations of the International
copper, molybdenum or cobalt X-rays are employed most Commission on Radiological Protection should be applied.
commonly for organic substances (the use of cobalt anodes
can be especially preferred to separate distinct X -ray lines). SPECIMEN PREPARATION AND MOUNTING
The choice of radiation to be used depends on the absorption The preparation of the powdered material and mounting
characteristics of the specimen and possible fluorescence by of the specimen in a suitable holder are critical steps in
atoms present in the specimen. The wavelengths used in many analytical methods, and are particularly so for XRPD
powder diffraction generally correspond to the Ka radiation analysis, sin ce they can greatly affect the quality of the data to
from the anode. Consequently, it is advantageous to make be collected(J2). The main sources of error due to specimen
the X-ray beam 'monochromatic' by eliminating all the other preparation and mounting are briefly discussed here for
components of the emission spectrum. This can be partly instruments in Bragg-Brentano parafocusing geometry.

(12) Similarly, changes in the specimen can occur during elata colleclion in thc case o[ a non-equilibrium specimen (temperature, hurnidity).

General Notices (1) apply to all monographs and other texts 341
2.9.33. Characterisation of crystalline solids by XRPD EUROPEAN PHARMACOPOEIA 8.0

SPECIMEN PREPARATION influences the quality of the results of an XRPD investigation.


In general, the morphology of many crystalline particles tends Therefore, the different components of the diffractometer
to give a specimen that exhibits sorne degree of preferred must be carefully adjusted (optical and mechanical systems,
orientation in the specimen holder. This is particularIy evident etc.) to minimise adequately systematic errors, while
for needle-like or plate-like crystals when size reduction optimising the intensities received by the detector. The search
yields finer needles or platelets. Preferred orientation in the for maximum intensity and maximum resolution is always
specimen influences the intensities of various reflections, antagonistic when aligning a diffractometer. Hence, the best
so that some are more intense and others are less intense, compromise must be sought whilst performing the alignl11ent
compared to what would be expected from a cornpletely procedure. There are many different conngurations and each
random specimen. Several techniques can be employed to supplier's equipment requires specinc alignment procedures.
improve randomness in the orientation of crystallites (and The overall diffractometer performance must be tested and
therefore to minimise preferred orientation), but further monitored periodically using suitable certified reference
reduction of particle size is often the best and simplest materials. Depending on the type of analysis, other
approach. The optimum number of crystallites depends well-denned reference materials may also be employed,
on the diffractometer geometry, the required resolution although the use of certified reference materials is preferred.
and the specimen attenuation of the X-ray beam. In some
cases, particle sizes as large as 50 !lm will provide satisfactory QUALITATIVE PHASE ANALYSIS (IDENTIFICATION OF
results in phase identification. However, excessive milling PHASES)
(crystallite sizes less than approximately 0.5 !lm) may cause The identification of the phase composition of an unknown
line broadening and significant changes to the sample itself sarnple by XRPD is usually based on the visual or
such as: computer-assisted comparison of a portian of its XRPD
- specimen contarnination by particles abraded from the pattern to the experimental or calculated pattern of a reference
milling instruments (mortar, pestle, balls, etc.); material. Ideally, these reference patterns are collected on
- reduced degree of crystallinity; well-characterised single-phase specirnens. This approach
makes it possible in most cases to identify a crystalline
- solid-state transition to another polymorph;
substance by its 28 diffracHon angles or d-spacings and by its
- chemical decomposition; relative intensities. The computer-aided comparison of the
- introduction of internal stress; diffraction pattern of the unknown sample to the comparison
data can be based either on a more-or-less extended 28-range
- solid -state reactions.
of the whole diffraction pattern or on a set of reduced data
Therefore, it is advisable to compare the diffraction pattern
derived from the pattern. For example, the list of d-spacings
of the non-ground specimen with that corresponding to a
and normalised intensities (Inmn,)' a so-called (d, InormHist
specimen of smaller particle size (e.g. a milled specimen). lf
extracted from the pattern, is the crystallographic fingerprint
the XRPD pattern obtained is of adequate quality considering
of the material, and can be compared to (d, InormHists of
its intended use, then grinding may not be required.
single-phase samples compiled in databases.
It should be noted that if a sample contains more than one
For most organic crystals, when using Cu radiation, it is
phase and if sieving is used to isolate particles to a specific
appropriate to record the diffraction pattern in a 28-range
size, the initial composition may be altered.
from as near 00 as possible to at least 40 0 • The agreement in
SPECIMEN MOUNTING the 28-diffraction angles between specimen and reference is
Effect of spedmen displacement A specimen surface that is within 0.2 for the same crystal form, while relative intensities
0

offset by D with reference to the diffractometer rotation axis between specimen and reference may vary considerably due
causes systematic errors that are very difficult to avoid entirely; to preferred orientation effects. By their very nature, variable
for the reflection mode, this results in absolute D·cos8 shifts(13) hydrates and solvates are recognised to have varying unit cell
in 28 positions (typically of the order of 0.01 ° in 28 at low dimensions and as such shifting occurs in peak positions of
angles (cos8 ~ 1) for a displacement D == 15 flm) and the measured XRPD patterns for these materials. In these
asymmetric broadening of the profile towards low 28 values. unique materials, variance in 28-positions of greater than 0.2 0

Use of an appropriate internal standard allows the detection is not unexpected. As such, peak position variances such as
and correction of this effect simultaneously with that arising 0.2 0 are not applicable to these materials. For other types of
from specimen transparency. This is by far the largest source samples (e.g. inorganic salts), it may be necessary to extend
of errors in data collected on well-aligned diffractometers. the 28-region scanned to well beyond 40°. It is generally
sufficient to scan past the 10 strongest reflections identified in
Effect of spedmen thickness and tnmsparency. When single phase XRPD database files.
the XRPD method in reflection mode is applied, it is often
preferable to work with specimens of 'infinite thickness'. It is sometimes difficult or even impossible to identify phases
To minimise the tral1sparency effect, it is advisable to use in the following cases:
a non-diffracting substrate (zero background holder), for - non-crystallised or amorphous substances;
exal11ple a plate of single crystalline sílicon cut parallel to - the components to be identified are present in low mass
the 510 lattice planes(14). One advantage of the transl11ission fractions of the analyte amounts (generally less than 10 per
mode is that problems with sample height and specimen cent m/m);
transparency are less important. The use of an appropriate
- pronounced preferred orientation effects;
internal standard allows the detection and correction of
this effect simultaneously with that arising from specimen - the phase has not been filed in the database used;
displacement. - formation of solid solutions;
- presence of disordered structures that alter the unit cell;
CONTROL OF THE INSTRUMENT PERFORMANCE
- the specimen comprises too many phases;
Goniometers and the corresponding incident and diffracted
X-ray beam optics have many mechanical parts that need - presence of lattice deformations;
adjustment. The degree of alignment or misalignment directly - structural similarity of different phases;

(13) Note that a goniometer zero alignment shift would result in constant shift on all observed 2e-Ene positions, in other words, the whole diffrachan pattern is in thi5 case translated by an
offset of ZO in 28.
(14) In the case of a thin with low attenuation, accurate measurements of line positions can be made witb diffractometer configurations in either transmission or
reflection measurements of line positions Oil specimens with low attenuation are preferably made using \'I11tl1 parallel beam optics. Ihis helps to
reduce the effects tbickness.

342 See the information section on general monographs (ca ver pages)
EUROPEAN PHARMACOPOEIA 8.0 2.9.34. Bulle density and tapped density of powders

QUANTITATIVE PHASE ANALYSIS - if the sample consists of crystalline fractions and an


If the sample under investigation is a mixture of 2 or more amorphous fraction of different chemical compositions,
known phases, of which not more than 1 is amorphous, the amounts of each of the individual crystalline phases
the percentage (by volume or by mass) of each crystalline may be estimated using appropriate standard substances as
phase and of the amorphous phase can, in many cases, be described above; the amorphous fraction is then deduced
determined. Quantitative phase analysis can be based on indirectly by subtraction;
the integrated intensities, on the peak heights of several - if the sample consists of one amorphous and one crystalline
individual diffraction lines(1S), or on the full pattern. These fraction, either as a 1- phase or a 2- phase mixture, with the
integrated intensities, peak heights or full-pattern data points same elemental composition, the amount of the crystalline
are compared to the corresponding values of reference phase ('the degree of crystallinity') can be estimated by
materials. These reference materials shall be single-phase or a measuring 3 are as of the diffractogram:
mixture of known phases. The difficulties encountered during A total are a of the peaks arising from diffraction
quantitative analysis are due to specimen preparation (the from the crystalline fraction of the sample;
accuracy and precision of the results require in particular
homogeneity of all phases and a suitable particle size B total are a below area A;
distribution in each phase) and to matrix effects. In favourable e background area (due to air scattering,
cases, amounts of crystalline phases as small as 10 per cent fluorescence, equipment, etc).
may be determined in solid matrices.
When these are as have been measured, the degree of
POLYMORPHlC SAMPLES crystallinity can be roughly estimated using the following
For a sample composed of 2 polymorphic phases a and b, the formula:
following expression may be used to quantify the fraction Fa
ofphase a: % crystallinity = 100A! (A +B - C)

It is noteworthy that this method do es 110t yield absolute


degree-of-crystallinity values and hence is generally used for
comparative purposes only.
The fraction is derived by measuring the intensity ratio More sophisticated methods are also available, such as the
between the 2 phases, knowing the value of the constant K. K is Ruland method.
the ratio of the absolute intensities of the 2 pure polymorphic
phases lo/lobo Its value can be determined by measuring SINGLE CRYSTAL STRUCTURE
standard samples. In general, the determination of crystal structures is performed
METHODS USlNG A STANDARD from X-ray diffraction data obtained using single crystals.
The most commonly used methods for quantitative analysis However, crystal structure analysis of organic crystals is a
are: challenging task, since the lattice parameters are comparatively
large, the symmetry is low and the scattering properties are
- the 'external standard method' ; normally very low.
- the 'internal standard method'; For any given crystalline form of a substance, knowledge of the
crystal structure allows the calculation of the corresponding
- the 'spiking method' (often also called the 'standard
XRPD pattern, thereby providing a 'preferred -orientation -free'
addition method').
reference XRPD pattern, which may be used for phase
The 'external standard method' is the most general method identification.
and consists of comparing the X-ray diffraction pattern of the
mixture, or the respective line intensities, with those measured
in a reference mixture or with the theoretical intensities of a
structural model, if it is fully known. 01/2013:20934

To limit errors due to matrix effects, an internal reference


material with crystallite size and X-ray absorption coefficient
2.9.34. BULK DENSITY AND TAPPED
comparable to those of the components of the sample, and DENSITY OF POWDERS
with a diffraction pattern that does not overlap at all that of
the sample to be analysed, can be used. A known quantity of Bulk density
this reference material is added to the sample to be analysed
and to each of the reference mixtures. Under these conditions, The bulk density of a powder is the ratio of the mass of
a linear relationship between line intensity and concentration an untapped powder sample to its volume, including the
exists. This application, called the 'internal standard method', contribution of the interparticulate void volume. Hence, the
requires a precise measurement of diffraction intensities. bulk density depends on both the density of powder particles
and the spatial arrangement of particles in the powder bed.
In the 'spiking method' (or 'standard addition method'), sorne
The bulk density is expressed in grams per millilitre despite
of the pure phase a is added to the mixture containing the
the International Unit being kilogram per cubic metre
unknown concentration of a. Multiple additions are made to
(1 g/mL = 1000 kg/m 3 ), because the measurements are made
prepare an intensity-versus-concentration plot in which the
using cylinders. It may also be expressed in grams per cubie
negative x intercept is the concentration of the phase a in the
centimetre.
original sample.
The bulking properties of a powder are dependent upon the
preparation, treatment and storage of the sample, i.e. how it
ESTIMATE OF THE AMORPHOUS AND CRYSTALLINE has been handled. The particles can be packed to have a range
FRACTIONS of bulk densities and, moreover, the slightest disturbance of
In a mixture of crystalline and amorphous phases, the the powder bed may result in a changed bulk density. Thus,
crystalline and amorphous fractions can be estimated in the bulk density of a powder is often very difficult to measure
several ways. The choice of the method used depends on the with good reproducibility and, in reporting the results, it is
nature of the sample: essential to specify how the determination was made.

(15) If the crystal structures of all components are known, the Rietveld method can be used to quantify them with good accuracy. Jf the crystal structures of the components are not known,
the Pawley or least squares methods can be used.

General Notices (1) apply to all monographs and other texts 343
2.9.34. Bulk density and tapped density of powders EUROPEAN PHARMACOPOEIA 8.0

A
The bulk density of a powder is determined either by
measuring the volume of a known mas s of powder sample,
which may have been passed through a sieve, in a graduated
cylinder (Method 1), or by measuring the mas s of a known
volume of powder that has been passed through a volumeter B
into a cup (Method 2) or has been introduced into a measuring
vessel (Method 3).
Methods 1 and 3 are favoured.
e
METHOD 1: MEASUREMENT IN A GRADUATED
CYLINDER
Procedure. Pass a quantity of powder sufficient to complete
the test through a sieve with apertures greater than or equal
to 1.0 mm, if necessary, to break up agglomerates that may
have formed during storage; this must be done gently to avoid
changing the nature of the material. Into a dry, graduated, D
250 mL cylinder (readable to 2 mL), gently introduce, without
compacting, approximately 100 g (m) ofthe test sample
weighed with 0.1 per cent accuracy. If necessary, carefully
level the powder without compacting, and read the unsettled
apparent volume (Vo) to the nearest graduated unit. Calculate E
the bulk density in grams per millilitre using the formula m/Vo'
Generally, replicate determinations are desirable for the
determination of this property.
If the powder density is too low or too high, such that the test
sample has an untapped apparent volume of more than 250 mL
or less than 150 mL, it is not possible to use 100 g of powder
F
sample. In this case, a different amount of powder is selected
as the test sample, such that its untapped apparent volume is
between 150 mL and 250 mL (apparent volume greater than
or equal to 60 per cent of the total volume of the cylinder); the
mass of the test sample is specified in the expression of results. A. 1.0 mm sieve E. glass baffle

B. powder funnel F. cup


For test samples having an apparent volume between 50 mL
and 100 mL, a 100 mL cylinder readable to 1 mL can be used; C. loading funnel G. stand
the volume of the cylinder is specified in the expression of
D. baffle box
results.
Figure 2.9.34.-1. - Volumeter
METHOD 2: MEASUREMENT IN A VOLUMETER METHOD 3: MEASUREMENT IN A VESSEL
Apparatus. The apparatus consists of a 100 mL cylindrical
Apparatus. The apparatus (Figure 2.9.34.-1) consists of a vessel of stainless steel with dimensions as specified in
top funnel fitted with a 1.0 mm sieve, mounted over a baffle
-54.0-- _57.0_
Figure 2.9.34.-2.

0
box containing 4 glass baffles over which the powder slides
and bounces as it passes. At the bottom of the baffle box is a
funnel that collects the powder and allows it to pour into a
cup mounted directly below it. The cup may be cylindrical
(25.00 ± 0.05 mL volume with an internal diameter of
50.5 52.i0 [50.5] 50.i 0
30.00 ± 2.00 mm) or cubical (16.39 ± 0;20 mL volume with
internal dimensions of 25.400 ± 0.076 mm) .. 2;01 10:0 540 ¡
Procedure. Allow an excess of powder to flow through the
apparatus into the sample receiving cup until it overflows, Figure 2.9.34.-2. - Measuring vessel (left) and cap (right)
using a minimum of 25 cm 3 of powder with the cubical cup Dimensions in millimetres
and 35 cm 3 of powder with the cylindrical cupo Carefully, Procedure. Pass a quantity of powder sufficient to complete
scrape excess powder from the top of the cup by smoothly the test through a 1.0 mm sieve, if necessary, to break up
moving the edge of the blade of a spatula perpendicular to agglomerates that may have formed during storage, and allow
and in contact with the top surface of the cup, taking care to the obtained sample to flow freely into the measuring vessel
keep the spatula perpendicular to prevent packing or removal until it overflows. Carefully scrape the excess powder from
of powder from the cupo Remove any material from the side the top of the vessel as described under Method 2. Determine
of the cup and determine the mass (M) of the powder to the the mass (Mo) ofthe powder to the nearest 0.1 per cent by
nearest 0.1 per cent. Calculate the bulk density in grams per subtracting the previously determined mass of the empty
millilitre using the formula M/Vo (where V o is the volume of measuring vessel. Calculate the bulk density in grams per
the cup) and record the average of 3 determinations using millilitre using the formula Mol1 00 and record the average of
3 different powder samples. 3 determinations using 3 different powder samples.

344 See the information section on general monographs (cover pages)


EUROPEAN PHARMACOPOEIA 8.0 2.9.34. Bulle density and tapped density of powders

Tapped density in increments of, for example, 1250 taps, until the difference
between successive measurements is less than or equal to
The tapped density is an increased bulk density attained after 2 mL. Fewer taps may be appropriate for sorne powders,
mechanically tapping a receptacle containing the powder when validated. Calculate the tapped density in grams per
sample. millilitre using the formula mi Vf (where 1'; is the final tapped
The tapped density is obtained by mechanically tapping a volume). Generally, replicate determinations are desirable for
graduated measuring cylinder or vessel containing the powder the determination of this property. Specify the drop height
sample. After observing the initial powder volume or mass, with the results.
the measuring cylinder or vessel is mechanically tapped, If it is not possible to use a 100 g test sample, use a reduced
and volume or mass readings are taken until little further amount and a suitable 100 mL graduated cylinder (readable
volume or mass change is observed. The mechanical tapping to 1 mL) weighing 130 ± 16 g and mounted on a support
is achieved by raising the cylinder or vessel and allowing it weighing 240 ± 12 g. The modified test conditions are
to drop, under its own mass, a specified distance by one of specified in the expression of the results.
3 methods as described below. Devices that rotate the cylinder
or vessel during tapping may be preferred to minimise any METHOD 2
possible separation of the mass during tapping down. Procedure. Proceed as directed under Method 1 except that
the mechanical tester provides a fixed drop of 3 ± 0.2 mm at
METHOD 1
a nominal rate of 250 taps per minute.
Apparatus. The apparatus (Figure 2.9.34.-3) consists of the
following: METHOD 3
- a 250 mL graduated cylinder (readable to 2 mL) with a Procedure. Proceed as described under Method 3 for
mass of 220 ± 44 g; measuring the bulk density, using the measuring vessel
- a settling apparatus capable of producing, per minute, equipped with the cap shown in Figure 2.9.34.-2. The
either nominally 250 ± 15 taps from a height of 3 ± 0.2 mm, measuring vessel with the cap is lifted 50-60 times per
or nominally 300 ± 15 taps from a height of 14 ± 2 mm. minute by the use of a suitable tapped density testero Carry
The support for the graduated cylinder, with its holder, has out 200 taps, remove the cap and carefully scrape excess
a mass of 450 ± 10 g. powder from the top of the measuring vessel as described
under Method 3 for measuring the bulk density. Repeat
Procedure. Proceed as described aboye for the determination the procedure using 400 taps. If the difference between the
of the bulk volume (Vo)' Secure the cylinder in the support. 2 mas ses obtained after 200 and 400 taps exceeds 2 per cent,
Carry out 10, 500 and 1250 taps on the same powder sample repeat the test using 200 additional taps until the difference
and read the corresponding volumes VlO' V soo and to the between successive measurements is les s than 2 per cent.
nearest graduated unit. If the difference between V soo and Calculate the tapped density in grams per millilitre using
V 1250 is less than or equal to 2 mL, V 1250 is the tapped volume. the formula MJlOO (where Mfis the mass of powder in the
If the difference between V soo and V 12S0 exceeds 2 mL, repeat measuring vessel). Record the average of 3 determinations

E
E
-' lú
E E (")
o

E :c
Ol
(")
e
('1 o ·iD ro
O
..::t:.. t ('1 ..c :Si
ro
Graduated cylinder -----¡-t-_±_ a. e
ro ro ~
-O ;2 O
..±. ID
Cií
:Si
rJJ
rJJ
E
-O
-.::t:- ::J
-O .!!1 e
Cylinder
support
i:
-+-
~
(')
-O
e

rD
Anvil
_lH This dimension is such
tha! the drop G) meets
.--J--¡-¡t;======~specifications and that, at
i the lowest point of the cam,
!he cylinder support is sitting
freely on the upper part of
the anvil.
Cam

Figure 2.9.34.-3. - Settling device for powder samples


Dimensians in millimetres

General Natices (1) apply ta all monographs and other texts 345
2.9.35. Powder fineness EUROPEAN PHARMACOPOEIA 8.0

using 3 different powder samples. The test conditions, Q,(x) = cumulative distribution of particles with a dimension
including tapping height, are specified in the expression of less than or equal to x where the subscript r reflects the
the results. distribution type.
r Distrlbution type
Measures of powder compressibility
O Nllmber
Because the interparticulate interactions influencing the
1 Length
bulking properties of a powder are also the interactions that
interfere with powder flow, a comparison of the bulk and 2 Area
tapped densities can give a measure of the relative importan ce
of these interactions in a given powder. Such a comparison is 3 Volllme
often used as an index of the ability of the powder to flow, for Therefore, by definition:
example the compressibility index or the Hausner ratio.
Q,(x) = 0.90 when x = x 90
The compressibility index and Hausner ratio are measures
of the propensity of a powder to be compressed as described Q,(x) = 0.50 when x = x so
aboye. As such, they are measures of the powder's ability Q,(x) = 0.10 when x = xlQ
to settle, and they permit an assessment of the relative An alternative but less informative method of classifying
importance of interparticulate interactions. In a free-flowing powder fineness is by use of the descriptive terms in
powder, such interactions are less significant, and the bulk Table 2.9.35.-1.
and tapped densities will be closer in value. For more-poorly
Table 2.9.35.-1.
flowing materials, there are frequently greater interparticulate
interactions, and a greater difference between the bulk and Classification of powders by fineness
tapped densities will be observed. These differences are
reflected in the compressibility index and the Hausner ratio. Cumulative
Descriptive term X 50 (¡.tm) distribution by
Compressibility index: volume basis, Q,(x)
100 (Va - Vi)
Coarse > 355 Q,(355) < 0.50
Va
Q,(180) < 0.50 and
Moderately fine 180 - 355
Vo unsettled apparent volume; Q,(355) ¿ 0.50

VJ final tapped volume. Q,(l25) < 0.50 and


Fine 125 - 180
Q3(l80) ¿ 0.50
Hausner Ratio:
Very fine S; 125 Q3(l25) ¿ 0.50
Va
Vi
01/2010:20936
Depending on the material, the compressibility index can be
determined using VIO instead of V o. lf VIO is used, it is clearly
stated with the results. 2.9.36. POWDER FLOW(16)
The widespread use of powders in the pharmaceutical industry
0712008:20935 has generated a variety of methods Íor characterising powder
flow. Not surprisingly, scores of references appear in the
2.9.35. POWDER FINENESS pharmaceuticalliterature, attempting to correlate the various
measures of powder flow to manufacturing properties. The
Particle-size distribution is estimated by analytical sieving development of such a variety of test methods was inevitable;
(2.9.38) or by application of other suitable methods where powder behavior is multifaceted and thus complicates the
appropriate. A simple descriptive classification of powder effort to characterise powder flow.
fineness is provided in this chapter. For practical reasons,
The purpose of this chapter is to review the methods for
sieves are commonly used to measure powder fineness. characterising powder flow that have appeared most frequently
Sieving is most suitable where a majority of the particles are in the pharmaceuticalliterature. In addition, while it is
larger than about 75 !lm, although it can be used for some
clear that no single and simple test method can adequately
powders having smaller particle sizes where the method can characterise the flow properties of pharmaceutical powders,
be validated. Light diffraction is also a widely used technique this chapter proposes the standardisation of test methods that
for measuring the size of a wide range of particles. may be valuable during pharmaceutical development.
Where the cumulative distribution has been determined by 4 commonly reported methods for testing powder flow are:
analytical sieving or by application of other methods, particle
size may be characterised in the following manner: - angle of repose,
- compressibility index or Hausner ratio,
x 90 particle size corresponding to 90 per cent of the
cumulative undersize distribution; - flow rate through an orifice,
- shear cell.
x so median particle size (i.e. 50 per cent of the particles
are smaller and 50 per cent of the particles are In addition, numerous variations of each of these basic
larger) ; methods are available. Given the number of test methods
and variations, standardising the test methodology, where
x lO particle size corresponding to 10 per cent of the possible, would be advantageous.
cumulative undersize distribution.
With this goal in mind, the most frequently used methods
It is recognised that the symbol d is also widely used to are discussed below. Important experimental considerations
designate these values. Therefore, the symbols dgO ' dso' dJO are identified and recommendations are made regarding
may be used. standardisation of the methods. In general, any method of
The following parameters may be defined based on the measuring powder flow must be practical, useful, reproducible
cumulative distribution. and sensitive, and must yield meaningful results. It bears

(16) Ihis chapter has undergone pharmacopoeial harmonisation. See chapter 5.8. Pharmacopoeial hcmnonisatiol1.

346 See the information section on general monographs (cover pages)


EUROPEA N PHARMACOPOEIA 8.0 2.9.36. Powder flow

repeating that no simple powder flow method will adequately Table 2.9.36.-1. - Flow properties and corresponding angles
or completely characterise the wide range of flow properties 01 repose(1?)
experienced in the pharmaceutical industry. An appropriate
F10w property Angle ofrepose (degrees)
strategy may well be the use of multiple standardised test
methods to characterise the various aspects of powder flow as Excellent 25-30
needed by the pharmaceutical scientist.
Good 31-35

Fair (aid not needed) 36-40

ANGLE OF REPOSE Passable (may hang up) 41-45

The angle of repose has been used in several branches of Poor (must agitate, vibrate) 46-55
science to characterise the flow properties of solids. Angle of
Very poor 56-65
repose is a characteristic related to interparticulate friction, or
resistance to movement between particles. Angle of repose test Very, very poor > 66
results are reported to be very dependent upon the method
used. Experimental difficulties arise due to segregation of Experimental considerations Cor angle of repose
material and consolidation or aeration of the powder as the
con e is formed. Despite its difficulties, the method continues Angle of repose is not an intrinsic property of the powder,
to be used in the pharmaceutical industry, and a number of that is to say, it is very much dependent upon the method
examples demonstrating its value in predicting manufacturing used to form the cone of powder. On this subject, the existing
problems appear in the literature. literature raises these important considerations:
- the peak of the cone of powder can be distorted by the
The angle of repose is the constant, three-dimensional angle impact of powder from aboye. By carefully building the
(relative to the horizontal base) assumed by a cone-like pile of powder cone, the distortion caused by impact can be
material formed by any of several different methods, described minimised;
briefly below.
- the nature of the base upon which the powder cone is
Bask methods for angle of repose formed influences the angle of repose. It is recommended
that the powder cone be formed 011 a 'common base', which
A variety of angle of repose test methods are described in the
can be achieved by forming the cone of powder on a layer
literature. The most common methods for determining the
of powder. This can be done by using a base of fixed
static angle of repose can be classified based on 2 important
diameter with a protruding outer edge to retain a layer of
experimental variables:
powder upon which the cone is formed.
- the height of the 'funnel' through which the powder passes Recommended procedure for angle of repose
may be fixed relative to the base, or the height may be
Form the angle of repose on a fixed base with a retaining lip
varied as the pile forms;
to retain a layer of powder on the base. The base must be
- the base upon which the pile forms may be of fixed diameter free of vibration. Vary the height of the funnel to carefully
or the diameter of the powder cone may be allowed to vary build up a symmetrical cone of powder. Care must be
as the pile forms. taken to prevent vibration as the funnel is moved. The
funnel height is maintained at approximately 2-4 cm from
Variations in angle of repose methods the top of the powder pile as it is being formed in order
to minimise the impact of falling powder on the tip of the
Variations of the aboye methods have also been used to some cone. If a symmetrical cone of powder cannot be successfully
extent in the pharmaceuticalliterature: or reproducibly prepared, this method is not appropriate.
Determine the angle of repose by measuring the height of the
- drained angle 01 repose: this is determined by allowing cone of powder and calculating the angle of repose, u, from
an excess quantity of material positioned aboye a fixed the following equation:
diameter base to 'drain' from the container. Formation
of a cone of powder on the fixed diameter base allows height
tan (Q ) = -,--_-=C-_ _
determination of the drained angle of repose; 0.5 x base

- dynamic angle 01 repose: this is determined by filling a


cylinder (with a clear, flat cover on one end) and rotating COMPRESSIBILITY INDEX AND HAUSNER RATIO
it at a specified speed. The dynamic angle of repose is the In recent years the compressibility index and the closely
angle (relative to the horizontal) formed by the flowing related Hausner ratio have become the simple, fast, and
powder. The internal angle of kinetic friction is defined by popular methods of predicting powder flow characteristics.
the plane separating those particles sliding down the top The compressibility index has been proposed as an indirect
layer of the powder and those particles that are rotating measure of bulk density, size and shape, surface area, moisture
with the drum (with roughened surface). content, and cohesiveness of materials, because all of these
General scale of flowability for angle of repose can influence the observed compressibility indexo The
compressibility index and the Hausner ratio are determined
While there is some variabon in the qualitative description by measuring both the bulk volume and tapped volume of
of powder flow using the angle of repose, much of the a powder.
pharmaceuticalliterature appears to be consistent with the Basic methods for compressibility index and Hausner ratio
classification by Carr(1?), which is shown in Table 2.9.36.-1.
There are examples in the literature of formulations with While there are some variations in the method of determining
an angle of repose in the range of 40-50 degrees that the compressibility index and Hausner ratio, the basic
manufactured satisfactorily. When the angle of repose exceeds procedure is to measure the unsettled apparent volume, (Vo)'
50 degrees, the flow is rarely acceptable for manufacturing and the final tapped volume, (V{l, ofthe powder after tapping
purposes. the material until no further vo ume changes occur. The

(17) Carr RL. Evaluating Ilow properties of solids. Chcl11. Eng 1965; 72:163-168.

General Notices (1) apply to all monographs and other texts 347
2.9.36. Powder flow EUROPEAN PHARMACOPOEIA 8.0

compressibility index and the Hausner ratio are calculated The flow rate through an orifice is generally measured as
as follows: the mass per time flowing from any of a number of types of
containers (cylinders, funnels, hoppers) Measurement of the
Va - VI
o

Compressibility Index = 100 x ---=-=-~ flow rate can be in discrete increments or continuous.
Va
Basic methods fúr flow through an orifice
· = -Va
H a'Usner R atw There are a variety of methods described in the literature. The
V¡ most common for determining the flow rate through an orifice
Alternatively, the compressibility index and Hausner ratio may can be classified based on 3 important experimental variables:
be calculated using measured values ofbulk density (Pbalk) and - the type of container used to contain the powder. Common
tapped density (Ptapped) as follows: containers are cylinders, funnels, and hoppers fram
production equipment;
ptapped - pbulk
Compressibility 1 ndex = 100 x :....:c:~""---'---'-"-'.:c. - the size and shape of the orifice used. The orífice djameter
ptapped
and shape are critical factors in determining powder flow
H a'Usner Ratio = Ptapped rate;
pbvlk - the method of measuring powder flow rateo Flow rate can
In a variation of these methods, the rate of consolidation is be measured continuously using an electronic balance
sometimes measured rather than, or in addition to, the change with sorne sort of recording device (strip chart recorder,
in volume that occurs on tapping. For the compressibility computer). It can also be measured in discrete samples
index and the Hausner ratio, the generally accepted scale of (for example, the time it takes for 100 g of powder to pass
flowability is given in Table 2.9.36.-2. through the orifice to the nearest tenth of a second or the
amount of powder passing through the orifice in 10 sto
Table 2.9.36.-2. - Scale offlowability 0 7J the nearest tenth of a gram).
Compressibility Flow character Hausner ratio Variati.ons i.n methods for flow through an ori.fice
inclex (per cent)
Either mass flow rate or volume flow rate can be determined.
1-10 Excellent 1.00-1.11 Mass flow rate is the easier of the methods, but it biases the
11-15 Good 1.12-1.18 results in favour of high -density materials. Since die fill is
volumetric, determining volume flow rate may be preferable.
16-20 Fair 1.19-1.25 A vibrator is occasionally attached to facilitate flow from the
21-25 Passable 1.26-1.34 container, however, this appears to complicate interpretation
of results. A moving orífice device has been proposed to
26-31 Poor 1.35-1.45 more closely simulate rotary press conditions. The minimum
32-37 Very poor 1.46-1.59 diameter orifice through which powder flows can also be
identified.
> 38 Very, very poor > 1.60
General scale of flowability for flow through an orífice
Experimental considerations for the compressibiJity index No general scale is available because flow rate is critically
and Hausner ratio dependent 011 the method used to measure it. Comparison
Compressibility index and Hausner ratio are not intrinsic between published results is difficult.
properties of the powder, that is to say, they are dependent Experimental considerations for flow through an orifice
upon the methodology used. The existing literature points out Flow rate through an orifice is not an intrinsic property of the
several important considerations affecting the determination powder. It is very much dependenl upon the methodology
of the unsettled apparent volume, V o' of the final tapped used. The existing literature points out several important
volume, Vi' of the bulk density, Pbalk' and of the tapped density, considerations affectjng these methods:
Ptapped: - the d¡ameter and shape of the orifice,
- the diameter of the cylinder used, - the type of container material (metal, glass, plastic),
- the number of times the powder is tapped to achieve the - the djameter and height of the powder bed.
tapped density,
Recommended pro ce dure for flow through an orifice
- the mass of material used in the test,
Flow rate through an orifice can be used only for materials
- rotation of the sample during tapping. that have sorne capacity to flow. 1t is not useful for cohesive
Recommended procedure for compressibility index and materials. Provided that the height of the powder bed (the
Hausner ratio 'head' of powder) is much greater than the díameter of the
orífice, the flow rate is virtually independent of the powder
Use a 250 mL volumetric cylinder with a test sample mass
head. It is advisable to use a cylinder as the container, because
of 100 g. Smaller amounts and volumes may be used, but
the waHs of the container must have little effect on flow. This
variations in the method must be described with the results.
configuration results in flow rate being determined by the
An average of 3 determinations is recommended.
movement of powder over powder, rather than powder along
FLOW THROUGH AN ORIFICE the wall of the container. Powder flow rate often increases
when the height of the powder column is less than twice the
The flow rate of a material depends upon many factors, sorne diameter of the column. The orifice must be circular and the
of which are particle-related and sorne related to the process. cylinder must be free of vibration. General guidelines for
Monitoring the rate of flow of material through an orifice dimensions of the cylinder are as follows:
has been proposed as a better measure of powder flowability.
- diameter of the opening greater than 6 times the diameter
Of particular significance is the utility of monitoring flow
of the partides,
continuously, since pulsating flow patterns have been observed
even for free-flowing materials. Changes in flow rate as the - diameter of the cylinder greater than twice the djameter of
container empties can also be observed. Empirical equations the opening.
relating flow rate to the diameter of the opening, particle Use of a hopper as the container may be appropriate and
size, and particle density have been determined. However, representative of flow in a productiol1 situation. It is not
determining the flow rate through an orifice is useful only advisable to use a funnel, particularly one with a stem, because
with free-flowing materials. flow rate will be determined by the size and length of the stem

348 See the information section on monographs (ca ver pages)


EUROPEAN PHARMACOPOEIA 8.0 2.9.37. Optical microscopy

as well as the friction between the stem and the powder. A 01/2010:20937
truncated cone may be appropriate, but flow will be influenced
by the powder-wall friction coefficient, thus, selection of an 2,9.37. OPTICAL MICROSCOPY(J8)
appropriate construction material is important.
Optical microscopy for partide characterisation can generally
For the opening in the cylinder, use a flat-faced bottom be applied to particles of 1 flm and greater. The lower limit
plate with the option to vary orifice diameter to provide is imposed by the resolving power of the microscope. The
maximum flexibility and better ensure a powder-over-powder upper limit is less definite and is determined by the increased
flow pattern. Rate measurement can be either discrete or difficulty associated with the characterisation of larger
continuous. Continuous measurement using an electronic partides. Various alternative techniques are available for
balance can more effectively detect momentary flow rate partide characterisation outside the applicable range of optical
variations. microscopy. Optical microscopy is particularly useful for
characterising partides that are not sphericaL This method
may also serve as a base for the calibration of faster and more
routine methods that may be developed.
SHEAR CELL METHODS
Apparatus. Use a microscope that is stable and protected
In an effort to put powder flow studies and hopper design on a from vibration. The microscope magnification (product of the
more fundamental basis, a variety of powder shear testers and objective magnification, ocular magnification, and additional
methods that permit more thorough and precisely defined magnifying components) must be sufficient to allow adequate
assessment of powder flow properties have been developed. characterisation of the smallest partides to be dassified in the
Shear ceH methodology has been used extensively in the study test sample. The greatest numerical aperture of the objective
of pharmaceutical materials. From these methods, a wide is sought for each magnification range. Polarising filters may
variety of parameters can be obtained, induding the yield loci be used in conjunction with suitable analysers and retardation
representing the shear stress-shear strain relationship, the plates. Colour filters of relatively narrow spectral transmission
angle of internal friction, the unconfined yield strength, the are used with achromatic objectives, and are preferable
tensile strength, and a variety of derived parameters such as with apochromats; they are required for appropriate colour
the flow factor and other flowability indices. Because of the rendition in photomicrography. Condensers, corrected at least
ability to control experimental parameters more precisely, for spherical aberration are used in the microscope substage
flow properties can also be determined as a function of and with the lampo The numerical aperture of the substage
consolidation load, time, and other environmental conditions. condenser matches that of the objective under the conditions
These methods have been successfully used to determine of use; this is affected by the actual aperture of the condenser
critical hopper and bin parameters. diaphragm and the presence of immersion oils.
Basic methods for shear ceH Adjustment. The precise alignment of al! elements of
the optical system and proper focusing are essential. The
One type of shear cel! is the cylindrical shear ceH which is focusing of the elements is done in accordance with the
split horizontally, forming a shear plane between the lower recommendations of the microscope manufacturero Critical
stationary base and the upper moveable portion of the shear axial alignment is recommended.
cel! ringo After powder bed consolidation in the shear ceH Ulumination. A requirement for good illumination is a
(using a well-defined procedure), the force necessary to shear uniform and adjustable intensity of light over the entire field
the powder bed by moving the upper ring is determined. of view; K6hler illumination is preferred. With coloured
Annular shear cell designs offer some advantages over the partides, choose the colour of the filters so as to control the
cylindrical shear cell design, induding the need for less contrast and detail of the image.
material, A disadvantage, however, is that because of its
design, the powder bed is not sheared as uniformly because Visual characterisation. The magnification and numerical
material on the outside of the annulus is sheared more aperture must be sufficiently high to allow adequate resolution
than material in the inner region. A third type of shear ceH of the images of the partides to be characterised. Determine
(plate-type) consists of a thin sandwich of powder between a the actual magnification using a calibrated stage micrometer
lower stationary rough surface and an upper rough surface to calibrate an ocular micrometer. Errors can be minimised if
that is moveable. the magnification is sufficient that the image of the partide is
at least 10 ocular divisions. Each objective must be calibrated
All of the shear cell methods have their advantages and separately. To calibrate the ocular scale, the stage micrometer
disadvantages, but a detailed review is beyond the scope of scale and the ocular scale must be aligned. In this way, a
this chapter. As with the other methods for characterising precise determination of the distance between ocular stage
powder flow, many variations are described in the literature. divisions can be made. Several different magnifications may
A significant advantage of shear cell methodology in general be necessary to characterise materials having a wide particle
is a greater degree of experimental control. The methodology size distribution.
generally is rather time-consuming and requires significant Photographic charaderisation. If partide size is to be
amounts of material and a well-trained operator. determined by photographic methods, take care to ensure
Recommendations for shear cell that the object is sharply focused at the plane of the
photographic emulsiono Determine the actual magnification
The many existing shear cell configurations and test methods by photographing a calibrated stage micrometer, using
provide a wealth of data and can be used very effectively photographic film of sufficient speed, resolving power, and
to characterise powder flow. Theyare also helpful in the contrasto Exposure and processing must be identical for
design of equipment such as hoppers and bins. Because of the photographs of both the test sample and the determination
diversity of available equipment and experimental pro ce dures, of magnification. The apparent size of a photographic image
no specific recommendations regarding methodology are is influenced by the exposure, development, and printing
presented in this chapter. It is recommended that the results processes as well as by the resolving power of the microscope.
of powder flow characterisation using shear ceH methodology Preparation of the mount. The mounting medium will
indude a complete description of equipment and methodology vary according to the physical properties of the test sample.
used. Sufficient, but not excessive, contrast between the sample and

(18) This chapter has undergone phannacopoeial harmonisation. See chapter 5.8. Pharmacopoeial harmo/lisalion.

General Notices (1) apply to all monographs and other texts 349
2.9.37. Optical microscopy EUROPEAN PHARMACOPOEIA 8.0

the mounting medium is required to ensure adequate detail - Feret's díameter: the distanee between imaginary parallel
of the sample edge. The particles must rest in one plane and lines tangent to a randomly oriented particle and
be adequately dispersed to distinguish individual particles of perpendicular to the ocular scale,
interest. Furthermore, the particles must be representative
of the distribution of sizes in the material and must not be - Martins diameter: the d¡ameter ofthe particle at the point
altered during preparation of the mount. Care must be taken that divides a randomly oriented particle into 2 equal
to ensure that this important requirement is met. Seleetion of projected areas,
the mounting medium must in dude a eonsideration of the
analyte solubility. - projected area diameter: the diameter of a circle that has
the same projected are a as the particle,
CrystalHnity characterisation. The crystallinity of a material
may be characterised to determine compliance with the - length: the longest dimension from edge to edge of a
crystallinity requirement where stated in the individual particle oriented parallel to the ocular seale,
monograph of a drug substance. Unless otherwise speeified
in the individual monograph, mount a few particles of the - width: the longest dimension of the particle measured at
sample in mineral oil on a clean glass slide. Examine the right angles to the length.
mixture using a polarising microseope: the particles show Partide shape characterisation. For irregularly shaped
birefringence (interference colors) and extinction positions particles, characterisation of particle size must also include
when the microscope stage is revolved. information on particle shape. The homogeneity of the
Limit test of partide size by microscopy. Weigh a suitable powder must be cheeked using appropriate magnifieation.
quantity of the powder to be examined (for example, The following defines some commonly used descriptors of
10-100 mg), and suspend it in 10 mL of a suitable medium particle shape (see Figure 2.9.37.-2).
in which the powder does not dissolve, adding, if necessary,
a wetting agent. A homogeneous suspension of partides - acicular: slender, needle-like particle of similar width and
can be maintained by suspending the particles in a medium thickness,
of similar or matching density and by providing adequate
- columnar: long, thin particle with a width and thickness
agitation. Introduce a portion of the homogeneous suspension
that are greater than those of an acicular particle,
into a suitable eounting cell, and sean under a microscope
an are a corresponding to not less than 10 flg of the powder - flake: thin, flai particle of similar length and width,
to be examined. Count a11 the particles having a maximum
dimension greater than the prescribed size limito The size - plate: flat particle of similar length and width but with
limit and the permitted number of particles exceeding the greater thickness than a flake particle,
limit are defined for each substance.
- lath: long, thin, blade-like particle,
Partide size characterisation. The measurement of particle
size varies in eomplexity depending on the shape of the - equant: particle of similar length, width, and thickness;
particle, and the number of particles characterised must be both cubical and spherical particles are included.
sufficient to ensure an aceeptable level of uneertainty in the
measured parameters. Additional information on particle size General observations. A particle is generally considered
measurement, sample size, and data analysis is available, for to be the smallest di serete unit. A particle may be a liquid
example, in ISO 9276. For spherical particles, size is defined by or semi -solid droplet; a single erystal or polycrystalline;
the diameter. For irregular particles, a variety of definitiol1s of amorphous or an agglomerate. Particles may be associated.
particle size existo In general, for irregularly shaped particles, This degree of association may be described by the following
characterisation of particle size must also include information terms:
on the type of diameter measured as well as information on
- lamellar: stacked plates,
particle shape. Several commonly used measurements of
particle size are defined in Figure 2.9.37.-1. - aggregate: mass of adhered partides,

- agglomerate: fused or cemented particles,

- conglomerate: mixture of 2 or more types of particles,

- spherulite: radial cluster,

- drusy: partide covered with tiny particles.

1 - 1 1 - - - - - - - - - '......., Feret's diameter Partide condition may be described by the following terms:

- edges: angular, rounded, smooth, sharp, fractured,

- optical: color (using proper color balancing filters),


transparent, translucent, opaque,
Martin's diameter
- defects: occlusions, inclusions.

Surface characteristics may be described as:

- cracked: partial split, break, or fissure,

- smooth: free of irregularities, roughness, or projections,

- porous: having openings or passageways,

- rough: bumpy, uneven, not smooth,


Figure 2.9.37.-1. - Commonly used measurements of particle
size pitted: small indentations.

350 See the information section on general monographs (cover pages)


EUROPEAN PHARMACOPOElA 8.0 2.9.38. Partide-size distribuHon estlmation by analytical sieving

Flake
Equant

~~
I~V
Plate

Columnar
I~
Figure 2.9.37.-2. - Commonly used descriptions of particle shape

0112010:20938 controlled to prevent moisture uptake or loss by the sample.


In the absence of evidence to the contrary, analytical test
2.9.38, PARTICLE-SIZE DISTRIBUTION sieving is normally carried out at ambient humidity. Any
special conditions that apply to a particular material must be
ESTIMATION BY ANALYTICAL detailed in the individual monograph.
SIEVING(J9) PrincipIes of analytical sieving. Analytical test sieves are
Sieving is one of the oldest methods of classifying powders constructed from a woven-wire mesh, which is of simple weave
and granules by particle-size distribution. When using a that is assumed to give nearly square apertures and is-joined to
woven sieve cloth, the sieving will essentially sort the particles the base of an open cylindrical container. The basic analytical
by their intermediate size dimension (Le. breadth or width). method involves stacking the sieves on top of one another in
Mechanical sieving is most suitable where the majority of the ascending degrees of coarseness, and then placing the test
particles are larger than about 75 tun. For smaller particles, powder on the top sieve. The nest of sieves is subjected to a
their light weight provides insufficient force during sieving to standardised period of agitation, and then the mass of material
overcome the surface forces of cohesion and adhesion that retained on each sieve is accurately determined. The test gives
cause the particles to stick to each other and to the sieve, and the mass percentage oí powder in each sieve size range.
thus cause particles that would be expected to pass through This sieving process for estimating the particle-size
the sieve to be retained. For such materials other means of distribution of a single pharmaceutical powder is generally
agitation such as air-jet sieving or sonic-sifter sieving may be intended for use where at least 80 per cent of the particles are
more appropriate. Nevertheless, sieving can sometimes be larger than 75 11m. The size parameter involved in determining
used for some powders or granules having median particle particle-size distributioll by anaIytical sieving is the length of
sizes smaller than 75 11m where the method can be validated. the side of the minimum square aperture through which the
In pharmaceutical terms, sieving is usually the method particle will pass.
of choice for classification of the coarser grades of single
powders or granules. It is a particularly attractive method in TEST SIEVES
that powders and granules are classified only on the basis of Test sieves suitable for pharmacopoeial tests conform to
particle size, and in most cases the analysis can be carried out the current edition of ISO 3310-1: Test sieves - Technical
in the dry state. requirements and testing Part 1: Test sieves of metal wire
Among the limitations of the sieving method are the need cloth (see Table 2.9.38.-1). Unless otherwise specified in the
for an appreciable amount of sample (normally at least 25 g, monograph, use those ISO sieves listed as principal sizes in
depending on the density of the powder or granule, and the Table 2.9.38.-1 that are recommended in the particular region.
diameter of the test sieves) and the difficulty in sieving oily Sieves are selected to cover the entire range of particle sizes
or other cohesive powders or granules that tend to clog the present in the test sample. A nest of sieves having a v'2
sieve openings. The method is essentially a two-dimensional progression of the area of the sieve openings is recommended.
estimate of size because passage through the sieve aperture is The nest of sieves is assembled with the coarsest screen at
frequently more dependent on maximum width and thickness the top and the finest at the bottom. Use micro me tres or
than on length. millimetres in denoting test sieve openings.
This methodis intended for estimation of the total Test sieves are made from stainless steel or, less preferably,
particle-size distribution of a single material. It is not intended from brass or another suitable non-reactive wire.
for determination of the proportion of particles passing or
Calibration and recalibration of test sieves is in accordance
retained on 1 or 2 sieves.
with the current edition ofISO 3310-1. Sieves are carefully
Estimate the particle-size distribution as described under Dry examined for gross distortions and fractures, especially at
sieving method, unless otherwise specified in the individual their screen frame joints, before use. Sieves may be calibrated
monograph. Where difficulty is experienced in reaching the optically to estimate the average opening size, and opening
endpoint (i.e. material does not readily pass through the variability, of the sieve mesh. Alternatively, for the evaluation
sieves) or when it is necessary to use the finer end of the of the effective opening of test sieves in the size range of
sieving range (below 75 11m), serious consideration must be 212-850 11m, standard glass spheres are available. Unless
given to the use of an alternative particle-sizing method. otherwise specified in the individual monograph, perform the
Sieving is carried out under conditions that do not cause the sieve analysis at controlled room temperature and at ambient
test sample to gain or lose moisture. The relative humidity of relative humidity.
the environment in which the sieving is carried out must be

(19) This chapter has undergone pharmacopoeial harmonisation. See chapter 5.8. Pharmacopoeial harmonisatiol1.

General Notices (1) apply to all monographs and other texts 351
2.9.38. Partide-size distribution estimation by analytical sieving EUROPEAN PHARMACOPOElA 8.0

Table 2.9.38.-1. ISO Nominal Aperture US Recom- European Japanese


Sieve mended Sieve Sieve
ISO Nominal Aperture US Recom- European Japanese Principal Supplementary No. USP Sieves No. No.
Sieve mended Sieve Sieve sizes sizes (flm)
Principal Supplementary No. USP Sieves No. No. R 20/3 R20 R40/3
sizes sizes Ü1m)
R 20/3 R20 R 40/3 280 flm

11.20 mm 11.20 mm 11.20 mm 11 200 250 11m 250 11m 250 flm 60 250 250 60

10.00mm 224 f1m

9.50 111m 212 ¡llll 70 70

9.00111111 200 11m

8.00 mm 8.00 mm 8.00111111 180 11m 180 11111 180 11m 80 180 180 83

7.10111111 160 ¡llll

6.70 111m 150 11m 100 100

6.30 111m 140 11m

5.60 mm 5.60 mm 5.60 mm 5600 3.5 125 11m 125 11m 125 flm 120 125 125 119

5.00111111 112 11m

4.75111111 4 106 11m 140 140

4.50111111 100 flm

4.00 mm 4.00 mm 4.00111111 5 4000 4000 4.7 90 flm 90 11m 90 11111 170 90 90 166

3.55111m 80 f1m

3.35111m 6 5.5 75 11111 200 200

3.15111m 71 11m

2.80 m111 2.80 111m 2.80111111 7 2800 2800 6.5 63 11m 63 11m 63 f1111 230 63 63 235

2.50111111 56 11m

2.36 mm 8 7.5 53 11m 270 282

2.24111m 50 11m

2.00111111 2.00 111m 2.00m111 10 2000 2000 8.6 45 11m 45 11m 45 ¡llll 325 45 45 330

1.80m111 40 11m

1.70111111 12 10 38 flm 38 391

1.60111111
Cleaning test sieves. ldeally, test sieves are cleaned using only
1.40111111 1.40 mm 1.40 m111 14 1400 1400 12 a low-pressure air jet or a liquid stream. lf sorne apertures
1.25111m
remain blocked by test particles, careful gentle brushing may
be used as a last resort.
1.18111m 16 14
Test sample. lf the test sample mass is not given in the
1.12 mm monograph for a particular material, use a test sample having
a mass of 25-100 g, depending on the bulk density of the
1.00 111m 1.00 mm 1.00 mm 18 1000 1000 16
material, for test sieves having a 200 mm diameter. Por 76 mm
900¡llll sieves, the amount of material that can be accommodated
is approximately 1/7 that which can be accommodated by
850 f1m 20 18
a 200 mm sieve. Determine the most appropriate mass for
800 f1m a given material by test sieving accurately weighed samples
of different masses, such as 25 g, 50 g and 100 g, for the
710 11m 710 11111 710 11m 25 710 710 22
same time period on a mechanical shaker (note: if the test
630 f1m results are similar for the 25 g and 50 g samples, but the 100 g
sample shows a lower percentage through the finest sieve,
600 flm 30 26
the 100 g sample size is too large). Where only a sample of
560 f1111 10-25 gis available, smaller diameter test sieves conforming
to the same mesh specifications may be substituted, but the
500 f1m 500 11m 500 11m 35 500 500 30
endpoint must be redetermined. The use of test samples
450 11m having a smaller mass (e.g. down to 5 g) may be needed. Por
materials with low apparent particle density, or for materials
425 f1m 40 36
mainly comprising particles with a highly iso-diametrical
400 11m shape, sample masses below 5 g for a 200 mm screen may be
necessary to avoid excessive blocking of the sieve. During
355 flm 355 11m 355 11m 45 355 355 42
validation of a particular sieve-analysis method, it is expected
315 11m that the problem of sieve blocking will have been addressed.
300 11m 50 50 lf the test material is prone to absorbing or losing significant
amounts of water with varying humidity, the test must be
carried out in an appropriately controlled environment.
Similarly, if the test material is known to develop an

352 See the information section on general monographs (cover pages)


EUROPEAN PHARMACOPOEIA 8,0 2.9.39. Water-solid interactions

electrostatic charge, careful observation must be made to normal agitation mechanism. It requires sequential analyses
ensure that such charging do es not influence the analysis. on individual sieves starting with the finest sieve to obtain a
An antistatic agent, such as colloidal silicon dioxide and/or particle-size distribution. Air-jet sieving often includes the
aluminum oxide, may be added at a 0.5 per cent (m/m) level use of finer test sieves than used in ordinary dry sieving. This
to minimise this effect. If both of the aboye effects cannot technique is more suitable where only oversize or undersize
be eliminated, an alternative particle-sizing technique must fractions are needed.
be selected. In the sonic-sifter method, a nest of sieves is used, and the
Agitation methods. Several different sieve and test sample is carried in a vertically oscillating column of air
powder-agitation devices are commercially available, aH of that lifts the sample and then carries it back against the mesh
which may be used to perform sieve analyses. However, the openings at a given number of pulses per minute. It may
different methods of agitation may give different results for be necessary to lower the sample amount to 5 g when sonic
sieve analyses and endpoint determinations because of the sifting is employed.
different types and magnitudes of the forces acting on the The air-jet sieving and sonic-sifter sieving methods may be
individual particles under test. Methods using mechanical useful for powders or granules when the mechanical sieving
agitation or electromagnetic agitation, and that can induce techniques are incapable of giving a meaningful analysis.
either a vertical oscillation or a horizontal circular motion,
These methods are highly dependent upon proper dispersion
or tapping or a combination of both tapping and horizontal
of the powder in the air current. This requirement may be
circular motion are available. Entrainment of the particles
hard to achieve if the method is used at the lower end of the
in an air stream may also be used. The results must indicate
sieving range (Le. below 75 flm), when the particles tend
which agitation method was used and the agitation parameters
to be more cohesive, and especially if there is any tendency
used (if they can be varied), since changes in the agitation
for the material to develop an electrostatic charge. For the
conditions will give different results for the sieve analysis and
aboye reasons endpoint determination is particularly critica!,
endpoint determination, and may be sufficiently different to
and it is very important to confirm that the oversize material
give a failing result under sorne circumstances.
comprises single particles and is not composed of aggregates.
Endpoint determination. The test sieving analysis is complete
when the mass on any of the test sieves does not change by INTERPRETATION
more than 5 per cent or 0.1 g (lO per cent in the case of 76 mm The raw data must include the mass of the test sample, the
sieves) of the previous mass on that sieve. Ifless than 5 per total sieving time, the precise sieving methodology, and the set
cent of the total sample mass is present on a given sieve, the values for any variable parameters, in addition to the masses
endpoint for that sieve is increased to a mas s change of not retained on the individual sieves and in the pan,
more than 20 per cent of the previous mass on that sieve.
It may be convenient to convert the raw data into a cumulative
lf more than 50 per cent of the total sample mass is found mass distribution, and if it is desired to express the distribution
on any one sieve, unless this is indicated in the monograph, in terms of a cumulative mass undersize, the range of sieves
the test is repeated, but with the addition to the sieve nest of used must include a sieve through which all the material
a more coarse sieve intermediate between that carrying the passes. If there is evidence on any of the test sieves that the
excessive mass and the next coarsest sieve in the original nest, material remaining 011 it is composed of aggregates formed
Le. addition of the ISO series sieve omitted from the nest of during the sieving process, the analysis is invalido
sieves.
SIEVING METHODS
Mechanical agitation (Dry sieving method). Tare each test 04/2011:20939
sieve to the nearest 0.1 g. Place an accurately weighed quantity
of test sample on the top (coarsest) sieve, and replace the lid. 2.9.39. WATER-SOLID INTERACTIONS:
Agitate the nest of sieves for 5 min, then carefully remove
each sieve from the nest without loss of material. Reweigh DETERMINATION OF SORPTION-
each sieve, and determine the mass of material on each one. DESORPTION ISOTHERMS AND OF
Determine the mass of material in the collecting pan in a
similar manner. Re-assemble the nest of sieves, and agitate for WATER ACTIVITY
5 mino Remove and weigh each sieve as previously described. INTRODUCTION
Repeat these steps until the endpoint criteria are met (see
Endpoint determination under Test sieves). Upon completion Pharmaceutical solids as raw materials or as constituents
of the analysis, reconcile the masses of material. Totalloss of dosage forms most often come in contact with water
must not exceed 5 per cent of the mass of the original test during processing and storage. This may occur (a) during
sample. crystallisation, lyophilisation, wet granulation, or spray
drying; and (b) because of exposure upon handling and
Repeat the analysis with a fresh sample, but using a single storage to an atmosphere containing water vapour or exposure
sieving time equal to that of the combined times used aboye. to other materials in a dosage form that contain water capable
Confirm that this sieving time conforms to the requirements of distributing it to other ingredients. Sorne properties known
for endpoint determination. When this endpoint has been to be altered by the association of solids with water include
validated for a specific material, then a single fixed time rates of chemical degradation in the "solid-state", crystal
of sieving may be used for future analyses, providing the growth and dissolution, dispersibility and wetting, powder
particle-size distribution falls within normal variation. flow, lubricity, powder compactibility, compact hardness and
If there is evidence that the particles retained on any sieve are microbial contamination.
aggregates rather than single particles, the use of mechanical Although precautions can be taken when water is perceived to
dry sieving is unlikely to give good reproducibility, and a be a problem, i.e. eliminating all moisture, reducing contact
different particle-size analysis method must be used. with the atmosphere, or controlling the relative humidity of
Air-entrainment methods (Air-jet and sonic-sifter sieving). the atmosphere, su eh precautions generally add expense to the
Different types of commercial equipment that use a moving process with no guarantee that during the life of the product
air current are available for sieving. A system that uses a single further problems associated with moisture will be avoided. It
sieve at a time is referred to as air-jet sieving. It uses the same is also important to recognise that there are many situations
general sieving methodology as that described under Dry where a certain level of water in a solid is required for proper
sieving method, but with a standardised air jet replacing the performance, e.g. powder compaction. 1t is essential for both

General Natices (1) apply ta all managraphs and other texts 353
2.9.39. Water-solid interactions EUROPEAN PHARMACOPOEIA 8.0

reasons, therefore, that as much as possible is known about the increasingly aboye the glass transition temperature, it is
effects of moisture on solids before strategies are developed not surprising that a number of important bulk properties
for their handling, storage and use. dependent on the rheology of the solid are affected by moisture
Sorne of the more critical pieces of required information contento Since amorphous solids are metastable relative to the
concerning water-solid interactíons are: crystalline form of the material, with small-molecular-mass
materials, it is possible for absorbed moisture to initiate
- total amount of water present;
reversion of the solid to the crystalline form, particularly
- the extent to which adsorption and absorption occur; if the solid is transformed by the sorbed water to a "fluid"
- whether or not hydrates form; state. This is the basis of "cake collapse" often observed
- speciflc surface area of the solid, as well as such properties during the lyophilisation process. An additional phenomenon
as degree of crystallinity, degree of porosity, and glass noted speciflcally with water-soluble solids is their tendency
transition and melting temperature; to deliquesce, i.e. to dissolve in their own sorbed water, at
- site of water interaction, the extent of binding, and the relative humidities, RH¡, in excess of the relative humidity of
degree of molecular mobility; a saturated solution of the solid, RHQ' Deliquescence arises
because of the high water solubility of the solid and the
- effects of temperature and relative humidity; signiflcant effect it has on the colligative properties of water. It
- essentially irreversible hydration; is a dynamic process that continues to occur as long as RHi is
- kinetics of moisture uptake; greater than RHo'
- various factors that might influence the rate at which water The key lo understanding the effects water can have on the
vapour can be taken up by a solid; properties of solids, and vice versa, rests with an understanding
- for water-soluble solids capable ofbeing dissolved by the of the location of the water molecule and its physical state.
sorbed water, under which conditions dissolution will take More specifically, water associated with solids can exist in a
place. state that is directly bound to the solid, as well as in a state of
mobility approaching that ofbulk water. This difference in
PHYSICAL STATES OF SORBED WATER
mobility has been observed through such measurements as
Water can physically interact with solids in different ways. It heats of sorption, freezing point, nuclear magnetic resonance,
can interact at the surface (adsorption) or it can penetrate the dielectric properties and diffusion. Such changes in mobility
bulk solid structure (absorption). When both adsorption and have been interpreted as arising because of changes in the
absorption occur, the term sorption is often used. Adsorption thermodynamic state of water as more and more water
is particularly critical in affecting the properties of solids is sorbed. Thus, water bound directly to a solid is often
when the speciflc surface are a is large. Large values of speciflc thought as unavailable to affect the properties of the solid,
surface are a are seen with solids having very small particles, whereas larger amounts of sorbed water may become more
as well as with solids having a high degree of intraparticle clustered and form water more Iike that exhibiting solvent
porosity. Absorption is characterised by an association of properties. In the case of crystal hydrates, the combination of
water per gram of solid that is much greater than that which intermolecular forces (hydrogen bonding) and crystal packing
can form a mono molecular layer on the available surface, can produce very strong water-solid interactions. Recognising
and an amount that is generally independent of the specific that the presence of water in an amorphous solid can affect
surface area. the glass transition temperature and hence the physical state
Most crystalline solids willnot absorb water into their bulle of the solid, at low levels of water, most polar amorphous
structures because of the close packing and high degree solids are in a highly viscous glassy state because of their
of order of the crystallattice. Indeed, it has been shown high values of Tg. Hence, water is "frozen" into the solid
that the degree of absorption into solids exhibiting partial structure and is rendered immobile by the high viscosity, e.g.
crystallinity and partia! amorphous structure is often inversely 10 13 Pa·s. As the amount of water sorbed increases and Tg
proportional to the degree of crystallinity. With sorne decreases, approaching ambient temperatures, the glassy state
crystalline solids, however, crystal hydrates may formo These approaches that of a "fluid" state and water mobility along
hydrates may exhibit a stoichiometric relationship, in terms with the mobility of the solid itself increases signiflcantly.
of water molecules bound per solid molecule, or they may At high RH, the degree of water plasticisation of the solid
be non-stoichiometric. Upon dehydration, crystal hydrates can be sufficiently high so that water and the solid can now
may either retain their original crystal structure, or lose their achieve signiflcant amounts of mobility. In general, therefore,
crystallinity and become amorphous, or transform into a new this picture of the nature of sorbed water helps to explain
anhydrous or less-hydrated crystal formo the rather signiflcant effect moisture can have on a number
Amorphous or partially amorphous solids are capable of taking of bulk properties of solids such as chemical reactivity and
up signiflcant amounts of water because there is sufflcient mechanical deformation. It suggests strongly that methods of
molecular disorder in the solid to permit penetration, evaluating chemical and physical stability of solids and solid
swelling or dissolution. Such behaviour is observed with dosage forms take into account the effects water can have on
most amorphous polymers and with small-molecular-mass the solid when it is sorbed, particularly when it enters the
solids rendered amorphous during preparation, e.g. by solid structure and acts as a plasticiser.
lyophilisation, or after milling. The introduction of defects Rafes of water uptake. The rate and extent to which solids
into highly crystalline solids will also produce this behaviour. exposed to the atmosphere might either sorb or desorb water
The greater the chemical afflnity of water for the solid, the vapour can be a critical factor in the handling of solids. Even
greater the total amount that can be absorbed. When water is the simple act of weighing out samples of solid on an analytical
absorbed by amorphous solids, the bulk properties of the solid balance and the exposure, therefore, of a thin layer of powder
can be signiflcantly altered. It is well established, for example, to the atmosphere for a few minutes can lead to significant
that amorphous solids, depending on the temperature, error in, for example, the estimation of 10ss on drying values.
can exist in at least one of 2 states, "glassy" or "fluid"; the It is well established that water-soluble solids exposed to
temperature at which one state transforms into the other is the relative humidities aboye that exhibited by a saturated solution
glass transition temperature, Tg• of that solid will spontaneously dissolve vía deliquescence
Water absorbed into the bulk solid structure, by virtue of its and continue to dissolve over a long time periodo The rate of
effect on the free volume of the solid, can act as an efflcient water uptake in general depends on a number of parameters
plasticiser and reduce the value of T a • Since the rheological not found to be critical in equilibrium measurements because
properties of "fluid" and "glassy" states are quite different, rates of sorption are primarily mass-transfer controlled
i.e. the "fluid" state eX1!.¡ibits much less viscosity as one goes with sorne contributions from heat-transfer mechanisms.

354 See the information section on general monographs (cover pages)


EUROPEAN PHARMACOPOEIA 8.0 2.9.39. Water-solid inferactions

Thus, factors such as vapour diffusion coefficients in air and Methods. Samples may be sto red in chambers at various
in the solid, convective airflow, and the surface area and relative humidities (Figure 2.9.39.-1). The mass gained or lost
geometry of the solid bed and surrounding environment, for each sample is then measured. The major advantage of
can play an important role. Indeed, the method used to this method is convenience, while the major disadvantages are
make measurements can often be the rate-determining factor the slow rate of reaching constant mass, particularly at high
because of these environmental and geometric factors. relative humidities, and the error introduced in opening and
closing the chamber for weighing.
DETERMINATION OF SORPTION-DESORPTION
ISOTHERMS Dynamic gravimetric water sorption systems allow the
PrincipIe. The tendency to take up water vapour is best on-line weighing of a sample in a controlled system to
assessed by measuring sorption or desorption as a function assess the interaction of the material with moisture at
of relative humidity, at constant temperature, and under various programmable levels of relative humidity at a
conditions where sorption or desorption is essentially constant temperature. The major benefit of a controlled
occurring independently of time, i.e. equilibrium. Relative system is that isothermal conditions can be more reliably
humidity, RH, is defined by the following expression: established and that the dynamic response of the sample to
changing conditions can be monitored. Data points for the
Pe determination of the sorption isotherm (e.g. from O per cent
Po X 100
to approximately 95 per cent RH, non condensing) are only
taken after a sufficiently constant signal indicates that the
Pe pressure of water vapour in the system; sample has reached equilibrium at a given level of humidity.
In some cases (e.g. deliquescence), the maximum time may be
Po saturation pressure of water vapour under the same
restricted although the equilibrium level is not reached. The
conditions.
apparatus must adequate!y control the temperature to ensure
The ratio PiPo is referred to as the relative pressure. Sorption a good baseline stability as well as accurate control of the
or water uptake is best assessed starting with dried samples relative humidity generation. The required relative humidities
and subjecting them to a known relative humidity. Desorption can be generated, e.g. by accurately mixing dry and saturated
is studied by beginning with a system already containing vapour gas with flow controllers. The electrostatic behaviour
sorbed water and reducing the relative humidity. As the name of the powder must also be considered. The verification of
indicates, the sorption-desorption isotherm is valid only for the temperature and the relative humidity (controlled with,
the reference temperature, hence a special isotherm exists for for example, a certified hygrometer, certified salt solutions
each temperature. Ordinarily, at equilibrium, moisture content or deliquescence points of certified salts over an adequate
at a particular relative humidity must be the same, whether range), must be consistent with the instrument specification.
determined from sorption or desorption measurements. The balance must provide a sufficient mass resolution and
However, it is common to see sorption-desorption hysteresis. long term stability.

A B

I D
L

H G F

A. Humidity controller D. Humidity regulated module G. Vapour humidifier

B. Temperature controlled chamber E. Reference H. Flow control module

C. Balance module F. Sample 1. Dry gas

Figure 2.9.39.-1. - Example of an apparatus for the determination of the water sorption (other designs are possible)

General NoNces (1) apply to all monographs and other texts 355
2.9.39. Water-soHd iníeractions EUROPEAN PHARMACOPOEIA 8.0

It is also possible to measure amounts of water uptake not appear more like that seen with adsorption processes. X-ray
detectable gravimetrically using volumetric techniques. In crystallographic analysis and thermal analysis are particularly
sorne cases, direct analysis of water content by different useful for the study of such systems.
methods such as determination of the boiling point, For situations where water vapour adsorption occurs
determination of water by distillation, 10ss on drying or predominantly, it is very helpful to measure the specific
gas chromatography may be advantageous. In the case of surface are a of the solid by an independent method and to
adsorption, to improve sensitivity, one can increase the specific express adsorption as mass of water sorbed per unít area of
surface area of the sample by reducing particle size or by solid surface. This can be very useful in assessing the possible
using larger samples to increase the total area. It is important, importance of water sorption in affecting solid properties.
however, that such comminution of the solid does not alter For example, 0.5 per cent m/m uptake of water could hardly
the surface structure of the solid or render it more amorphous cover the bare surface of 100 m 2 /g, whiIe for 1.0 m 2 /g this
or otherwise less ordered in crystallinity. For absorption, amounts to 100 times more surface coverage. In the case of
where water uptake is independent of specific surface area, pharmaceutical solids which have a specific surface are a in the
only increasing sample size will help. Increasing sample size, range of 0.01 m 2 /g to 10 m 2/g, what appears to be low water
however, will increase the time to establish sorne type of content could represent a significant amount of water for the
equilibrium. To establish accurate values, it is important to get available surface. Since the "dry surface area" is not a factor
desolvation of the sample as thoroughly as possible. Higher in absorption, sorption of water with amorphous or partially
temperatures and lower pressures (vacuum) facilitate this amorphous solids can be expressed on the basis of unit mass
process; however, one must be aware of any adverse effects corrected for crystallinity, when the crystal form does not
this might have on the solid such as dehydration, chemical sorb significant amounts of water relative to the amorphous
degradation or sublimation. Using higher temperatures to regions.
induce desorption, as in a thermogravimetric apparatus,
Iikewise must be carefully carried out because of these possible DETERMINATION OF THE WATER ACTIVITY
pitfalls. PrincipIe. Water activity, is the ratio of vapour pressure
Report and interpretation of the data. Sorption data are of water in the product (P) to saturation pressure of water
usually reported as a graph of the apparent mass change in per vapour (Po) at the same temperature. It is numerically equal
cent of the mass of the dry sample as a function of relative to 11100 of the relative humidity (RH) generated by the
humidity or time. Sorption isotherms are reported both in product in a c10sed system. RH can be calculated from direct
tabular form and as a graph. The measurement method must measurements of partial vapour pressure or dew point, or
be traceable with the data. from indirect measurement by sensors whose physical or
electric characteristics are altered by the RH to which they
Adsorption-desorption hysteresis can be interpreted, for are exposed. Ignoring activity coeffieients, the relationship
example, in terms of the porosity of the sample, its state between Aw and equilibrium relative humidity (ERH) are
of agglomeration (capillary condensation), the formation represented by the following equations:
of hydrates, polymorphic change, or liquefying of the
sample. Certain types of systems, particularly those with P
Aw= -
microporous solids and amorphous solids, are capable of Po
sorbing large amounts of water vapour. Here, the amount
of water associated with the solid as relative humídity is ERH (per cent) = Aw x 100
decreased, is greater than the amount that originally sorbed as
Method. The water activity is determined by placing the
the relative humidity was increased. For microporous solids,
sample in a small airtight cup inside which the equilibrium
vapour adsorptíon-desorption hysteresis is an equilibrium
between the water in the solid and the headspace can be
phenomenon associated with the process of capillary
established. The volume of the headspace must be small in
condensation. This takes place because of the high degree of
relation to the sample volume in order not to change the
irregular curvature of the micropores and the fact that they
sorption state of sample during the test. The equilibration as
"fill" (adsorption) and "empty" (desorption) under different
a thermodynamic process takes time but may be accelerated
equilibrium conditions. For non-porous solids capable of
by forced circulation within the eell. The acquired water
absorbing water, hysteresis occurs because of a change in the
activity value is only valid for the simultaneously determined
degree of vapour-solid interaction due to a change in the
temperature. This requires a precise temperature-measuring
equilibrium state of the solid, e.g. conformation of polymer
device as part of the equipment. Furthermore, the probe must
chains, or because the time scale for structural equilibrium is
be thermally insulated to guarantee a constant temperature
longer than the time scale fOI water desorption. In measuring
during the test. The sensor measuring the humidity of
sorption-desorption isotherms, it is therefore important to
the headspace air aboye the sample is a key component.
establish that something close to an equilibrium state has
Theoretically, all types of hygrometers can be used, but for
be en reached. Particularly with hydrophilic polymers at high
analytical purposes miniaturisation and robustness are a
relative humidities, the establishment of water sorption or
precondition. The Aw measurement may be conducted using
desorption values independent of time is quite difficult, since the dew point/chilled mirror method(20). A polished, chilled
one is usually dealing with a polymer plasticised into its "fluid"
mirror is used as a condensing surface. The cooling system is
state, where the solid is undergoing significant change.
electronically linked to a photoelectric cell into which light
In the case of crystal hydrate formation, the plot of water is reflected from the condensing mirror. An air stream, in
uptake versus pressure or relative humidity will in these equilibrium with the test sample, is directed at the mirror,
cases exhibit a sharp in crease in uptake at a particular which cools until condensation occurs on the mirror. The
pressure and the amount of water taken up will usually temperature at which this condensation begins is the dew point
exhibit a stoichiometric mole:mole ratio of water to solido from which the ERH is determined. Commercially available
In sorne cases, however, crystal hydrates will not appear to instruments using the dew point/chilled mirror method
undergo a phase change or the anhydrous form will appear or other technologies need to be evaluated for suitability,
amorphous. Consequently, water sorption or desorption may qualified, and calibrated when used to make water activity

(20) AOAC Internationa! Oflicia! Method 978.18.

356 See the information section on general monographs (cover pages)


EUROPEAN PHARMACOPOEIA 8.0 2.9.40. Uniformity of dosage units

determinations. These instruments are typically calibrated apply to each active substance being comprised in dosage units
over an adequate range, for exampIe, using sorne saturated salt containing one or more active substances, unless otherwise
solutions at 25 oC such as those listed in Table 2.9.39.-1. specified elsewere in this Pharmacopoeia.
Table 2.9.39.-1. - Standard saturated salt solutions The uniformity of dosage units can be demonstrated by either
of 2 methods: content uniformity or mass variation (see
Saturated salts solutions ERH Table 2.9.40.-1).
at 25 oC (per cent)
Aw
The test for content uniformity of preparations presented in
Potassium sulfate dosage units is based on the assay of the individual contents of
97.3 0.973 active substance(s) of a number of dosage units to determine
(K,S04)
whether the individual contents are within the limits set. The
Barium chloride content uniformity method may be applied in all cases.
90.2 0.902
(BaCl,)
The test for mass variation is applicable for the following
Sodium chloride
75.3 0.753
dosage forms:
(NaCl) (l) solutions enclosed in single-dose containers and in soft
Magnesium nitrate capsules;
52.9 0.529
(Mg(NO,}2) (2) solids (including powders, granules and sterile solids) that
Magnesium .chloride
are packaged in single-dose containers and contain no added
32.8 0.328 active or inactive substances;
(MgCI,)
(3) solids (inc!uding sterile solids) that are packaged in
Lithium chloride single-dose containers, with or without added active or
11.2 0.112
(LiCI) inactive substances, that have been prepared from true
solutions and freeze-dried in the final containers and are
labelled to indicate this method of preparation;
(4) hard capsules, uncoated tablets, or film-coated tablets,
0412012:20940 containing 25 mg or more of an active substance comprising
25 per cent or more, by mass, of the dosage unit Of, in the case
of hard capsules, the capsule contents, except that uniformity
2.9.40. UNIFORMITY OF of other active substances present in lesser proportions is
DOSAGE UNITS demonstrated by meeting content uniformity requirements.
The test for content uniformity is required for all dosage forms
To ensure the consistency of dosage units, each unít in a batch not meeting the aboye conditions for the mass variation test.
should have an active substance content within a narrow Alternatively, products that do not meet the 25 mg/25 per cent
range around the label claim. Dosage units are defined as threshold limit may be tested for uniformity of dosage units
dosage forms containing a single dos e or a part of adose of an by mass variation instead of the content uniformity test
active substance in each dosage unit. Unless otherwise stated, on the following conditiol1: the concentration Relative
the uniformity of dosage units specification is not intended Standard Deviation (RSD) of the active substance in the final
to apply to suspensions, emulsions or gels in single-dose dosage units is not more than 2 per cent, based on process
container s intended for cutaneous administration. The test validation data and development data, and if there has been
for content uniformity is not required for multivitamin and regulatory approval of such a change. The concentration RSD
trace-eIement preparations. is the RSD of the concentration per dosage unit (m/m or m/V),
The term 'uniformity of dosage unit' is defined as the degree where COl1centration per dosage unit equals the assay result
of uniformity in the amount of the active substance among per dosage unit divided by the individual dosage unit mass.
dosage units. Therefore, the requirements of this chapter See the RSD formula in Table 2.9.40.-2.

Table 2.9.40.-1. - Application of Content Un iform ity (CU) and Mass Variation (MV) test for dosage forms
Dosage forms Type Sub-Type Dose and ratio of active substance

" 25 mg and " 25 per cent < 25 mg or < 25 per cent

Tablets uncoated MV CU

coated film-coated MV CU

others CU CU

Capsules hard MV CU

soft suspensions, emulsions, gels CU CU

solutions MV MV

Solids in single-dose
single component MV MV
containers

solution freeze-dried in final


multiple components MV MV
container

others CU CU

Solutions enclosed in MV MV
single-dose containers

Others CU CU

General Notices (1) apply to all monographs and other texts 357
2.9.40. Uniformity of dosage units EUROPEAN PHARMACOPOEIA 8.0

CONTENT UNIFORMITY is removed from an individual container in conditions of


normal use. Express the results as delivered dose. Calculate
Select not fewer than 30 units, and proceed as follows for the the acceptance value (see Table 2.9.40.-2).
dosage form designated. Where different procedures are used Calculation of Acceptance Value
for assay of the preparation and for the content uniformity
test, it may be necessary to establish a correction factor to be Calculate the Acceptance Value (A V) using the formula:
applied to the results of the latter.
1M-Xl +ks
Solid dosage forms. Assay 10 units individually using an
appropriate analytical method. Calculate the acceptance value for which the terms are as defined in Table 2.9.40.-2.
(see Table 2.9.40.-2). MAS S VARIATION
Liquid or semi-solid dosage forms. Assay 10 units Carry out an assay for the active substance( s) on a
individually using an appropriate analytical method. Carry representative sample of the batch using an appropriate
out the assay on the amount of well-mixed material that analytical method. This value is result A, expressed as

Table 2.9.40.-2.
Variable Definition Conditions Value
-
X Mean of individual contents (Xl'
x" ... , x,), expressed as a percentage
of the label claim

xl' x2 )",) XII


Individual contents of the
dosage units tested, expressed
as a percentage of the label claim

11 Sample size (number of dosage units


in a sample)

k Acceptability constant If 11 = 10, then 2.4

If 11 = 30, then 2.0

s Sample standard deviation


[iE Xl']'"
"
n-1

RSD Relative standard deviation 100s


~-

M (case 1) Reference value If 98.5 per cent <; X <; 101.5 per M=X
To be applied when T <; 101.5 cent, then (AV= ks)

If X < 98.5 per cent, then M = 98.5 per cent


(A V = 98.5 - X + ks)

IfX> 101.5 per cent, then M = 101.5 per cent


(AV= X -101.5 + ks)

M (case 2) Reference value If 98.5 per cent <; X <; T, then M=X
To be applied when T> 101.5 (AV=ks)

If X < 98.5 per cent, then M = 98.5 per cent


(AV= 98.5 - X + ks)

IfX> T, then M= T per cent


(AV=X-T+ks)

Acceptance value (A V) General formula:


IM- + ks
Calculations are specified above
for the difIerent cases.

L1 Maximum allowed acceptance value L1 = 15.0 un les s otherwise specified


L2 Maximum allowed range for On the low side, no dosage unit L2 = 25.0 un les s otherwise specified
deviation of each dosage unit tested result can be les s than 0.75 M while
from the calculated value of M on the high side, no dosage unit
result can be greater than 1.25 M
(This is based on L2 value of 25.0)

T Target content per dosage unit at


time of manufacture, expressed
as a percentage of the labe! claim.
Unless otherwise stated, T is
equal to 100 per cent or T is the
manufacturer's approved target
content per dosage unit

358 See the information section on general monographs (cover pages)


EUROPEAN PHARMACOPOEIA 8.0 2.9.41. Friability of granules and spheroids

percentage of label claim (see Calculation of Acceptance tha11 (1 - L2 x O.Ol)M or more than (l + L2 x O.Ol)M in
Value). Assume that the concentration (mass of active calculation of acceptance value under content uniformity or
substance per mass of dosage unit) is uniformo Select not under mass variation. Unless otherwise specified, L1 is 15.0
fewer than 30 dosage units, and proceed as follows for the and L2 is 25.0.
dosage form designated.
Uncoated or film-coated tablets. Accurately weigh 10 tablets 0412012:20941
individually. Calculate the active substance content, expressed
as percentage of label claim, of each tablet from the mass of 2.9.41. FRIABILITY OF GRANULES
the individual tablets and the result of the assay. Calculate the
acceptance value. AND SPHEROIDS
Hard capsules. Accurately weigh 10 capsules individually, This chapter describes 2 methods for determination of the
taking care to preserve the identity of each capsule. Remove friability of granules and spheroids, which may be used during
the contents of each capsule by suitable means. Accurately development studies. It is recognised, however, that many
weigh the emptied shells individually, and calculate for each methods with equal suitability may be used.
capsule the net mass of its contents by subtracting the mass of This test is intended to determine, under defined conditions,
the shell from the respective gross mass. Calculate the active the friability of granules and spheroids. Friability is defined as
substance content in each capsule Írom the mass of product a reduction in the mass of the granules or spheroids or in the
removed from the individual capsules and the result oí the formation of fragments of granules or spheroids, occurring
assay. Calculate the acceptance value. when the granules ol' spheroids are subjected to mechanical
Soft capsules. Accurately weigh 10 intact capsules individually strain during handling (tumbling, vibration, fluidisation, etc.).
to obtain their gross masses, taking care to preserve the Examples of changes are abrasion, breakage or deformation of
identity of each capsule. Then cut open the capsules by means granules or spheroids.
of a suitable clean, dry cutting instrument such as scissors or a
sharp open blade, and remove the contents by washing with a METHODA
suitab1e solvent. Allow the occluded solvent to evaporate from Apparatus (fluidised-bed apparatus). The apparatus (see
the shells at room temperature over a period of about 30 min, Figure 2.9.41.-1) consists of a glass cylinder (A) with a
taking precautions to avoid uptake or 10ss of moisture. Weigh conicallower part. The cylinder is provided with a sieve
the individual shells, and calculate the net contents. Calculate lid (B) having al1 aperture size of 500 flm or any other
the active substance content in each capsule from the mass of suitable sieve. The conical end is connected to a U -shaped
product removed from the individual capsules and the result glass tube (C) that can be disconnected from the cylinder
of the assay. Calculate the acceptance value. for removal ofthe granules or spheroids. The U-tube is
Solid dosage forms other than tablets and capsules. Proceed attached to a T-coupling (D). One inlet ofthe T-coupling
as directed for hard capsules, treating each unit as described is joined by a silicone tube to a manometer for regulating
therein. Calculate the acceptance value. the compressed-air flow (use compressed air complying with
the test for water in the monograph Medicinal air (1238)),
Liquid 01' semi-solid dosage forms. Accurately weigh the the other one is connected via a silicone tube to a by-pass
amount ofliquid or semi-solid that is removed from each of 10 flowmeter (E) (0.10-1.00 m 3.h- 1).
individual containers in conditions of normal use. lf necessary,
compute the equivalent volume after determining the density. Procedure. The following procedure is usually suitable.
Calculate the active substance content in each container from Remove the fine particles by sieving (sieve having an aperture
the mass of product removed from the individual containers size of 710 flm or any other suitable sieve). Introduce about
and the result of the assay. Calculate the acceptance value. 8.0 g (m j ) of granules or spheroids into the cylinder (A). Close
the apparatus with the sieve lid (B). Adjust the flow rate of
Cakulation of Acceptance Value. Calculate the acceptance the compressed air to 0.45 m 3 ·h- 1 After 15 min, remove the
value (A V) as shown in content uniformity, except that granules or spheroids from the apparatus by disconnecting
the individual contents of the units are replaced with the the U-tube and weigh again (m 2 ). Test 3 samples and calculate
individual estimated contents defined below. the mean value. 1t is recommended to spray the inside of the
individual estimated contents of the apparatus with an antistatic agent every 3 determinations in
dosage units tested; order to prevent electrostatic charging.
L055 on drying. Dry in an oven at 105 oC, unless otherwise
where
prescribed. Alternatively, other drying conditions as described
A in general chapter 2.2.32 may be used.
Xi = Wi X
W Cakulation

individual masses of the dosage units F = mI (100 - TI) - m2 (100 - T 2 ) x 100


tested; mI
A content of active substance (percentage of
F friability;
label claim) obtained using an appropriate
analytical method (assay); percentage loss on drying befare the test (mean of
w mean of individual masses (Wl' w2, ... , w,,). 2 determinations);
T2 percentage 10s5 on drying after the test (mean of
CRITERIA 2 determinations);
Apply the following criteria, unless otherwise specified. mj mas s of the granules or spheroids before the test,
in grams;
Solid, semi-soHd and liquid dosage forms. The requirements
for dosage uniformity are met if the acceptance value of m2 mass of the granules or spheroids after the test, in
the first 10 dosage units is les s than or equal to Ll per grams.
cent. If the acceptance value is greater than L1 per cent,
test the next 20 dosage units and calculate the acceptance METHOD B
value. The requirements are met if the fina! acceptance Apparatus (oscillating apparatus). The apparatus (see
value of the 30 dosage units is less than or equal to L1 per Figure 2.9.41.-2) consists of a glass container, containing the
cent and no individual content of the dosage unit is less granules or spheroids to be examined, which is subjected

General Notices (1) apply lo all monographs and other texts 359
2.9.41. Friability of granules and spheroids EUROPEAN PHARMACOPOEIA 8.0

5cm~

~
E §
l!)

55 cm .
JII L NS 145/23
4.5 cm

-
5 cm

lNS 145/2j
--<-1--1-0--'-1 cm
o
E
()

E
()

E
()
r-;:;-"1
~
~
'"
~

~ ~

Figure 2.9.41.-1. - Fluidised-bed apparatus

D
Glass container

Time Frequency
107 ± 3 mL glass container,
42.0 ± 0.5 mm in internal diameter
and 85.0 ± 1 mm in height,
with a twist-off cap.

Figure 2.9.41.-2. - Oscillating apparatus

to horizontal oscillations. The frequency and duration of or for 120 S at a lower frequency (e.g. 1400scillations/min)
the oscillations can be varied continuously. The frequency for soft granules or spheroids. Sieve (355 11m, or the same
can be adjusted, using a scale, to a value in the range sieve as used previously) and weigh the granules or spheroids
0-400 oscillations/min. The duration can be set to a value in again (m 2 ). Test 3 samples and calculate the mean value.
the range 0-9999 s.
L055 on drying. Dry in an oven at 105 oC, unless otherwise
Procedure. The following procedure is usually suitable. prescribed. Alternatively, other drying conditions as described
Remove the fine particles by sieving (sieve having an aperture in general chapter 2.2.32 may be used.
size of 355 flm or any other suitable sieve). In the glass
container, weigh about 10.00 g (m,) of the granules or Calculation
spheroids. Install the container in the apparatus. Shake for F = ml (100 ~ Td ~ m2 (100 ~ T 2 ) x 100
240 s at the highest frequency for hard granules or spheroids, ml

360 See the information section on general monographs (cover pages)


EUROPEAN PHARMACOPOEIA 8.0 2.9.43. Apparent dissolution

F friability; spreads through the dissolution medium according to its


physico-chemical properties.
T¡ percentage loss on drying before the test (mean of
2 determinations); In justified and authorised cases, representative fractions of
large volume suppositories may be tested.
T2 percentage 10ss on drying after the test (mean of
2 determinations); SAMPLING AND EVALUATION
m¡ mass of the granules or spheroids before the test, in Samples are always collected at the outlet of the ceH,
grams; irrespective of whether the circuit is opened or closed.
m2 mass of the granules OI spheroids after the test, in Filter the liquid removed using an inert fiIter of appropriate
grams. pore size that do es not cause significant adsorption of the
active substance from the solution and does not contain
substances extractable by the dissolution medium that would
0112008:20942
interfere with the prescribed analytical method. Proceed with
analysis of the filtrate as prescribed.
2.9.42. DISSOLUTION TEST FOR The quantity of the active substance dissolved in a specified
LIPOPHILIC SOLID DOSAGE FORMS time is expressed as a percentage of the content stated on the
label.
APPARATUS
The apparatus (see Figure 2.9.42.-1) consists of:
A reservoir for the dissolution medium.
- A pump that forces the dissolutíon medium upwards (3)
through the flow-through cell.
A flow-through ceH shown in Figure 2.9.42.-2 specifically Metal grid
Capillary
intended for lipophilic solid dosage forms such as
suppositories and soft capsules. It consists of 3 transparent (2)
parts which fit into each other. The lower part (1) is made
up of 2 adjacent chambers connected to an overflow device.
The dissolutíon medium passes through chamber A and is
subjected to an upwards flow. The flow in chamber B is
downwards directed to a smaH-size bore exit which leads
upwards to a filter assembly. The middle part (2) of the cel! L{)
L{)

has a cavity designed to collect lipophilic excipients which


float on the dissolution medium. A metal grill serves as a (1 )
rough filter. The upper part (3) holds a filter unit for paper,
glass fibre or cellulose filters.
A water-bath that will maintain the dissolution medium at
37 ± 0.5 oc.

Sieve with
point

el 1.6

Figure 2.9.42.-2. - Flow-through cell


Dimensions in millimetres
Reservoir for Pump Thermostatically controlled Collecting receptacles
dissolution medium flow-through cel! and filter for analysis

01/2008:20943
Figure 2.9.42.-1. - Flow-through apparatus
corrected 6.1

Dissolution medium. If the dissolution medium is buffered,


adjust its pH to within ± 0.05 units of the prescribed value.
2.9.43. APPARENT DISSOLUTION
Remove any dissolved gases from the dissolution medium This method is mainly used to determine the apparent
before the test since they can cause the formatíon of bubbles dissolution rate of pure solid substances. It may also be used
that significantly affect the results. for the determination of the apparent dissolution rate of active
substances in preparations presented as powders or granules.
METHOD APPARATUS
Place 1 unít of the preparation to be examined in chamber A. AH parts of the apparatus that may come into contact with the
Close the ceU with the prepared filter assembly. At the sample or the dissolution medium are chemically inert and do
beginning of the test, chamber A requires air removal via not adsorb, react with, or interfere with the test sample. No
a small orifice connected to the filter assembly. Heat the part of the assembly or its envíronment contributes significant
dissolution medium to an appropriate temperature taking the motion, agitation or vibration beyond that resulting from the
melting point of the preparation into consideration. Using a flow-through system.
suitable pump, introduce the warmed dissolution medium Apparatus that permits observatíon of the sample is preferable.
through the bottom of the ceH to obtain a suitable continuous
The apparatus (see Figure 2.9.43.-1) consists of:
flow through an open or closed circuit at the prescribed
rate (± 5 per cent). When the dissolution medium reaches - a reservoir for the dissolutíon medium;
the overflow, air starts to escape through the capillary and - a pump that force s the dissolution medium upwards
chamber B fills with the dissolution medium. The preparation through the flow-through cel!;

General Notices (1) apply to all monographs and other texts 361
2.9.43. Apparent dissolution EUROPEAN PHARMACOPOEIA 8.0

r-----
r--

j
I
I

A B e o

A. reservoir for dissolution medium B. pump C. thermostatically control!ed flow-through cel! and filter D. col!ecting vessels for analysis

Figure 2.9.43.-1. - Flow-through apparatus

- a flow-through ceU, preferably of transparent material, cOl1tain substances extractable by the dissolution medium
mounted vertical!y with a filter system preventing escape of that would interfere with the prescribed analytical method.
undissolved particles; Proceed with the analysis of the filtrate as prescribed.
a water-bath that will maintain the dissolution medium at
the chosen temperature (generally 37 ± 0.5 OC). ASSESSMENT OF THE RESULTS
The flow-through cel! shown in Figure 2.9.43.-2 consists of When the test is performed for batch releas e purposes, an
3 parts that fit into each other. The lower part supports a adequate number of replicates is carried out.
system of grids and filters on which the powder is placed. The The results are expressed as:
middle part, which fits onto the lower part, contains an insert
that sieves the sample when the dissolution medium flows - the amount of dissolved substance by time unit (if the
through the cel!. This insert is made up of 2 parts: a conical dissolution is linear) ;
sieve that is placed on the sample and a clip placed midway - the dissolution time of the whole sample and at appropriate
down the middle part to hold the sieve in place when the intermediate stages.
dissolution medium passes through. A 2nd filtration assembly
(grid and filter) is placed on top of the middle part befo re
fitting the upper part through which the dissolution medium
flows out of the cell.
~ ___ F
DISSOLUTION MEDIUM
If the dissolution medium is buffered, adjust its pH to within
± 0.05 units. Remove any dissolved gases from the dissolution
medium before the test, sin ce they can cause the formation of
bubbles, which significantly affect the results. ____- - - E
METHOD ____- - - c
co
L()
Place a bead of 5 ± 0.5 mm diameter at the bottom of the
con e of the lower part followed by glass beads of suitable
size, preferably of 1 ± 0.1 mm diameter. Place a sieve (with
0.2 mm apertures), a suitable filter and a 2nd sieve on top of ____- - - B
the lower parto Fit the middle part onto the lower part. Weigh

~I
the assembly. Place the sample on the filtration assembly and
weigh the sample in the cel!. Place the sieve of the insert,
cone upwards, on the sample, and position the clip midway
down the middle part. Place a sieve (with 0.2 mm apertures)
and a suitable filter on top of the middle part. Fit the upper
part. Heat the dissolution medium to the chosen temperature.
Using a suitable pump, introduce the dissolution medium
through the bottom of the cell to obtain a suitable continuous
flow through an open or closed circuit at the prescribed 0.8
rate ± 5 per cent.

SAMPLING
A. lower part C. clip E. middle part
Samples of dissolutiol1 medium are collected at the outlet of
the ceH, irrespective of whether the circuit is opened or closed. B. sieve D. insert F. upper part

Immediately filter the liquid removed using an inert filter


Figure 2.9.43.-2. - Flow-through ceU
of appropriate pore size that does 110t cause significant
adsorptiol1 of the substances from the solution and does 110t Dimensions in millimetres

362 See the information section on general monographs (caver pages)


EUROPEAN PHARMACOPOEIA 8.0 2.9.44. Preparations for nebulisation: characterisation

0112012:20944 Filter system. A suitably validated low-resistance filter,


capable of quantitatively collecting the aerosol and enabling
recovery of the active substance with an appropriate solvent,
2.9.44. PREPARATIONS is used for the test. The dead volume of the filter casing do es
FOR NEBULISATION: not exceed 10 per cent ofthe tidal volume used in the breath
simulation.
CHARACTERISATION
METHOD
Products used for nebulisation and intended for pulmonary Attach the filter (contained in the fiIter holder) (A) to the
delivery are characterised using the following tests: breath simulator (B) according to Figure 2.9.44.-1. Fill the
- Active substance delivery rate and total active substance nebuliser (C) with the volume of the medicinal product as
delivered; specified in the patient instructions. Attach the mouthpiece
of the nebuliser to the inhalation filter using a mouthpiece
- Aerodynamic assessment of nebulised aerosols. adapter if required, ensuring that connections are airtight.
These tests standardise the approach given to the assessment Make sure the nebuliser is positioned in the same orientation
of the dose that would be delivered to a patient but are as intended for use; this may require tilting the breathing
not intended to provide assessment of the nebuliser simulator and filter holder. Set the breathing simulator to
device itself, which is described in the European standard generate the specified breathing pattern.
EN 13544-1 :2007 +Al:2009, Respiratory therapy equipment -
Part 1: Nebulizing systems and their components.
The mass- rather than the number-weighted size distribution
is more appropriate to evaluate product performance. Indeed,
active substance mass as a function of aerodynamic diameter
is more indicative of therapeutic effect within the respiratory
tract. A B
A. inhalation filter and filter holder B. breathing simulator C. nebuliser
ACTIVE SUBSTANCE DELIVERY RATE AND TOTAL
ACTIVE SUBSTANCE DELIVERED Figure 2.9.44.-1. - Experimental set-up for breathing simulator
These tests are performed to assess the rate of delivery to testing
the patient and the total active substance delivered to the Start the breathing simulator then, at the beginning of an
patient, using standardised conditions of volumetric flow inhalation cycle, start the nebuliser. Operate the nebuliser
rateo It is essential that breath-enhanced and breath-actuated for a defined initial time periodo The time chosen, usually
nebulisers be evaluated by a breathing simulator, as the output 60 ± 1 s, must allow sufficient active substance deposition
of these types of device is highly dependent on inhalation on the inhalation filter to allow quantitative analysis. If the
flow rateo The methodology below describes the use of a quantity of active substance deposited on the inhalation filter
standard breathing pattern defined for adults. Should a in 60 s is insufficient for this analysis, the length of the time
particular product for nebulisation only be indicated for interval for aerosol collection can be increased. If the filter is
paediatric (i.e. neonate, infant or child) use, then paediatric soaked with the preparation, this time can be decreased. At
breathing pattern(s) must be used. Breathing patterns are the end of this initial period, stop the nebuliser.
used, rather than continuous flow rates, to provide a more
appropriate measure of the mass of active substance that Place a fresh filter and filter holder in position and continue
would be delivered to patients. until nebulisation ceases. Interrupt nebulisation and exchange
filters if necessary, to avoid filter saturation.
Active substance delivery rate and total active substance
delivered are appropriate characteristics because they allow RESULTS
the mass delivered to be characterised in a standard way Using a suitable method of analysis, determine the mass of
regardless of the nebuliser used. Accordingly, the test active substance collected on the filters and filter holders
methodology described below allows that the mass of active during each time interva!. Determine the active substance
substance delivered in the P' period (typically 1 min) is delivery rate by dividing the mass of active substance collected
measured (consequently giving an assessment of active on the first inhalation filter by the time interval used for
substance delivery rate) as well as capturing the total mass collection. Determine the total mass of active substance
of active substance delivered. delivered by summing the mas s of active substance collected
on all inhalation filters and filter holders.
APPARATUS
Breathing simulator. A commercially available breathing AERODYNAMIC ASSESSMENT OF NEBULISED
simulator, which is able to generate the breathing profiles AEROSOLS
specified in Table 2.9.44.-1, is used for the test. The breathing Nebulised products need to be size-characterised at flow
pro file indicated for adults is used unless the medicinal product rates lower than the range that is normally used for powder
is specifically intended for use in paediatrics, where alternate inhalers and metered-dose inhalers. A flow rate of 15 L/min
patterns should be used, as indicated in Table 2.9.44.-l. is recommended in the European standard because this value
represents a good approximation to the mid-inhalation flow
Table 2.9.44.-1. - Breathing simulator specifications rate achievable by a tidally breathing healthy adult (500 mL
Item Specification tidal volume).
Adult Neonate Infant Child Although low-angle laser light scattering instruments
(laser diffractometers) can provide rapid size-distribution
Tidal 500 mL 25 mL 50 mL 155 mL measurements of nebuliser-generated aerosols, these
volume
techniques do 110t detect the active substance; rather they
Frequency 15 40 30 25 measure the size distribution of the droplets irrespective of
cycles/min cycles/min cycles/min cycles/min
their content. This may not be a problem with homogeneous
Waveform sinusoidal sinusoidal sinusoidal sinusoidal solutions, but can result in significant error if the product
Inhala- 1:1 1:3 1:3 1:2 to be nebulised is a suspension, or if droplet evaporation is
tion/exha- significant as can be the case with certain nebuliser types.
lation ratio Cascade impactors enable the aerosol to be characterised

General Notices (1) apply to all monographs and other texts 363
2.9.44. Preparations for nebulisation: characterisation EUROPEAN PHARMACOPOEIA 8.0

unambiguously in terms of the mass of active substance as a the sampling period (Ta) is the most effective way to avoid
function of aerodynamic diameter. Laser diffraction may be overloading in any given system, balancing overloading with
used if validated against a cascade impaction method. analytical sensitivity.
Apparatus E (see below under Apparatus), a cascade impactor, Re-entrainment. Droplet bounce and re-entrainment are
has been calibrated at 15 L/min specifically to meet the less likely with nebuliser-produced droplets than with solid
recommendation of the European standard, and is therefore partic!es from inhalers and for that reason coating would not
used for this test. Determining mass balance in the same normally be required.
way as for powder inhalers and metered-dose inhalers is METHOD
not straightforward, in that the dose is being captured as Pre-cool the assembled impactor and induction port in a
a continuous output, and hence is not inc!uded. As part refrigerator (set at about 5 OC) for not less than 90 min and
of method development, recovery experiments must be start the determination within about 5 min of removal of the
performed to validate the method. impactor from the refrigerator. Other methods that maintain
the impactor at a constant temperature (for example, use of a
It is also recognised that the control of evaporation of
cooling cabinet) can also be employed when validated.
droplets produced by nebulisers may be critical to avoid
bias in the droplet size assessment process. Evaporation can Set up the nebuliser with a supply of driving gas (usually
be minimised by cooling the impactor to a temperature of air or oxygen), or use a compressor, at the pressure and
about 5 oC, typically achieved by cooling the impactor in a flow rate specified by the manufacturer of the nebuliser.
refrigerator for about 90 mino Typically, at least after each Take precautions to ensure that the gas supply line does not
day of use, the apparatus must be fully c!eaned, inc!uding the beco me detached from the nebuliser when under pressure.
inter-stage passageways, in view of the greater risk of corrosion Fill the nebuliser with the volume of the medicinal product as
caused by the condensation/accumulation of saline-containing specified in the patient instructions.
droplets on inter-stage metalwork associated with cooling Remove the impactor from the refrigerator. Attach the
the impactor. It is recommended to dry al! surfaces of the induction port to the impactor, and connect the outlet of the
apparatus after each test, for examp!e with compressed airo impactor/external fiIter to a vacuum source that is capable of
Note: the micro-orifice collector (MOC) should not be dried drawing air through the system at 15 L/min as specified in
with compressed air. Figure 2.9.44.-2. Turn on the flow through the impactor.
APPARATUS Connect a flow meter, calibrated for the volumetric flow
A detailed description of Apparatus E and the induction port leaving the meter, to the induction port. Adjust the flow
is contained in general chapter 2.9.18, and includes details control valve located between the impactor and the vacuum
of critica! dimensions and the qualification process for the source to achieve a steady flow through the system at 15 L/min
impactor (stage mensuration). (± 5 per cent). Remove the flow meter.
Make sure the nebuliser is positioned in the same orientation
A back-up filter in addition to the micro-orifice as intended for use then attach the mouthpiece of the
collector (MOC) must be used to ensure quantitative recovery nebuliser to the induction port, using a mouthpiece adapter if
of active substance from the nebulised aerosol at the specified required, ensuring that connections are airtight. Switch 011 the
flow rate of 15 L/min. The filter is located below the MOC flow/compressor for the nebuliser. Sample for a predetermined
(internal filter option) or a filter in holder, externa! to the time (To)' Once determined, this time (To) must be defined
impactor, is used to capture any fine drop!ets that pass beyond and used in the analyticalmethod for a particular medicinal
the last size fractionating stage. product to ensure that mass fraction data can be compared.
A pre-separator is not used for testing nebuliser-generated At the end of the sampling period, switch off the driving gas
aerosols. flow/compressor to the nebuliser, remove the nebuliser from
the induction port and switch off the flow from the vacuum
METHOD VALIDATION source to the impactor.
Impactor stage ovedoading. During method development Dismantle the impactor and, using a suitable method of
and validation, it is important to confirm that the volume analysis, determine the mass of active substance collected
of liquid sampled from the nebuliser does not overload the in the induction port, on each stage and on the back-up
impactor. Visual inspection of the collection surfaces on filter/external fiIter as described for Apparatus E in general
stages collecting most of the droplets may reveal streaking chapter 2.9.18. Add the mass of active substance collected in
if overloading has occurred. This phenomenon is usually the MOC to that deposited on the back-up filter/externa! filter
also associated with an increase in mass of active substance and treat as a single sample for the purpose of subsequent
collected on the final stage and back-up filter. Reducing calculations.

e o
A. nebuliser B. induction port C. impactor (apparatus E) D. f]ow control valve E. VaCUU111 source

Figure 2.9.44.-2. - Apparatus E for measuring the size distribution of preparations for nebulisation

364 See the information section on general monographs (cover pages)


EUROPEAN PHARMACOPOEIA 8.0 2.9.45. Wettability of porous solids induding powders

Calculate the mass fraction (Fm,comp) of the active substance 07/2009:20945


deposited on each component of the impactor, commencing
with the induction port and proceeding in order through the
impactor, using the following expression: 2.9.45. WETTABILITY OF POROUS
F _ m comp SOLIDS INCLUDING POWDERS
rn,cornp - NI
INTRODUCTION
mass associated with the component under
evaluation; The wettability of solid surfaces is commonly characterised by
M total mass collected by the system. direct or indirect contact angle measurements. The contact
angle (8) between a liquid and a solid is the angle naturally
formed when a drop of a liquid is placed on a solid surface.
Present Fm,comp in order of location within the measurement This is depicted in Figure 2.9.45.-l. For a given liquid, wettable
equipment, beginning at the induction port and ending with
solids show a low contact angle and non-wettable solids show
the back-up filter of the impactor (see Figure 2.9.44.-3). Where
a contact angle of 90° or more.
appropriate, Fm,comp for adjacent stages of the impactor may be
combined in order to report the mass fraction collected on a
group of stages as a single value.

0.3

Figure 2.9.45.-l. - Contact angle (8) of a sessile drop observed


"-
011 a non-porous surface
~ 0.2
LLÉ 2 methods for the determination of wettability are deseribed
below. The methods are eapable of measuring the wettability
0.1
of porous solids Iike powders or granules. Both methods
express the wettability by a contact angle measurement
between the porous solid and a given liquido
The sessile drop method is based on direct measurement of a
1:: C\l (") '<t L!) <O r-- Ü contact angle of a sessile drop on a compaeted powder dise.
o (J) (J) (J) (J) (J) (J) (J) O
o... Ol Ol Ol Ol Ol Ol Ol :;¡;
e ro ro ro ro ro ro With the Washbufll method the contact angle is indirectly
o Uí 2 -.:::
Uí U) Uí Uí Uí Uí
:o
:::J
~
LL
measured. The method is based on the capillary effect of
u the powder pores, The effect (mass gain) is recorded by
E
special electronic balances starting the moment when the
powder sample touches the surface of a liquid, preferably not
Figure 2.9.44.-3. - Example of mass fraetíon of droplets dissolving or poorly dissolving the sample. The measurement
presented ín terms of location within the sampling system has very little or no effect on the state of the powder.
Determine the cumulative mass-weighted particle-size Any pre-treatment of the sample to be examined is
distribution of the aerosol size-fractionated by the impactor in disadvantageous, since the properties may be significantly
accordance with the procedure given in general chapter 2.9.18. altered. For example, the compaction of a powder as a dise
Starting at the filter, derive a cumulative mass versus effective may decrease the surface free energy when the crystalline
cut-off diameter of the respective stages (see Table 2.9.44.-2 state ofthe powder is changed (e.g. metastable forms), 01'
for the appropriate cut-off diameters at 15 L/min). Plot the may increase surface free energy by creating crystal defects
cumulative fraction of active substance versus cut -off diameter (disadvantage of the sessile drop method since compacted
in a suitable format,for example logarithmic or log-probability powder dises are tested).
format. Where appropriate, determine by interpolation the
The methods are usually applied to examine the following
fraction either below a given size or between an upper and
parameters:
a lower size limit.
- batch-to-batch consistency of samples in terms of
Table 2.9.44.-2. - Cut-off sizes for Apparatus E at 15 Llmín wettability;
Stage Cut-off diameter (f1m) - effect of liquid viscosity on wettability;
14.1 - effect of surface tension of a liquid on wettability;
2 8.61 - alteration of surface properties of samples.
5.39
SESSILE DROP METHOD
4 3.30
This method may be used to characterise directly the
5 2.08 wettability of coatings and compacted formulations sueh as
tablets. Moreover, it is sometimes possible to use the sessile
6 1.36
drop instrument in a dynamic measurement (dynamic contact
7 0.98 angle measurement, Figure 2.9.45.-2) of porous solid/liquid
systems where the contact angle decreases. By taking several
If necessary, and where appropriate, determine values for eontact angle measurements as a function of time, the rate of
the mass median aerodynamic diameter (MMAD) and the spreading accompanied by penetration of a liquid droplet into
geometric standard deviation (GSD), as appropriate. a slightly porous solid may be studied.

General Notices (1) apply to all mOl1ographs and other texts 365
2.9.45. Wettability of porous solids induding powders EUROPEAN PHARMACOPOEIA 8.0

A= r¡
gas e x p2 x , X cos g (2)

II viscosity of the liquid;


p density of the liquid;
Ys
y surface tension of the liquid;
8 contact angle between the solid and the liquid;
e material constant, dependent on the porous texture
of the solido

Equations (1) and (2) lead to equation (3):

m2 r¡
cos g = - x ~--:o--
t e x p2 x, (3)
Figure 2.9.45.-2. - Sessile drop determination with visual
inspection of the droplet In setting up a Washburn determination, a liquid with known
Under equilibrium conditions the contact angle of a sessile density (p), viscosity (ll), and surface tension (y) is used.
drop depends on 3 interrelated surface tensions and is Under these conditions, when the mass of liquid rising into
determined using Young's equation (see Figure 2.9.45.-2, the porous solid is monitored as a function of time (such that
2
1" part): capillary penetration rate (~ ) is the experimental data),
,s = ,SL + ,L cos g 2 unknowns remain according to equation (3): the contact
angle (8) of the liquid on the solid, and the solid material
ys surface tension of the solid with air; constant (e).
Determination ofthe material constant (e). The material
YSL interfacial tension of the solid with the liquid;
constant for a porous solid is determined by the following
surface tension of the liquid with airo equation, considering cylindrical pores:
PROCEDURE
Since powders are unable to form a completely flat surface, (4)
2
the powder is usually compacted as a disc in an attempt to
make the surface smoother. A liquid drop with a given volume r
is placed on the disc (see Figure 2.9.45.-2) allowing direct average capillary radius within the porous solid;
measurement of the contact angle using a goniometer fitted N number of capillaries per volumetric unit.
with an eyepiece protractor, or by geometric construction
on a photomicrograph. Other physical and mathematical lf a Washburn determination is performed with a liquid
procedures of data analysis may also be appropriate. The drop considered to have a contact angle of 0° (cos 0 = 1) on the 0

volume may influence the resulto Several determinations of


solid, then the solid material constant (e) is the only remaining
the contact angle (8) (n = 6) are usually carried out and the unknown in equation (3) and can thus be determined.
average is calculated. n-Heptane is the liquid of choice for determining material
WASHBURN METHOD constants because of its low surface tension (20.14 mN.m- 1
at 25 OC). n-Hexane may also be used (18.43 mN·m~ 1 at
The Washburn method is able to measure the contact angle of
25 OC) but is more volatile. If the powder dissolves too
porous solids with a contact angle in the range of 0-90 0

quickly in these liquids, hexamethyldisiloxane may be used
The tested material is the combination of the sample, the instead (15.9 mN-m~ 1 at 25 OC). Replicate determinations are
holder and the filter system. Therefore, an estimation or performed (n = 6) and the average value calculated.
determination of the true value is not possible and only
apparent values of the contact angle can be determined. Once the material constant (e) has been determined for the
However, the contact angle of the sample is the functional solid to be examined, a sample of the solid can be tested
property on which the result is significantly dependent. The for wettability by another liquido The material constant
outcome of the test is a ranking order listing the wettability determined by the n-heptane test is used in the Washburn
of different substances or formulations characterised by an equation, in combination with the capillary penetration
apparent contact angle. 2
rate (.~ ) data obtained while testing the substance to be
PRINCIPLE examined in the prescribed liquido This allows calculation of
lf a porous solid is brought into contact with a liquid, such the contact angle.
that the solid is not submerged in the liquid, but rather is just
touching the liquid surface, then the rise of liquid into the NOTE: if a series of liquids (at least 2liquids in addition to
pores of the solid due to capillary action will be governed by the liquid used to determine the material constant) is tested
the following equations: against a given solid then the resultant contact angle data can
be used to calculate the surface energy of the porous solido
2 t
m =- APPARATUS
A (1)
Figure 2.9.45.-3 shows the principal components of the
m mass of liquid sucked into the solid; apparatus. The main device is an electronic balance with
a suitable processor ensuring a suitable resolution in force
time elapsed sine e the solid and the liquid were measurement and a suitable resolution in lifting up the
brought into contact; immersion liquid towards the sample.
A constant, dependent on the properties of the liquid
and the solid to be examined, calculated using the Table 2.9.45.-1 indicates parameters of the electronic balance
following equation: that are generally considered suitable.

366 See the informatiol1 section 011 general monographs (cover pages)
EUROPEAN PHARMACOPOEIA 8.0 2.9.45. Wettability of porous solids induding powders

Table 2.9.45.-1. - Technical parameters of the eleetronie balance PROCEDURE


Lift Mass measurement FiHing of the sample holder. Place a disc of filter paper in
Range > 110 mm o - 210 g the bottom of the aluminium or glass sample holder. This
prevents powder from leaking out of the bottom of the celI.
Resolution 0.1 flm 10 flg The filter does not have to be made of paper, but it must be
Speed 0.099 - 500 mm/min a material that is easily wetted by the liquid to be tested. A
black-band filter (used for reverse osmosis) is recommended
because of its high porosity and minimum flow resistance.
Place a known amount of powder into the cel!. The
reproducibility of material constants and contact angles will
depend on the ability to weigh out the same amount of powder
for each test when a sufficient and adjusted amount of powder
is compacted in a uniform way (i.e. tapping/compaction of
the powder).
For most powders, a correct amount is in the range of a few

-lr-
----
grams, typically filling about 2/3 of the capacity of the holder.
Place a second piece of filter paper on top of the powder in the
cel!. This will prevent powder from rising through the holes
in the pisto n during the compaction process and/or during
-----------
----------
---------- - - E the determination.
------,.....,-----
------------- Tapping/ compaction of the powder. A bulk powder bed is
very porous and thus very sensitive to smal! influences that
--F
can easily alter the porosity and consequently the e-constant.
Therefore a tapped powder may be advantageous and will
A. electronic balance C. sample holder E. immersion liquid show more reproducible results. The appropriate l1umber
B. computer D. filter
oftaps must first be evaluated: 50-100 taps are usually
F. lift
appropriate.
Figure 2.9.45.-3. - Apparatus for contact angle measurement lf the aluminium sample holder is used then it may be
by the Washburn method mounted in the cylinder of a stamp volumeter, which can run
the evaluated number of taps.
Sample holders. The sample holder may be a small glass
cylinder with a sintered-glass filter at one end. rf tapping is not appropriate, the powder bed is compacted by
Powder material holders (see Figure 2.9.45-4) may also be screwing the piston of the aluminium sample holder applying
made of aluminium; they are less fragile than those made of a specified pressure.
glass and have small holes in the bottom that render them A further possibility is centrifugation under defined
easier to clean than a sintered-glass filter. The cover for the conditions. Where applicable, a compacted disc of the powder
cell is equipped with 2 screw threads. One connects it with sample may also be mounted on the electronic balance. A
the sample chamber while the other allows the user to guide sample holder is omitted in this case.
a pisto n down onto the sample itself and compact it. The After connecting to the balance, the sample holder is
apparatus is similar to an automatic tensiometer, except for positioned with the porous solid just aboye the surface of the
the sample holder. liquid (see Figure 2.9.45.-3), using the lift.
The liquid is raised further until it just touches the bottom of
the porous sample. Mass-versus-time data is then collected
as liquid penetrates into the solido Data can be presented
in either graphical or tabular format. The apparatus may
perform the whole determination automatically.
CRITICAL PARAMETERS
The following points must be considered.
Sample properties:
- water content of the sample;
- crystalline or solid-state properties of the sample
(polymorphic form, type of solvate).
Sample preparation:
- homogeneity of any powder blend to be examined;
- particle-size distribution; before testing it is sometimes
advisable to sieve the sample (e.g. using a 250 flm sieve);
the optimal compaction parameters (amount of sample,
number of taps or pisto n mass) must be determined;
- the compaction state of the different powder samples must
be uniform;
View 01 plunger bottom View 01 apparatus bottom - the sample holder OI, if used, the glass frit must be carefully
cleaned;
A. fixing C. thread E. capillary hales
- uniformity of the results is improved by using a sample
B. cover D. plunger F. capillary hales holder made of aluminium.
Figure 2.9.45.-4. - Example of sample holder with plunger for Immersion liquid:
compactíon of a powder - specifications of the immersion liquid must be indicated.

General Notiees (1) apply to all monographs and other texts 367
2.9.47. Uniformity of dosage units using large sample sizes EUROPEAN PHARMACOPOEIA 8.0

0412013:20947 Unless otherwise specified, Ll is 15.0 and L2 is 25.0.


Table 2.9.47.-l. is to be interpreted as follows:
2.9.47. DEMONSTRATION OF
- for a sample size of n = 400, enter the table at n 2 385:
UNIFORMITY OF DOSAGE UNITS k = 2.23 and e2 = 3;
USING LARGE SAMPLE SIZES - for a sample size of n = 450, enter the table at n 2 407:
The proeedure is intended far, but nat limited ta, the evaluatian k = 2.24 and c2 = 3;
af medicinal products that are manufactured using praeess - for a sample size of n = 500, enter the table at n 2 490:
analytical teehnalagy (PAT) methadalagy. k = 2.24 and e2 = 4.
Compliance with general chapter 2.9.40. Un ifarm ity af
dasage units can be demonstrated by the following procedure, ALTERNATIVE 2 (NON-PARAMETRIC)
when large samples (sample size n 2 100) are evaluated.
Application of this chapter does not constitute a mandatory Select not fewer than 100 units according to a predefined
requirement. It presents 2 alternative tests (Alternative 1 and sampling plan.
Alternative II). Fulfilling the requirements of either of the The consistency of dosage units is evaluated by content
2 alternatives is considered as evidence that the medicinal uniformity or mass variation as prescribed in Table 2.9.40.-1.
product tested complies with general chapter 2.9.40. The Assay individually or weigh the units and calculate individual
2 alternatives are considered equivalent in their demonstration contents as prescribed in general chapter 2.9.40. Count the
of compliance with general chapter 2.9.40. number of individual dosage units with a content outside
(l ± Ll x O.Ol)M and the number of individual dosage units
ALTERNATIVE 1 (PARAMETRIC) with a content outside (l ± L2 x O.Ol)M. Evaluate if the values
Select not fewer than 100 units according to a predefined are within the limits defined in Table 2.9.47.-2.
sampling plan.
CRITERIA
The consistency of dosage units is evaluated by content
uniformity OI mass variation as prescribed in Table 2.9.40.-l. Apply the following criteria, unless otherwise specified.
Calculate the acceptance value (A V) using the following The requirements for dosage form uniformity are met if:
expression: l. the number of individual dosage units outside
(1 ± L1 x O.Ol)M is less than or equal to el; and
IM-xl+ks 2. the number of individual dosage units outside
fOI which the terms are defined in Table 2.9.40.-2, but using the (1 ± L2 x O.Ol)M is less than or equal to e2.
sample size-dependent value for k defined in Table 2.9.47.-1.
el and e2 for a given sample size n are defined in Table 2.9.47.-2.
CRITERIA Unless otherwise specified, L1 is 15.0 and L2 is 25.0.
Apply the following criteria, unless otherwise specified.
Table 2.9.47.-2 is to be interpreted as follows:
The requirements for dosage form uniformity are met if:
1. the acceptance value (A V) is less than or equal to L1; and - for a sample size of n = 400, enter the table at n 2 394:
el = 11 and e2 = 3;
2. in the calculation of acceptance value (A V) under content
uniformity or under mass variation, the number of - for a sample size of n = 450, enter the table at n 2 434:
individual dosage units outside (l ± L2 x O.Ol)M is less el = 12 and e2 = 3;
than or equal to e2 as defined for a given sample size n in - for a sample size of n = 500, enter the table at n 2 490:
Table 2.9.47.-1. el = 13 and e2 = 4.

368 See the informatían seetían an general manographs (ca ver pages)
EUROPEAN PHARMACOPOEIA 8.0 2.9.47. Uniformity of dosage units using large sample sizes

Table 2.9.47.-1. - Acceptability constant (k) and acceptable number of dosage units
with a content outside (1 ± L2 x O.Ol)M (= e2) for a given sample size n

100 2.15 804 2.26 2480 2.29 23 4366 2.30 41 6252 2.31 59 8243 2.31 78
7
105 2.16 905 2.27 2585 2.29 24 4471 2.30 42 6357 2.31 60 8347 2.31 79

120 2.17 O 908 2.27 8 2690 2.29 25 4576 2.30 43 6462 2.31 61
8452 2.31 80

139 2.18 1013 2.27 9 2794 2.29 26 4680 2.30 44 6566 2.31 62
8557 2.31 81
1118 2.27 10 2899 2.29 27 4785 2.30 45 6671 2.31 63
161 2.19
8662 2.31 82
1223 2.27 3004 2.29 28 4890 2.30 46 6776 2.31 64
176 2.19 11 8767 2.31 83
1276 2.28 3109 2.29 4995 2.30 47 6881 2.31 65
189 2.20 29
1 8871 2.31 84
1328 2.28 12 3171 2.30 5099 2.30 48 6985 2.31 66
224 2.21
1432 2.28 13 3213 2.30 30 5204 2.30 49 7090 2.31 67 8976 2.31 85
270 2.22
1537 2.28 14 3318 2.30 31 5309 2.30 50 7195 2.31 68 9081 2.31 86
280 2.22
2 1642 2.28 15 3423 2.30 32 5414 2.30 51 7300 2.31 69 9186 2.31 87
328 2.23
1747 2.28 16 3528 2.30 33 5519 2.30 52 7404 2.31 70
9290 2.31 88
385 2.23
3 1851 2.28 3633 2.30 34 5623 2.30 53 7509 2.31 71
17 9395 2.31 89
407 2.24
1918 2.29 3737 2.30 35 5728 2.30 54 7614 2.31 72
9500 2.31 90
490 2.24
1956 2.29 18 3842 2.30 36 5833 2.30 55 7719 2.31 73
4
9605 2.31 91
516 2.25 2061 2.29 19 3947 2.30 37 5938 2.30 56 7824 2.31 74

594 2.25 9710 2.31 92


2166 2.29 20 4052 2.30 38 6042 2.30 7928 2.31 75
5 57
672 2.26 2270 2.29 21 4156 2.30 39 6136 2.31 8033 2.31 76 9814 2.31 93

699 2.26 6 2375 2.29 22 4261 2.30 40 6147 2.31 58 8138 2.31 77 9919 2.31 94

General Notiees (1) apply to all monographs and other texts 369
2.9.47. Uniformity of dosage units using large sample sizes EUROPEAN PHARMACOPOEIA 8.0

Tabie 2.9.47.-2. - Acceptable number of individual dosage units with a cantent outside (1 ± L1 )( O.Ol)M (= el) and
(1 ± L2 x O.Ol)M (= c2) respectively, for a given sample size n

I n P-J I c1 I C21 ~I
P-J I---'-------'
n I C211
C1 n P-J I c1 I C211 n P-) I c1 I C211 n P-J I c1 I C21 ~I
P-) I---'-------'
n I C211
C1 n P-) I c1 I C21
100 3 1432 35 2899 67 4366 98 5833 129 7300 160 8767 191
123 4 O 1476 36 13 2935 68 27 4377 99 41 5835 130 7304 161 8780 192 83
55 69
159 5 1521 37 2981 69 4424 100 5883 131 7351 162
8828 193
176 5 1537 37 3004 69 4471 101 5930 132 7399 163
8871 193
196 6 1566 38 14 3027 70 28 4518 102 42 5938 132 7404 163
1 8875 194
1611 39 3073 71 4565 103 5977 133 56 7447 164 70 84
234 7
1642 39 8923 195
273 8 3109 71 4576 103 6024 134 7494 165
1656 40 3120 72 6042 134 7509 165 8971 196
280 8 15 4612 104 43
29
1701 41 3166 73 6072 135 57 7542 166 71 8976 196
313 9 2 4658 105
1746 42 3212 74 6119 136 7589 167 9019 197 85
353 10 4680 105
1747 42 3213 74 6147 136 7614 167
385 10 4705 106 44 9066 198
1791 43 16 3259 75 30 6166 137 58 7637 168 72
394 11 4752 107 9081 198
3 1836 44 138 7684 169
3305 76 6214
434 12 4785 107 9114 199 86
1851 44
3318 76 6252 138 7719 169
476 13 4799 108 45
1882 45 17 9162 200
3351 77 31 6261 139 7732 170 73
490 13 4846 109 59
1927 46 9186 200
3398 78 6308 140 7779 171
517 14 4 4890 109
1956 46 9210 201 87
3423 78 6355 141 7824 171
559 15 4893 110
1972 47 18 46
3444 79 32 6357 141 7827 172 9257 202
594 15 2018 48 4940 111 74
3491 80 6403 142 60 7875 173 9290 202
601 16 2061 48 4987 112
5 3528 80 6450 143 7922 174 9305 203 88
644 17 2063 49 4995 112
19 3537 81 6462 143 7928 174
686 18 2109 33 5034 113 47 9353 204
50
3584 82 6498 144 61 7970 175 75
699 18 2154 51 5081 114 9395 204
3630 83 6545 145 8017 176
729 19 6 2166 51 5099 114 9401 205
3633 83 6566 145 8033 176 89
772 20 2200 52 20 5128 115 48 9449 206
3677 84 34 6592 146 62 8065 177 76
804 20 2246 53 5175 116
3723 85 6640 147 8113 178 9496 207
815 21 2270 53 5204 116
7 3737 85 6671 147 8138 178 9500 207
858 22 2291 54 21 5222 117 49
3770 86 35 6687 148 63 8160 179 77 9544 208 90
2337 55 5269 118
902 23
3817 87 6734 149 8208 180 9592 209
2375 55
908 23 5309 118
3842 87 6776 149 8243 180
2383 56 9605 209
945 24 8 22 5317 119
3863 88 36 50 6782 150 8256 181 78
2429 57 64 9640 210 91
989 25 5364 120
3910 89 6829 151 8303 182
2475 58 9688 211
1013 25 5411 121
3947 89 6877 152 8347 182
2480 58
1033 26 9 5414 121 9710 211
3956 90 6881 152 8351 183
2520 59 23 37 79
1077 27 5458 122 51 9735 212 92
2566 60 4003 91 6924 153 65 8399 184
1118 27 5505 123 154 185 9783 213
2585 60 4050 92 6972 8446
1121 28 4052 92 5519 123 6985 154 8452 185
2612 61 24 9814 213
10
1165 29 4097 93 38 5552 124 52 7019 155 66 8494 186 80
2658 62 9831 214 93
1209 30 4143 94 5599 125 7067 156 8542 187
2690 62 9879 215
1223 30 4156 94 5623 125 7090 156 8557 187
2704 63 25
9919 215
1253 31 11 4190 95 39 5647 126 53 7114 157 67 8589 188 81
2750 64
9927 216
1298 32 2794 64 4237 96 5694 127 7161 158 8637 189
5728 127 9975 217 94
1328 32 2796 65 4261 96 7195 158 8662 189
26 10023 218
1342 33 12 2843 66 4284 97 40 5741 128 54 7209 159 68 8685 190 82
1387 34 2889 67 4330 98 5788 129 7256 160 8732 191 10070 219

370 See the information section on general monographs (caver pages)


EUROPEAN PHARMACOPOEIA 8.0

3. Materials for containers and


containers

General No/ices (1) apply to all monographs and other tex/s 371
EUROPEAN PHARMACOPOEIA 8.0

372 See the information scction on general monographs (cover pages)


EUROPEAN PHARMACOPOEIA 8.0

Materials used for the


manufacture of containers
3.1. Materials used for the manufacture of containers ........ 375 3.1.7. Poly(ethylene - vinyl acetate) for containers and tubing
3.1.1. Materials for containers for human blood and blood for total parenteral nutrition preparations .......................... 391
components ............................................................................. 375 3.1.8. Silicone oil used as a lubricant.. ................................... 393
3.1.1.1. Materials based on plasticised poly(vinyl chloride) for 3.1.9. Silicone elastomer for closures and tubing ................. 394
containers for human blood and blood components ......... 375 3.1.10. Materials based onnon-plasticised poly(vinyl chloride)
3.1.1.2. Materials based on plasticised poly(vinyl chloride) for for containers for non-injectable, aqueous solutions ......... 395
tubing used in sets for the transfusion of blood and blood 3.1.11. Materials based on non-plasticised poly(vinyl
components ............................................................................. 378 chloride) for containers for dry dosage forms for oral
3.1.3. Polyolefins ....................................................................... 380 administration ......................................................................... 397
3.1.4. Polyethylene without additives for containers for 3.1.13. Plastic additives ............................................................ 398
parenteral preparations and for ophthalmic preparations .. 383 3.1.14. Materials based on plasticised poly(vinyl chloride)
3.1.5. Polyethylene with additives for containers for parenteral for containers for aqueous solutions for intravenous
preparations and for ophthalmic preparations ................... 384 infusion .................................................................................... 401
3.1.6. Polypropylene for containers and closures for parenteral 3.1.15. Polyethylene terephthalate for container s for
preparations and ophthalmic preparations ......................... 388 preparations not for parenteral use ...................................... 403

General Natices (1) apply ta all managraphs and ather texts 373
EUROPEAN PHARMACOPOEIA 8.0

374 See the information section on general monographs (ca ver pages)
EUROPEAN PHARMACOPOEIA 8.0 3.1.1.1. Plastidsed PVC materials for containers for blood

0112013:30100 Vinyl chloride. Head-space gas chromatography (2.2.28).


Internal standard solution. Using a micrasyringe, inject
3.1. MATERIALS USED FOR THE 10 flL of ether R into 20.0 mL of dimethylacetamide R,
immersing the tip of the needle in the solvent. lmmediately
MANUFACTURE OF CONTAINERS before use, dilute the solution to 1000 times its volume with
The materials described in this chapter are used for the dimethylacetamide R.
manufacture of container s for pharmaceutical use. Their use Test solution. Place 1.000 g of the material to be examined in a
may also be considered for the manufacture of part or all of 50 mL vial and add 10.0 mL of the internal standard solution.
objects used for medico-surgical purposes. Close the vial and secure the stopper. Shake, avoiding contact
Materials and polymers other than those described in the between the stopper and the liquido Place the vial in a
Pharmacopoeiamay be used subject to approval in each case water-bath at 60 ± 1 oC fOI 2 h.
by the competent authority responsible for the licensing for Vinyl chloride primary solution. Prepare in a fume cupboard.
sale of the preparation in the container. Place 50.0 mL of dimethylacetamide R in a 50 mL vial, stopper
the vial, secure the stopper and weigh to the nearest 0.1 mg.
Transmissible spongiform encephalopathies (5.2.8). A
Fill a 50 mL polyethylene or polypropylene syringe with
risk assessment of the product with respect to transmissible
gaseous vinyl chloride R, allow the gas to remain in contact
spongiform encephalopathies is carried out, and suitable
with the syringe for about 3 min, empty the syringe and fill
measures are taken to minimise any such risk.
again with 50 mL of gaseous vinyl chloride R. Fit a hypodermic
needle to the syringe and reduce the volume of gas in the
0112008:30101 syringe from 50 mL to 25 mL. lnject the remaining 25 mL
of vinyl chloride slowly into the vial shaking gently and
avoiding contact between the liquid and the needle. Weigh
3,1.1. MATERIALS FOR CONTAINERS the vial again; the increase in mass is about 60 mg (1 flL of the
FOR HUMAN BLOOD AND BLOOD solution thus obtained contains about 1.2 flg of vinyl chloride).
COMPONENTS Allow to stand for 2 hours. Keep the primar y solution in a
refrigerator.
NOTE: for materials based on plasticised poly(vinyl chloride) Vinyl chloride standard solution: vinyl chloride primary
for containers for aqueous solutions for intravenous infusion, solution, dimethylacetamide R (1:3 V/V).
see text 3.1.14.
Reference solutions. Place 10.0 mL of the internal standard
Plastic containers for the collection, storage, processing solution in each of six 50 mL vials. Close the vials and
and administration of blood and its components may be secure the stoppers. lnject 1 flL, 2 flL, 3 flL, 5 flL and 10 flL,
manufactured from one or more polymers, if necessary with respectively, of the vinyl chloride standard solution into five of
certain additives. the vials. The six solutions thus obtained contain, respectively,
lf al! or part of the container consists of a material described O flg, about 0.3 flg, 0.6 flg, 0,9 flg, 1.5 flg and 3 Ilg ofvinyl
in a text of the Pharmacopoeia, the quality of the material chloride. Shake, avoiding contact between the stopper and the
is controlled by the methods indicated in that text. (See liquido Place the vials in a water-bath at 60 ± 1 oC for 2 h.
3.1.1.1. Materials based on plasticised poly(vinyl chloride) for Column:
containersfor human blood and blood components).
- material: stainless steel;
In normal conditions of use the materials and containers
- size: 1 = 3 m, 0 = 3 mm;
made from such materials do not release monomers, or other
substances, in amounts likely to be harmful nor do they - stationary phase: silanised diatomaceous earth for gas
lead to any abnormal modifications of the blood or blood chromatography R impregnated with 5 per cent m/m of
components. dimethylstearamide R and 5 per cent m/m of macrogol
400 R.
Carrier gas: nitrogen for chromatography R.
01/2008:90001 Flow rate: 30 mLlmin.
corrected 7.5
Temperature:
3.1.1.1. MATERIALS BASED ON - column: 45 oC;
- injection port: 100 oC;
PLASTICISED POLY (VINYL
- detector: 150 oc.
CHLORIDE) FOR CONTAINERS Detection: flame ionisation.
FOR HUMAN BLOOD AND BLOOD Injection: 1 mL of the headspace.
COMPONENTS Limit:
DEFlNITlON - vinyl chloride: maximum 1 ppm.
Materialsbased on plasticised poly(vinyl chloride) contain Additives
not less than 55 per cent of poly(vinyl chloride) and contain A certain number of additives are added to the polymers to
various additives, in addition to the high-molecular-mass optimise their chemical, physica! and mechanical properties in
polymer obtained by polymerisation ofvinyl chloride. order to adapt them for the intended use. All these additives
Materials based on plasticised poly(vinyl chloride) for are chosen fram the following list which specifies for each
containers for human blood and blood components are product the maximum allowable content:
defined by the nature and the proportions of the substances - di(2-ethylhexyl)phthalate (pi as tic additive 01): maximum
used in their manufacture. 40 per cent;
PRODUCTION - zinc octano ate (zinc 2-ethylhexanoate) (plastic additive 02):
maximum 1 per cent;
Materials based on plasticised poly(vinyl chloride) are
produced by polymerisation methods that guarantee a residual - calcium stearate or zinc stearate: maximum 1 per cent or
vinyl chloride content of less than 1 ppm. The manufacturing 1 per cent of a mixture of the two;
process is validated to demonstrate that the product complies - N,N'-diacylethylenediamines (plastic additive 03):
with the following test. maximum 1 per cent;

General Notices (1) apply to all monographs and other texts 375
3.1.1.1. Plastidsed PVC maíerials for containers for blood EUROPEAN PHARMACOPOEIA 8.0

- one of the following epoxidised oils: maximum 10 per cent Solution S2. Place 25 g of the material to be examined in a
or 10 per cent of a mixture of the two: borosilicate-glass flask. Add 500 mL of water for injections R
- epoxidised soya oil (plastic additive 04), of which the and cover the neck of the flask with a borosilicate-glass beaker.
oxiran oxygen content is 6 per cent to 8 per cent and the Heat in an autoclave at 121 ± 2 oC for 20 mino Allow to cool
iodine value is not greater than 6; and decant the solution. Make the volume up to 500 mL.
- epoxidised linseed oil (plastic additive 05), of which the Appearance of solution S2. Solution S2 is clear (2.2.1) and
oxiran oxygen content is not greater than 10 per cent colourless (2.2.2, Method II).
and the iodine value is not greater than 7. Addity or alkalinity. To 100 mL of solution S2, add 0.15 mL
Very low amounts of antioxidants added to the vinyl chloride of BRP indicator solution R. Not more than 1.5 mL of 0.01 M
monomer may be detected in the polymer. sodium hydroxide is required to change the colour of the
No antioxidant additive may be added to the polymer. indicator to blue. To 100 mL of solution S2 add 0.2 mL of
methyl orange solution R. Not more than 1.0 mL of 0.01 M
Ultramarine blue is the only colouring material permitted to
hydrochloric acid is required to initiate the colour change of
be added.
the indicator from yellow to orange.
The supplier of the material must be able to demonstrate
that the qualitative and quantitative composition of the type Absorbance (2.2.25). Evaporate 100.0 mL of solutioll S2 to
sample is satisfactory for each production batch. dryness. Dissolve the residue in 5.0 mL of hexane R. From
250 nm to 310 nm the absorbance is 110t greater than 0.25.
CHARACTERS Redudng substances. Carry out the test within 4 h of
Colourless or pale yellow powder, beads, granules or, after preparation of solution S2. To 20.0 mL of solution S2 add 1 mL
transformation, translucent sheets of varying thickness or of dilute sulfuric acid R and 20.0 mL of 0.002 M potassium
containers, with a slight odour. On combustion it gives off permanganate. Boil under a reflux condenser for 3 min and
dense, black smoke. cool immediately. Add 1 g of potassium iodide R and titrate
immediately with 0.01 M sodium thiosulfate, using 0.25 mL of
IDENTIFICATION starch solution R as indicator. Carry out a blank titration using
{f necessary, before use, cut the samples of the material to be 20 mL of water for injections R. The difference between the
examined into pieces of maximum dimension on a side of not two titration volumes is not more than 2.0 mL.
greater than 1 cm. Primary aromatic amines: maximum 20 ppm.
To 2.0 g of the material to be examined add 200 mL of To 2.5 mL of solution Al obtained during the identification,
peroxidefree ether R and heat under a reflux condenser for
add 6 mL of water R and 4 mL of 0.1 M hydrochloric acid.
8 h. Separate the residue B and the solution A by filtration.
Shake vigorously and discard the upper layer. To the aqueous
Evaporate solution A to dryness under reduced pressure in a layer add 0.4 mL of a freshly prepared 10 giL solution of
water-bath at 30 oc. Dissolve the residue in 10 mL of toluene R sodium nitrite R. Mix and allow to stand for 1 mino Add
(solution Al). Dissolve the residue B in 60 mL of ethylene 0.8 mL of a 25 giL solution of ammonium sulfamate R,
chloride R, heating on a water-bath under a re flux condenser. allow to stand for 1 min and add 2 mL of a 5 giL solution of
Filter. Add the solution obtained dropwise and with vigorous naphthylethylenediamine dihydrochloride R. After 30 min,
shaking to 600 mL of heptane R heated almost to boiling. any colour in the solution is not more intense than that in
Separate the coagulum B1 and the organic solution by hot a standard prepared at the same time in the sal11e manner
filtration. Allow the latter to cool; separate the precipitate B2 replacing the aqueous layer with a mixture of 1 mL of a
that forms and filter through a tared sintered-glass filter (40) 0.01 giL solution of naphthylamine R in 0.1 M hydrochloric
(2.1.2). acid, 5 l11L of water R and 4 mL of 0.1 M hydrochloric acid
A. Infrared absorption spectrophotometry (2.2.24). instead of the aqueous layer.
Preparation. Dissolve the coagulum Bl in 30 mL of Plastic additives 01, 04 and 05. Thin-layer chromatography
tetrahydrofuran R and add, in small volumes with shaking, (2.2.27).
40 mL of anhydrous ethanol R. Separate the precipitate B3 Reference solutions. Prepare 0.1 mg/mL solutiol1s of
by filtration and dry in vacuo at a temperature not exceeding plastic additive 01 CRS, plastic additive 04 CRS and plastic
50 oC over diphosphorus pentoxide R. Dissolve a few additive 05 CRS, respectively, in toluene R.
milligrams of precipitate B3 in 1 mL of tetrahydrofuran R,
place a few drops of the solution obtained 011 a sodium Plate: TLC silica gel GF254 plate R.
chloride plate and evaporate to dryness in an oven at Mobile phase: toluene R.
100-105 oc.
Application: 0.5 mL of solution Al obtained during the
Comparison: poly(vinyl chloride) CRS. identification as a band 30 mm by 3 mm and 5 flL of each
B. Infrared absorption spectrophotometry (2.2.24). reference solution.
Examine the residue C obtained in the test for pi as tic Development: over a path of 15 cm.
additives 01, 04 and 05. Drying: in airo
Comparison: plastic additive 01 CRS. Detection A: examine in ultraviolet light at 254 nm.
TESTS Locate the zone corresponding to plastic additive 01
1f necessary, before use, cut the samples of the material to be (R p = about 0.4). Remove the area of silica gel corresponding
examined into pieces of maximum dimension on a side of not to this zone and shake with 40 mL of ether R for 1 mino Filter,
greater than 1 cm. rinse with two quantities, each of 10 mL of ether R, add the
rinsings to the filtrate and evaporate to dryness. The residue C
Solution SI. Place 5.0 g of the material to be examined in weighs not more than 40 mg.
a combustion flask. Add 30 mL of sulfuric acid R and heat
until a black, syrupy mass is obtained. Cool and add carefully Detection B: expose the plate to iodine vapour for 5 mino
10 mL of strong hydrogen peroxide solution R. Heat gently. Examine the chromatogram and locate the band corresponding
Allow to cool and add 1 mL of strong hydrogen peroxide to plastic additives 04 and 05 (R F = O). Remove the afea of
solution R; repeat by alternating evaporation and addition silica gel corresponding to this zone. Similarly remove a
of hydrogen peroxide solution until a colourless liquid is corresponding are a of silica gel as a blank reference. Separately
obtained. Reduce the volume to about 10 mL. Cool and dilute shake both samples for 15 l11in with 40 mL of methanol R.
to 50.0 mL with water R. Filter, rinse with 2 quantities, each of 10 mL of methanol R,

376 See ¡he information section on general monographs (caver pages)


EUROPEAN PHARMACOPOEIA 8.0 3.1.1.1. Plasticised PVC materials for coníainers for blood

add the rinsings to the filtrate and evaporate to dryness. The Test solution. Dilute solution SI 100 times with 0.1 M
difference between the masses of both residues is not more hydrochloric acid.
than 10 mg. Reference solutions. Prepare the reference solutions using
PlasHc additive 03. zinc standard solution (100 ppm Zn) R, diluting with 0.1 M
hydrochloric acid.
Wash precipitate B2 obtained during the identification and
contained in the tared sintered-glass filter (40) (2.1.2) with Source: zinc hollow-cathode lampo
anhydrous ethanol R. Dry to constant mass over diphosphorus Wavelength: 213.9 nm.
pentoxide R and weigh the filter. The residue weighs not more Atomisation device: air-acetylene flameo
than 20 mg. Verify the absence of zinc in the hydrochloric acid used.
Infrared absorption spectrophotometry (2.2.24). Heavy metals (2.4.8): maximum 50 ppm.
Preparation: the residue obtained aboye. To 10 mL of solution SI add 0.5 mL of phenolphthalein
Comparison: plastic additive 03 CRS. solution R and then strong sodium hydroxide solution R until
Barium: maximum 5 ppm. a pale pink colour is obtained. Dilute to 25 mL with water R.
12 mL of the solution complies with test A. Prepare the
Inductively coupled plasma-atomic emission spectrometry reference solution using lead standard so/ution (2 ppm Pb) R.
(2.2.57).
Water extractable substances: maximum 0.3 per cent.
Test solution. Ignite 1.0 g of the substance to be examined in a
silica crucible. Take up the residue with 10 mL of hydrochloric Evaporate 50 mL of solution S2 to dryness on a water-bath
add R and evaporate to dryness on a water-bath. Take up the and dry in an oven at 100-105 oC to constant mass. Carry
residue with 20 mL of 0.1 M hydrochloric acid. out a blank test with 50.0 mL of water for injections R. The
residue weighs not more than 7.5 mg taking into account the
Reference solution. A solution containing 0.25 ppm of barium blank test.
prepared by dilution of barium standard solution (50 ppm
Ba) R with 0.1 M hydroch/oric add. ASSAY
Wavelength: use the emission of barium at 455040 nm, the Carry out the oxygen-flask method (2.5.10) using 50.0 mg.
spectral background being taken at 455.30 nm. Absorb the combustion products in 20 mL oí 1 M sodium
Verify the absence ofbarium in the hydrochloric acid used. hydroxide. To the solution obtained add 1 mL of dibuty/
phthalate R, 2.5 mL of nitric acid R, 5 mL of ferric ammonium
Cadmium: maximum 0.6 ppm. sulfate solution R2 and 10.0 mL of 0.1 M si/ver nitrate. Titrate
Atomic absorption spectrometry (2.2.23, Method I). with 0.05 M ammonium thiocyanate until a reddish-yellow
Test so/ution. Evaporate 10 mL of solution SI to dryness. colour is obtained. Carry out a blank test.
Take up the residue using 5 mL of a 1 per cent V/V solution 1 mL of 0.1 M silver nitrate is equivalent to 6.25 mg of
of hydrochloric acid R, filter and dilute the filtrate to 10.0 mL poly(vinyl chloride).
with the same acid solution. In addition, the following tests are carried out on the sterile
Reference so/utions. Prepare the reference solutions using and empty containers.
cadmium standard so/ution (0.1 per cent Cd) R, diluting with a Solution S3. If the container to be examined contains an
1 per cent V/V solution of hydrochloric acid R. anticoagulant solution, empty the container and wash the
Source: cadmium hollow-cathode lampo inside with 250 mL of water for injections R at 20 ± 1 oC and
discard the washings before the preparation of solution S3.
Wavelength: 228.8 nm.
Introduce into the container a volume of water for injections R
Atomisation device: air-acetylene flameo corresponding to the volume of solutiol1. Close the container
Verify the absence of cadmium in the hydrochloric acid used. and heat in an autoclave so that the temperature of the liquid
is maintained at 110 oC for 30 mino After cooling, fin the
Caldum: maximum 0.07 per cent.
container with water for injections R to its nominal volume
Inductively coupled plasma-atomic emission spectrometry and homogenise.
(2.2.57).
Reference solution. Heat water for injections R in a
Test solution. Use the test solution prepared for the borosilicate-glass flask in an autoclave at 110 oC for 30 mino
determination of barium.
Reducing substances. Immediately after preparation of
Reference solution. A solution containing 50.0 ppm of calcium solution S3, transfer to a borosilicate-glass flask a volume
prepared by dilution of calcium standard solution (400 ppm corresponding to 8 per cent of the nominal volume of the
Ca) R with 0.1 M hydrochloric acid. container. At the same time, prepare a blank using an equal
Wavelength: use the emission of calcium at 315.89 nm, the volume of the freshly prepared reference solution in another
spectral background being taken at 315.60 nm. borosilicate-glass flask. To each solution add 20.0 mL of
Verify the absence of calcium in the hydrochloric acid used. 0.002 M potassium permanganate and 1 mL of dilute sulfuric
acid R. Allow to stand protected from light far 15 mino To
Tin maximum 20 ppm. each solution add 0.1 g of potassium iodide R. Allow to stand
Inductively coupled plasma-atomic emission spectrometry protected from light for 5 min and titrate immediately with
(2.2.57). 0.01 M sodium thiosulfate, using 0.25 mL of starch solution R
Test solution. Dilute solution SIlO times with water R as indicator. The difference between the two titrations is not
immediately before use. more than 2.0 mL.
Reference solution. Introduce 2 mL of tin standard solution Acidity or alkalinity. To a volume of solution S3
(5 ppm Sn) R into a 50 mL flask containing 5 mL of a 20 per corresponding to 4 per cent of the nominal capacity of the
cent V/V solution of sulfuric acid R and dilute to 50 mL with container add 0.1 mL of phenolphthalein solution R. The
water R immediately befare use. solution remains colourless. Add 004 mL of 0.01 M sodium
Wavelength: use the emission of tin at 189.99 nm, the spectral hydroxide. The solution is pink. Add 0.8 mL of 0.01 M
background being taken at 190.10 nm. hydroch/oric acid and 0.1 mL of methyl red so/ution R. The
solution is orange-red or red.
Verify the absence of tin in the sulfuric acid used.
Chlorides (2.4.4): maximum 004 ppm, determined on solution
Zinc maximum 0.2 per cent. S3. Prepare the standard using a mixture of 1.2 mL of chloride
Atomic absorption spectrometry (2.2.23, Method I). standard solution (5 ppm Cl) R and 13.8 mL of water R.

General Notices (1) app/y to all monographs and other texts 377
3.1.1.2. Plastidsed PVC materials for transfusion ofblood EUROPEAN PHARMACOPOEIA 8.0

Ammonium (2.4.1, Method A): maximum 2 ppm. 01/2008:90002


corrected 7.5
Dilute 5 mL of solution S3 to 14 mL with water R.
Water extractable substances. Evaporate 100 mL of
3.1.1.2. MATERIALS BASED ON
solution S3 to dryness on a water-bath. Dry in an oven to PLASTICISED POLY(VINYL
constant mass at 100-105 oc. Carry out a blank test using CHLORIDE) POR TUBING USED
100 mL of the reference solution. The residue from solution S3
weighs not more than 3 mg, taking into account the blank test. IN SETS POR THE TRANSFUSION OP
Absorbance (2.2.25): maximum 0.30, determil1ed between BLOOD AND BLOOD COMPONENTS
wavelel1gths of 230 nm and 250 11m 011 soIution S3; maximum DEFIN1TION
0.10, determined between wavelengths of 251 11m and 360 nm
on soIution S3. Use the reference solution as the compensation Content: minimum 55 per cent ofpoly(vinyl chloride).
liquido The plasticiser used is di(2-ethylhexyl) phthalate (plastic
additive 01).
Extractable plastic additive 01. Use as the extraction
solvent, ethanol (96 per cent) R diluted with water R to have PRODUCTION
a relative density (2.2.5) of 0.9389 to 0.9395, measured with Materials based 011 plasticised poly(vinyl chloride) are
a del1simeter. produced by polymerisation methods that guarantee a residual
vinyl chloride content of less than 1 ppm. The manufacturing
Stock solution. Dissolve 0.100 g of plastic additive 01 CRS in process is validated to demonstrate that the product complies
the extraction solvent al1d dilute to 100.0 mL with the same with the following test.
solvent. Vinyl chloride. Head-space gas chromatography (2.2.28).
Internal standard solution. Using a microsyringe, inject
Standard solutions. 1nto 5 separate 100 mL volumetric flasks, 10 flL of ether R into 20.0 mL of dimethylacetamide R,
introduce respectively 1.0 mL, 2.0 mL, 5.0 mL, 10.0 mL, and immersing the tip of the needle in the solvent. Immediately
20.0 mL of stock solution. before use, dilute the solution to 1000 times its volume with
dimethylacetamide R.
Measure the absorbances (2.2.25) of the standard solutions Test solution. Place 1.000 g of the material to be examined in a
at the absorption maximum at 272 nm, USi11g the extraction 50 mL vial and add 10.0 mL of the internal standard solution.
solvent as compensation liquid and plot a curve of absorbance Close the vial and secure the stopper. Shake, avoiding contact
against the COl1centration of plastic additive Ol. between the stopper and the liquido Place the vial in a
water-bath at 60 ± 1 oC for 2 h.
Extraetion procedure. Using the donor tubing and the needle Vinyl chloride primary solution. Prepare in a fume cupboard.
or adapter, fill the empty container with a volume equal Place 50.0 mL of dimethylacetamide R in a 50 mL vial, stopper
to half the nominal volume with the extraction solvent, the vial, secure the stopper and weigh to the nearest 0.1 mg. .
previously heated to 37 oC in a well-stoppered flask. Expel Fill a 50 mL polyethylene or polypropylene syringe with
the air completely from the container and seal the donor gaseous vinyl chloride R, allow the gas to remain in contact
tubing. Immerse the filled container in a horizontal position with the syringe for about 3 min, empty the syringe and fill
in a water-bath maintained at 37 ± 1 oC for 60 ± 1 min again with 50 mL of gaseous vinyl chloride R. Fit a hypodermic
without shakil1g. Remove the container from the water-bath, needle to the syringe and reduce the volume of gas in the
invert it gentIy 10 times and transfer the contents to a syringe from 50 mL to 25 mL. Inject the remaining 25 mL of
glass flask. Immediately measure the absorbance at the vinyl chloride slowly into the vial shaking gently and avoiding
absorption maximum at 272 11m, using the extraction solvent contact between the liquid and the needle. Weigh the vial
as compensation liquido again; the increase in mass is about 60 mg (1 flL of the solution
thus obtained contains about 1.2 flg ofvinyl chloride). Allow
Determine the concentration of plastic additive 01 in to stand for 2 h. Keep the primary solution in a refrigerator.
milligrams per 100 mL of extract from the calibration curve. Vinyl chloride standard solution: vinyl chloride primary
The concel1tratiol1 does 110t exceed: solution, dimethylacetamide R (1:3 V/V).
Reference solutions. Place 10.0 mL of the internal standard
- 10 mg per 100 mL for containers of nominal volume greater solution in each of six 50 mL vials. Close the vials and
than 300 mL but not greater than 500 mL; secure the stoppers. Inject 1 flL, 2 flL, 3 flL, 5 flL and 10 flL,
respectively, of the vinyl chloride standard solution into 5 of
- 13 mg per 100 mL for containers of nominal volume greater the vials. The 6 solutions thus obtained contain respectively,
than 150 mL but not greater than 300 mL; O flg, about 0.3 flg, 0.6 flg, 0.9 flg, 1.5 flg and 3 flg of vinyl
chloride. Shake, avoiding contact between the stopper and the
- 14 mg per 100 mL for containers of nominal volume up liquido Place the vials in a water-bath at 60 ± 1 oC for 2 h.
to 150 mL. Column:
- material: stainless steel;
Where containers contain an anticoagulant solution, this - size: 1 = 3 m, 0 = 3 mm;
solution complies with the monograph on Anticoagulant and
preservative solutions for human blood (0209) and the following - stationary phase: silanised diatomaceous earth for gas
additional test. chromatography R impregnated with 5 per cent m/m of
dimethylstearamide R and 5 per cent m/m of macrogol
Absorbance (2.2.25): maximum 0.5, by measuring at the 400 R.
absorption maximum at 280 nm. Carrier gas: nitrogen for chromatography R.
Flow rate: 30 mL/min.
Measure the absorbance of the anticoagulant solution from
Temperature:
the container between 250 nm and 350 nm, using as the
compensation liquid an anticoagulant solution of the same - column: 45 oC;
composition that has not been in contact with a plastic - injeetion port: 100 oC;
material. - detector: 150 oc.

378 See the information section on general monographs (cover pages)


EUROPEAN PHARMACOPOEIA 8.0 3.1.1.2. Plastidsed PVC materials for transfl1sion ofblood

Detection: flame ionisation. Limit: locate the zone corresponding to plastic additive 01.
Injection: 1 mL of the head space. Remove the are a of silica gel corresponding to this zone and
Limit: shake with 40 mL of ether R. Filter without 105s and evaporate
to dryness. The residue weighs not more than 40 mg.
- vinyl chloride: maximum 1 ppm.
The supplier of the material must be able to demonstrate Barium: maximum 5 ppm.
that the qualitative and quantitative composition of the type Inductively coupled plasma-atomic emission spectrometry
sample is satisfactory for each production batch. (2.2.57).
Test solution. Ignite 1.0 g of the substance to be examined in a
CHARACTERS silica crucible. Take up the residue with 10 mL of hydroch/oric
Almost colourless or pale-yellow material in the form of acid R and evaporate to dryness on a water-bath. Take up the
powder, beads, granules or, after transformation, tubes with a residue with 20 mL of 0.1 M hydrochloric acid.
slight odour. Reference so/ution. A solution containing 0.25 ppm ofbarium
On combustion it gives off dense, black smoke. prepared by dilution of barium standard solution (50 ppm
Ba) R with 0.1 M hydroch/oric acid.
IDENTIFICATION
If necessary, cut the samples of the material to be examined Wavelength: use the emission ofbarium at 455.40 nm, the
into pieces with a maximum dimension on a síde of not greater spectral background being taken at 455.30 nm.
than 1 cm. Verify the absence ofbarium in the hydrochloric acid used.
A. To 0.5 g add 30 mL of tetrahydrofuran R. Heat with stirring Cadmium: maximum 0.6 ppm.
on a water-bath in a fume cupboard for 10 mino The Atomic absorption spectrometry (2.2.23, Method 1).
material dissolves completely. Add methanol R dropwise Test solution. Evaporate 10.0 mL of solution SI to dryness.
with stirring. A granular precipitate is formed. Filter the Take up the residue using 5 mL of a 1 per cent V/V solution
precipitate and dry at 60 oc. Examine the precipitate by of hydroch/oric acid R, filter and dilute the filtrate to 10.0 mL
infrared absorption spectrophotometry (2.2.24). Dissolve with the same acid.
50 mg in 2 mL of tetrahydrofuran R and pour on a glass
slide. Dry in an oven at 80 oC, remove the film and fix Reference solutions. Prepare the reference solutions using
on a suitable mount. Examine by infrared absorption cadmium standard so/ution (0.1 per cent Cd) R, diluting with a
spectrophotometry (2.2.24), comparing with the spectrum 1 per cent V/V solution of hydrochloric acid R.
obtained with poly(vinyl chloride) CRS. Source: cadmium hollow-cathode lampo
B. Infrared absorption spectrophotometry (2.2.24). Examine Wave/ength: 228.8 nm.
the residue obtained in the test plastic additive Ol. Atomisation device: air-acetylene flameo
Comparison: p/astic additive al CRS. Verify the absence of cadmium in the hydrochloric acid used.
TESTS Tin: maximum 20 ppm.
If necessary, cut the samples of the material to be examined Inductively coupled plasma-atomic emission spectrometry
ínto pieces wíth a maximum dimension on a side of not greater (2.2.57).
than 1 cm. Test solution. Dilute solution SI 10 times with water R
Solution SI. Place 5.0 g of the material to be examined in immediately befo re use.
a combustion flask. Add 30 mL of sulfuric acid R and heat Reference solution. Introduce 2 mL of tin standard so/ution
until a black, syrupy mas s is obtained. Cool and add carefully (5 ppm Sn) R into a 50 mL flask containing 5 mL of a 20 per
10 mL of strong hydrogen peroxide solution R. Heat gently. cent V/V solution of sulfuric acid R and dilute to 50 mL with
Allow to cool and add 1 mL of strong hydrogen peroxide water R immediately before use.
solution R; repeat by alternating evaporation and addition Wavelength: use the emission oftin at 189.99 nm, the spectral
of hydrogen peroxide solution until a colourless liquid is background being taken at 190.10 nm.
obtained. Reduce the volume to about 10 mL. Cool and dilute
Verify the absence of tin in the sulfuric acid used.
to 50.0 mL with water R.
Heavy metals (2.4.8): maximum 50 ppm.
8olution 82. Place 25 g of the material to be examined in a
borosilicate-glass flask. Add 500 mL of water R and cover the To 10 mL of solution SI add 0.5 mL of pheno/phthalein
neck of the flask with a borosilicate-glass beaker. Heat in an so/ution R and then strong sodium hydroxide so/ution R until
autoclave at 121 ± 2 oC for 20 min. Allow to co01 then decant a pale pink colour is obtained. Dilute to 25 mL with water R.
the solution and make up to a volume of 500 mL. 12 mL of the solution complíes with test A. Prepare the
reference solution using lead standard solution (2 ppm Pb) R.
Appearance of solution 82. Solution S2 is clear (2.2.1) and
colourless (2.2.2, Method II). ASSAY
Plastic additive 01. Thin-layer chromatography (2.2.27). To 0.500 g add 30 mL of tetrahydrofuran R and heat with
Test solution. To 2.0 g of the material to be examined add stirring on a water-bath in a fume cupboard for 10 min. The
200 mL of peroxide-free ether R and heat under a reflux material dissolves completely. Add 60 mL of methano/ R
condenser for 8 h. Separate the residue and the solution by dropwise with stirring. A granular precipitate of poly(vinyl
filtration and evaporate the solution to dryness under reduced chloride) is formed. Allow to stand for a few minutes.
pressure in a water-bath at 30 oc. Dissolve the residue in Continue addition of methanol R until no further precipitation
10 mL of to/uene R. is observed. Transfer to a sintered-glass filter (40) (2.1.2),
Reference so/ution. Disso1ve 0.8 g of plastic additive 01 CRS in using three small quantities of methanol R to aid transfer and
to/uene R and dilute to 10 mL with the same solvento to wash the precipitate. Dry the filter and the precipitate to
constant mass at 60 oC and weigh.
Plate: TLC silica gel G plate R.
In addition, carry out the following tests on sterilised sets.
Mobile phase: toluene R.
Application: 0.5 mL of the test solution and 5 flL of the 8olution S3. Make a closed circulation system from 3 sets
reference solution, as a band 30 mm by 3 mm. and a 300 mL borosilicate-glass vessel. Fit to the vessel a
suitable thermostat device that maintains the temperature
Development: over a path of 15 cm. of the liquid in the vessel at 37 ± 1 oc. Circulate 250 mL of
Drying: in airo water for injections R through the system in the direction
Detection: in ultraviolet light at 254 nm. used for transfusion for 2 h at arate of 1 L/h (for example

General Notices (1) apply to all monographs and other texts 379
3.1.3. Polyolefins EUROPEAN PHARMACOPOEIA 8.0

using a peristaltic pump applied to as short a piece of suitable - 4,4',4"- (2,4,6-trimethylbenzene-l ,3,5-triyltrismethy-
silicone elastomer tubing as possible). Collect the whole of lene )tris [2,6-bis( 1, l-dimethylethyl)phenol] (plastic
the solution and allow to cool. additive 10): maximum 0.3 per cent;
Appearance of solution. Solution S3 is clear (2.2.1) and - 2,2'-bis( octadecyloxy)-5,5'-spirobi[1,3,2-dioxa-
colourless (2.2.2, Method JI). phosphinane] (plastic additive 14): l11aximum 0.3 per cent;
Addity or alkalinity. To 25 mL of solution S3 add 0.15 mL - didodecyl 3,3'-thiodipropionate (plastic additive 16):
of BRP indicator solution R. Not more than 0.5 mL of 0.01 M maximum 0.3 per cent;
sodium hydroxide is required to change the colour of the - dioctadecyl 3,3'-thiodipropionate (plastic additive 17):
indicator to blue. To 25 mL of solution S3 add 0.2 mL of maximum 0.3 per cent;
methyl orange solution R. Not more than 0.5 mL of 0.01 M - tris[2,4-bis(1,l-dimethylethyl)phenyl] phosphite (plastic
hydrochloric acid is required to initiate the colour change of additive 12): maximum 0.3 per cent;
the indicator from yellow to orange. - pi as tic additive 18: maxil11um 0.1 per cent;
Ahsorbance (2.2.25): maximum 0.30, determined between - copolymer of dimethyl succinate and (4-hydroxy-2,2,6,6-
wavelengths of 230 nm and 250 nm on solution S3; maximum tetramethylpiperidin-1-yl)ethanol (plastic additive 22):
0.15, determined between wavelengths of 251 nm and 360 nm maximum 0.3 per cent.
on solution S3. The total of antioxidant additives listed aboye do es not exceed
Redudng substances. Carry out the test within 4 h of 0.3 per cent.
preparation of solution S3. To 20.0 mL of solution S3 add 1 mL - hydrotaleite: maximum 0.5 per cent;
of diluíe sulfuric acid R and 20.0 mL of 0.002 M potassium - alkanamides: maximul11 0.5 per cent;
permanganate. Boil for 3 min and cool immediately. Add
1 g of potassium iodide R and titrate with 0.01 M sodium - alkenamides: maximum 0.5 per cent;
thiosulfate using 0.25 mL of starch solution R as indicator. - sodium silico-aluminate: maximum 0.5 per cent;
Carry out a blank test using 20 mL of water for injections R. - silica: maximum 0.5 per cent;
The difference between the titration volumes is not greater - sodium benzoate: maximum 0.5 per cent;
than 2.0 mL. - fatty acid esters or salts: maximum 0.5 per cent;
Water extractable substances. Evaporate 50.0 mL of - trisodium phosphate: maximum 0.5 per cent;
solution S3 to dryness on a water-bath and dry to constant
- liquid paraffin: maximum 0.5 per cent;
mass in an oven at 100-105 0e. Carry out a blank test using
50.0 mL of water for injections R. The residue obtained with - zinc oxide: maximum 0.5 per cent;
solution S3 is not greater than 1.5 mg, taking account of the - tale: maximum 0.5 per cent;
blank test. - magnesium oxide: maximum 0.2 per cent;
- caleium stearate or zinc stearate or a mixture of both:
Ol/2008:30103 maximum 0.5 per cent;
corrected 7.5 - titanium dioxide: maximum 4 per cent.
The supplier of the material must be able to demonstrate
3.1.3. POLYOLEFINS that the qualitative and quantitative composition of the type
sample is satisfactory for each production batch.
DEFINITION
Polyolefins are obtained by polymerisation of ethylene or CHARACTERS
propylene or by copolymerisation of these substances with Appearance: powder, beads, granules or, after transformation,
not more than 25 per cent of higher homologues (C 4 to C 10 ) sheets of varying thickness or containers.
or of carboxylic acids or of esters. Certain materials may be Solubility: practically insoluble in water, soluble in hot
mixtures of polyolefins. aromatic hydrocarbons, practically insoluble in anhydrous
ethanol, in hexane and in methanol.
PRODUCTION
They soften at temperatures between 65 oC and 165 0e. They
A certain number of additives are added to the polymer in bum with a blue flameo
order to optimise their chemical, physical and mechanical
properties in order to adapt them for the intended use. Al! IDENTIFICATION
of these additives are chosen from the appended list which 1f necessary, cut the samples of the material to be examined
specifies for each product the maximum allowable content. into pieces of maximum dimension on a side of not greater
They may contain at most 3 antioxidants, 1 or several than 1 cm.
lubricants or antiblocking agents as well as titanium dioxide A. Infrared absorption spectrophotometry (2.2.24).
as an opacifying agent when the material must provide
Preparatian: to 0.25 g add 10 mL of toIuene R and boíl
protection from light.
under a re flux condenser for about 15 min; place a few
- butylhydroxytoluene (plastic additive 07): drops of the solution obtained on a sodium chloride slide
maximum 0.125 per cent; and evaporate the solvent in an oven at 80 0e.
- pentaerythrityl tetrakis[3-(3,5-di-tert-butyl-4- Absarption maxima: at 2920 cm- I , 2850 cm- 1, 1475 cm- J,
hydroxyphenyl)propionate] (plastic additive 09): maximum 1465 cm- 1, 1380 cm' 1, 1170 cm- I , 735 cm'- 1 and 720 cm- l.
0.3 per cent;
The spectrum obtained is identical to the spectrum
- 1,3,5-tris( 3,5-di - tert- butyl-4-hydroxybenzyl)-S- obtained with the material selected for the type sample. If
triazine-2,4,6(lH,3H,5H)-trione, (plastic additive 13): the material to be examined is in the form of sheets, the
maximum 0.3 per cent; identification may be determined directly on a cut piece
- octadecyl 3-( 3,5-di - tert- butyl-4-hydroxyphenyl)propionate of suitable size.
(plastic additive 11): maximum 0.3 per cent; B. It complies with the supplementary tests corresponding to
- ethylene bis[3,3-bis[3-(1,1-dimethylethyl)-4- the additives presento
hydroxyphenyl]butanoate] (plastic additive 08): maximum e. In a platinum crucible, mix about 20 mg with 1 g of
0.3 per cent; potassium hydrogen sulfate R and heat until completely
- dioctadecyl disulfide (plastic additive 15): maximum 0.3 per melted. Allow to cool and add 20 mL of dilute sulfuric
cent; acid R. Heat gently. Filter the resulting solution. To the

380 See the information section on general monographs (ca ver pages)
EUROPEAN PHARMACOPOEIA 8.0 3.1.3. Polyolefins

filtrate add 1 mL of phosphoric acid R and 1 mL of strong Extractable titanium: maximum 1 ppm.
hydrogen peroxide solution R. If the substance is opacified Inductively coupled plasma-atomic emissio11 spectrometry
with titanium dioxide, an orange-yellow colour develops. (2.2.57).
TESTS Test solution. Use solution S3.
Jf necessary, cut the samples of the material to be examined Reference solutions. Prepare the reference solutions using
into pieces of maximum dimension on a side of not greater titanium standard solutian (100 ppm Ti) R, diluting with 0.1 M
than 1 cm. hydrochloric acid.
Wavelength: use the emission oftitanium at 336.12 11m, the
Solution SI. Use solution S1 within 4 h of preparation. Place
spectral background being taken as 336.16 nm.
25 g in a borosilicate-glass flask with a ground-glass neck.
Add 500 mL of water for injections R and boil under a reflux Verify the absence of titanium in the hydrochloric acid used.
condenser for 5 h. Allow to cool and decant. Reserve a portion Extractable zinc: maximum 1 ppm.
of the solution for the test for appearance of solution SI and Atomic absorption spectrometry (2.2.23, Method J).
filter the rest through a sintered-glass filter (16) (2.1.2).
Test solution. Use solution S3.
Solution 52. Place 2.0 g in a conical borosilicate-glass flask
Reference solutions. Prepare the reference solutions using
with a ground-glass neck. Add 80 mL of toluene R and boíl
zinc standard solution (lO ppm Zn) R, diluting with 0.1 M
under a reflux condenser with constant stirring for 90 mino
hydrochloric acid.
Allow to cool to 60 oC and add with continued stirring 120 mL
of methanol R. Filter the solution through a sintered-glass Source: zinc hollow-cathode lampo
filter (16) (2.1.2). Rinse the flask and the filter with 25 mL of a Wavelength: 213.9 nm.
mixture of 40 mL of toluene R and 60 mL of methanol R, add Atomisation device: air-acetylene flameo
the rinsings to the filtrate and dilute to 250 mL with the same
Verify the absence of zinc in the hydrochloric acid used.
mixture of solvents. Prepare a blank solution.
Extractable heavy metals (2.4.8): maximum 2.5 ppm.
Solution S3. Place 100 g in a conical borosilicate-glass flask
with a ground-glass neck. Add 250 mL of 0.1 M hydrochloric Evaporate 50 mL of solution S3 to about 5 mL on a water-bath
acid and boil under a reflux condenser with constant stirring and dilute to 20.0 mL with water R. 12 mL of the solution
for 1 h. Allow to cool and decant the solution. complies with test A. Prepare the reference solution using
2.5 mL of lead standard solution (10 ppm Pb) R.
Appearance of soludon SI. Solution SI is clear (2.2.1) and
colourless (2.2.2, Method JI). Sulfated ash (2.4.14): maximum 1.0 per cent, determined
on 5.0 g. This limit does not apply to material that has been
Addity or alkalinity. To 100 mL of solution SI, add 0.15 mL opacified with titanium dioxide.
of BRP indicator solution R. Not more than 1.5 mL of 0.01 M
sodium hydroxide is required to change the coiour of the SUPPLEMENTARY TESTS
indicator to blue. To 100 mL of solution SI add 0.2 mL of These tests are to be carried out, in whole or in part, only if
methyl orange solution R. Not more than 1 mL of 0.01 M required by the stated eompasition or the use of the material.
hydrochloric acid is required to initiate the colour change of
the indicator from yellow to orange. PhenoHc antioxidants. Liquid chromatography (2.2.29).
Absorbance (2.2.25): maximum 0.2, determined between 50lvent mixture: acetonitrile R, tetrahydrofuran R (50:50 V/V).
wavelengths of 220 nm and 340 nm 011 solution Sl. Test solution 521. Evaporate 50 mL of solution S2 to dryness
in vacuo at 45 oc. Dissolve the residue in 5.0 mL of the solvent
Reducing substances. To 20 mL of solution SI add 1 mL
mixture. Prepare a blank solution from the blank solution
of dilute sulfuric acid R and 20 mL of 0.002 M potassium
corresponding to solution S2.
permanganate. Boíl under a reflux condenser for 3 min and
cool immediately. Add 1 g of potassium iodide R and titrate Test solution 522. Evaporate 50 mL of solution S2 to dryness in
immediately with 0.01 M sodium thiosulfate, using 0.25 mL vacuo at 45 oc. Dissolve the residue with 5.0 mL of methylene
of starch solution R as indicator. Carry out a blank titration. chloride R. Prepare a blank solution from the blank solution
The difference between the titration volumes is not more than corresponding to solution S2.
3.0 mL. Test solution 523. Evaporate 50 mL of solution S2 to dryness
Substances soluble in hexane. Place 10 g in a 250 mL conical in vacuo at 45 oc. Dissolve the residue in 5.0 mL of a mixture
borosilicate-glass flask with a ground-glass neck. Add 100 mL of equal volumes of acetonitrile R and a 10 giL solution of
of hexane R and boil under a reflux condenser for 4 h, stirring tert-butylhydroperoxide R in tetrahydrofuran R. Close the flask
constantly. Cool in iced water and filter rapidly (the filtration and allow to stand for 1 h. Prepare a blank solution using the
time must be less than 5 min; if necessary the filtration may blank of solution S2.
be accelerated by applying pressure to the solution) through Of the following referenee solutions, prepare only those that are
a sintered-glass filter (16) (2.1.2) maintaining the solution necessary for the analysis of the phenolic antioxidants stated in
at about O oc. Evaporate 20 mL of the filtrate in a tared the composition of the substance to be examined.
borosilicate-glass dish on a water-bath. Dry the residue in an Reference solution (a). Dissolve 25.0 mg of butylhydraxy-
oven at 100-105 oC for 1 h. The mass ofthe residue obtained toluene CRS (plastic additive 07) and 60.0 mg of plastic
must be within 10 per cent of that of the residue obtained with additive 08 CR5 in 10.0 mL of the solvent mixture. Dilute
the type sample and do es not exceed 5 per cent. 2.0 mL of this solution to 50.0 mL with the solvent mixture.
Extradable aluminium: maximum 1 ppm. Reference solution (b). Dissolve 60.0 mg of plastic
Inductively coupled plasma-atomic emission spectrometry additive 09 CRS and 60.0 mg of plastic additive 10 CR5 in
(2.2.57). 10.0 mL of the solvent mixture. Dilute 2.0 mL of this solution
to 50.0 mL with the solvent mixture.
Test solution. Use solution S3.
Reference solution (e). Dissolve 60.0 mg of plastic
Reference solutions. Prepare the reference solutions using additive 11 CR5 and 60.0 mg of plastic additive 12 CRS in
aluminium standard solution (200 ppm Al) R, diluting with 10.0 mL of methylene chloride R. Dilute 2.0 mL of this solution
0.1 M hydrochloric acid. to 50.0 mL with methylene chloride R.
Wavelength: use the emission of aluminium at 396.15 nm, the Reference solution (d). Dissolve 25.0 mg of plastic
spectral background being taken as 396.25 11m. additive 07 CRS in 10.0 mL of the solvent mixture. Dilute
Verify the absence of aluminium in the hydrochloric acid used. 2.0 mL of this solution to 50.0 mL with the solvent mixture.

General Notices (1) apply to all monographs and other texts 381
3.1.3. Polyoleflns EUROPEAN PHARMACOPOElA 8.0

Reference so/utíon (e). Dissolve 60.0 mg of plastic System suitability:


additive 08 CRS in 10,0 mL of the solvent mixture, Dilute - resolutian: minimum 2,0 between the peaks due
2,0 mL of this solution to 50,0 mL with the solvent mixture, to pI as tic additive 09 and plastic additive 10 in the
Reference so/ution (jJ Dissolve 60,0 mg of plastic chromatogram obtained with reference solution (b);
additive 13 CRS in 10,0 mL of the solvent mixture, Dilute - the chromatogram obtained with test solution S21
2,0 mL of this solution to 50,0 mL with the solvent mixture, only show peaks due to antioxidants stated in the
Reference so/ution (g), Dissolve 60,0 mg of plastic composition and minor peaks that also appear in the
additive 09 CRS in 10,0 mL of the solvent mixture, Dilute chromatogram corresponding to the blank solution,
2,0 mL of this solution to 50,0 mL with the solvent mixture, Limit: the are as of the peaks in the chromatogram obtained
Reference solution (h), Dissolve 60,0 mg of plastic with test solution 521 are less than the corresponding areas
additive 10 CRS in 10,0 mL ofthe solvent mixture, Dilute of the peaks in the chromatograms obtained with reference
2,0 mL of this solution to 50,0 mL with the solvent mixture, solutions of the antioxidants on the list aboye that are
Reference so/ution (i), Dissolve 60,0 mg of plastic stated in the composition,
additive 11 CRS in 10,0 mL of methy/ene chloride R, Dilute C, lf the substance to be examined contains plastic additive 11
2,0 mL of this solution to 50,0 mL with methylene chloride R, and/or plastic additive 12, carry out the test as described
Reference solution (j), Dissolve 60,0 mg of plastic for plastic additive 07 and/or plastic additive 08 with the
additive 12 CRS in 10,0 mL of methylene chloride R, Dilute following modifications,
2,0 mL of this solution to 50,0 mL with methylene chloride R, Mobile phase: water R, 2-propanol R, methanol R
Reference solution (k), Dissolve 20.0 mg of plastie (5:45:50 V/V/V),
additive 18 CRS in 10,0 mL of a mixture of equal volumes of Flow rate: 15 mL/min,
acetonitrile R and a 10 giL solution of tert-butylhydroperoxide R Injection: 20 flL of the test solution 522, the corresponding
in tetrahydrofuran R, Allow to stand in a closed container for blank solution, the reference solution (c), and either the
1 h, Dilute 2,0 mL of this solution to 50,0 mL with the solvent reference solution (i) or (j) or the reference solutions (i)
mixture, and (j),
A lf the substance to be examined contains pI as tic additive 07 System suitability:
and/or plastic additive 08, carry out the test as follows.
- resolution: minimum 2,0 between the peaks due
Column: to plastic additive 11 and plastic additive 12 in the
- size: l = 0,25 m, 0 = 4,6 mm; chromatogram obtained with reference solution (c);
- stationary phase: octadecylsilyl silica gel for - the chromatogram obtained with test solution 522
chromatography R (5 flm), only show peaks due to antioxidants stated in the
Mobile phase: water R, acetonitrile R (30:70 V/V), composition and minor peaks that also appear in the
Flow rate: 2 mL/min, chromatogram corresponding to the blank solution,
Deteetian: spectrophotometer at 280 nm, Limit: the areas of the peaks in the chromatogram obtained
with test solution 522 are less than the corresponding are as
Injection: 20 flL of the test solution 521, the corresponding of the peaks in the chromatograms obtained with reference
blank solution, the reference solution (a), and either the solutions (i) and/or (j),
reference solutions (d) or (e) or the reference solutions (d)
and (e), D, lf the substance to be examined contains plastic additive 18,
carry out the test as described for pIastic additive 07 and/or
Run time: 30 min, plastic additive 08 with the following modifications,
System suitability: Mobile phase: tetrahydrofuran R, acetonitrile R (20:80 V/V).
- resolution: minimum 8,0 between the peales due Flow rate: 1,5 mL/min,
to pIastic additive 07 and plastic additive 08 in the
chromatogram obtained with reference solution (a); Deteetion: spectrophotometer at 270 nm,
- the chromatogram obtained with test solution 521 Injection: 20 flL of the test solution S23, the corresponding
only show peaks due to antioxidants stated in the bIank solution and the reference solution (k),
composition and minor peaks that also appear in the System suitability:
chromatogram corresponding to the bIank solution, - resolution: minimum 6,0 between the 2 principal peales
Limit: the areas of the peaks in the chromatogram obtained (approximate retention times of 35 and 5,8) in the
with test solution S21 are less than the corresponding are as chromatogram obtained with reference solution (le);
of the peaks in the chromatograms obtained with reference - the chromatogram obtained with test solution S23
solutions (d) and/or (e), only show peaks due to antioxidants stated in the
B, lf the substance to be examined contains one or more of composition and minor peaks that also appear in the
the following antioxidants: chromatogram corresponding to the blank solution,
- plastic additive 09; Limit: the are as of the peaks in the chromatogram obtained
- pI as tic additive 10; with test solution 523 are Iess than the corresponding areas
of the peaks in the chromatograms obtained with reference
- plastic additive 11; solution (k),
- plastic additive 12;
Non-phenolic antioxidants, Thin-Iayer chromatography
- plastic additive 13; (22,27),
carry out the test as described aboye with the following Test solution S24, Evaporate 100 mL of solutiol1 S2 to dryness
modifications, in vacua at 45 oC, Dissolve the residue in 2 mL of acidified
Mobile phase: water R, tetrahydrofuran R, acetonitrile R methylene chloride R,
(10:30:60 V/V/V), Reference solution (1), Dissolve 60 mg of plastie additive 14 CRS
Flow rate: 15 mL/min, in 10 mL of methylene chloride R, mlute 2 mL of this solution
Injeetion: 20 ~lL of the test solution 521, the corresponding to 10 mL with acidified methylene chloride K
blank solution, the reference solution (b) and the reference Reference solution (m), Dissolve 60 mg of plastic
solutions of the antioxidants on the list aboye that are additive 15 CRS in 10 mL of methylene chloride R, DiIute 2 mL
stated in the composition, of this solution to 10 mL with acidified methylene chlaride R,

382 See the infarmatial1 sectian on general manographs (caver pages)


EUROPEAN PHARMACOPOEIA 8.0 3.1.40 Polyethylene without additives for coníainers

Reference solution (n). Dissolve 60 mg of plastic additive 16 CRS Reference solution (r). Dissolve 40 mg of oleamide (plastie
in 10 mL of methylene chloride R. Dilute 2 mL of this solution additive 20 CRS) in 20 111L of methylene chloride R.
to 10 mL with acidified methylene chloride R. Referenee solution (s). Dissolve 40 mg of erucamide (plastic
Reference solution (o). Dissolve 60 mg of plastic additive 17 CRS additive 21 CRS) in 20 mL of methylene chloride R.
in 10 mL of methylene chloride R. Dilute 2 mL of this solution Plate: TLC silica gel GFCS4 plate R (2 plates).
to 10 mL with acidified methylene chloride R.
A. Mobile phase: anhydrous ethanol R, trimethylpentane R
Referenee solution (p). Dissolve 60 mg of plastie additive 16 CRS (25:75 V/V).
and 60 mg ofplastie additive 17 CRS in 10 mL of methylene
Application: 10 flL of the test solution S24 and reference
chloride R. Dilute 2 mL of this solution to 10 mL with acidified
solution (q).
methylene chloride R.
Development: over a path of 10 cm.
Plate: TLC silica gel GF254 plate R.
Drying: in air.
Mobi/e phase A: hexane R.
Detection: spray with a 2 giL solution of
Mobi/e phase B: methylene chloride R.
diehlorophenolindophenol, sodium salt R in anhydrous
Application: 20 flL of the test solution S24, the reference ethanol R and heat in an oven at 120 oC for a few minutes
solution (p) and the reference solutions corresponding to all to intensify the spots.
the phenolic and non-phenolic antioxidants mentioned in the
Limit: any spot corresponding to plastic additive 19 in the
type composition of the material to be examined.
chromatogram obtained with test solution S24 is identical
Development A: over a path of 18 cm with mobile phase A. in position to (R F = about 0.5) but not more intense than
Drying A: in airo the spot in the chromatogram obtained with reference
Development B: over a path of 17 cm with mobile phase B. solution (q).
Drying B: in airo B. Mobile phase A: hexane R.
Deteetion: examine in ultraviolet Iight at 254 nm; spray with Mobile phase B: methanal R, methylene chloride R
aleoholic iodine solution R and examine in ultraviolet light at (5:95 V/V).
254 nm after 10-15 mino Application: 10 flL of the test solution S24 and the reference
System suitability: reference solution (p) : solutions (r) and (5).
- the chromatogram shows 2 c1early separated spots. Development A: over a path of 13 cm with mobile phase A.
Limit: any spots in the chromatogram obtained with test Drying A: in airo
solution S24 are not more intense than the spots in the Development B: over a path of 10 cm with mobile phase B.
corresponding positions in the chromatograms obtained with Drying B: in airo
the reference solutions.
Deteetian: spray with a 40 giL solution of phosphomolybdic
Plastic additive 22. Liquid chromatography (2.2.29). acid R in anhydrous ethanol R. Heat in an oven at 120 oC
Test solution. Evaporate 25 mL of solution S2 to dryness in until spots appear.
vacuo at 45 oc. Dissolve the residue in 10 mL of toluene R and Limit: any spots corresponding to plastic additive 20 or
10 mL of a 10 giL solution of tetrabutylammonium hydroxide R plastic additive 21 in the chromatogram obtained with test
in a mixture of 35 volumes of toluene R and 65 volumes of solution S24 are identical in position to (R F = about 0.2)
anhydrous ethanol R. Boil under a reflux condenser for 3 h. but not more intense than the corresponding spots in
Allow to cool and filter if necessary. the chromatograms obtained with reference solutions (r)
Reference solution. Dissolve 30 mg of plastic additive 22 CRS and (s).
in 50 mL of toluene R. Add 1 mL of this solution to 25 mL
of blank solution S2 and evaporate to dryness in vacuo at
45 oc. Dissolve the residue in 10 mL of toluene R and 10 mL 01/2008:30104
of a 10 giL solution of tetrabutylammonium hydroxide R corrected 6.0
in a mixture of 35 volumes of toluene R and 65 volumes of
anhydrous ethanol R. Boil under a reflux condenser for 3 h. 3.1.4. POLYETHYLENE WITHOUT
Allow to cool and filter if necessary.
ADDITIVES FOR CONTAINERS FOR
Column:
- size: 1 = 0.25 m, 0 = 4.6 mm;
PARENTERAL PREPARATIONS AND
- stationary phase: aminopropylsilyl si/ica gel for FOR OPHTHALMIC PREPARATIONS
chromatography R (5 flm). DEFINITION
Mabile phase: anhydrous ethanol R, hexane R 01:89 V/V). Polyethylene without additives is obtained by the
Flow rate: 2 mLlmin. polymerisation of ethylene under high pressure in the presence
Detection: spectrophotometer at 227 nm. of oxygen or free-radical-forming initiators as catalyst.
Injection: 20 flL. CHARACTER5
Run time: 10 mino
Appearance: beads, granules, powder or, after transformation,
System suitability: translucent sheets of varying thickness or containers.
- resolution: minimum 7 between the peaks due to the "diol" Solubility: practically insoluble in water, soluble in hot
component and to the diluent of the reference solution. aromatic hydrocarbons, practically insoluble in anhydrous
Limit: the area of the peak due to the "diol" component ethanol, in hexane and in methanol.
from plastic additive 22 in the chromatogram obtained with It softens at temperatures beginning at 65 oc.
the test solution is less than the corresponding peak in the
chromatogram obtained with the reference solution. Relative density: 0.910 to 0.937.
Amides and stearates. Thin-layer chromatography (2.2.27). IDENTIFICATION
Test solution. Use test solution 524 described in the test for If necessary, cut the samples of the material to be examined
non -phenolic antioxidants. into pieces af maximum dimension on a side of not greater
Reference solution (q). Dissolve 20 mg of stearic acid (plastic than 1 cm.
additive 19 CRS) in 10 mL of methylene chloride R. A. Infrared absorption spectrophotometry (2.2.24).

General Notices (1) apply to all manographs and other texts 383
3.1.5. Polyethylene with additives fOl" containers EUROPEAN PHARMACOPOEIA 8.0

Preparation: to 0.25 g add 10 mL of toluene R and boil methylene chloride R. Prepare a blank solution from the blank
under a reflux condenser for about 15 mino Place a few solution corresponding to solution S2.
drops of the solution on a sodium chloride disc and Reference soluíion. Dissolve 20 mg of plastic addiíive 15 CRS
evaporate the solvent in an oven at 80 oc. and 20 mg of plastic additive 08 CRS in methylene chloride R
Absorption maxima: at 2920 cm- l, 2850 cm- l, 1465 cm- l, and dilute to 10 mL with the same solvent.
730 cm- l and 720 cm-l. Plate: TLC si/ica gel G plate R.
The spectrum obtained is identical to that obtained with Mobile phase A: hexane R.
the material selected for the type sample. If the material to Mobile phase B: methanol R, methylene chloride R (5:95 V/V).
be examined is in the form of sheets, the identification may
be performed directly on a cut piece of suitable size. Application: 10 11L.
B. Additives (see Tests). Development A: over a path of 13 cm using mobile phase A.
Drying A: in airo
TESTS Development B: over a path of 10 cm using mobile phase B.
If necessary, cut the samples of the material to be examined Drying B: in airo
into pieces of maximum dimension on a side of not greater
than 1 cm. Deíection: spray with a 40 giL solution of phosphomolybdic
acid R in ethanol (96 per cent) R and heat at 120 oC until the
Solution SI. Place 25 g in a borosilicate-glass flask with a spots appear in the chromatogram obtained with the reference
ground-glass neck. Add 500 mL of water for injections R and solution.
heat under a reflux condenser for 5 h. Allow to cool and
System suitability: reference solution:
decant. Keep part of the solution for the test for appearance
of solution. Filter the rest through a sintered glass filter (16) - the chromatogram shows 2 separated spots.
(2.1.2). Use solution Sl within 4 h of preparation. Limit: no spot appears in the chromatogram obtained with
SoluHon S2. Place 2.0 g in a conical borosilicate-glass flask the test solution, except for a spot which may be at the solvent
with a ground-glass neck. Add 80 mL of toluene R and boil front from the first development and which corresponds
under a reflux condenser with constant stirring for 1 h 30 mino to oligomers. Disregard any spots corresponding to those
Allow to cool to 60 oC and add with continued stirring 120 mL obtained in the chromatogram with the blank solution.
of methanol R. Filter the solution through a sintered-glass Extractable heavy rnetals (2.4.8): maximum 2.5 ppm.
filter (16) (2.1.2). Rinse the flask and the filter with 25 mL of a Evaporate 50 mL of solution S3 to about 5 mL on a water-bath
mixture of 40 mL of toluene R and 60 mL of methanol R, add and dilute to 20 mL with water R. 12 mL of solution complies
the rinsings to the filtrate and dilute to 250 mL with the same with test A. Prepare the reference solution using 2.5 mL of
mixture of solvents. Prepare a blank solution. lead standard solution (10 ppm Pb) R.
Solution 53. Place 100 g in a conical borosilicate-glass flask 5ulfated ash (2.4.14): maximum 0.02 per cent, determined
with a ground-glass neck. Add 250 mL of 0.1 M hydrochloric on 5.0 g.
acid and boil under a re flux condenser with constant stirring
for 1 h. Allow to cool and decant the solution.
0112008:30105
Appearance of solution. Solution SI is clear (2.2.1) and
corrected 7.5
coIourless (2.2.2, Method II).
Acidity or alkalinity. To 100 mL of solution SI add 0.15 mL
of BRP indicator solution R. Not more than 1.5 mL of 0.01 M
3.1.5. POLYETHYLENE WITH
sodium hydroxide is required to change the colour of the ADDITIVES FOR CONTAINERS FOR
indicator to bIue. To 100 mL of solution SI add 0.2 mL of PARENTERAL PREPARATIONS AND
methyl orange solution R. Not more than l.0 mL of 0.01 M
hydrochloric acid is required to reach the beginning of the FOR OPHTHALMIC PREPARATIONS
colour change of the indicator from yellow to orange. DEFINITION
Absorbance (2.2.25): maximum 0.2, determined between Polyethylene with additives is obtained by the polymerisation
wavelengths of 220 nm and 340 nm on solution S l. of ethylene under pressure in the presence of a catalyst or by
Redudng substances. To 20 mL of solution SI add 1 mL copolymerisation of ethylene with not more than 25 per cent
of dilute sulfuric acid R and 20 mL of 0.002 M potassium ofhigher alkene homologues (C 3 to CID)'
permanganate. Boil under a re flux condenser for 3 min and
cool immediately. Add 1 g of potassium iodide R and titrate PRODUCTION
immediately with 0.01 M sodium thiosulfate, using 0.25 mL A certain number of additives are added to the polymer in
of starch solution R as indicator. Carry out a blank titration. order to optimise their chemical, physical and mechanical
The difference between the titration volumes is not more than properties in order to adapt them for the intended use. AH
0.5 mL. these additives are chosen from the appended list which
specifies for each product the maximum allowable content.
Substances soluble in hexane. Place 10 g in a 250 mL conicaI
borosilicate-glass flask with a ground-glass neck. Add 100 mL They may contain at 1110St 3 antioxidants, 1 or several
of hexane R and boil under a reflux condenser for 4 h, stirring lubricants or antiblocking agents as well as titanium dioxide
constantly. Cool in iced water and filter rapidly through a as an opacifying agent when the material must provide
sintered-glass filter (16) (2.1.2) maintaining the solution at protection from light.
O oC (the filtration time must be Iess than 5 min; if necessary - butylhydroxytoluene (plastic additive 07): maximum
the filtration may be acceIerated by applying pressure to the 0.125 per cent;
solution). Evaporate 20 mL of the filtrate in a tared glass dish - pentaerythrityl tetrakis[3-(3,5-di-tert-butyl-4-
on a water-bath. Dry the residue in an oven at 100-105 oC hydroxyphenyl)propionate] (plastic additive 09): maximum
for 1 h. The mass of the residue obtained is within 10 per 0.3 per cent;
cent of the residue obtained with the type sample and does - 1,3,5-tris( 3,5-di - tert- butyl-4-hydroxybenzyl) -5- triazine-
not exceed 5 per cent. 2,4,6(lH,3H,5H)-trione (plastic additive 13): maximum
Additives. Thin-layer chromatography (2.2.27). 0.3 per cent;
Test solution. Evaporate 50 mL of solution 52 to dryness in - octadecy! 3- (3,S-di -tert-butyl-4- hydroxyphenyl)propionate,
vacuo at 45 oc. Dissolve the evaporation residue with 5 mL of (plastic additive 11): maximul11 0.3 per cent;

384 See the information section 011 general monographs (caver pages)
EUROPEAN PHARMACOPOEIA 8.0 3.1.5. Polyethylene with additives fOl" containel"s

- ethylene bis[3,3-bis[3-(1,l-dimethylethyl)-4- acid R. Heat gently. Filter the resulting solution. To the
hydroxyphenyl]butanoate] (plastic additive 08): maximum filtrate add 1 mL of phosphoric acid R and 1 mL of strong
0.3 per cent; hydrogen peroxide solution R. lf the substance is opacined
- dioctadecyl disulfide (plastic additive 15): maximum with titanium dioxide, an orange-yellow colour develops.
03 per cent;
TESTS
- 4,4',4"-(2,4,6-trimethylbenzene-l ,3,5-triyltrismethylene)-
If necessary, cut the samples of the material to be examined
tris [2,6-bis(l, l-dimethylethyl)phenol] (plastic additive 10):
into pieces of maximum dimension on a si de of not greater
maximum 0.3 per cent;
than 1 cm.
- 2,2'-bis( octadecyloxy)-5,5'-spirobi[1,3,2-dioxa-
phosphinane] (plastic additive 14): maximum 0.3 per cent; Solution SI. Place 25 g in a borosilicate-glass flask with a
ground-glass neck. Add 500 mL of water for injections R and
- didodecyl 3,3'-thiodipropionate (plastic additive 16): boil under a reflux condenser for 5 h. Allow to cool and decanto
maximum 0.3 per cent; Reserve a portion of the solution for the test for appearance of
- dioctadecyl 3,3'-thiodipropionate (plastic additive 17): solution and filter the rest through a sintered -glass filter (16)
maximum 0.3 per cent; (2.1.2). Use within 4 h of preparation.
- tris [2,4-bis(l,1-dimethylethyl)phenyl] phosphite (plastic Solution S2. Place 2.0 g in a conical borosilicate-glass flask
additive 12): maximum 0.3 per cent. with a ground-glass neck. Add 80 mL of toluene R and boil
The total of antioxidant additives listed aboye does not exceed under a reflux condenser with constant stirring for 90 mino
0.3 per cent. Allow to cool to 60 oC and add with continued stirring 120 mL
- hydrotalcite: maximum 0.5 per cent; of methanol R. Filter the solution through a sintered-glass
alkanamides: maximum 0.5 per cent; filter (16) (2.1.2). Rinse the flask and the filter with 25 mL of
a mixture of 40 mL of toluene R and 60 mL of methanol R,
alkenamides: maximum 0.5 per cent;
add the rinsings to the filtrate and dilute to 250.0 mL with the
sodium silico-aluminate: maximum 0.5 per cent; same mixture of solvents. Prepare a blank solution.
silica: maximum 0.5 per cent;
Solution 53. Place 100 g in a conical borosilicate-glass flask
sodium benzoate: maximum 0.5 per cent; with a ground-glass neck. Add 250 mL of 0.1 M hydrochloric
fatty acid esters or salts: maximum 0.5 per cent; acid and boil under a reflux condenser with constant stirring
trisodium phosphate: maximum 0.5 per cent; for 1 h. Allow to cool and decant the solution.
liquid paraffin: maximum 0.5 per cent; Appearance of solution. Solution SI is clear (2.2.1) and
zinc oxide: maximum 0.5 per cent; colourless (2.2.2, Method II).
magnesium oxide: maximum 0.2 per cent; Addity Ol" alkalinity. To 100 mL of solution SI add 0.15 mL
calcium stearate or zinc stearate ar a mixture of both: of BRP indicator solution R. Not more than 1.5 mL of 0.01 M
maximum 0.5 per cent; sodium hydroxide is required to change the c010ur of the
indicator to blue. To 100 mL of s01ution SI add 0.2 mL of
- titanium dioxide only for materials far containers for
methyl orange solution R. Not more than 1.0 mL of 0.01 M
ophthalmic use: maximum 4 per cent.
hydrochloric acid is required to reach the beginning of the
The supplier of the material must be able to demonstrate colour change of the indicator from yellow to orange.
that the qualitative and quantitative composition of the type
sample is satisfactary for each praductian batch. Absorbance (2.2.25): maximum 0.2, determined between
wavelengths of 220 nm and 340 nm on solution Sl.
CHARACTERS Redudng substances. To 20 mL of solution SI add 1 mL
Appearance: powder, beads, granules Of, after transformation, of dilute sulfuric acid R and 20 mL of 0.002 M potassium
translucent sheets of varying thicknesses ar containers. permanganate. Boil under a reflux condenser for 3 min and
Solubility: practically insoluble in water, soluble in hot cool immediately. Add 1 g of potassium iodide R and titrate
aromatic hydrocarbons, practically insoluble in anhydrous immediately with 0.01 M sodium thiosulfate, using 0.25 mL
ethanol, in hexane and in methanol. of starch solution R as indicator. Carry out a blank titration.
It softens at temperatures between 70 oC and 140 0e.
The difference between the titration volumes is not more than
0.5 mL.
Relative density: 0.890 to 0.965.
Substances soluble in hexane. Place 10 g in a 250 mL conical
IDENTIFICATION borosilicate-glass flask with a ground-glass neck. Add 100 mL
If necessary, cut the samples of the material to be examined of hexane R and boil under a reflux condenser for 4 h, stirring
into pieces of maximum dimension on a side of not greater constantly. Cool in iced water and fiIter rapidly through a
than 1 cm. sintered-glass filter (16) (2.1.2) maintaining the solution at
A. Infrared absorption spectrophotometry (2.2.24). O oC (the filtration time must be less than 5 min; if necessary
the filtration may be accelerated by applying pressure to
Preparation: to 0.25 g add 10 mL of toluene R and boil the solution). Evaporate 20 mL of the filtrate in a tared
under a reflux condenser for about 15 mino Place a few borosilicate-g1ass dish on a water-bath. Dry the residue in an
drops of the solution on a sodium chloride disc and oven at 100-105 oC for 1 h. The mass of the residue obtained
evaporate the solvent in an oven at 80 0e. must be within 10 per cent of the residue obtained with the
Absorption maxima: at 2920 cm- l , 2850 cm-l, 1465 cm- l , type sample and does not exceed 5 per cent.
1375 cm-l, 1170 cm-l, 730 cm- l and 720 cm- l.
Extractable aluminium: maximum 1 ppm.
The spectrum obtained is identical to the spectrum
Inductively coupled plasma-atomic emission spectrometry
obtained with the material selected for the type sample. lf
(2.2.57).
the material to be examined is in the form of sheets, the
identincation may be performed directly on a cut piece of Test solution. Use solution S3.
suitable size. Reference solutions. Prepare the reference solutions using
B. It complies with the supplementary tests corresponding to aluminium standard solution (200 ppm Al) R, diluting with
the additives present (see Tests). 0.1 M hydrochloric acid.
e. In a platinum crucible, mix about 20 mg with 1 g of Wavelength: use the emission of aluminium at 396.15 nm, the
potassium hydrogen sulfate R and heat until completely spectral background being taken as 396.25 nm.
melted. Allow to co01 and add 20 mL of dílute sulfuric Verify the absence of aluminium in the hydrochloric acid used.

General Notices (1) apply to all monographs and other texts 385
3.1.5. Polyethylene with additives for containers EUROPEAN PHARMACOPOEIA 8.0

Extractable chromium: maximum 0.05 ppm. SUPPLEMENTARY TESTS


Inductively coupled plasma-atomic emission spectrometry These tests are to be carried out, in whole or in part, only if
(2.2.57). required by the stated eomposition of the material.
Test solution. Use solution S3. Phenolic antioxidants. Liquid chromatography (2.2.29).
ReÍerence solutions. Prepare the reference solutions using 501vent mixture: acetonitrile R, tetrahydrofuran R (50:50 V/V).
chromium standard solution (100 ppm Cr) R, diluting with a
Test solution 521. Evaporate 50 mL of solution S2 to dryness
mixture of 2 volumes of hydrochloric acid R and 8 volumes in vacuo at 45 oc. Dissolve the residue with 5.0 mL of the
of water R. solvent mixture. Prepare a blank solution from the blank
Wavelength: use the emission of chromium at 205.55 nm, the solution correspondi-ng to solution S2.
spectral background being taken as 205.50 nm. Test solution S22. Evaporate 50 mL of solution S2 to dryness in
Verify the absence of chromium in the hydrochloric acid used. vacuo at 45 oc. Dissolve the residue with 5.0 mL of methylene
Extractable titanium: maximum 1 ppm. chloride R. Prepare a blank solution ffom the blank solution
corresponding to solution S2.
Inductively coupled plasma-atomic emission spectrometry
(2.2.57). OÍ the Íollowing reÍerence solutions, only prepare those that are
necessary Íor the analysis oÍ the phenolic antioxidants stated in
Test solution. Use solution S3. the composition oÍ the substanee to be examined.
ReÍerence solutions. Prepare the reference solutions using ReÍerence solution (a). Dissolve 25.0 mg of butylhydroxy-
titanium standard solution (100 ppm Ti) R, diluting with 0.1 M toluene CRS (plastic additive 07) and 60.0 mg of plastic
hydrochloric acid. additive 08 CRS in 10.0 mL of the solvent mixture. Dilute
Wavelength: use the emission of titanium at 336.12 nm, the 2.0 mL of this solution to 50.0 mL with the solvent mixture.
spectral background being taken as 336.16 nm. ReÍerence solution (b). Dissolve 60.0 mg of plastic
Verify the absence of titanium in the hydrochloric acid used. additive 09 CR5 and 60.0 mg of plastic additive 10 CRS in
Extractable vanadium: maximum 0.1 ppm. 10.0 mL of the solvent mixture. Dilute 2.0 mL of this solution
to 50.0 mL with the solvent mixture.
Inductively coupled plasma-atomic emission spectrometry
(2.2.57). ReÍerence solution (e). Dissolve 60.0 mg of plastic
additive 11 CRS and 60.0 mg of plastic additive 12 CRS in
Test solution. Use solution S3. 10.0 mL of methylene chloride R. Dilute 2.0 mL of this solution
ReÍerence solutions. Prepare the reference solutions using to 50.0 mL with methylene chloride R.
vanadium standard solution (1 giL V) R, diluting with a ReÍerence solution (d). Dissolve 25.0 mg of butylhydroxy-
mixture of 2 volumes of hydrochlorie aeid R and 8 volumes toluene CRS (plastic additive 07) in 10.0 mL of the solvent
of water R. mixture. Dilute 2.0 mL of this solution to 50.0 mL with the
Waveiength: use the emission of vanadium at 292.40 nm, the solvent mixture.
spectral background being taken as 292.35 nm. Reference solution (e). Dissolve 60.0 mg of plastic
Verify the absence of vanadium in the hydrochloric acid used. additive 08 CRS in 10.0 mL of the solvent mixture. Dilute
Extractable zinc: maximum 1 ppm. 2.0 mL of this solution to 50.0 mL with the solvent mixture.
Atomic absorption spectrometry (2.2.23, Method I). ReÍerence solution m. Dissolve 60.0 mg of plastic
additive 13 CRS in 10.0 mL of the solvent mixture. Dilute
Test solution. Use solution S3. 2.0 mL of this solution to 50.0 mL with the solvent mixture.
ReÍerence solutions. Prepare the reference solutions using ReÍerence solution (g). Dissolve 60.0 mg of plastic
zinc standard solution (lO ppm Zn) R, diluting with 0.1 M additive 09 CR5 in 10.0 mL of the solvent mixture. Dilute
hydrochlorie acid. 2.0 mL of this solution to 50.0 mL with the solvent mixture.
50urce: zinc hollow-cathode lampo Reference solution (h). Dissolve 60.0 mg of plastic
Wavelength: 213.9 nm. additive 10 CRS in 10.0 mL of the solvent mixture. Dilute
Atomisation de vice : air-acetylene flameo 2.0 mL of this solution to 50.0 mL with the solvent mixture.

Extractable zirconium: maximum 0.1 ppm. ReÍerence solutiol1 (i). Dissolve 60.0 mg of plastic
additive 11 CRS in 10.0 mL of methylene chloride R. Dilute
Inductively coupled plasma -atomic emission spectrometry 2.0 mL of this solution to 50.0 mL with methylene ehloride R.
(2.2.57).
ReÍerenee solution (j). Dissolve 60.0 mg of plastic
Test solution. Use solution S3. additive 12 CRS in 10.0 mL of methylene chloride R. Dilute
ReÍerence solutiol1S. Prepare the reference solutions using 2.0 mL of this solution to 50.0 mL with methylene chloride R.
zirconium standard solution (1 giL Zr) R, diluting with a A. If the substance to be examined contains plastic additive 07
mixture of 2 volumes of hydrochloric acid R and 8 volumes and/or plastic additive 08, proceed as follows.
of water R.
Column:
Wavelength: use the emission ofzirconium at 343.82 nm, the
spectral background being taken as 343.92 nm. - size: 1 = 0.25 m, 0 = 4.6 mm;

Verify the absence of zirconium in the hydrochloric acid used. - stationary phase: octadecylsilyl si/ica gel Íor
chromatography R (5 flm).
Extractable heavy metals (2.4.8): maximum 2.5 ppm.
Mobile phase: water R, aeetonitrile R (30:70 V/V).
Evaporate 50 mL of solution S3 to about 5 mL on a water-bath
and dilute to 20.0 mL with water R. 12 mL of the solution Flow rate: 2 mL/min.
complies with test A. Prepare the reference solution using Detectiol1: spectrophotometer at 280 11m.
2.5 mL of lead standard solution (lO ppm Pb) R. Injection: 20 flL of test solution S21, of the corresponding
Sulfated ash (2.4.14): maximum 1.0 per cent, determined on blank solution, of reference solution (a), and either
5.0 g. reference solution (d) or (e), or reference solutions (d)
This limit does 110t apply to material opacified with titanium and (e).
dioxide. Run time: 30 mino

386 See the inÍormation section on general monographs (ca ver pages)
EUROPEAN PHARMACOPOEIA 8.0 3.1.5. Polyethylene with additives for coníainers

System suitability: Reference solution (k). Dissolve 60 mg of plastic additive 14 CR5


- resolution: minimum 8.0 between the peaks due in methylene chloride R and dilute to 10 mL with the same
to plastie additive 07 and plastie additive 08 in the solvento Dilute 2 mL of this solution to 10 mL with acidified
ehromatogram obtained with referenee solution (a); methylene chloride R.
- the chromatogram corresponding to test solution 521 Reference solution (1). Dissolve 60 mg of plastic additive 15 CR5
only show peaks due to antioxidants stated in the in methylene chloride R and dilute to 10 mL with the same
composition and minor peaks that also appear in the solvento Dilute 2 mL of this solution to 10 mL with acidified
chromatogram corresponding to the blank solution. methylene chloride R.
Limit: the areas of the peaks of test solution S21 are less than Reference solution (m). Dissolve 60 mg of plastic
the are as of the eorresponding peaks in the ehromatograms additive 16 CR5 in methylene chloride R and dilute to 10 mL
obtained with referenee solutions (d) and/or (e). with the same solvent. Dilute 2 mL of this solution to 10 mL
with acidified methylene chloride R.
B. lf the substance to be examined contains one or more of
the following antioxidants: Reference solution (n). Dissolve 60 mg of plastic additive 17 CRS
- plastic additive 09; in methylene chloride R and dilute to 10 mL with the same
solvent. Dilute 2 mL of this solution to 10 mL with acidified
- plastic additive 10; methylene chloride R.
- plastic additive 11; Reference solution (o). Dissolve 60 mg of plastic additive 16 CR5
- plastie additive 12; and 60 mg of plastic additive 17 CR5 in methylene chloride R
- plastic additive 13; and dilute to 10 mL with the same solvent. Dilute 2 mL ofthis
solution to 10 mL with acidified methylene chloride R.
proeeed as deseribed aboye with the following
modifications. Plate: TLC silica gel GF254 plate R.
Mobile phase: water R, tetrahydrofuran R, acetonitrile R Mobile phase A: hexane R.
(10:30:60 V/V/V). Mobile phase B: methylene chloride R.
Flow rate: 1.5 mLlmin. Application: 20 flL of test solution S23, of reference solution (o)
Injection: 20 flL of test solution 521, of the corresponding and of the reference solutions eorresponding to al! the
blank solution, of referenee solution (b) and reference phenolie and non-phenolie antioxidants mentioned in the
solutions of the antioxidants on the list aboye that are type eomposition of the material to be examined.
stated in the composition.
Development A: over a path of 18 cm with mobile phase A.
5ystem suitability:
Drying A: in airo
- resolution: minimum 2.0 between the peaks due
to plastie additive 09 and plastic additive 10 in the Development B: over a path of 17 cm with mobile phase B.
ehromatogram obtained with reference solution (b); Drying B: in airo
- the chromatogram corresponding to test solution 521 Detection: examine in ultraviolet light at 254 nm, spray with
only show peaks due to antioxidants stated in the alcoholic iodine solution R and examine in ultraviolet light at
composition and minor peaks that also appear in the 254 nm after 10-15 mino
chromatogram eorresponding to the blank solution.
5ystem suitability: reference solution (o):
Limit: the are as of the peaks of test solution 521 are less than
- the chromatogram shows 2 clearly separated spots.
the are as of the eorresponding peaks in the ehromatograms
obtained with reference solutions of the antioxidants on the Limits: any spots in the ehromatogram obtained with test
list aboye that are stated in the eomposition. solution S23 are not more intense than the spots in the same
e. lf the substanee to be examined eontains plastic additive 11 loeations in the ehromatograms obtained with the referenee
and/or plastic additive 12, carry out the test as deseribed solutions.
for plastic additive 07 and/or plastie additive 08 with the Amides and stearates. Thin-Iayer ehromatography (2.2.27).
following modifications. Test solution. Use test solution 523 described in the test for
Mobile phase: water R, 2-propanol R, methanol R non-phenolic antioxidants.
(5:45:50 V/V/V). Reference solution (p). Dissolve 20 mg of stearic acid CRS
Flow rate: 1.5 mLlmin. (plastie additive 19) in methylene chloride R and dilute to
Injection: 20 flL of test solution S22, of the eorresponding 10 mL with the same solvent.
blank solution, of referenee solution (e), and either of Reference solution (q). Dissolve 40 mg of plastic additive 20 CR5
reference solution (i) or (j), or referenee solutions (i) and (j). in methylene chloride R and dilute to 20 mL with the same
System suitability: solvent.
- resolution: minimum 2.0 between the peaks due Reference solution (r). Dissolve 40 mg of plastic additive 21 CRS
to plastic additive 11 and plastic additive 12 in the in methylene ehloride R and dilute to 20 mL with the same
chromatogram obtained with referenee solution (e); solvento
- the ehromatogram eorresponding to test solution S22 Plates: TLC silica gel GF254 plate R (2 plates).
only show peaks due to antioxidants stated in the
A. Mobile phase: anhydrous ethanol R, trimethylpentane R
eomposition and minor peaks that also appear in the
(25:75 V/V).
ehromatogram eorresponding to the blank solution.
Applieation: 10 ~lL of test solution S23 and reference
Limit: the areas of the peaks of test solution S22 are less than
solution (p).
the are as of the eorresponding peaks in the ehromatograms
obtained with referenee solutions (i) and/or (j). Development: over a path of 10 cm.
Non-phenolic antioxidants. Thin-layer ehromatography Drying: in airo
(2.2.27). Deteetion: spray with a 2 giL solution of
Test solution 523. Evaporate 100 mL of solution 52 to dryness dichlorophenolindophenol, sodium salt R in anhydrous
in vacuo at 45 0e. Dissolve the residue in 2 mL of acidified ethanol R and heat in an oven at 120 oC for a few minutes
methylene chloride R. to intensify the spots.

General Notices (1) apply to all monographs and other texts 387
3.1.6. Polypropylene for containers and dosures EUROPEAN PHARMACOPOEIA 8.0

Limit: any spot corresponding to plastic additive 19 in the - 2,2' -bis( octadecyloxy) -5,5' -spirobi [1 ,3,2-dioxa-
chromatogram obtained with test solution S23 is identical phosphinane] (plastic additive 14): maximum 0.3 per cent;
in position (R p = about 0.5) but not more intense than the - didodecyl 3,3'-thiodipropionate (plastic additive 16):
spot in the same location in the chromatogram obtained maximum 0.3 per cent;
with reference solution (p).
- dioctadecyI3,3'-thiodipropionate (plastic additive 17):
B. Mobile phase A: hexane R. maximum 0.3 per cent;
Mobile phase B: methanol R, methylene chloride R - tris(2,4-di-tert-butylphenyl) phosphite (plastic additive 12):
(5:95 VIV). maximum 0.3 per cent;
Application: 10 I1L of test solution S23 and reference The total of antioxidant additives listed aboye does not exceed
solutions (q) and (r). 0.3 per cent.
Development A: over a path of 13 cm with mobile phase A. - hydrotalcite: maximum 0.5 per cent;
Drying A: in airo - alkanamides: maximum 0.5 per cent;
Development B: over a path of 10 cm with mobile phase B. - alkenamides: maximum 0.5 per cent;
Drying B: in airo
- sodium silico-aluminate: maximum 0.5 per cent;
Detection: spray with a 40 giL solution of phosphomolybdic
- silica: maximum 0.5 per cent;
acid R in anhydrous ethanol R and heat in an oven at 120 oC
until spots appear. - sodium benzoate: maximum 0.5 per cent;
Limit: any spots corresponding to plastic additive 20 or - fattyacid esters or salts: maximum 0.5 per cent;
plastic additive 21 in the chromatogram obtained with the - trisodium phosphate: maximum 0.5 per cent;
test solution S23 are identical in position (R p = about 0.2) - liquid paraffin: maximum 0.5 per cent;
but not more intense than the corresponding spots in
the chromatograms obtained with reference solutions (q) - zinc oxide: maximum 0.5 per cent;
and (r). - tale: maximum 0.5 per cent;
- magnesium oxide: maximum 0.2 per cent;
0112008:30106
- calcium stearate or zinc stearate or a mixture of both:
corrected 7.5 maximum 0.5 per cent;
- titanium dioxide, only for materials for containers for
ophthalmic use: maximum 4 per cent.
3.1.6. POLYPROPYLENE FOR
The supplier of the material must be able to demonstrate
CONTAINERS AND CLOSURES that the qualitative and quantitative composition of the type
FOR PARENTERAL PREPARATIONS sample is satisfactory for each production batch.
AND OPHTHALMIC PREPARATIONS CHARACTERS
DEFINITION Appearance: powder, beads, granules or, after transformation,
Polypropylene consists of the homopolymer of propylene translucent sheets of varying thicknesses or containers.
or of a copolymer of propylene with not more than 25 per Solubility: practically insoluble in water, soluble in hot
cent of ethylene or of a mixture (alloy) of polypropylene with aromatic hydrocarbons, practically insoluble in anhydrous
not more than 25 per cent of polyethylene. 1t may contain ethanol, in hexane and in methanol.
additives. 1t softens at temperatures beginning at about 120 De.
PRODUCTION IDENT1FICATION
A certain number of additives are added to the polymer in 1f necessary, cut the material ta be examined into pieces of
order to optimise their chemical, physical and mechanical maximum dimension on a side of not greater than 1 cm.
properties in order to adapt them for the intended use. Al!
A. Infrared absorption spectrophotometry (2.2.24).
these additives are chosen from the appended list which
specifies for each product the maximum allowable content. Preparation: to 0.25 g add 10 mL of toluene R and boíl
under a reÍlux condenser for about 15 mino Place a few
They may contain at most 3 antioxidants, one or several
lubricants or antiblocking agents as well as titanium dioxide as drops of the hot solution on a sodium chloride disc and
evaporate the solvent in an oven at 80 De.
opacifying agent when the material must provide protection
from Iight. Absorption maxima: at 1375 cm-l, 1170 cm- l, 995 cm- l
and 970 cm-l.
- butylhydroxytoluene (pI as tic additive 07): maximum
0.125 per cent; The spectrum obtained is identical to the spectrum
- pentaerythrityl tetrakis[3-(3,5-di-tert-butyl-4- obtained with the material selected for the type sample. lf
hydroxyphenyl)propionate] (plastic additive 09): maximum the material to be examined is in the form of sheets, the
0.3 per cent; identification may be performed directly on a cut piece of
suitable size.
- 1,3,5-tris( 3,5-di - tert- butyl-4-hydroxybenzyl) -5- triazine-
2,4,6(lH,3H,5H)-trione (plastic additive 13): maximum B. It complies with the supplementary tests corresponding to
0.3 per cent; the additives present (see Tests).
- octadecyl 3-( 3,5-di - tert- butyl-4-hydroxyphenyl)propionate, e. In a platinum crucible, mix about 20 mg with 1 g of
(plastic additive 11): maximum 0.3 per cent; potassium hydrogen sulfate R and heat until completely
melted. Allow to cool and add 20 mL of dilute sulfuric
- ethylene bis[3,3-bis[3-(l,1-dimethylethyl)+
acid R. Heat gently. Filter the resulting solution. To the
hydroxyphenyl]butanoate] (plastic additive 08): maximum filtrate add 1 mL of phosphoric acid R and 1 mL of strong
0.3 per cent; hydrogen peroxide solutian R. lf the substance is opacified
- dioctadecyl disulfide (plastic additive 15): maximum with titanium dioxide, an orange-yellow colour develops.
0.3 per cent;
- 2,2',2",6,6',6"-hexa - tert-butyl-4,4',4"- [(2,4,6-trimethyl- TESTS
1,3,5-benzenetriyl)trismethylene] triphenol (plastic If necessary, cut the material to be examined into pieces of
additive 10): maximum 0.3 per cent; maximum dimension 011 a side of not greater than 1 cm.

388 See the informatian section on general monographs (ca ver pages)
EUROPEAN PHARMACOPOEIA 8.0 3.1.6. Polypropylene for container!> and dosures

Solution SI. Use solution 51 within 4 h of preparation. Place Reference solutions. Prepare the reference solutions using
25 g in a borosilicate-glass flask with a ground-glass neck. chromium standard solution (100 ppm Cr) R, diluting with a
Add 500 mL of water for injections R and boil under a reflux mixture of 2 volumes of hydrochloric acid R and 8 volumes
condenser for 5 h. Allow to cool and decanto Reserve a portion of water R.
of the solution for the test for appearance of solution and filter Wavelength: use the emission of chromium at 205.55 11m, the
the rest through a sintered-glass filter (16) (2.1.2). spectral background being taken as 205.50 nm.
Solution S2. Place 2.0 g in a conical borosilicate-glass flask Verify the absence of chromium in the hydrochloric acid used.
with a ground-glass neck. Add 80 mL of toluene R and boil
under a reflux condenser with constant stirring for 1 h 30 mino Extractable titanium: maximum 1 ppm.
Allow to cool to 60 oC and add with continued stirring 120 mL Inductively coupled plasma-atomic emission spectrometry
of methanol R. Filter the solution through a sintered-glass (2.2.57).
filter (16) (2.1.2). Rinse the flask and the filter with 25 mL of Test solution. Use solution 53.
a mixture of 40 mL of toluene R and 60 mL of methanol R,
Reference solutions. Prepare the reference solutions using
add the rinsings to the filtrate and dilute to 250.0 mL with the
titanium standard solution (100 ppm Ti) R, diluting with 0.1 M
same mixture of solvents. Prepare a blank solution.
hydrochloric acid.
Solution S3. Place 100 g in a conical borosilicate-glass flask Wavelength: use the emission oftitanium at 336.12 nm, the
with a ground-glass neck. Add 250 mL of 0.1 M hydrochloric spectral background being taken as 336.16 nm.
acid and boil under a reflux condenser with constant stirring
for 1 h. Allow to coo1 and decant the solution. Verify the absence of titanium in the hydrochloric acid used.
Appearance of solution. Solution SI is not more opalescent Extractable vanadium: maximum 0.1 ppm.
than reference suspension II (2.2.1) and is colourless (2.2.2, Inductively coupled plasma-atomic emission spectrometry
Method JI). (2.2.57).
Addity 01" alkalinity. To 100 mL of solution SI add 0.15 mL Test solution. Use solution S3.
of BRP indicator solution R. Not more than 1.5 mL of 0.01 M Reference solutions. Prepare the reference solutions using
sodium hydroxide is required to change the colour of the vanadium standard solution (1 giL V) R, diluting with a
indicator to blue. To 100 mL of solution SI add 0.2 mL of mixture of 2 volumes of hydrochloric acid R and 8 volumes
methyl orange solution R. Not more than 1.0 mL of 0.01 M of water R.
hydrochloric acid is required to reach the beginning of the
Wavelength: use the emission of vanadium at 292.40 nm, the
colour change of the indicator from yellow to orange.
spectral background being taken as 292.35 nm.
Absol"bance (2.2.25): maximum 0.2, determined between Verify the absence of vanadium in the hydrochloric acid used.
wavelengths of 220 nm to 340 nm on solution SI.
Extractable zinc: maximum 1 ppm.
Redudng substances. To 20 mL of solution SI add 1 mL
of dilute sulfuric acid R and 20 mL of 0.002 M potassium Atomic absorption spectrometry (2.2.23, Method I).
permanganate. Boil under a reflux condenser for 3 min and Test solution. Use solution S3.
cool immediately. Add 1 g of potassium iodide R and titrate Reference solutions. Prepare the reference solutions using
immediately with 0.01 M sodium thiosulfate, using 0.25 mL zinc standard solution (la ppm Zn) R, diluting with 0.1 M
of starch solution R as indicator. Carry out a blank titration. hydrochloric acid.
The difference between the titration volumes is not more than
0.5 mL. 50urce: zinc hollow-cathode lampo
Wavelength: 213.9 nm.
Substances soluble in hexane. Place 10 g in a 250 mL conical
borosilicate-glass flask with a ground-glass neck. Add 100 mL Atomisation device: air-acetylene flameo
of hexane R and boíl under a reflux condenser for 4 h, stirring Verify the absence of zinc in the hydrochloric acid used.
constantly. Cool in iced water and filter rapidly through a
Extractable heavy metals (2.4.8): maximum 2.5 ppm.
sintered-glass filter (16) (2.1.2) maintaining the solution at
O oC (the filtration time must be less than 5 min; if necessary Concentrate 50 mL of solution S3 to about 5 mL on a
the filtration may be accelerated by applying pressure to the water-bath and dilute to 20.0 mL with water R. 12 mL of the
solution). Evaporate 20 mL of the filtrate in a tared glass dish solution complies with test A. Prepare the reference solution
on a water-bath. Dry the residue in an oven at 100-105 oC for using 2.5 mL of lead standard solution (10 ppm Pb) R.
1 h. The mass of the residue obtained must be within 10 per Sulfated ash (2.4.14): maximum 1.0 per cent, determined
cent of the residue obtained with the type sample and does on 5.0 g. This limit does not apply to material that has been
not exceed 5 per cent. opacified with titanium dioxide.
Extractable aluminium: maximum 1 ppm.
SUPPLEMENTARY TESTS
Inductively coupled plasma-atomic emission spectrometry These tests are to be carried out, in whole or in part, only if
(2.2.57). required by the stated composition of the material.
Test solution. Use solution 53. Phenolic antioxidants. Liquid chromatography (2.2.29).
Reference solutions. Prepare the reference solutions using 50lvent mixture: acetonitrile R, tetrahydrofuran R (50:50 VIV).
aluminium standard solution (200 ppm Al) R, diluting with Test solution 521. Evaporate 50 mL of solution S2 to dryness
0.1 M hydrochloric acid. in vacuo at 45 oc. Dissolve the residue with 5.0 mL of the
Wavelength: use the emission of aluminium at 396.15 nm, the solvent mixture. Prepare a blank solution from the blank
spectral background being taken as 396.25 nm. solution corresponding to solution S2.
Test solution 522. Evaporate 50 mL of solution S2 to dryness in
Verify the absence of aluminium in the hydrochloric acid used. vacuo at 45 oc. Dissolve the residue with 5.0 mL of methylene
Extractable chromium: maximum 0.05 ppm. chloride R. Prepare a blank solution from the blank solution
corresponding to solution S2.
Inductively coupled plasma-atomic emission spectrometry
(2.2.57). Of the following reference solutions, only prepare those that
are necessary for the analysis of the phenolic antioxidants
Test solution. Use solution S3. stated in the composition of the substance to be examined.

General Notices (1) apply to all monographs and other texts 389
3.1.6. Polypropylene for containers and do sures EUROPEAN PHARMACOPOEIA 8.0

Referenee solution (a). Dissolve 25.0 mg of butylhydroxy- - plastic additive 13;


taluene CRS (plastic additive 07) and 60.0 mg of plastie carry out the test as described aboye with the following
additive 08 CRS in 10.0 mL of the solvent mixture. Dilute modifications.
2,0 mL of this solution to 50.0 mL with the solvent mixture,
Mobi/e phase: water R, tetrahydrofuran R, acetanitrile R
Referenee solution (b), Dissolve 60,0 mg of plastie (10:30:60 V/V/V).
additive 09 CRS and 60.0 mg of plastie additive 10 CRS in
10.0 mL of the solvent mixture, Dilute 2,0 mL of this solution Flow rate: 1.5 mL/min.
to 50,0 mL with the solvent mixture, Injection: 20 flL of test solution S21, corresponding blank
Reference solution (e), Dissolve 60.0 mg of plastie solution, reference solution (b) and reference solutions
additive 11 CRS and 60.0 mg of plastic additive 12 CRS in of the antioxidants on the list aboye that are stated in the
10 mL of methylene ehloride R. Dilute 2.0 mL of this solution composition.
to 50.0 mL with methylene ehloride R. System suitability:
Referenee solution (d). Dissolve 25,0 mg of buty/hydroxy- - reso/utian: minimum 2.0 between the peaks due
toluene CRS (plastic additive 07) in 10,0 mL of the solvent to plastic additive 09 and plastic additive 10 in the
mixture, Dilute 2,0 mL of this solution to 50,0 mL with the chromatogram obtained with reference solution (b);
solvent mixture, - the chromatogram corresponding to test solution S21
Reference solution (e), Dissolve 60,0 mg of plastic only show peaks due to antioxidants stated in the
additive 08 CRS in 10.0 mL of the solvent mixture, Dilute composition and minor peaks that also appear in the
2,0 mL of this solution to 50,0 mL with the solvent mixture, chromatogram corresponding to the blank solution.
Referenee solution (f), Dissolve 60,0 mg of plastie Limit: the areas of the peaks in the chromatogram obtained
additive 13 CRS in 10,0 mL of the solvent mixture. Dilute with test solution S21 are less than the are as of the
2.0 mL of this solution to 50.0 mL with the solvent mixture, corresponding peaks in the chromatograms obtained with
Reference salution (g), Dissolve 60.0 mg of plastic reference solutions of the antioxidants on the list aboye that
additive 09 CRS in 10,0 mL of the solvent mixture. Dilute are stated in the composition.
2.0 mL of this solution to 50,0 mL with the solvent mixture, e. lf the substance to be examined contains plastic additive 11
Referenee solutian (h). Dissolve 60,0 mg of plastic and/or pI as tic additive 12, carry out the te,st as described
additive 10 CRS in 10,0 mL of the solvent mixture, Dilute for plastic additive 07 and/or plastic additive 08 with the
2.0 mL of this solution to 50,0 mL with the solvent mixture, following modifications,
Referenee solutian (i), Dissolve 60,0 mg of p/astic Mobile phase: water R, 2-propanal R, methanol R
additive 11 CRS in 10.0 mL of methylene eh/oride R. Dilute (5:45:50 V/V/V).
2,0 mL of this solution to 50,0 mL with methy/ene ehloride R. Flow rate: 1.5 mL/min.
Referenee solution (j). Dissolve 60.0 mg ofplastic Injection: 20 flL of test solution S22, corresponding blank
additive 12 CR5 in 10,0 mL of methylene ehloride R. Dilute solution, reference solution (c), and either reference
2,0 mL of this solution to 50,0 mL with methy/ene chloride R. solution (i) or (j), or reference solutions (i) and (j),
A, If the substance to be examined contains plastic additive 07 suitability:
and/or plastic additive 08, carry out the test as follows,
- teso/ution: minimum 2.0 between the peaks due
Column: to plastic additive 11 and plastic additive 12 in the
- size: l = 0.25 m, (2) = 4,6 mm; chromatogram obtained with reference solution (c);
- statianary phase: octadecylsilyl silica gel for - the chromatogram corresponding to test solution S22
chromatography R (5 11m), only show peaks due to antioxidants stated in the
composition and minor peaks that also appear in the
Mabile phase: water R, acetonitrile R (30:70 V/V),
chromatogram corresponding to the blank solution,
Flow rate: 2 mL/min.
Limit: the areas of the peaks in the chromatogram obtained
Deteetion: spectrophotometer at 280 nm, with test solution S22 are less than the areas of the
Injection: 20 I1L of test solutiol1 S21, corresponding blank corresponding peaks in the chromatograms obtained with
solution and reference solution (a), and either reference reference solutions (i) and/or (j),
solution (d) or (e), or reference solutions (d) and (e), Non-phenoHc antioxidants, Thin-layer chromatography
Run time: 30 min, (2,2.27),
System suitability: Test solution 523, Evaporate 100 mL of solution S2 to dryness
- reso/ution: minimum 8,0 between the peaks dne in vacuo at 45 0e. Dissolve the residue with 2 mL of acidified
to plastic additive 07 and plastic additive 08 in the methylene eh/aride R.
chromatogram obtained with reference solution (a); Reference solution (k), Dissolve 60 mg of p/astic additive 14 CRS
- the chromatogram corresponding to test solution S21 in methylene ehloride R and dilute to 10 mL with the same
only show peaks due to antioxidants stated in the solvent Dilute 2 mL of the solution to 10 mL with acidified
composition and minor peaks that also appear in the methylene ehloride R.
chromatogram corresponding to the blank solution. Referenee so/ution (1), Dissolve 60 mg of p/astic additive 15 CR5
Limit: the areas of the peaks in the chromatogram obtained in methylene ehloride R and dilute to 10 mL with the same
with test solution S21 are less than the are as of the solvent Dilute 2 mL of the solution to 10 mL with aeidified
corresponding peaks in the chromatograms obtained with methy/ene ehlaride R.
reference soiutions (d) and/or (e). Reference so/utian (m), Dissolve 60 mg of plastie
B. rf the substance to be examined contains one or more of additive 16 CR5 in methylene chloride R and dilute to 10 mL
the following antioxidants: with the same solvento Dilute 2 mL of the solution to 10 mL
with acidifled methylene ehloride R.
- plastic additive 09;
- plastic additive 10;
Referenee solutian (n), Dissolve 60 mg of plastic additive 17 CRS
in methylene ehlaride R and dilute to 10 mL with the same
- plastic additive 11; solvent Dilute 2 mL ofthe solution to 10 mL with acidified
- plastic additive 12; rnethylene ehloride R.

390 See the information seetion on monographs (caver pages)


EUROPEAN PHARMACOPOEIA 8.0 3.1.7. Poly(ethylene - vinyl acetate) for containers and tubing

Referenee solution (o). Dissolve 60 mg of plastie additive 16 CRS Limit: any spots corresponding to plastic additive 20 or
and 60 mg of plastie additive 17 CRS in methylene ehloride R plastic additive 21 in the chromatogram obtained with
and dilute to 10 mL with the same solvent. Dilute 2 mL of the test solution S23 are identical in position (R p about 0.2)
solution to 10 mL with aeidified methylene eh/oride R. but not more intense than the corresponding spots in
Plate: TLC si/ica gel GF254 plate R. the chromatograms obtained with reference solutions (q)
and (r).
Mobile phase A: hexane R.
Mobile phase B: methylene eh/oride R.
Application: 20 IlL of test solution S23, reference solution (o) 0112008:30107
and reference solutions corresponding to all the phenolic and
non-phenolic antioxidants mentioned in the type composition
of the material to be examined.
3.1.7. POLY(ETHYLENE - VINYL
Development A: over a path of 18 cm with mobile phase A. ACETATE) FOR CONTAINERS AND
Drying A: in airo TUBING FOR TOTAL PARENTERAL
Development B: over a path of 17 cm with mobile phase B. NUTRITION PREPARATIONS
Drying B: in air. DEFINITION
Detection: examine in ultraviolet light at 254 nm; spray with Poly(ethylene - vinyl acetate), complying with the following
alcoholic iodine solution R and examine in ultraviolet light at requirements, is suitable for the manufacture of containers
254 nm after 10-15 mino and tubing for total parenteral nutrition preparations. It is
System suitability: reference solution (o): obtained by copolymerisation of mixtures of ethylene and
- the chromatogram shows 2 clearly separated spots. vinyl acetate.
Content of vinyl acetate:
Limits: any spots in the chromatogram obtained with test
solution S23 are not more intense than the spots in the same - material used for containers: a defined quantity of not more
positions in the chromatograms obtained with the reference than 25 per cent;
solutions. - material used for tubing: a defined quantity of not more
than 30 per cent.
Amides and stearates. Thin-layer chromatography (2.2.27).
Test solution. Use solution S23 described in the test for PRODUCTION
non-phenolic antioxidants. A certain number of additives are added to the polymer in
Reference solution (p). Dissolve 20 mg of stearic acid CRS order to optimise their chemical, physical and mechanical
(pI as tic additive 19) in methylene chloride R and dilute to properties in order to adapt them for the intended use. AH
10 mL with the same solvent. these additives are chosen from the appended list which
Reference solution (q). Dissolve 40 mg of plastic additive 20 CRS
specifies for each product the maximum allowable contento
in methylene ehloride R and dilute to 20 mL with the same Poly(ethylene - vinyl acetate) may contain not more than 3
solvent. of the following antioxidants:
Reference so/ution (r). Dissolve 40 mg of plastic additive 21 CRS - butylhydroxytoluene (plastic additive 07): maximum
in methylene chloride R and dilute to 20 mL with the same 0.125 per cent;
solvent. - pentaerythrityl tetrakis[3-(3,5-di-tert-butyl-4-
Plate: TLC siliea gel GF254 plate R (2 plates). hydroxyphenyl)propionate1(plastic additive 09): maximum
0.2 per cent;
A. Mobile phase: anhydrous ethanol R, trimethylpentane R
- octadecyI3-(3,5-di-tert-butyl-4-hydroxyphenyl)propionate
(25:75 V/V).
(plastic additive 11): maximum 0.2 per cent;
Application: 10 IlL of solution S23 and reference
- tris(2A-di-tert-butylphenyl) phosphite (plastic additive 12):
solution (p).
maximum 0.2 per cent;
Development: over a path of 10 cm. - 2,2',2",6,6',6"-hexa -tert- butyl-4A'A" -[(2A,6-trimethyl-
Drying: in airo 1,3,5-benzenetriyl)trismethylene1triphenol (plastic
Detection: spray with a 2 giL solution of additive 10): maximum 0.2 per cent.
dichlorophenolindophenol, sodium salt R in anhydrous It may also contain:
ethanol R and heat in an oven at 120 oC for a few minutes - oleamide (plastic additive 20): maximum 0.5 per cent;
to intensify the spots. - erucamide (plastic additive 21): maximum 0.5 per cent;
Limit: any spot corresponding to plastic additive 19 in the - calcium stearate or zinc stearate or a mixture of both:
chromatogram obtained with test solution S23 is identical maximum 0.5 per cent;
in position (R F about 0.5) but not more intense than the
- calcium carbonate or potassium hydroxide: maximum
spot in the same position in the chromatogram obtained
0.5 per cent;
with reference solution (p).
- colloidal silica: maximum 0.2 per cent.
B. Mobile phase A: hexane R.
The supplier of the material must be able to demonstrate
Mobile phase B: methanol R, methylene chloride R that the qualitative and quantitative composition of the type
(5:95 V/V). sample is satisfactory for each production batch.
Application: 10 flL of solution S23 and reference
solutions (q) and (r). CHARACTERS
Development A: over a path of 13 cm with mobile phase A. Appearanee: beads, granules or, after transformation,
translucent sheets or tubing of varying thickness or samples of
Drying A: in air.
finished objects.
Development B: over a path of 10 cm with mobile phase B. Solubility: practically insoluble in water, soluble in hot
Drying B: in airo aromatic hydrocarbons, practically insoluble in anhydrous
Detection: spray with a 40 giL solution of phosphomolybdic ethanol, in methanol and in hexane, which dissolves, however,
acid R in anhydrous ethanol R; heat in an oven at 120 oC low molecular mass polymers.
until spots appear. It burns with a blue flameo

General Notices (1) apply to all monographs and other texts 391
3.1.7. Poly(ethylene - vinyl acetate) for containers and tubing EUROPEAN PHARMACOPOEIA 8.0

The temperatul'e at which the substance softens changes with Reference solution (b). Dissolve 40 mg of plastie additive 20 CRS
the vinyl acetate content: fl'om about 100 oC fol' contents of a in 10 mL of methylene chloride R. Dilute 1 mL of this solution
few pel' cent to about 70 oC fOl' contents of 30 per cent. to 5 mL with methylene chloride R.
Reference solution (e). Dissolve 40 mg of plastie additive 21 CRS
IDENTIFICATION in 10 mL of methylene chloride R. Dilute 1 mL of this solution
1f necessary, cut the samples of material to be examined into to 5 111L with methylene chloride R.
pieces of maximum dimension on a side of not greater than Plates: TLC siliea gel GF254Plate R (2 plates).
1 cm. A. Mobile phase: anhydrous ethanol R, trimethylpentane R
Infrared absorption spectrophotometry (2.2.24). (25:75 V/V).
Preparation: to 0.25 g add 10 mL of toluene R and boíl undel' Application: 10 [1L.
a reflux condenser for about 15 mino Place a few drops of the Development: over a path of 10 cm.
solutio11 obtained 011 a disc of sodium chloride and evaporate Drying: in airo
the solvent in an oven at 80 oc.
Deteetion: spray with a 2 giL solution of
Absorption maxima due to vinyl acetate: at 1740 cm-1, dichlorophenolindophenol, sodium salt R in anhydrous
1375 cm- \ 1240 cm- l, 1020 cm- l and 610 cm-l. ethanol R and heat in an oven at 120 oC for a few minutes
Absorption maxima due to ethylene: at 2920-2850 cm- 1, to intensify the spots.
1470 cm-1, 1460 cm- l, 1375 cm- l, 730 cm- I and 720 cm-l. Limit: any spot corresponding to plastic additive 19 in the
The spectrum obtained is identical to the spectrum obtained chromatogram obtained with the test solution is not more
with the type sample provided by the manufacturero If the intense than the spot in the chromatogram obtained with
material to be examined is in the form of sheets, the spectrum reference solution (a).
may be determined directly on a cut piece of suitable size. B. Mobile phase A: hexane R.
Mobile phase B: methanol R, methylene chloride R
TESTS (5:95 V/V).
If necessary, cut the samples of the material to be examined Applieation: 10 [1L.
into pieces of maximum dimension on a side of not greater Development A: over a path of 13 cm with mobile phase A.
than 1 cm.
Drying A: in airo
Solution SI. Place 2.0 g in a borosilicate-glass flask with a
Development B: over a path of 10 cm with mobile phase B.
ground-glass neck. Add 80 mL of toluene R and heat under a
reflux condensel' with constant agitation for 90 mino Allow to Drying B: in airo
cool to 60 oC and add 120 mL of methanol R to the flask with Deteetion: spray with a 40 giL solution of phosphomolybdic
constant stirring. Filter the solution through a sintered-glass aeid R in anhydrous ethanol R and heat in an oven at 120 oC
filter (16) (2.1.2). Rinse the flask and the filter with 25 mL of a until spots appear.
mixture of 40 mL of toluene R and 60 mL of methanol R, add Limit: any spots corresponding to plastic additive 21 or
the rinsing mixture to the filtrate and dilute to 250 mL with plastic additive 20 in the chromatogram obtained with
the same mixture of solvents. the test solution are not more intense than the spots in
50lution S2. Use within 4 h of preparation. Place 25 g in a the chromatograms obtained with reference solutions (b)
borosilicate-glass flask with a ground-glass neek. Add 500 mL and (e) respectively.
of water for injections R and boil under a reflux condenser Phenolic antioxidants. Liquid chromatography (2.2.29).
for 5 h. Allow to eool and decanto Reserve a portian of the
Solvent mixture: acetonitrile R, tetrahydrofuran R (50:50 V/V).
solution fol' the test fol' appearance of solution S2 and filter
the rest through a sintered-glass filter (16) (2.1.2). Test solution (a). Evaporate 50 mL of solution SI to dryness
in vacuo at 45 oC and dissolve the residue in 5.0 mL of the
Appearance of solution 52. Solution S2 is clear (2.2.1) and solvent mixture.
colourless (2.2.2, Method II).
Test solution (b). Evaporate 50 mL of solution Sl to dryness in
Addity al' alkalinity. To 100 mL of solution S2 add 0.15 mL vaeuo at 45 oC and dissolve the residue in 5.0 mL of methylene
of BRP indieator solution R. Not more than 1.0 mL of 0.01 M ehloride R.
sodium hydroxide is required to ehange the colour of the
Reference solution (a). Dissolve 25 mg of butylhydroxy-
indieator to blue. To 100 mL of solution S2 add 0.2 mL of toluene CRS (plastic additive 07), 40 mg of plastic
methyl orange solution R. Not more than 1.5 mL of 0.01 M
additive 10 CRS, 40 mg of plastic additive 09 CRS and 40 mg of
hydrochloric acid is required to reach the beginning of the
plastie additive 11 CRS in 10 mL of the solvent mixture. Dilute
colour change of the indicatOl' from yellow to orange. 2 mL of this solution to 50.0 mL with the solvent mixture.
Absorbance (2.2.25): maximum 0.2 determined between Referenee solution (b). Dissolve 40 mg of plastic additive 11 CRS
wavelengths of 220 mn and 340 nm on solution S2. and 40 mg of plastic additive 12 CRS in 10 mL of methylene
Redudng suhstances. To 20 mL of solution S2 add 1 mL ehloride R. Dilute 2 mL of this solution to 50.0 mL with
of dilute sulfuric acid R and 20 mL of 0.002 M potassium methylene chloride R.
permanganate. Boil under a re flux condenser for 3 min and Column:
cool immediately. Add 1 g of potassium iodide R and titrate - size: 1= 0.25 m; 0 = 4.6 mm;
immediately with 0.01 M sodium thiosulfate, using 0.25 mL
of starch solution R as indicator. Carry out a blank titration. - stationary phase: oetadeeylsilyl silica gel for
The difference between the titration volumes is not more than chromatography R (5 [1m).
0.5 mL. Mobile phase: water R, tetrahydrofuran R, acetonitrile R
(10:30:60 V/V/V).
Amides and steark aeid. Thin-layer chromatography
(2.2.27). Flow rate: 1.5 mL/min.
Test solution. Evaporate 100 mL of solution SI to dryness Deteetion: spectrophotometer at 280 nm.
in vaeuo at 45 oc. Dissolve the residue in 2 mL of aeidified Injection: 20 [1L of test solution (a) and reference solution (a).
methylene chloride R. System suitability: reference solution (a):
Reference solution (a). Dissolve 20 mg of stearic acid CRS - resolution: minimum 2.0 between the peaks due to plastic
(plastie additive 19) in 10 mL of methylene chloride R. additive 09 and plastic additive 10;

392 See the information section on general monographs (cover pages)


EUROPEAN PHARMACOPOEIA 8.0 3.1.8. Silicone oH used as a lubdcant

- number oi theoretical plates: minimum 2500, calculated for 0112008:30108


the peak due to plastic additive 07.
Limits: 3.1.8. SILICONE OIL USED AS A
- the chromatogram obtained with test solution (a) shows LUBRICANT
only principal peaks corresponding to the peaks in the
chromatogram obtained with reference solution (a) with a
retention time greater than 2 min;
- the areas of the peaks in the chromatogram obtained
with test solution (a) are not greater than those of the
corresponding peaks in the chromatogram obtained with DEFINITION
reference solution (a), except fo1' the last peak eluted in the Silicone oil used as a lubricant is a poly(dimethylsiloxane)
chromatogram obtained with reference solution (a). obtained by hydrolysis and polycondensation of
If the chromatogram obtained with test solution (a) shows dichlorodimethylsilane and chlorotrimethylsilane. Different
a peak with the same retention time as the last antioxidant grades exist which are characterised by a number indicating
eluted from reference solution (a), carry out the test as the nominal viscosity placed after the name.
described with the following modifications. Silicone oils used as lubricants have a degree of polymerisation
Mobile phase: water R, 2-propanol R, methanol R (n '" 400 to 1200) such that their kinematic viscosities are
(5:45:50 V/V/V). nominally between 1000 mm 2·s- 1 and 30 000 mm 2.s- L.
Injection: 20 !lL of test solution (b) and reference solution (b). CHARACTERS
System suitability: reference solution (b): Appearance: clear, colourless liquid of various visco sities.
- resolution: minimum 2.0 between the peaks due to plastic So/ubility: practically insoluble in water and in methanol,
additive 11 and plastic additive 12. very slightly soluble in anhydrous ethanol, miscible with ethyl
Limits: acetate, with methyl ethyl ketone and with toluene.
- the chromatogram obtained with test solution (b) shows IDENTIFICATION
only principal peaks corresponding to the peaks in the
A. Kinematic viscosity at 25 oC (see Tests).
chromatogram obtained with reference solution (b) with a
retention time greater than 3 min; B. Infrared absorption spectrophotometry (2.2.24).
- the areas of the peaks in the chromatogram obtained Comparison: silicone oi! CRS.
with test solution (b) are not greater than those of the The region of the spectrum from 850-750 cm- 1 is not taken
corresponding peaks in the chromatogram obtained with into account since it may show slight differences depending
reference solution (b). on the degree of polymerisation.
Substances soluble in hexane. Place 5 g in a borosilicate-glass C. Heat 0.5 g in a test-tube over a small flame until white
flask with a ground-glass neck. Add 50 mL of hexane R, fit a fumes begin to appear. Invert the tube over a 2nd tube
condenser and boil under reflux on a water-bath with constant containing 1 mL of a 1 giL solution of chromotropic acid,
stirring for 4 h. Cool in iced-water; a gel may formo Adapt a sodium salt R in sulfuric acid R so that the fumes reach the
cooling jacket filled with iced water to a sintered -glass filter solution. Shake the 2nd tube for about 10 S and heat on a
(16) (2.1.2) fitted with a device al!owing pressure to be applied water-bath for 5 mino The solutiol1 is violet.
during filtration. AlIow the filter to cool for 15 mino Filter D. In a platinum crucible, prepare the sulfated ash (2.4.14)
the hexane solution applying a gauge pressure of 27 kPa and using 50 mg. The white powder obtained gives the reaction
without washing the residue; the filtration time must not of silicates (2.3.1).
exceed 5 mino Evaporate 20 mL of the solution to dryness on a
water-bath. Dryat 100 oC for 1 h. The mass of the residue is TESTS
not greater than 40 mg (2 per cent) if copolymer is used for Acidity. To 2.0 g add 25 mL of a mixture of equal volumes
containers and not greater than 0.1 g (5 per cent) if copolymer of anhydrous ethanol R and ether R, previously neutralised
is used for tubing. to 0.2 mL of bromothymol blue solution R1, and shake. Not
more than 0.15 mL of 0.01 M sodium hydroxide is required to
Sulfated ash (2.4.14): maximum 1.2 per cent, determined on
change the colour of the soIution to bIue.
5.0 g.
Viscosity (2.2.10). Determine the dynamic viscosity at 25 oc.
ASSAY Calculate the kinematic viscosity taking the relative density to
Introduce 0.250 g to 1.000 g of the substance to be examined, be 0.97. The kinematic viscosity is within the range 95 per cent
according to the vinyl acetate content of the copolymer to be to 105 per cent of the nominal viscosity stated on the label.
examined, into a 300 mL conical flask with a ground-glass Mineraloils. Place 2 mL in a test-tube and examine in
neck containing a magnetic stirrer. Add 40 mL of xylene R. ultraviolet light at 365 nm. The fluorescence is not more
Boíl under a reflux condenser with stirring for 4 h. Stirring intense than that of a solution containing 0.1 ppm of quinine
continuously, allow to cool until precipitation begins before sulfate R in 0.005 M sulfuric acid examined in the same
slowly adding 25.0 mL of alcoho/ic potassium hydroxide conditions.
so/ution R1. Boíl again under a reflux condenser with stirring
Phenylated compounds. The refractive index (2.2.6) is not
for 3 h. Allow to cool with continued stirring, rinse the
greater than 1.410.
condenser with 50 mL ofwater R and add 30.0 mL of 0.05 M
sulfuric acid to the flask. Transfer the contents of the flask into Heavy metals: maximum 5 ppm.
a 400 mL beaker; rinse the flask with two quantities, each of Solvent mixture: dilute ammonia R2, 2 giL solution of
50 mL, of a 200 giL solution of anhydrous sodium sulfate R hydroxylamine hydrochloride R (1:9 V/V).
and three quantities, each of 20 mL, of water R and add al! Mix 1.0 g with methylene chloride R and dilute to 20 mL with
the rinsings to the beaker containing the initial solution. the same solvent. Add 1.0 mL of a freshly prepared 0.02 giL
Titrate the excess sulfuric acid with 0.1 M sodium hydroxide, solution of dithizone R in methylene ch/oride R, 0.5 mL of
determining the end-point potentiometrically (2.2.20). Carry water R and 0.5 mL of the solvent mixture. At the same
out a blank titration. time, prepare the reference solution as follows: to 20 mL of
1 mL of 0.05 M sulfuric acid is equivalent to 8.609 mg of vinyl methylene chloride R add 1.0 mL of a freshly prepared 0.02 giL
acetate. solution of dithizone R in methylene chloride R, 0.5 mL of lead

General Notices (1) apply to all monographs and other texts 393
3.1.9. Silicone elastomer for do sures and tubing EUROPEAN PHARMACOPOEIA 8.0

standard solution (10 ppm Pb) R and 0.5 mL ofthe solvent Solution S. Place 25 g in a borosilicate-glass flask with a
mixture. Immediately shake each solution vigorously for ground-glass neck. Add 500 mL of water R and boil under a
1 mino Any red colour in the test solution is not more intense reflux condenser for 5 h. Allow to cool and decant the solution.
than that in the reference solution. Appearance of solution. Solution S is clear (2.2.1).
Volatile matter: maximum 2.0 per cent, determined on 2.00 g Acidity or alkalinity. To 100 mL of solution S add 0.15 mL
by heating in an oven at 150 oC for 24 h. Carry out the test of bromothymol blue solution R1. Not more than 2.5 mL of
using a dish 60 mm in diameter and 10 mm deep. 0.01 M sodium hydroxide is required to change the colour of
LABELLING the indicator to blue. To a further 100 mL of solution S, add
0.2 mL of methyl orange solution R. Not more than 1.0 mL of
The label states:
0.01 M hydrochloric acid is required to reach the beginning of
- the nominal viscosity by a number placed after the name of the colour change of the indicator from yellow to orange.
the product;
Relative density (2.2.5): 1.05 to 1.25, determined using a
- that the contents are to be used as a lubricant.
density bottle with anhydrous ethanol R as the immersion
liquido
01/2008:30109
Reducing substances. To 20 mL of solution S add 1 mL
of dilute sulfuric acid R and 20 mL of 0.002 M potassium
3.1.9. SILICONE ELASTOMER FOR permanganate. Allow to stand for 15 mino Add 1 g of
CLOSURES AND TUBING potassium iodide R and titrate immediatelywith 0.01 M sodium
thiosulfate using 0.25 mL of starch solution R as indicator.
DEFINITION Carry out a blank titration using 20 mL of water R instead of
Silicone elastomer complying with the following requirements solution S. The difference between the titration volumes is
is suitable for the manufacture of closures and tubing. not more than 1.0 mL.
Silicone elastomer is obtained by cross-linking a linear Substances soluble in hexane: maximum 3 per cent.
polysiloxane constructed mainly of dimethylsiloxy units with
small quantities of methylvinylsiloxy groups; the chain ends Evaporate 25 mL of the solution obtained in the test for
are blocked by trimethylsiloxy or dimethylvinylsiloxy groups. phenylated compounds in a glass evaporating dish on a
water-bath and dry in an oven at 100-105 oC for 1 h. The
The general formula of the polysiloxane is: residue weighs not more than 15 mg.

H3 C,
M { O/Si
P1{3 <C~~H3
O/Si O/ M'
Phenylated compounds. Place 2.0 g in a borosilicate-glass
flask with a ground-glass neck and add 100 mL of hexane R.
Boil under a reflux condenser for 4 h. Cool, then filter rapidly
n n'
through a sintered-glass filter (16) (2.l.2). Collect the filtrate
and close the container immediately to avoid evaporation. At
H3C, PH3 wavelengths from 250 nm to 340 nm, the absorbance (2.2.25)
/Si~CH2
is not greater than 0.4.
The cross-linking is carried out in the hot state: Mineraloils. Place 2 g in a 100 mL conical flask containing
- either with: 30 mL of a mixture of 5 volumes of ammonia R and 95 volumes
- 2,4-dichlorobenzoyl peroxide for extruded products; or of pyridine R. Allow to stand for 2 h, shaking frequently.
Decant the pyridine solution and examine in ultraviolet light
- 2,4-dichlorobenzoyl peroxide or dicumyl peroxide or
at 365 nm. The fluorescence is not greater than that of a
OO-(l,l-dimethylethyl) O-isopropyl monoperoxy-
carbonate or 2,5-bis [(1, 1-dimethylethyl)dioxy]-2,5- solution containing 1 ppm of quinine sulfate R in 0.005 M
sulfuric acid examined in the same conditions.
dimethylhexane for moulded products;
- or by hydrosilylation by means of polysiloxane with -SiR Volatile matter: maximum 0.5 per cent for silicone elastomer
groups using platinum as a catalyst. prepared using peroxides; maximum 2.0 per cent for silicone
elastomer prepared using platinum.
In all cases, appropriate additives are used such as silica
and sometimes small quantities of organosilicon additives Weigh 10.0 g ofthe substance previously stored for 48 h in a
(a,w-dihydroxypolydimethylsiloxane) . desiccator over anhydrous calcium chloride R. Heat in an oven
at 200 oC for 4 h, allow tocool in,a desiccator and weigh again.
CHARACTERS Silicone elastomer prepared using peroxides complies with the
Appearance: transparent or translucent material. following additional test:
Solubility: practically insoluble in organic solvents, sorne Residual peroxides: maximum 0.08 per cent calculated as
of which, for example cyclohexane, hexane and methylene dichlorobenzoyl peroxide.
chloride, cause a reversible swelling of the material.
Place 5 g in a borosilicate-glass flask, add 150 mL of methylene
IDENTIFICATION chloride R and close the flask. Stir with a mechanical stirrer
A. Infrared absorption spectrophotometry (2.2.24) by the for 16 h. Filter rapidly, collecting the filtrate in a flask with
multiple reflection method for solids. a ground-glass neck. Replace the air in the container with
oxygen-free nitrogen R, introduce 1 mL of a 200 giL solution
Comparison: silicone elastomer CRS. of sodium iodide R in anhydrous acetic acid R, close the flask,
B. Heat 1.0 g in a test-tube over a small flame until white shake thoroughly and allow to stand protected from light for
fumes begin to appear. Invert the tube over a 2nd tube 30 mino Add 50 mL of water R and titrate immediately with
containing 1 mL of a 1 giL solution of chromotropic acid, 0.01 M sodium thiosulfate, using 0.25 mL of starch solution R
sodium salt R in sulfuric acid R so that the fumes reach the as indicator. Carry out a blank titration. The difference
solution. Shake the 2nd tube for about 10 s and heat on a between the titration volumes is not greater than 2.0 mL.
water-bath for 5 mino The solution is violeto
Silicone elastomer prepared using platinum complies with the
C. 50 mg of the residue of combustion gives the reaction of following additional test:
silicates (2.3.1).
Platinum: maximum 30 ppm.
TESTS In a quartz crucible, ignite 1.0 g of the material to be examined,
If necessary, cut the material into pieces of maximum dimension raising the temperature gradually until a white residue is
on a side of not greater than 1 cm. obtained. Transfer the residue to a graphite crucible. To the

394 See the information section on general monographs (cover pages)


EUROPEAN PHARMACOPOEIA 8.0 3.1.10. Non-plastidsed PVC materials for non-injectable solutions

quartz crucible add 10 mL of a freshly prepared mixture of Fill a 50 mL polyethylene or polypropylene syringe with
1 volume of nitric acid R and 3 volumes of hydrochloric acid R, gaseous vinyl chloride R, allow the gas to remain in contact
heat 011 a water-bath for 1-2 min al1d transfer to the graphite with the syringe fOI about 3 min, empty the syringe and fill
crucible. Add 5 mg of potassium chloride R and 5 mL of again with 50 mL of gaseous vinyl chloride R. Fit a hypodermic
hydrofluoric acid R and evaporate to dryness on a water-bath. needle to the syringe and reduce the volume of gas in the
Add 5 mL of hydrofluoric acid R and evaporate to dryness syringe from 50 mL to 25 mL. Inject these 25 mL of vinyl
again; repeat this operation twice. Dissolve the residue in chloride slowly into the vial, shaking gently and avoiding
5 mL of 1 M hydrochloric acid, warming on a water-bath. contact between the liquid and the needle. Weigh the vial
Allow to cool and add the solution to 1 mL of a 250 giL again; the increase in mass is about 60 mg (1 [1L of the solution
solution of stannous chloride R in 1 M hydrochloric acid, rinse thus obtained contains about 1.2 ~lg of vinyl chloride). AlIow
the graphite crucible with a few millilitres of 1 M hydrochloric to stand for 2 h. Keep the primary solution in a refrigerator.
acid and dilute to 10.0 mL with the same acid. Vinyl chloride standard solutiol1: vinyl chloride primary
Prepare simultaneously the reference solution as follows: solution, dimethylacetamide R 0:3 V/V).
to 1 mL of a 250 giL solution of stannous chloride R in 1 M Reference solutions. Place 10.0 mL of the internal standard
hydrochloric acid, add 1.0 mL of platinum standard solution solution in each of six 50 mL vials. Close the vials and
(30 ppm Pt) R and dilute to 10.0 mL with 1 M hydrochloric secure the stoppers. Inject 1 [1L,2 ¡.tL, 3 ¡.tL. 5 ¡.tL and 10 flL,
acid. respectively, of the vinyl chloride standard solution into 5 of
The colour of the test solution is not more intense than that of the vials. The 6 solutions thus obtained contain respectively,
the standard. O ¡.tg, about 0.3 ¡.tg, 0.6 ¡.tg, 0.9 flg, 1.5 ¡.tg and 3 ¡.tg of vinyl
chloride. Shake, avoiding contact between the stopper and the
LABELLING liquido Place the vials in a water-bath at 60 ± 1 oC Íor 2 h.
The label states whether the material was prepared using
Column:
peroxides or platinum.
- material: stainless steel;
0112008:30110 - size: 1 = 3 m, 0 = 3 mm;
corrected 7.5 - stationary phase: silanised diatomaceous earth for gas
chromatography R impregnated with 5 per cent m/m oí
3.1.10. MATERIALS BASED ON dimethylstearamide R al1d 5 per cent m/m of macrogol400 R.
NON-PLASTICISED POLY(VINYL Carder gas: nitrogen for chromatography R.
CHLORIDE) FOR CONTAINERS Flow rate: 30 mL/min.
Temperature:
FOR NON -INJECTABLE, AQUEOUS
- column: 45 oC;
SOLUTIONS - injection port: 100 oC;
DEFINITION - detector: 150 oc.
Materials based on non-plasticised poly(vinyl chloride) that Detectiol1: flame ionisatiol1.
comply with the following specifications are suitable for Injection: 1 mL of the head space.
the manufacture of containers for non-injectable aqueous
solutions. They may also be used for solid forms for oral Limit:
administration and in some cases, subject to special studies - vinyl chloride: maximum 1 ppm.
on the compatibility of the container with its contents, these Additives
materials may be suitable Íor the preparation of containers
In order to obtain the required mechanical and stability
for suppositories. They consist of 1 or more poly(vinyl
characteristics, material s based on non-plasticised poly(vinyl
chloride/vinyl acetate) or of a mixture oí poly(vinyl chloride)
chloride) may contain:
and poly(vinyl acetate) or of poly(vinyl chloride).
- epoxidised soya oil of which the oxiran oxygen content is
The chlorine content expressed as poly(vinyl chloride) is not
6 per cent to 8 per cent and the iodine value is not greater
less than 80 per cent.
than 6: maximum 8 per cent;
They may contain not more than 15 per cent of copolymers
based on acrylic and/or methacrylic acids and/or their esters, - calcium salt or zinc salts of aliphatic fatty acids with more
and/or on styrene and/or butadiene. than 7 carbon atoms: maximum 1.5 per cent or maximum
1.5 per cent of their mixture;
PRODUCTION - liquid paraffin: maximum 1.5 per cent;
Materials based on non-plasticised poly(vinyl chloride) are - waxes: maximum 1.5 per cent;
produced by polymerisation methods that guarantee a residual
- hydrogenated oils or esters of aliphatic fatty acids:
vinyl chloride content of less than 1 ppm. The manufacturing
maximum 2 per cent;
process is validated to demonstrate that the product complies
with the following test. - macro gol esters: maximum 1.5 per cent;
Vinyl chloride. Head-space gas chromatography (2.2.28). - sorbitol: maximum 1.5 per cent;
Internal standard solution. Using a microsyringe, inject - 2,4-dinonylphenyl phosphite, or di(4-nonylphenyl)
10 [1L of ether R into 20.0 mL of dimethylacetamide R, phosphite or tris(nonylphenyl) phosphite: maximum 1 per
immersing the tip of the needle in the solvent. Immediately cent.
before use, dilute the solution to 1000 times its volume with They may contain one oí the following groups of stabilisers:
dimethylacetamide R. - tin as di(isooctyl) 2,2'-[(dioctylstannylene)bis(thio)]-
Test solution. Place 1.000 g oí the material to be examined in a diacetate containing about 27 per cent of tri(isooctyl)
50 mL vial and add 10.0 mL of the internal standard solution. 2,2',2" -[(monooctylstannylidyne )tris( thio)] triacetate:
Close the vial and secure the stopper. Shake, avoiding contact maximum 0.25 per cent;
between the stopper and the liquido Place the vial in a - tin as a mixture containing not more than 76 per cent of
water-bath at 60 ± 1 oC for 2 h. di(isooctyl) 2,2'- [( dimethylstannylene )bis( thio)] diacetate
Vinyl chloride primary solution. Prepare in a fume cupboard. and not more than 85 per cent of tri(isooctyl)
Place 50.0 mL of dimethylacetamide R in a 50 mL vial, stopper 2,2',2" -[(monomethylstannylidyne)tris(thio) ]triacetate;
the vial, secure the stopper and weigh to the nearest O.lmg. (isooctyl is e.g. 2-ethylhexyl): maximum 0.25 per cent;

General Notices (1) apply to all monographs and other texts 395
3.1.10. Non-plasticised PVC materials for non-injectable solutions EUROPEAN PHARMACOPOEIA 8.0

- 1- phenyleicosane-1,3 -dione (benzoylstearoylmethane) Rejerence so/ution. A solution containing 0.1 ppm of


or 2-( 4-dodecylphenyl)indole or didodecyl barium prepared by dilution of barium standard so/ution
1, 4-dihydropyridine-2,6 -dimethyl-3,5 -dicarboxylate : (50 ppm Ba) R with 0.1 M hydrochloric acid.
maximum 1 per cent or 1 per cent of a mixture of two of Wavelength: use the emission of barium at 455.40 nm, the
these. spectral background being taken at 455.30 nm.
They may contain a colorant or pigment and may be opacified Verify the absence of barium in the hydrochloric acid used.
by titanium dioxide.
Examined at 455.40 nm, the emission of the test solution is
The supplier of the material must be able to demonstrate 110t greater than that of the reference solution.
that the qualitative and quantitative composition of the type
sample is satisfactory for each production batch. Extractable cadmium: maximum 0.6 ppm.
Atomic absorption spectrometry (2.2.23, Method J).
CHARACTERS Test so/ution. Solution S3.
Appearance: powder, beads, granules, sheets of varying Rejerence solution. A solution containing 0.03 ppm of
thicknesses or samples taken from finished objects. cadmium prepared by diluting cadmium standard so/ution
Solubi/ity: insoluble in water, soluble in tetrahydrofuran, (0.1 per cent Cd) R with 0.1 M hydroch/oric acid.
slightly soluble in methylene chloride, insoluble in anhydrous Verify the absence of cadmium in the hydrochloric acid used.
ethanol.
Examined at 228.8 11m, the absorbance of the test solution is
They bum with an orange-yellow flame edged with green, not greater than that of the reference solution.
giving off thick black smoke.
Tin-stabilised materials: maximum 0.25 per cent of S11.
IDENTIFICATION Tin stock so/ution. Dilute 81 mg of plastic additive 23 CRS to
Infrared absorption spectrophotometry (2.2.24). 100.0 mL with tetrahydrojuran R.
Preparation. Dissolve the residue A (see Tests: solution S2) in Tin standard solution. Dilute 20 mL of the tin stock solution
5 mL of tetrahydrojuran R. Apply a few drops of the solution to 100.0 mL with ethano/ (96 per cent) R.
to a sodium chloride plate and evaporate to dryness in an To 0.10 mL of solution S2 in a test tube add 0.05 mL of 1 M
oven at 100-105 oc. hydroch/oric acid, 0.5 mL of potassium iodide solution R and
Absorption maxima: at 2975 cm- 1, 2910 cm- \ 2865 cm - 1, 5 mL of ethanol (96 per cent) R. Mix thoroughly and wait for
1430 cm- l , 1330 cm- l , 1255 cm- l , 690 cm- l and 615 cm- L. 5 mino Add 9 mL of water R and 0.1 mL of a 5 giL solution of
sodium su/jite R and mix thoroughly. Add 1.5 mL of dithizone
The spectrum obtained is identical to that of the material so/ution R freshly diluted 100-fold with methylene ch/oride R,
selected for the type sample. shake for 15 s and allow to stand for 2 mino At the same time
prepare a reference solution in the same manner using 0.1 mL
TESTS
of the tin standard solution.
Jj necessary, cut the samples oj the materia/ to be examined
Any violet colour in the lower layer obtained with solution S2
into pieces with a maximum dimension on a side oj not greater
is not more intense than that obtained with the reference
than 1 cm.
solution. The greenish-blue colour of dithizone solution tums
Solution SI. Place 25 g in a borosilicate-glass flask. Add pink in the presence of tino
500 mL of water R and cover the neck of the flask with
Non-Hn stabilised materials: maximum 25 ppm of Sn.
aluminium foil or a borosilicate-glass beaker. Heat in an
autoclave for 121 ± 2 oC for 20 mino Allow to cool and allow To 5 mL of solution S2 in a test tube add 0.05 mL of 1 M
the solids to settle. hydrochloric acid and 0.5 mL of potassium iodide solution R.
Mix thoroughly and wait for 5 mino Add 9 mL of water R
Soll.ltion S2. Dissolve 5.0 g in 80 mL of tetrahydrojuran R and 0.1 mL of a 5 giL solution of sodium sulfite R and mix
and dilute to 100 mL with the same solvento Filter if necessary thoroughly. If the solution obtained is 110t colourless, add the
(the solution may remain opalescent). To 20 mL ofthe sodium sulfite solution in 0.05 mL fractions. Add 1.5 mL of
solution add, dropwise and with gentle shaking, 70 mL of dithizone so/ution R freshly diluted 100-fold with methylene
ethano/ (96 per cent) R. Cool in ice for 1 h. Filter or centrifuge chloride R, shake for 15 s and allow to stand for 2 mino At the
(residue A). Wash the residue A with ethanol (96 per cent) R, same time prepare a reference solution in the same manner
add the washings to the filtrate or the centrifugation liquid using 0.05 mL of the tin standard solution (see test aboye).
and, dilute to 100 mL with ethanol (96 per cent) R.
Any violet colour in the lower layer obtained with solution S2
Solution S3. Place 5 g in a borosilicate-glass flask with a is not more intense than that obtained with the reference
ground-glass neck. Add 100 mL of 0.1 M hydrochloric acid solution.
and boil under a reflux condenser for 1 h. Allow to cool and
allow the solids to settle. Extractable heavy metals (2.4.8): maximum 20 ppm.
Appearance of solution SI. Solution SI is not more 12 mL of solution S3 complies with test A. Prepare the
opalescent than reference suspension II (2.2.1) and is reference solution using 10 mL of /ead standard solution
colourless (2.2.2, Method JI). (1 ppm Pb) R.

Absorbance of solution SI (2.2.25). Evaporate 100 mL Extractable zinc: maximum 100 ppm.
of solution SI to dryness. Dissolve the residue in 5 mL of Atomic absorption spectrometry (2.2.23, Method I).
hexane R. Filter if necessary through a filter previously rinsed Test so/ution. Solution S3 diluted lO-fold with water R.
with hexane R. At wavelengths from 250 nm to 310 nm, the Rejerence solution. A solution containing 0.50 ppm of zinc
absorbance of the filtrate is not greater than 0.25. prepared by dilution of zinc standard solution (5 mg/mL Zn) R
Absorbance of solution 52 (2.2.25): maximum 0.2 for with 0.01 M hydroch/oric acid.
tin-stabilised materials or 0.4 for other materials determined Verify the absence of zinc in the hydrochloric acid used.
between wavelengths of 250 nm and 330 nm 011 solution S2.
Examined at 214.0 nm, the absorbance of the test solution is
Extradable barium: maximum 2 ppm. not greater than that of the reference solution.
Inductively coupled plasma-atomic emission spectrometry Sulfated ash (2.4.14): maximum 1.0 per cent, determined on
(2.2.57). 1.0 g; maximum 4.0 per cent when the materials are opacified
Test solution. 501ution 53. using titanium dioxide.

396 See the injormation section on monographs (ca ver pages)


EUROPEAN PHARMACOPOEIA 8.0 3.1.11. Non-plastidsed PVC maíerials for dry dosage forms (oral)

ASSAY Reference solutions. Place 10.0 mL of the internal standard


Carry out the oxygen-flask method (2.5.10) using 50.0 mg of solution in each of six 50 mL vials. Close the vials and
the material to be examined. Absorb the combustion products secure the stoppers. Inject 1 flL, 2 [1L, 3 [1L, 5 [1L and 10 ¡.tL,
in 20 mL of 1 M sodium hydroxide. To the solution obtained respectively, of the vinyl chloride standard solution into 5 of
add 1 mL of dibutyl phthalate R, 2.5 mL of nitric acid R, 5 mL the viaIs. The 6 solutions thus obtained contain respectively,
offerric ammonium sulfate solution R2 and 10.0 mL of 0.1 M O [1g, about 0.3 ¡.tg, 0.6 [1g, 0.9 [1g, 1.5 [1g and 3 [1g of vinyl
silver nitrate. Titrate with 0.05 M ammonium thioeyanate chloride. Shake, avoiding contact between the stopper and the
until a reddish-yellow colour is obtained. Carry out a blank liquido Place the vials in a water-bath at 60 ± 1 oC for 2 h.
titration. Column:
1 mL of 0.1 M silver nitrate is equivalent to 6.25 mg of - material: stainless steel;
poly(vinyl chloride). - size: 1 = 3 m, 0 = 3 mm;
- stationary phase: silanised diatomaceous earth for gas
chromatography R impregnated with 5 per cent m/m of
04/2009:30111 dimethylstearamide R and 5 per cent m/m of macrogol
correded 7.0 400 R.
Carrier gas: nitrogen for chromatography R.
3.1.11. MATERIALS BASED ON Flow rate: 30 mUmin.
NON-PLASTICISED POLY(VINYL Temperature:
CHLORIDE) FOR CONTAINERS FOR - column: 45 oC;
- injection port: 100 oC;
DRY DOSAGE FORMS FOR ORAL
- detector: 150 oc.
ADMINISTRATION
Deteetion: flame ionisation.
DEFINITION Injection: 1 mL of the head space.
Materials based on non-plasticised poly(vinyl chloride) for Limit:
containers for dry dosage forms for oral administration are - vinyl ehloride: maximum 1 ppm.
suitable for the manufacture of sheets OI containers, and
consist of 1 or more poly(vinyl chloride/vinyl acetate) OI of a Additives
mixture of poly(vinyl chloride) and poly(viny! acetate) or of In order to obtain the required mechanical and stability
poly(vinyl chloride). characteristics, materials based on non-plasticised poly(vinyl
The chlorine content expressed as poly(vinyl chloride) is not chloride) may contain:
less than 80 per cent. - epoxidised soya oil of which the oxiran oxygen content is
They may contain not more than 15 per cent of copolymers 6 per cent to 8 per cent and the iodine value is not greater
based on acrylic and/or methacrylic acids and/or their esters, than 6 for tin -stabilised materials: maximum 2 per cent;
and/or on styrene and/or butadiene. - epoxidised soya oil of which the oxiran oxygen content is
6 per cent to 8 per cent and the iodine value is not greater
PRODUCTION than 6 for non-tin-stabilised materials: maximum 3 per
Materials based on non-plasticised poly(vinyl chloride) are cent;
produced by polymerisation methods that guarantee a residual - calcium, magnesium or zinc salts of aliphatic fatty acids
vinyl chloride content of less than 1 ppm. The manufacturing with more than 7 carbon atoms: maximum 1.5 per cent or
process is validated to demonstrate that the product complies maximum 1.5 per cent of their mixture;
with the following test for vinyl chloride. - waxes: maximum 4 per cent;
Vinyl chloride. Head-space gas chromatography (2.2.28). - liquid paraffin: maximum 1.5 per cent;
Internal standard solution. Using a microsyringe, inject - hydrogenated oils or esters of aliphatic fatty acids:
10 ¡.tL of ether R into 20.0 mL of dimethylacetamide R, maximum 2 per cent;
immersing the tip of the needle in the solvent. Immediately
- the percentage sum of the 3 lubricants above:
before use, dilute the solution to 1000 times its volume with
maximum 4 per cent;
dimethylacetamide R.
- macrogol esters: maximum 1.5 per cent;
Test solution. Place 1.000 g of the material to be examined in a
50 mL vial and add 10.0 mL of the internal standard solution. - sorbitol: maximum 1.5 per cent;
Close the vial and secure the stopper. Shake, avoiding contact - 2,4-dinonylphenyI phosphite, or di(4-nonylphenyl)
between the stopper and the liquido Place the vial in a phosphite or tris(nonylphenyl) phosphite: maximum 1 per
water-bath at 60 ± 1 oC for 2 h. cent;
Vinyl chloride primary solution. Prepare in a fume cupboard. - calcium carbonate: maximum 1 per cent;
Place 50.0 mL of dimethylacetamide R in a 50 mL vial, - silica: maximum 1 per cent.
stopper the vial, secure the stopper and weigh to the nearest
They may contain one of the 3 following groups of stabilisers
0.1 mg. Fill a 50 mL polyethylene or polypropylene syringe
(where isooctyI is, for example, 2-ethylhexyl):
with gaseous vinyl chloride R, allow the gas to remain in
contact with the syringe for about 3 min, empty the syringe - tin as di(isooctyl) 2,2'-[(dioctylstannylene)bis(thio)]-
and fill again with 50 mL of gaseous vinyl ehloride R. Fit a diacetate containing about 27 per cent of tri(isooctyl)
hypodermic needle to the syringe and reduce the volume of 2,2'2" -[(monooctylstannylidyne )tris( thio)] triacetate:
gas in the syringe from 50 mL to 25 mL. Inject the 25 mL of maximum 0.25 per cent;
vinyl chloride slowly into the vial, shaking gently and avoiding - tin as a mixture containing not more than 76 per cent of
contact between the liquid and the needle. Weigh the vial di(isooctyl) 2,2'- [( dimethylstannylene )bis( thio)] diacetate
again; the increase in mass is about 60 mg (1 flL of the solution and not more than 85 per cent of tri(isooctyl)
thus obtained contains about 1.2 flg of vinyl chloride). Allow 2,2' ,2" - [(monomethylstannylidyne )tris( thio)] triacetate:
to stand for 2 h. Keep the primary solution in a refrigerator. maximum 0.25 per cent;
Vinyl chloride standard solution: vinyl chloride primary - 1-phenyleicosane-l ,3-dione (benzoyIstearoylmethane):
solution, dimethylacetamide R (1:3 V/V). maximum 1 per cent.

General Notices (1) apply to all monographs and other texts 397
3.1.13. Plastk additives EUROPEAN PHARMACOPOEIA 8.0

They may contain a colorant or pigment and may be opacified To 0.10 mL of solution S2 in a test tube add 0.05 mL of 1 M
by titanium dioxide. hydrochloric acid, 0.5 mL of potassium iodide solution R and
The supplier of the material must be able to demonstrate 5 mL of ethanol (96 per cent) R. Mix thoroughly and wait for
that the qualitative and quantitative composition of the type 5 mino Add 9 mL of water R and 0.1 mL of a 5 giL solution of
sample is satisfactory for each production batch. sodium sulfite R and mix thoroughly. Add 1.5 mL of dithizone
solution R freshly diluted 100-foId with methylene chloride R,
CHARACTERS shake for 15 s and allow to stand for 2 mino At the same time
prepare a reference solution in the same manner using 0.1 mL
Appearance: powder, beads, granules, sheets of varying of the tin standard solution.
thicknesses ar samples taken from finished objects.
Any violet colour in the lower layer obtained with solution S2
Solubility: insoluble in water, soluble in tetrahydrofuran, is not more intense than that obtained with the reference
slightly soluble in methylene chloride, insoluble in anhydrous
solution. The greenish-blue colour of dithizone solution turns
ethanol. pink in the presence of tino
They bum with an orange-yellow flame edged with green,
Non-tin-stabiJised materials: maximum 25 ppm of Sn.
giving off thick black smoke.
To 5 mL of solution S2 in a test tube add 0.05 mL of 1 M
IDENTIFICATION hydrochloric acid and 0.5 mL of potassium iodide solution R.
Infrared absorption spectrophotometry (2.2.24). Mix thoroughly and wait for 5 mino Add 9 mL of water R
and 0.1 mL of a 5 giL solution of sodium sulfite R and mix
Preparation. Dissolve residue A (see Tests: solution S2) in thoroughly. lf the solution obtained is not colourless, add the
5 mL of tetrahydrofuran R. Apply a few drops of the solution sodium sulfite solution in 0.05 mL fractions. Add 1.5 mL of
to a sodium chloride plate and evaporate to dryness in an dithizone solution R freshly diluted 100-fold with methylene
oven at 100-105 oc. chloride R, shake for 15 s and allow to stand for 2 mino At the
Absorption maxima: at 2975 cm- l, 2910 cm-l, 2865 cm- \ same time prepare a reference solution in the same manner
1430 cm- l, 1330 cm- \, 1255 cm- l, 690 cm- l and 615 cm-l. using 0.05 mL of the tin standard solution (see test aboye).
The spectrum obtained is identical to that of the material Any violet colour in the lower layer obtained with solution S2
selected fol' the type sample. is not more intense than that obtained with the reference
solution.
TESTS
Extractable heavy metals (2.4.8): maximum 20 ppm.
1f necessary, cut the samples of the material to be examined
12 mL of solution S3 complíes with test A. Prepare the
into pieces with a maximum dimension on a side of not greater
reference solution using 10 mL of lead standard solution
than 1 cm.
(1 ppm Pb) R.
Solution SI. Place 25 g in a borosilicate-glass flask. Add
Extractable zinc: maximum 100 ppm.
500 mL of water R and cover the neck of the flask with
aluminium foil or a borosilicate glass beaker. Heat in an Atomic absorption spectrometry (2.2.23, Method I).
autoclave for 121 ± 2 oC for 20 mino Allow to cool and allow Test solution. Solutiol1 S3 diluted 10-fold with water R.
the solids to settle.
Reference solution. A solutiol1 containing 0.50 ppm of zinc
Solution S2. Dissolve 5.0 g in 80 mL of tetrahydrofuran R prepared by dilution of zinc standard so/ution (5 mg/mL Zn) R
and dilute to 100 mL with the same solvent. Filter if necessary with 0.01 M hydrochloric acid.
(the solution may remain opalescent). To 20 mL ofthe
Verify the absence of zinc in the hydrochloric acid used.
solution add, dropwise and with gentle shaking, 70 mL of
ethanol (96 per cent) R. Cool in ice for 1 h. Filter or centrifuge Examined at 214.0 nm, the absorbance ofthe test solution is
(residue A). Wash residue A with ethanol (96 per cent) R, add not greater than that of the reference solution.
the washings to the filtrate or the centrifugation liquid and Sulfated ash (2.4.14): maximum 1.0 per cent, determined on
dilute to 100 mL with ethanol (96 per cent) R. 1.0 g; maximum 4.0 per cent when the materials are opacified
Solution 53. Place 5 g in a borosilicate-glass flask with a using titanium dioxide.
ground-glass neck. Add 100 mL of 0.1 M hydrochloric acid
and boil under a reflux condenser for 1 h. Allow to cool and ASSAY
allow the solids to settle. Carry out the oxygen-flask method (2.5.10) using 50.0 mg
Appearance of solution SI. Solution SI is not more of the material to be examined. Absorb the combustion
opalescent than reference suspension II (2.2.1) and is products in 20 mL of 1 M sodium hydroxide. To the solution
colourless (2.2.2, Method II). obtained add 2.5 mL of nitric acid R, 10.0 mL of 0.1 M si/ver
nitrate, 5 mL of ferric ammonium sulfate so/ution R2 and
Absorbance of solution SI (2.2.25). Evaporate 100 mL 1 mL of dibutyl phthalate R. Titrate with 0.05 M ammonium
of solution SI to dryness. Dissolve the residue in 5 mL of thiocyanate until a reddish-yellow colour is obtained. Carry
hexane R. Filter if necessary through a filter previously rinsed out a blank titration.
with hexane R. At wavelengths from 250 nm to 310 nm, the
absorbance of the filtrate is not greater than 0.3. 1 mL of 0.1 M si/ver nitrate is equivalent to 6.25 mg of
poly(vinyl chloride).
Absorbance of solution S2 (2.2.25): maximum 1.0,
determined between wavelengths of 250 nm and 330 nm
on solution S2 for material that do es not contain
1-phenyleicosane-1,3-dione; maximum 0.4, determined
between wavelengths of 250 nm and 330 nm on a 10-fold 0112008:30113
dilution of solution S2 in ethanol (96 per cent) R for material corrected 6.2
that contains l-phenyleicosane-1,3-dione.
Tin-stabiHsed materials: maximum 0.25 per cent ofSn. 3.1.13. PLASTIC ADDITIVES
Tin stock solution. Dilute 81 mg of plastic additive 23 CRS to NOTE: the nomenclature given first is according to the 1UPAC
100.0 mL with tetrahydrofuran R. rules. The synonym given in bold corresponds io the na me
Tin standard solution. Dilute 20 mL of the tin stock solution given in the texts of Chapter 3. The synonym corresponding to
to 100.0 mL with ethanol (96 per cent) R. the rules of the texts of "Chemical Abstracts" is also given.

398 See the information section on general monographs (cover pages)


EUROPEAN PHARMACOPOEIA 8.0 3.1.13. Plastic additives

addOl. C24H3S0.¡. [117-81-7]. PM RN 74640. ethylene bis[3,3-bis[3-(1,l-dimethylethyl)-4-


hydroxyphenyl]butanoate]
O~CH3 synonyms: - ethylene bis[3,3-bis[3-(l,1-dimethylethyl)-
~O CH 3 4-hydroxyphenyl] butano ate ],

~~CH3
- butanoic acid, 3,3-bis[3-(l,1-dimethylethyl)-
4-hydroxyphenyl]-, 1,2-ethanediyl ester,
- ethylene bis [3,3-bis(3-tert-butyl-4-
(2RS)-2-ethylhexyl benzene-1 ,2-dicarboxylate hydroxyphenyl) butyrate].
synonyms: - di(2-ethylhexyl) phthalate,
add09. C 73 H lO sÜ12' [6683-19-8]. PM RN 71680.
- 1,2-benzenedicarboxylic acid,
bis(2-ethylhexyl) estero o

\O~OR
add02. C16H3uÜ4Zn. [136-53-8]. PM RN 54120.
o o R"

H3C~O,zn'O~CH3 RO~
OR

CH 3 CH 3
methanetetryltetramethyl tetrakis[3- [3,5-bis( 1,1-
zinc (2RS)-2-ethylhexanoate
dimethylethyl) -4-hydroxyphenyl] propanoate]
synonyms: - zinc octano ate,
synonyms: - pentaerythrityl tetrakis [3- (3,5- di- tert-
- 2-ethylhexanoic acid, zinc salt (2:1), butyl-4-hydroxyphenyl)propionate],
- zinc 2-ethylcaproate. - 2,2-bis[ [[3- [3,5-bis(1, l-dimethylethyl)-4-
hydroxyphenyl] propanoyl] oxy] methyl]propane-
add03. [05518-18-3]/[00110-30-5]. PM RN 53440/53520. l,3-diyl 3- [3,5-bis( l,l-dimethylethyl)-4-
o hydroxyphenyl]propanoate,
~M~ 1, ,CH 3 - benzenepropanoic acid, 3,5-bis(l,1-dimethyl-
~Cl Jn~ ~/ ~ ethyl)-4-hydroxy-2,2-bis(hydroxymethyl)-
o propane-l,3-diol ester (4: 1),
N,N'-ethylenedialcanamide (with n and m '" 14 or 16) - 2,2-bis(hydroxymethyl)propane-l,3-diol
synonyms: - N,N'-diacylethylenediamines, tetrakis [3-( 3,5-di - tert- butyl-4-hydroxyphenyl)-
propionate ].
- N,N' -diacylethylenediamine (in this context
acyl means in particular palmitoyl and stearoyl). addlO. C s4 H 7sÜ3' [1709-70-2]. PM RN 95200.
add04. [8013-07-8]. PM RN 88640.
epoxidised soya oi!
add05. [8016-11-3]. PM RN 64240.
R-
epoxidised linseed oil
add06. [57455-37 -5](TSCA)/[101357-30-6]
(EINECS)/Pigment blue 29 (Cl 77007)
ultramarine blue 4,4',4" -[( 2,4,6-trimethylbenzene-l ,3,5-triyl)tris(methylene)]-
add07. C 1s H 2p. [128-37-0] PM RN 46640. tris [2,6-bis( 1, l-dimethylethyl)phenol]
synonyms: _ 2,2',2",6,6',6"-hexa-tert-butyl-
4,4',4"- [(2,4,6-trimethyl-l,3,5-
benzenetriyl) trismethylene] triphenol,
CH 3
- 1,3,5-tris[3,5-di-tert-butyl-4-hydroxybenzyl]-
H3 C CH 3 2,4,6-trimethylbenzene,
CH 3 - phenol, 4,4',4"- [(2,4,6-trimethyl-1,3,5-
2,6-bis( 1, 1-dimethylethyl)-4-methylphenol benzenetriyl)tris(methylene) ]tris[2,6-bis(l, 1-
dimethylethyl) -.
synonyms: - butylhydroxytoluene,
addll. C 35 H 6P3' [2082-79-3]. PM RN 68320.
- 2,6-bis( 1, l-dimethylethyl)-4-methylphenol,
- 2,6-di-tert-butyl-4-methylphenol.

add08. C SO H 6 PS' [32509-66-3]. PM RN 53670.

octadecyl 3- [3,5-bis(l,1-dimethylethyl)-4-
hydroxyphenyl]propanoate
synonyms: - octadecyl 3-(3,5-di-tert-butyl-4-
hyrlroxyphenyl)propionate,
- propanoic acid, 3- [3,5-bis(l, l-dimethylethyl)-
4-hydroxyphenyl]-, octadecyl estero

General Natíces (1) apply ta all managraphs and ather texts 399
3.1.13. Plastic additives EUROPEAN PHARMACOPOEIA 8.0

add12. C42H6P3P [31570-04-4]. PM RN 74240. add17. C4zHsP4S. [693-36-7]. PM RN 93280.


H3C CH 3 H3C CH 3 o o
~ ~ S ~ o /'-l[-J16
CH3 H3C~CH3 H3C --r.
[-J'6 o
J-CH3

H3 C
H 3C I
~ oN
I
dioctadecyl 3,3' -sulfanediyldipropanoate
synonyms: - dioctadecyl 3,3'-thiodipropionate,
- dioctadecyl 3,3' -sulfanediyldipropanoate,
H3C CH 3 I CH 3
- propanoic acid, 3,3'-thiobis-, octadecyl diester,
/
- stearyl thiodipropionate.
CH 3
H3C CH 3
add18. [119345-01-6]. PM RN 92560.
tris[2,4-bis( l,l-dimethylethyl)phenyl] phosphite mixture of seven products corresponding to reaction
product of di-tert-butyl phosphonite with biphosphorous
synonyms: - tris(2,4-di-tel"t-butylphenyl) phosphite,
trichloride, reaction products with biphenyl and
- pheno!, 2,4-bis( l,l-dimethylethyl)-, 2,4-bis( 1, l-dimethylethyl)phenol:
phosphite (3:1),
- 2,4-bis(l,1-dimethylethyl)phenyl, phosphite.

add13. C 4s H 69 NP6' [27676-62-6]. PM RN 95360.


R/ = H3 C

H 3C
M eH 3 H3C
CH 3

CH 3

component I
RO,~pR
R~ R/ '--/. . \=.1 \lR
2,4-bis(l, 1-dimethylethyl)phenyl biphenyl-4,4'-
diyldiphosphonite
1,3,5-tris [3,5-bis( 1, 1-dimethylethyl)-4-hydroxybenzyl]-1,3,5- component JI
triazine-2,4,6( 1H,3 H,5 H) -trione RO,
P-OR
synonyms: - 1,3,5-tris(3,5-di-tert-butyl-4-
hydroxybenzyl) -5- triazine-2,4,6( lH,3H,5H)- R\ ;=\ rl
trione, RÓ~
- 1,3,5-triazine-2,4,6( 1H,3H,5H)-trione, 2,4-bis( 1, l-dimethylethyl)phenyl biphenyl- 3,4'-
1,3,5-tris [[3,5-bis(1,1-dimethylethyl)-4- diyldiphosphonite
hydroxyphenyl] methyl]-.
component JII
add14. C41HsP6P2' [3806-34-6]. PM RN 50080. RO,
P-OR

RO-P\
p-d
OR
3,9-bis( octadecyloxy) -2,4,8,10-tetraoxa -3,9-diphospha-
spiro[5.5]undecane 2,4-bis(l, J -dimethylethyl)phenyl biphenyl-3,3'-
diyldiphosphonite
synonyms: - 2,2'-bis( octadecyloxy) -5,5'-spirobi[ 1,3,2-
dioxaphosphinane] , component IV

- 2,4,8,1 0-tetraoxa-3,9-diphosphaspiro [5.5]un- ~pR


decane, 3,9-bis( octadecyloxy)-. ~\R
add15. C36H74S2. [2500-88-1]. PM RN 49840. 2,4-bis( 1, 1-dimethylethyl)phenyl biphenyl-4-ylphosphonite
component V
S
I OR
S /
RO-P\
1,1' -disulfanediyldioctadecane OR

synonyms: - dioctadecyl disulfide, 2,4-bisO, l-dimethylethyl)phenyl phosphite


component VI
- octadecane, 1,1'-dithio-.
RO\~pR
add16. C30 H sgÜ,¡S. [123-28-4]. PM RN 93120. R/ '--/-\J~"OR
o o
2,4-bisO,1-dimethylethyl)phenyl 4'- [bis[2,4-bis(1, 1-
H3C --r.
[-JlO
~ o ~ S ~ o /'-l[-JlO
J-CH3
dimethylethyl)phenoxy]phosphanyl]biphenyJ-4-
ylphosphonate
didodecyl 3,3' -sulfanediyldipropanoate
component VJI
synonyms: - didodecyI3,3'-thiodipropionate, R-OH: 2,4-bis(l, l-dimethylethyl)phenol
- didodecyl 3,3' -sulfanediyldipropanoate, addl9. C¡SH 36 0 Z' [57-11-4]. PM RN 24550.
- propanoic acid, 3,3' -thiobis-, dodecyl diester,
H3 CN C0 2H
8
- lauryl thiodipropionate. octadecanoic acid

400 See the information section on general monographs (cover pages)


EUROPEAN PHARMACOPOEIA 8.0 3.1.14. Plasticised PVC maíerials for iníravenous solutions

synonyms: - steark acid, 0112008:30114


corrected 7.5
- octadecanoic acid.

add20. C 1s H 3S NO. [301-02-0]. PM RN 68960. 3.1.14. MATERIALS BASED ON


o PLASTICISED POLY(VINYL
H3Cr ,~r,ti CHLORIDE) FOR CONTAINERS
M7 H "NH 2

(9Z)-octadec-9-enamide POR AQUEOUS SOLUTIONS POR


synonyms: - oleamide, INTRAVENOUS INFUSION
- 9-octadecenamide, (Z)-, DEFINITION
- 9-cis-oleamide. Materials based on plasticised poly( vinyl chloride) contain
not less than 55 per cent of poly(vinyl chloride) and contain
add21. C 22 H 43 NO. [112-84-5]. PM RN 52720. various additives, in addition to the high-molecular-mass
polymer obtained by polymerisation of vinyl chloride.
o
Materials based 011 plasticised poly(vinyl chloride) for
H3C~NH2 container s for aqueous solutions for intravenous infusion are
( 13Z)-docos-13-enamide defined by the nature and the proportions of the substances
used in their manufacture.
5ynonyms: - erucamide,
PRODUCTION
- 13-docosenamide, (Z)-,
Materials based on plasticised poly( vinyl chloride) are
- 13-cis-docosenamide. produced by polymerisation methods which guarantee a
residual vinyl chloride cOlltent of less than 1 ppm. The
add22. [65447-77-0]. PM RN 60800. production method used is validated in order to demonstrate
that the product complies with the following test.
Vinyl chloride. Head space gas chromatography (2.2.28).
Internal standard solution. Using a microsyringe, inject
10 f1L of ether R into 20.0 mL of dimethylacetamide R,
immersing the tip of the needle in the solvent. Immediately
copolymer of dimethyl butanedioate and 1-(2-hydroxyethyl)- before use, dilute the solution to 1000 times its volume with
2,2,6,6-tetramethylpiperidin -4-01 dimethylacetamide R.
synonyms: - copolymer of dimethyl succinate and Test solution. Place 1.000 g of the material to be examined in a
(4- hydroxy- 2,2,6,6- tetramethylpiperidin -1- 50 mL vial and add 10.0 mL of the internal standard solution.
yl)ethanoL Close the vial and secure the stopper. Shake, avoiding contact
between the stopper and the liquido Place the vial in a
add23. water-bath at 60 ± 1 oC for 2 h.
mixture of component 1 and about 27 per cent of component II Vinyl chloride primary solution. Prepare in a fume cupboard.
Place 50.0 mL of dimethylacetamide R in a 50 mL vial, stopper
component I [26401-97-8] the vial, secure the stopper and weigh to the nearest 0.1 mg.
Fill a 50 mL polyethylene 01' polypropylene syringe with
gaseous vinyl chloride R, allow the gas to remain in contact

e- o-8'yo~~", with the syringe for about 3 min, empty the syringe and fill
again with 50 mL of gaseous vinyl chloride R. Fit a hypodermic
needle to the syringe and reduce the volume of gas in the
o
syringe from 50 mL to 25 mL. Inject the remaining 25 mL of
bis [( 2RS) - 2-ethylhexyl] 2,2'- [( dioctylstannanetriyl) bis- vinyl chloride slowly into the vial shaking gently and avoiding
(sulfanediyl)] diacetate contact between the liquid and the needle. Weigh the vial
again; the increase in mass is about 60 mg (1 f1L of the solution
5ynonym5: - di(isooctyl) 2,2'- [( dioctylstannylene)- thus obtained contains about 1.2 f1g of vinyl chloride). Allow
bis(thio) ]diacetate, to stand for 2 h. Keep the primary solution in a refrigerator.
Vinyl chloride standard solution: vinyl chloride primary
- bis(isooctyloxycarbonyJmethylthio)- solution, dimethylacetamide R (1:3 VIV).
dioctylstannane.
Reference solutions. Place 10.0 mL of the internal standard
component JI [26401-86-5] solution in each of six 50 mL vials. Close the vials and
secure the stoppers. Inject 1 I1L, 2 f1L, 3 f-lL, 5 f1L and 10 f1L,
respectively, of the vinyl chloride standard solution into 5 of
the vials. The 6 solutions thus obtained contain, respectively,
O f1g, about 0.3 I1g, 0.6 f1g, 0.9 f1g, 1.5 f1g and 3 f1g of vinyl
chloride. Shake, avoiding contact between the stopper and the
liquido Place the vials in a water-bath at 60 ± 1 oC for 2 h.
Column:
tris[ (2RS)-2-ethylhexyl] 2,2',2"- [( octylstannanetriyl)tris-
- material: stainless 5teel;
(sulfanediyl)] triacetate
- size: 1 = 3 m, 0 = 3 mm;
syl1ol1yms: - tri(isooctyl) 2,2',2"- [(monooctyl- - stationary phase: silanised diatomaceous earth for gas
stannylidyne )tris( thio)] triacetate, chromatography R impregnated with 5 per cent mlm of
dimethylstearamide R and 5 per cent mlm of macrogol400 R.
- 2,2',2" [(octylstanl1ylidyne)tris(thio)]tri- Carrier gas: nitrogen for chromatography R.
acetic acid, triisooctyl ester.
Flow rate: 30 mL/min.

General Notices (1) apply to all monographs and other texts 401
3.1.14. Plasticised PVC materials for intravenous solutions EUROPEAN PHARMACOPOEIA 8,0

Temperature: milligra:TIS of precipitate B3 in 1 mL of tetrahydrofuran R,


- calumn: 45 oC; place a íew drops of the solution obtained on a sodium
- injection port: 100 oC; chloride plate and evaporate to dryness in an oven at
100-105 oc,
- detector: 150 oc,
eh/oride) CRS,
Detection: flame ionisation,
B. Infrared absorption spectrophotometry (2224),
Injection: 1 mL of the head-space,
Examine the residue C obtained in the test for plastic
Limit: additives 01, 04 and 05,
- vinyl chloride: maximum 1 ppm,
Comparisol1: plastic additive 01 CRS,
Additives
A certain number of additives is added to the polymers to TESTS
optimise their chemical, physical and mechanical properties in 1f necessary, before use, cut the samples of the material to be
order to adapt them for the intended use, Al! these additives examined inío pieces of maximum dimension on a side of not
are chosen from the following list which specifies for each greater than 1 cm,
product the maximum allowable content: Solution SI. Place 5,0 g in a combustion flask Add 30 mL
- di(2-ethylhexyl)phthalate (plastic additive Ol): maximum of sulfuric acid R and heat until a black, syrupy mas s is
40 per cent; obtained, Cool and add carefully 10 mL of strong hydrogen
- zinc octanoate (zinc 2-ethylhexanoate) (plastic additive 02): peroxide solution R Heat gently, Allow to cool and add 1 mL
maximum 1 per cent; of strong hydrogen peroxide solution R; repeat by alternating
evaporation and addition of strong hydrogen peroxide solution
- calcium stearate or zinc stearate: maximum 1 per cent or
until a colourless liquid is obtained, Reduce the volume to
1 per cent of a mixture of the two;
about 10 mL Coo! and dilute to 50,0 mL with water R.
- N,N' -diacylethylenediamines (plastic additive 03):
maximum 1 per cent; SalutXon S2, Place 25 g in a borosilicate-glass flask Add
500 mL of water for injections R and cover the neck of the flask
- maximum 10 per cent of one of the following epoxidised
with aluminium foil 01' a borosilicate-glass beaker, Heat in an
oils or 10 per cent of a mixture of the two:
autoclave at 121 ± 2 oC for 20 min, Allow to cool and decant
- epoxidised soya oil (plastic additive 04) of which the the solutiol1,
oxiran oxygen content is 6 per cent to 8 per cent and the
iodine value is not greater than 6; Appearance of salutian S2, 501ution S2 is clear (22,1) and
colourless (2,22, Method II),
- epoxidised linseed oil (plastic additive 05) of which the
oxiran oxygen content is not greater than 10 per cent and Addity ar alkalinity, To 100 mL of solution 52, add 0,15 mL
the iodine value is not greater than 7, of BRP indicator solution R. Not more than 15 mL of 0,01 M
sodium hydroxide is required to change the colour of the
When colouring materials are added, ultramarine blue is
us~d, Other inorganic pigments may be added, provided the
indicator to bIue, To 100 mL of solution S2 add 0.2 mL of
methyl orange solution R Not more than LO mL of 0,01 M
satety of the material is demonstrated to the satisfaction of
hydrochloric acid is required to initiate the colour change of
the competent authority, Very low amounts of antioxidants
added to the vinyl chloride monomer used may be detected in the mdlcator trom yellow to orange,
the polymer, Absorbance Evaporate 100,0 rnL of solution S2 to
The supplier of the material must be able to demonstrate dryness, Dissolve the residue in 5,0 mL of hexane R From
that the qualitative and quantitative composition of the type 250 nm to 310 11m the absorbance is not greater than 0,25,
sample is satisfactory for each production batch, Redudng substances, out the test within 4 h of
preparatíon of solution 52. 20,0 mL of solution S2 add 1 mL
CHARACTERS of dilute sulfuric acid R and 20,0 mL of 0,002 M potassium
Colourless or pale yellow material in the form of powder, permanganate, Boil under a reflux condenser for 3 min and
beads, granules Of, after transformation, translucent sheets of cool immediately Add 1 g of potassium iodide R and titrate
varying thicknesses, with a slight odour, On combustion it immediately with 0,01 M sodium thiosulfate, using 0.25 mL of
gives off dense, black smoke, starch solution R as indicataL Carry out a blank titration using
20 mL of water for injections R. The difference between the
IDENTIFICATION
titration volumes is not more than 2,0 mL
If necessary,before use, cut the samples of the material to be
examined into pieces of maximum dimension on a side of not Primary aromatk amines: maximum 20 ppm,
greater than 1 cm, To 2,5 mL of solution Al obtained during the identification,
To 2,0 g of the material to be examined add 200 mL of add 6 mL of water R and 4 mL of 0,1 M hydrochloríc acid,
peroxide-free ether R and heat under a reflux condenser for Shake vigorously and discard the upper layer. To the lower
8 h, Separate the residue B and the solution A by filtration, layer add 004 mL of a freshly prepared 10 giL solution of
sodium nitrite R. Mix and allow to stand for 1 min, Add
Evaporate solution A to dryness under reduced pressure in a
0;8 mL of a 25 giL solution of ammonium sulfamate R,
water-bath at 30 oc, Dissolve the residue in 10 mL of toluene R
allow to stand for 1 min and add 2 mL of a 5 giL solution of
(solution Al), Dissolve the residue B in 60 mL of ethylene
naphthylethylenediamine dihydrochloride R After 30 min,
chloride R, heating 011 a water-bath under a reflux condenser.
any colour in the solution is 110t more intense than that in a
FilteL Add the obtained solution dropwise and with vigorous
standard prepared at the same time in the same manner using
shakll1g to 600 mL of heptane R heated almost to boiling,
a mIxture of 1 mL of a 0,01 giL solution of naphthylamine R
Separate by hot filtration the coagulum B 1 and the organic
in 0,1 M hydrochloric acid, 5 mL of water R and 4 mL of 0,1 M
solution, Allow the latter to cool; separate the precipitate B2
hydrochloric acid instead of the aqueous layer,
that forms al1d filter through a tared sintered-glass filter (40)
(2,1,2), Plastic additives 01, 04 and 05, Thin-layer chromatography
A. Infrared absorption spectrophotometry (2224), (2,227),
Preparation, Dissolve the coagulum Bl in 30 mL of Reference solutions, Prepare 0,1 mg/mL solutions of
tetrahydrofuran R and add, in small volumes with shaking, plastic additive 01 CRS, plastic additive 04 CRS and plastic
40 mL of anhydrous ethanol R Separate the precipitate B3 additive 05 CR5, respectively, in toluene R.
by filtration and dry in vacuo at a temperature not exceeding Plate: TLC silica gel plate R
50 oC over diphosphorus pentoxide R. Dissolve a few Mobile phase: toluene R.

402 See the information section on general monographs (caver pages)


EUROPEAN PHARMACOPOEIA 8.0 3.1.15. Polyethylene terephthalate for containel's

Applieation: 0.5 mL of solution Al obtained during the Verify the absence of calcium in the hydrochloric acid used.
identification as a band 30 mm by 3 mm and 5 IlL of each Tin: maximum 20 ppm.
reference solution.
Inductively coupled plasma-atomic emission spectrometry
Development: over a path of 15 cm. (2.2.57).
Drying: in airo Test solution. Dilute solution SI 10 times with water R
Deteetíon A: examine in ultraviolet light at 254 nm. immediately before use.
Locate the zone corresponding to plastic additive 01 (R p about Reference solution. Introduce 2 mL of tin standard solution
0.4). Remove the afea of silica gel corresponding to this zone (5 ppm Sn) R into a 50 mL flask containing 5 mL of a 20 per
and shake with 40 mL of ether R for 1 mino Filter, rinse with cent V/V solution of sulfuric acid R and dilute to 50 mL with
2 quantities, each of 10 mL of ether R, add the rinsings to the water R immediately before use.
filtrate and evaporate to dryness. The residue C weighs not Wavelength: use the emission oftin at 189.99 nm, the spectral
more than 40 mg. background being taken at 190.10 nm.
Deteetíon B: expose the plate to iodine vapour for 5 mino Verify the absence of tin in the hydrochloric acid used.
Examine the chromatogram and locate the band corresponding Zinc: maximum 0.2 per cent.
to plastic additives 04 and 05 (R p = O). Remove the area of Atomic absorption spectrometry (2.2.23, Method I).
silica gel corresponding to this zone. Similarly remove a
corresponding are a of silica gel as a blank reference. Separately Test solution. Dilute solution SIl 00 times with 0.1 M
shake both samples for 15 min with 40 mL of methanol R. hydrochloric acid.
Filter, rinse with 2 quantities, each of 10 mL of methanol R, Reference solutions. Prepare the reference solutions using
add the rinsings to the filtrate and evaporate to dryness. The zinc standard solution (100 ppm Zn) R, diluting with 0.1 M
difference between the masses of both residues is not more hydrochloric acid.
than 10 mg. Source: zinc hollow-cathode lampo
Plastk additive 03. Infrared absorption spectrophotometry Wavelength: 213.9 nm.
(2.2.24). Atomisation device: air-acetylene flameo
Preparatíon. Wash precipitate B2 obtained during the Verify the absence of zinc in the hydrochloric acid used.
identification and contained in the tared sintered-glass filter Heavy metals (2.4.8): maximum 50 ppm.
(40) (2.1.2) with anhydrous ethanol R. Dry to constant mass
To 10 mL of solution SI add 0.5 mL of phenolphthaleín
over diphosphorus pentoxíde R and weigh the filter. The
solution R and then strong sodium hydroxide solution R until
residue weighs not more than 20 mg.
a pale pink colour is obtained. Dilute to 25 mL with water R.
Comparison: plastic additive 03 CRS. 12 mL of solution complies with test A. Prepare the reference
Barium: maximum 5 ppm. solution using lead standard solutíon (2 ppm Pb) R.
Inductively coupled plasma-atomic emission spectrometry Water extractable substances: maximum 0.3 per cent.
(2.2.57). Evaporate 50.0 mL of solution S2 to dryness on a water-bath
Test solution. Ignite 1.0 g of the substance to be examined in a and dry at 100-105 oC until constant mass. Carry out a blank
silica crucible. Take up the residue with 10 mL of hydroehloric titratiol1 with 50.0 mL af water for injections R. The residue
acid R and evaporate to dryness Oil a water-bath. Take up the weighs not more than 7.5 mg taking inta account the blank
residue with 20 mL of 0.1 M hydrochloric acid. test.
Reference solution. A solution containing 0.25 ppm ofbarium ASSAY
prepared by dilution of baríum standard solution (50 ppm
Ba) R with 0.1 M hydrochloric acid. Carry out the oxygen-flask method (2.5.10) using 50.0 mg.
Absorb the combustion products in 20 mL of 1 M sodium
Wavelength: use the emission of barium at 455.40 nm, the hydroxide. To the solution obtained add 1 mL of dibutyl
spectral background being taken at 455.30 nm. phthalate R, 2.5 mL of nitric acid R, 5 mL of ferric ammonium
Verify the absence ofbarium in the hydrochloric acid used. sulfate solution R2 and 10.0 mL of 0.1 M silver nitrate. Titrate
Cadmium: maximum 0.6 ppm. with 0.05 M ammonium thiocyanate until a reddish-yellow
Atomic absorption spectrometry (2.2.23, Method I). colour is obtained. Carry out a blank test.
1 mL of 0.1 M silver nitrate is equivalent to 6.25 mg of
Test solutíon. Evaporate 10 mL of solution SI to dryness.
poly(vinyl chloride).
Take up the residue using 5 mL of a 1 per cent V/V solution
of hydrochloric acid R, filter and dilute the filtrate to 10.0 mL
01/2008:30115
with the same acid.
corrected 7.5
Reference solutions. Prepare the reference solutions using
cadmium standard solution (0.1 per cent Cd) R, diluting with a
1 per cent V/V solution of hydrochloric acid R.
3.1.15. POLYETHYLENE
Source: cadmium hollow-cathode lampo TEREPHTHALATE FOR CONTAINERS
Wavelength: 228.8 nm. FOR PREPARATIONS NOT FOR
Atomisation device: air-acetylene flameo PARENTERAL USE
Verify the absence of cadmium in the hydrochloric acid used.
Calcium: maximum 0.07 per cent.
Inductively coupled plasma-atomic emission spectrometry
(2.2.57).
Test solution. Use the test solution prepared for the
r
to~°yV
0 J ",,'"'""
determination of barium.
Reference solution. A solution containing 50.0 ppm of calcium DEFINITION
prepared by dilution of calcium standard solution (400 ppm Polyethylene terephthalate is obtained fram the
Ca) R with 0.1 M hydrochloric acid. polymerisation of terephthalic acid or dimethyl
Wavelength: use the emission of calcium at 315.89 nm, the terephthalate with ethylene gIyco!' Isophthalic acid,
spectral background being taken at 315.60 nm. dimethyl isophthalate, 1,4- bis(hydroxymethyl)cyclohexane

General Norices (1) apply to all monographs and other texts 403
3.1.15. Polyethylene terephthalate for containers EUROPEAN PHARMACOPOEIA 8.0

(cyclohexane-l,4-dimethanol) or diethylene glycol may be The solution turns yellow. Not more than 0.5 mL of 0.01 M
used in the polymerisation. It may contain not more than hydrochloric acid is required to reach the beginning of the
0.5 per cent of silica or silicates and colouring matter approved colour change of the indicator to orange.
by the competent authority. Absorbance of solution SI (2.2.25): maximum 0.20 between
220 nm and 340 nm. In addition, for coloured polyethylene
PRODUCTION terephthalate: maximum 0.05 between 400 nm to 800 nm.
The manufacturing process is validated to demonstrate that Absoroance of solution S2 (2.2.25): maximum 0.05 between
the residual acetaldehyde content is not greater than 10 ppm 400 nm and 800 nm.
in the granules.
Reducing substances. Add 2 mL of 0.5 M sulfuric acid and
CHARACTERS 20.0 mL of 0.002 M potassium permanganate to 20.0 mL of
solution S1. Boil for 3 mino Cool immediately to ambient
Appearance: clear or opaque granules. temperature. Add 1 g of potassium iodide R, 0.25 mL of
Solubility: practically insoluble in water, in ethanol (96 per starch solution R as indicator and titrate with 0.01 M sodium
cent) and in methylene chloride. It is hydrolysed by strong thiosulfate. Perform a blank titration using 20.0 mL of water R.
bases. The difference in volume used in the 2 titrations is not greater
than 0.5 mL.
IDENTIFICATION Substances soluble in dioxan: maximum 3 per cent.
A. Place 0.10 g of the material to be examined into a Place 2 g of the material to be examined in a borosilicate glass
borosilicate glass flask with a ground-glass neck. Add flask with a ground-glass neck. Add 20 mL of dioxan R and
25 mL of a 200 giL solution of potassium hydroxide R in a heat under reflux for 2 h. Evaporate 10 mL of the solution
50 per cent V/V solution of anhydrous ethanol R. Reflux for to dryness on a water-bath and then dry the residue at
30 mino Allow to cool and dilute to 100 mL with water R. 100-105 oc. The residue weighs a maximum of 30 mg.
Filter if necessary. Dilute 1.0 mL of the filtrate to 100 mL
with water R. Examined between 210 nm and 330 nm Extractable aluminium: maximum 1 ppm.
(2.2.25), the solution shows an absorption maximum at Inductively coupled plasma-atomic emission spectrometry
240 nm. (2.2.57).
B. Dissolve 0.05 g of the material to be examined in 2 mL Test solution. Solution S3.
of 1, 1, 1,3,3,3-hexafluoropropan-2-01 R. Apply to a glass Reference solutions. Prepare the reference solutions using
plate on a water-bath in a fume cupboard several drops of aluminium standard solution (200 ppm Al) R, diluting with
the solution to produce a film of about 15 mm by 15 mm. 0.1 M hydrochloric acid.
Allow the solvent to evaporate and remove the film using a
Wavelength: 396.15 nm, the spectral background being taken
stream of water and a scraper. Dry in an oven at 100-105 oC
at 396.25 nm.
for 1-2 h. Examine the film by infrared absorption
spectrophotometry (2.2.24). The spectrum of the material Verify the absence of aluminium in the 0.1 M hydrochloric
to be examined shows maxima in particular at 1725 cm - 1, acid used.
1410 cm- l , 1265 cm- I , 1120 cm- I , 1100 cm- J, 1020 cm-\ Extractable antimony: maximum 1 ppm.
875 cm- J, 725 cm- J. The spectrum obtained, in addition, is
Inductively coupled plasma-atomic emission spectrometry
identical to that of the material selected for the type sample.
(2.2.57).
TESTS Test solution. Solution S4.
If necessary, cut out samples for testing to a maximum size of Reference solutions. Prepare the reference solutions using
1 cm per side. antimony standard so/ution (100 ppm Sb) R, diluting with
0.01 M sodium hydroxide.
Solution SI. Place 10.0 g of the material to be examined in a
borosilicate glass flask witha ground-glass neck. Add 200 mL Wavelength: 231.15 nm or 217.58 nm, the spectral background
of water R and heat at 50 oC for 5 h. Allow to cool and decant being taken at 231.05 nm.
the solution. Use solution SI within 4 h of its preparation. Extractable barium: maximum 1 ppm.
Solution S2. Place 10 g of the material to be examined in a Inductively coupled plasma-atomic emission spectrometry
borosilicate glass flask with a ground-glass neck. Add 100 mL (2.2.57).
of ethanol (96 per cent) R and heat at 50 oC for 5 h. Allow to Test solution. Solution 53.
cool and decant the solution. Use solution S2 within 4 h of its
Reference solutions. Prepare the reference solutions using
preparation.
barium standard solutian (50 ppm Ba) R, diluting with 0.1 M
Solution 53. Place 20 g of the material to be examined in a hydrochloric acid.
borosilicate glass flask with a ground-glass neck. Add 50 mL Wavelength: 455.40 nm, the spectral background being taken
of 0.1 M hydrochloric acid and heat at 50 oC for 5 h. Allow at 455.30 11m.
to cool and. decant the solution. Use solution S3 within 4 h
of its preparation. Verify the absence of barium in the 0.1 M hydrochloric acid
used.
Solution 54. Place 20 g of the material to be examined into a
borosilicate glass flask with a ground-glass neck. Add 50 mL Extractable cobalt: maximum 1 ppm.
of 0.01 M sodium hydroxide and heat at 50 oC for 5 h. Allow to Inductively coupled plasma -atomic emission spectrometry
cool and decant. Use solution S4 within 4 h of its preparation. (2.2.57).
Appearam:e of solution SI. Solution SI is clear (2.2.1). Test solution. Solution S3.
Appearance of solution S2. Solution S2 is clear (2.2.1) and Reference solutions. Prepare the reference solutions using
colourless (2.2.2, Method II). cobalt standard solutian (lOO ppm Co) R, diluting with 0.1 M
hydrochloric acid.
Acidity or alkalinity. To 50 mL of solution SI add 0.15 mL
of BRP indicator solution R. The solution turns yellow. Not Wavelength: 228.62 nm, the spectral background being taken
more than 0.5 mL of 0.01 M sodium hydroxide is required at 228.50 nm.
to change the colour of the indicator to blue. To another Verify the absence of cobalt in the 0.1 M hydrochloric acid
50 mL of solution SI add 0.2 mL of methyl orange solution R. used.

404 See the information section on general monographs (cover pages)


EUROPEAN PHARMACOPOEIA 8.0 3.1.15. Polyethylene terephthalate for containers

Extractable germanium: maximum 1 ppm. Test solution. Solution S3.


Inductively coupled plasma-atomic emission spectrometry Reference solutions. Prepare the reference solutions using
(2.2.57). titanium standard solution (100 ppm Ti) R, diluting with 0.1 M
Test solution. Solution S4. hydrochloric acid.
Reference solutions. Prepare the reference solutions using
germanium standard solution (100 ppm Ge) R, diluting with Wavelength: 323.45 nm or 334.94 nm, the spectral background
0.01 M sodium hydroxide. being taken at 323.35 nm.
Wavelength: 206.87 nm or 265.12 nm, the spectral background Verify the absence of titanium in the O.lM hydrochloric acid
being taken at 206.75 nm. used.
Extractable manganese: maximum 1 ppm. Extractable zinc: maximum 1 ppm.
Inductively coupled plasma-atomic emission spectrometry
Inductively coupled plasma-atomic emission spectrometry
(2.2.57).
(2.2.57).
Test solution. Solution S3.
Reference solutions. Prepare the reference solutions using Test solution. Solution S3.
manganese standard solution (lOO ppm Mn) R, diluting with Reference solutions. Prepare the reference solutions using
0.1 M hydrochloric acid. zinc standard solution (lOO ppm Zn) R, diluting with 0.1 M
Wavelength: 257.61 nm, the spectral background being taken hydrochloric acid.
at 257.50 nm. .
Wavelength: 213.86 nm, the spectral background being taken
Verify the absence of manganese in the 0.1 M hydrochloric
at 213.75 nm.
acid used.
Extractable titanium: maximum 1 ppm. Verify the absence of zinc in the 0.1 M hydrochloric acid used.
Inductively coupled plasma-atomic emission spectrometry Sulfated ash (2.4.14): maximum 0.5 per cent determined on
(2.2.57). l.0 g.

General Notices (1) apply ta all monographs and other texts 405
EUROPEAN PHARMACOPOEIA 8.0

406 See the information sectian an general manographs (ca ver pages)
EUROPEAN PHARMACOPOEIA 8.0

3.2. Containers ........ ,................ ,................................................ 409 3.2.5, Sterile containers of plasticised poly(vinyl chloride) for
3.2.1. Glass containers for pharmaceutical use ..................... 409 human blood containing anticoagulant solution ............... 418
3.2.2. Plastic containers and closures for pharmaceutical 3.2,6. Sets for the transfusion of blood and blood
use ........................ ,....... ,................................................. ,.......... 414 components .... ,................ ,....................................................... 418
3.2.2.1. Plastic containers for aqueous solutions for 3.2.8, Sterile single-use plastic syringes ................................. 419
infusion .................................................................................... 414 3.2.9. Rubber closures for containers fo!' aqueous
3.2.3. Sterile plastic containers for human blood and parenteral preparations, for powders and for
blood components ............. ,.................................................... 415 freeze-dried powders ........................................... ,.................. 421
3.2.4. Empty sterile containers of plasticised poly(vinyl
chloride) for human blood and blood components ........... 417

General Natices (1) apply ta all managraphs and ather texts 407
EUROPEAN PHARMACOPOEIA 8.0

408 See the information section on general monographs (cover pages)


EUROPEAN PHARMACOPOEIA 8.0 3.2.1. Glass containers for pharmaceutical use

0112008:30200 of contact between the inner surface of the container or


glass grains and water. The hydrolytic resistan ce is evaluated
by titrating released alkali. According to their hydrolytic
resistance, glass containers are classified as follows:
3.2. CONTAINERS
- Type 1 glass containers: neutral glass, with a high hydrolytic
A container for pharmaceutical use is an article that contains resistance due to the chemical composition of the glass
or is intended to contain a product and is, or may be, in direct itself;
contact with it. The closure is a part of the container.
- Type II glass containers: usually of soda -lime-silica glass
The container (see General Notices section 1.3) is so designed with a high hydrolytic resistance resulting from suitable
that the contents may be removed in a manner appropriate treatment of the surface;
to the intended use of the preparation. It provides a varying
degree of protection depending on the nature of the product - Type III glass containers: usually of soda-lime-silica glass
and the hazards of the environment, and minimises the 10ss with only moderate hydrolytic resistance.
of constituents. The container does not interact physically or
The following italicised statements constitute general
chemically with the contents in a way that alters their quality
recommendations concerning the type of glass container
beyond the limits tolerated by officia1 requirements.
that may be used for different types of pharmaceutical
Single-dose container. A single-dose container holds a preparations. The manufacturer of a pharmaceutical product
quantity of the preparation intended for total or partial use is responsible for ensuring the suitability of the chosen
on 1 occasion only. container.
Multidose container. A multidose container holds a quantity Type f glass containers are suitable for most preparations
of the preparation suitable for 2 or more doses. whether or not for parenteral administration.
Well-closed container. A well-closed container protects the Type JI glass containers are suitable for most acidic and
contents from contamination with extraneous solids and neutral, aqueous preparations whether or not for parenteral
liquids and from loss of contents under ordinary conditions of administration.
handling, storage and transport.
Type fJI glass containers are in general suitable for non-aqueous
Airtight container. An airtight container is impermeable
preparations for parenteral administration, for powders for
to solids, liquids and gases under ordinary conditions of
parenteral administration (except for freeze-dried preparations)
handling, storage and transport. lf the container is intended
and for preparations not for parenteral administratian.
to be opened on more than 1 occasion, it must be so designed
that it remains airtight after re-dosure. Glass containers with a hydrolytic resistance higher than that
Sealed container. A sealed container is a container closed by recommended aboye for a particular type of preparation may
fusion of the material of the container. gene rally also be used.

Tamper-proof container. A tamper-proof container is a closed The container chosen for a given preparation shall be such
container fitted with a device that reveals irreversibly whether that the glass material does not releas e substances in quantities
the container has been opened. sufficient to affect the stability of the preparation or to present
a risk of toxicity. In justified cases, it may be necessary to have
Child-proof container. A container that is fitted with a closure detailed information on the glass composition, so that the
that prevents opening by children. potential hazards can be assessed.
Preparations for parenteral administration are normally
presented in colourless glass, but coloured glass may be
used for substances known to be light-sensitive. Colourless
0712010:30201 or coloured glass is used for the other pharmaceutical
preparations. It is recommended that aH glass containers
for liquid preparations and for powders for parenteral
3.2. L GLASS CONTAINERS FOR administration permit the visual inspection of the contents.
PHARMACEUTICAL USE The inner surface of glass containers may be specially treated
to improve hydrolytic resistance, to confer water-repellancy,
Glass containers for pharmaceutical use are glass articles etc. The outer surface may also be treated, for example to
intended to come into direct contact with pharmaceutical reduce friction and to improve resistance to abrasion. The
preparations. outer treatment is such that it does not contaminate the inner
Colourless glass is highly transparent in the visible spectrum. surface of the container.
Coloured glass is obtained by the addition of small amounts Except for type 1 glass containers, glass container s for
of metal oxides, chosen according to the desired spectral pharmaceutical preparations are not to be re-used. Containers
absorban ce. for human blood and blood components must not be re-used.
Neutral glass is a borosilicate glass containing signitlcant Glass containers for pharmaceutical use comp1y with the
amounts ofboric oxide, aluminium oxide alkali and/or relevant test or tests for hydrolytic resistance. When glass
alkaline earth oxides. Due to its composition neutral glass containers have non-glass components, the tests apply only to
has a high hydrolytic resistan ce and a high thermal shock the glass part of the container.
resistance.
To define the quality of glass containers according to the
Soda-lime-silica glass is a silica glass containing alkali metal intended use, one or more of the following tests are necessary.
oxides, mainly sodium oxide and alkaline earth oxides, mainly
calcium oxide. Due to its composition soda-lime-silica glass Tests for hydrolytic resistan ce are carried out to define the type
has only a moderate hydrolytic resistance. of glass (1, II OI III) and to control its hydrolytic resistance.
The hydrolytic stability of glass containers for pharmaceutical In addition, containers for aqueous parenteral preparations
use is expressed by the resistance to the release of soluble are tested for arsenic releas e and coloured glass containers are
mineral substances into water under the prescribed conditions tested for spectral transmission.

General Natices (1) apply to all monographs and other texts 409
3.2.1. Glass containers for pharmaceutical use EUROPEAN PHARMACOPOEIA 8.0

HYDROLYTIC RESISTANCE the mean value. This volume, expressed to 1 decimal place, is
the filling volume for the particular ampoule 101. The filling
volume may also be determined by weighing.
Table 3.2.1.-1. - Types of glass

Type of container

Type 1 and type II glass containers


Test to be performed

Test A (surface test)


\
(to distinguish from type III glass
containers)
Type 1 glass containers (to distinguish Test B (glass grains test) or test e
from type II and type III glass (etching test)
containers)
Type 1 and type II glass containers Tests A and B, or tests A and e
where it is necessary to determine A
whether the high hydrolytic resistance
is due to the chemical campo sitian or
to the surface treatment

The test is carried out by titration of the extract solutions


obtained under the conditions described for tests A, B and C.
EQUIPMENT
I I
an autoclave capable of maintaining a temperature of ¡
121°C ± 1°C, equipped with a thermometer OI a calibrated ;-~-

thermocouple recorder, a pressure gauge, a vent cock and a


tray, of sufficient capacity to accommodate aboye the water
level the number of containers needed to carry out the Figure 3.2.1.-1. - Filling volume of ampoules (up to point A)
test; e/ea n the autoclave vessel and all ancillary equipment TEST A. HYDROLYTIC RESISTANCE OF THE INNER
thoroughly before use with water R; SURFACES OF GLASS CONTAINERS (SURFACE TEST)
The determination is carried out 011 unused containers. The
- burettes with a suitable capacity; volumes of the test liquid necessary for the final determination
are indicated in Table 3.2.1.-2.
- one-mark volumetric flasks, with a capacity of 1000 mL; Table 3.2.1.-2. - Volume of test ¡iquid and number of titrations
Filling volume (mL) Volume of test liquid Number of titrations
- pipettes and beakers; for one titration
(mL)

- conical flasks with a capacity of 100 mL and 250 mL; Up to 3 25.0

Above 3 and up \0 30 50.0 2


- a water-bath;
Above 30 and up to 100 100.0 2

- a metal foil (e.g. aluminium, stainless steel). Above 100 100.0

Cleaning. Remove any debris or dust. Shortly before the


Flasks and beakers shall have been already used for the test test, rinse each container carefully at least twice with water R
or have been filled with water R and kept in an autoclave at and allow to stand. Immediately before testing empty the
121°C at least fOIl h before being used. containers, rinse once with water R then with water Rl and
DETERMINA TION OF THE FILLING VOLUME allow to drain. Complete the cleaning procedure from the first
rinsing in not less than 20 min and 110t more than 25 mino
The filling volume is the volume of water to be filled in the
container for the purpose of the test. For vials and bottles Heat closed ampoules on a water-bath or in an air-oven at
the filling volume is 90 per cent of the brimful capacity. For about 50 oC for approximately 2 min before opening; do not
ampoules it is the volume up to the height of the shoulder. rinse before testing.
Filling and heating. The containers are filled with water Rl up
Vials and bottles. Select, at random, 6 containers from the
to the filling volume. Containers in the form of cartridges or
sample lot, or 3 if their capacity exceeds 100 mL, and remove
prefilled syringes are closed in a suitable manner with material
any dirt or debris. Weigh the empty containers with an
that does not interfere with the test. Each container including
accuracy of 0.1 g. Place the containers on a horizontal surface
ampoules shall be loosely capped with an inert material
and fill them with distilled water R until about the rim edge,
such as a dish of neutral glass or aluminium foil previously
avoiding overflow and introduction of air bubbles. Adjust the
rinsed with water R. Place the containers on the tray of the
liquid levels to the brimfulline. Weigh the filled containers to
autoclave. Place the tray in the autoclave containing a quantity
obtain the mass of the water expressed to 2 decimal places for
of water R such that the tray remains clear of the water. Close
containers having a nominal volume less or equal to 30 mL,
the autoclave and carry out the following operations:
and expressed to 1 decimal place for containers having a
nominal volume greater than 30 mL. Calculate the mean - heat the autoclave to 100 oC and allow the steam to issue
value of the brimful capacity in millilitres and multiply it from the vent cock for 10 min;
by 0.9. This volume, expressed to 1 decimal place, is the filling - close the ventcod: and raise the temperature from 100 oC
volume for the particular container 101. to 121 oC at arate of 1 oC per min;
- maintain the temperature at 121 ± 1 oC for 60 ± 1 min;
Ampoules. Place at least 6 dry ampoules on a flat, horizontal
surface and fill them with distilled water R from a burette, - lower the temperature from 12l oC to 100 oC at arate of
until the water reaches point A, where the body of the 0.5 oC per min, venting to prevent vacuum;
ampoule declines to the shoulder (see Figure 3.2.1.-1). Read - do not open the autoclave before it has cooled down to
the capacities (expressed to 2 decimal places) and calculate 95 oC;

410 See the information section on general monographs (cover pages)


EUROPEAN PHARMACOPOEIA 8.0 3.2.1. Glass containers for pharmaceutical use

- remove the containers from the autoclave using normal - a desiccator;


precautions, place them in a water-bath at 80 oC, and fUn - an ultrasonic bath.
cold tap water, taking care that the water does not contact
the loase foil caps to avoid contamination of the extraction Method. Rinse the containers to be tested with water R and
solution; dry in the oven. Wrap at least 3 of the glass articles in clean
paper and crush to produce 2 samples of about 100 g each
- cooling time does not exceed 30 mino in pieces not more than 30 mm across. Place 30-40 g of the
The extraction solutions are analysed by titration according to pieces between 10-30 mm across taken from 1 of the samples
the method described below. in the mortar, insert the pestle and strike it heavily once only
Method. Carry out the titration within 1 h of removal of the with the hammer. Transfer the contents of the mortar, to the
containers from the autoclave. Combine the liquids obtained coarsest sieve (a) of the set. Repeat the operation until all
from the containers and mix. Introduce the prescribed volume fragments have been transferred to the sieve. Shake the set
(Table 3.2.1.-2) into a conical flask. Place the same volume of of sieves a short time by hand and remove the glass which
water R1 into a second similar flask as a blank. Add to each remains on sieves (a) and (b). Submit these portions to
flask 0.05 mL of methyl red solution R for each 25 mL ofliquid. further fracture, repeating the operation until about 10 g of
Titrate the blank with 0.01 M hydrochloric acid. Titrate the glass remains on sieve (a). Reject this portion and the portion
test liquid with the same acid until the colour of the resulting which passes through sieve (c). Reassemble the set of sieves
solution is the same as that obtained for the blank. Subtract and shake for 5 mino Transfer to a weighing bottle those glass
the value found for the blank titration from that found for grains which passed through sieve (b) and are retained on
the test liquid and express the results in millilitres of O. 01 M sieve (c). Repeat the crushing and sieving procedure with the
hydrochloric acid per 100 mL. Express titration values of less other glass sample and thus 2 samples of grains, each of which
than 1.0 mL to 2 decimal places and titration values of more shall be in excess of 10 g, are obtained. Spread each sample on
than or equal to 1.0 mL to 1 decimal place. a piece of clean glazed paper and remove any iron particles by
passing the magnet over them. Transfer each sample into a
Limits. The results, or the average of the results if more than beaker for cleaning. Add to the grains in each beaker 30 mL
one titration is performed, is not greater than the values stated of acetone R and scour the grains by suitable means, such as a
in Table 3.2.1.-3. rubber or plastic-coated glass rod. After scouring the grains,
Table 3.2.1.-3. - Limit values in the test for surface hydrolytic allow to settle and decant as much acetone as possible. Add
resistance another 30 mL of acetone R, swirl, decant again and add a new
portion of acetone R.
Maximum vo!ume ofO.01 M He! per
100 mL of test liquid (mL)
Glass containers

Filling volume (mL) Types 1 and U TypeIH


Up to 1 2.0 20.0

Above 1 and up to 2 1.8 17.6

Above 2 and up to 5 1.3 13.2

Aboye 5 and up to 10 1.0 10.2

Above 10 and up to 20 0.80 8.1

Aboye 20 and up to 50 0.60 6.1

Aboye 50 and up to 100 0.50 4.8

Aboye 100 and up to 200 0.40 3.8

Aboye 200 and up to 500 0.30 2.9

Aboye 500 0.20 2.2

TEST B. HYDROLYTIC RESISTANCE OF GLASS GRAINS


(GLASS GRAINS TEST)
Check that the articles as received have been annealed to a
commercially acceptable quality.
The test may be performed on the canes used for the
manufacture of tubing glass containers or on the containers.
Equipment
- a mortar, pestle (see Figure 3.2.1.-2) and hammer in
tempered, magnetic steel; Figure 3.2.1.-2. - Apparatus for glass grains method
- a set of 3 square-mesh sieves of stainless steel, mounted on (dimensions in millimetres)
frames of the same material and consisting of the following: Fill the bath of the ultrasonic vessel with water at room
(a) sieve no. 710; temperature, then place the beaker in the rack and immerse it
until the leve! of the acetone is at the level of the water; apply
(b) sieve no. 425;
the ultrasound for 1 mino Swirl the beaker, allow to settle and
(c) sieve no. 300; decant the acetone as completely as possible and then repeat
- a permanent magnet; the ultrasonic cleaning operation. If a fine turbidity persists,
- a metal foil (e.g. aluminium, stainless steel); repeat the ultrasonic cleaning and acetone washing until the
solution remains clear. Swirl and decant the acetone then
- a hot-air oven, capable of maintaining a temperature of dry the grains, first by putting the beaker on a warm plate
140 ± 5 oC; to remove excess acetone and then by heating at 140 oC for
- a balance, capable of weighing up to 500 g with an accuracy 20 min in the drying oven. Transfer the dried grains from each
ofO.005g; beaker into separate weighing botdes, insert the stoppers and

General NoNces (1) apply to all monographs and other texts 411
3.2.1. Glass containers for pharmaceutical use EUROPEAN PHARMACOPOElA 8.0

cool in the desiccator. Weigh 10.00 g of the cleaned and dried Distinction between type 1 and type JI glass coníainers
grains into 2 separate conical flasks. Add 50 mL of water Rl The results obtained in Test C are compared to those obtained
into each by means of a pipette (test solutions). Pipette 50 mL in Test A. The interpretation of the result is shown in
of water Rl into a third conica! flask which will serve as Table 3.2.1.-4.
a b!ank. Distribute the grains evenly over the flat bases of
the flasks by gentle shaking. Close the flasks with neutral Table 3.2.1.-4. - Distinction between Types 1 and 11 glass
glass dishes or aluminium foil rinsed with water R or with containers
inverted beakers so that the inner surface of the beakers fit Type 1 Type H
snugly down onto the top rims of the flasks. Place all 3 flasks
in the rack in the autoclave containing the water at ambient The values are closely similar The values greatly exceed those found
to those found in the test for in the test for surface hydrolytic
temperature, and ensure that they are he!d aboye the leve! of surface hydrolytic resistan ce for resistan ce and are similar but not
the water in the vessel. Carry out the autoclaving procedure in type I glass containers. larger than those for type III glass
a similar manner to that described under test A, but maintain containers.
the temperature of 121 ± 1 oC only for 30 ± 1 mino Do not
open the autoclave until it has cooled to 95 oc. Remove the ARSENIC
hot samples from the autoclave and cool the flasks in running The test applies to glass containers for aqueous parenteral
tap water as soon as possible, avoiding thermal shock. To each preparations.
of the 3 flasks add 0.05 mL of methyl red solution R. Titrate the
Hydride generation atomic absorption spectrometry (2.2.23,
blank solution immediately with 0.02 M hydrochloric acid then
Method 1).
titrate the test solutions until the colour matches that obtained
with the blank solution. Subtract the titration volume for the Test solution. Use the extract solution obtained from
blank solution from that for the test solutions. containers of types 1 and II, after autoclaving at 121 oC for 1 h
as described under test A for surface hydrolytic resistan ce.
NOTE: where necessary to obtain a sharp end-point, the clear
Transfer 10.0 mL to a 100 mL volumetric flask. Add 10 mL
solution is to be decanted into a separate 250 mL flask. Rinse
of hydrochloric acid R and 5 mL of a 200 giL solution of
the grains with 3 quantities, each of 15 mL, of water Rl by
potassium iodide R. Heat on a water-bath at 80 oC for 20 min,
swirling and add the washings to the main solution. Add
allow to cool and dilute to 100.0 mL with water R.
0.05 mL of the methyl red solution R. Titrate and calculate as
described below. In this case also add 45 mL of water Rl and Reference solutions. Prepare the reference solutions using
0.05 mL of methyl red solution R to the blank solution. arsenic standard solution (1 ppm As) R. Add 10 mL of
hydrochloric acid R and 5 mL of a 200 giL solution of
Calculate the mean value of the results in millilitres of 0.02 M
potassium iodide R. Heat on a water-bath at 80 oC for 20 min,
hydrochloric acid per gram of the sample and if required its
allow to cool and dilute to 100.0 mL with water R. The
equivalent in alkali extracted, calculated as micrograms of
concentration range of the reference solutions is typically
sodium oxide per gram of glass grains.
0.005 ppm to 0.015 ppm of As.
1 mL of 0.02 M hydrochloric acid is equivalent to 620 ¡.tg of
Acid reservo ir. Hydrochloric acid R.
sodium oxide.
Reducing reservoir. Sodium tetrahydroborate reducing
Repeat the test if the highest and lowest observed values differ solution R.
by more than 20 per cent.
Use a hydride generation device to introduce the test solution
Limits. Type I glass containers require not more than 0.1 mL into the cuvette of an atomic absorption spectrometer.
of 0.02 M hydrochloric acid per gram of glass, type II and Establish and standardise instrumental operating conditions
type III glass containers require not more than 0.85 mL of according to the manufacturer's instructions, optimise the
0.02 M hydrochloric acid per gram of glass. uptake rate of the peristaltic pump tubings, then connect
TEST C. TO DETERMINE WHETHER THE CONTAINERS tubings to the acid reservoir, the reducing reservoir and the
HA VE BEEN SURFACE-TREATED (ETCHING TEST) test solution.
When it is necessary to determine if a container has been Source: hollow-cathode lampo
surface-treated, and/or distinguish between type I and Wavelength: 193.7 nm.
type II glass containers, test e is used in addition to test A.
Atomisation device: air-acetylene flameo
Alternative!y, test A and B may be used. Test C may be carried
out either on unused samples or on samples previously tested Limit: maximum 0.1 ppm of As.
for test A.
SPECTRAL TRANSMISSlON FOR COLOURED GLASS
Vials and botdes. The volumes of test liquid required are CONTAlNERS
shown in Table 3.2.1.-2.
Equipment. A UV -VIS spectrophotometer, equipped with a
Rinse the containers twice with water R and fill to the brimful photodiode detector or equipped with a photomultiplier tube
point with a mixture of 1 volume of hydrofluoric acid R and coupled with an integrating sphere.
9 volumes of hydrochloric acid R and allow to stand for 10 mino
Empty the containers and rinse carefully 5 times with water R. Preparation of the spedmen. Break the glass container or
Immediately before the test, rinse once again with water R. cut it with a circular saw fitted with a wet abrasive wheel,
Submit the containers thus prepared to the same autoclaving such as a carborundum or a bonded-diamond wheel. Select
and determination procedure as described in test A for surface sections representative of the wall thickness and trim them as
hydrolytic resistance. rf the results are considerably higher suitable for mounting in a spectrophotometer. lf the specimen
than those obtained from the original surfaces (by about a is too small to cover the opening in the specimen holder, mask
factor of 5 to 10), the samples have been surface-treated. the uncovered portion with opaque paper or tape, provided
that the length of the specimen is greater than that of the slit.
Ampoules Before placing in the holder, wash, dry and wipe the specimen
NOTE: ampoules made from glass tubing are not normally with lens tissue. Mount the specimen with the aid of wax,
subjected to internal surface treatment because their high or by other convenient means, taking care to avoid leaving
chemical resistance is dependent upon the chemical composition fingerprints or other marks.
of the glass as a material. Method. Place the specimen in the spectrophotometer with
Apply the test method as described aboye for vials and bottles. its cylindrical axis parallel to the slit and in su eh a way that the
lf the ampoules are not surface-treated, the new values are light beam is perpendicular to the surface of the section and
slightly lower than those obtained in previous tests. that the losses due to reflection are at a minimum; Measure

412 See the information section on general monographs (cover pages)


EUROPEAN PHARMACOPOEIA 8.0 3.2.1. Glass cantainers far pharmaceutical use

the transmission of the specimen with reference to air in the Instructions on determination of the filling volume, cleaning
spectral regio n of 290-450 nm, continuously 01' at intervals of the containers, filling and heating are given aboye under
of20 nm. Hydrolytic resistance and Test A. Hydrolytic resistance of the
Limits. The observed spectral transmission for coloured inner surfaces of glass containers.
glass containers for preparations that are not for parenteral SOLUTIONS
administration do es not exceed 10 per cent at any wavelength Spectrochemieal buffer solution. Dissolve 80 g of caesium
in the range of 290 nm to 450 nm, irrespective of the type ehloride R in about 300 mL of water Rl, add 10 mL of
and the capacity of the glass container. The observed spectral 6 M hydrochloric acid R and transfer to a 1000 mL volumetric
transmission in coloured glass containers for parenteral !lask. Dilute to volume with v,;ater Rl and mix.
preparations do es not exceed the limits given in Table 3.2.1.-5. Stock solutions:
Table 3.2.1.-5. - Limits of spectral transmission for coloured - sodium oxide, c(NaP) = 1 mg/mL;
glass containers for parenteral preparations - potassium oxide, c(K 20) = 1 mg/mL;
- calcium oxide, c(CaO) = 1 mg/mL.
Maximum percentage of spectral transmission
at any wavelength between 290 nm and 450 nm Commercially available stock solutions may also be used.
Nominal volume (mL) Flame-sealed Containers with
Standard solutions. Prepare standard solutions by diluting
containers c10sures the stock solutions with water Rl to obtain concentrations
suitable for establishing the reference solutions in appropriate
Up lo 1 50 25
manner, e.g. with concentrations of 20 flg/mL of sodium
Aboye 1 and up lo 2 45 20 oxide, potassium oxide and calcium oxide, respectively.
Commercially available standard solutions may also be used.
Aboye 2 and up lo 5 40 15
Reference solutions. Prepare the reference solutions for
Aboye 5 and up lo 10 35 13 establishing the calibration graph (set of calibration solutions)
by diluting suitable concentrated standard solutions with
Aboye 10 and up lo 20 30 12
water Rl, so that the normal working ranges of the specific
Aboye 20 25 10 elements are covered, taking into account the instrument used
for the measurement. Typical concentration ranges of the
reference solutions are:
Annex - test for surface hydrolytic resistance - for determination by atomic emission spectrometry of
- determination by flame atomic absorption sodium oxide and potassium oxide: up to 10 Ilg/mL;
- for determination by atomic absorption spectrometry of
spectrometry (faas) sodium oxide and potassium oxide: up to 3 Ilg/mL;
The surface hydrolytic resistance of glass of types I and JI may - for determination by atomic absorption spectrometry of
be determined by analysis of the leaching solution by fiame calcium oxide: up to 7 Ilg/mL.
atomic absorption spectrometry. A number of elements that, Use reference solutions containing 5 per cent V/V of the
when present as oxides in glass, contribute to the alkalinity spectrochemical buffer solution.
of the solution, are determined and used to express an alkali METHOD
equivalent. The spectrometrie method has the advantage of Carry out preliminary measurements of the potassium oxide
allowing the use of a much smaller sample of extraet so that and calcium oxide concentrations on one of the extraction
it can be applied to small individual containers. This enables solutions. lf, for one container type, the concentration of
an evaluation of the uniformity of the eontainers in a given potassium oxide is less than 0.2 Ilg/mL and if the concentration
batch where this is eritical. The results of this measurement are of calcium oxide is less than 0.1 ~lg/mL, the remaining
not equivalent to those of titrimetry and the 2 methods cannot extraction SOlutiOl1S of this container type need not be
be considered interchangeable. A correlation between the 2 is analysed for these ions. Aspirate the extraction solution
dependent on the type of glass and the size and shape of the from each sample directly into the !lame of the atomic
container. The titrimetrie method is the reference method of absorption or atomic emission instrument and determine the
the Pharmacopoeia; the spectrometric method may be used in approximate concentrations of sodium oxide (and potassium
justified and authorised cases. oxide and calcium oxide, if present) by reference to calibration
A method suitable for this type of analysis is shown below. graphs produced from the reference solutions of suitable
concentration.
The determination is carried out on unused containers.
The number of containers to be examined is indicated FINAL DETERMINATION
in Table 3.2.1.-6. If dilution is unnecessary add to each container a volume of
the spectrochemical buffer solution equivalent to 5 per cent
Table 3.2.1.-6. - Number of eontainers to be examined for the of the filling volume, mix well and determine sodium oxide,
speetrometric method calcium oxide and potassium oxide, if present, by reference
to calibration graphs. For the determination of the calcium
Filling volume (mL) Number of containers Additional containers
to be measured for preliminary
oxide concentration by Dame atomic spectrometry, the nitrous
separately measurements oxide/acetylene !lame shall be used.
lf dilution is necessary, determine sodium oxide, calcium oxide
Up lo 2 20 2
and potassium oxide, if present, following the procedures
Aboye 2 and up lo 5 15 2 as described aboye. The measuring solutions shall contain
5 per cent V/V of the spectrochemical buffer solution.
Aboye 5 and up lo 30 10 2
Concentration values less than 1.0 flg/mL are expressed to
Aboye 30 and up lo 100 5 2 decimal places, values greater than or equal to 1.0 Ilg/mL
to 1 decimal place. Correct the result for the buffer addition
Above 100 3
and for dilution, if any.

General Notices (1) apply to all monographs and other texts 413
3.2.2. Plastic containers and dosures for pharmaceutical use EUROPEAN PHARMACOPOEIA 8.0

CALCULA TION For selection of a suitab1e plastic container, it is necessary to


Calculate the mean value of the concentration of individual know the full manufacturing formula of the plastic, including
oxides found in each of the samples tested, in micrograms of all materials added during formation of the container so that
the oxide per millilitre of the extraction solution and calculate the potential hazards can be assessed. The plastic container
the sum of the individual oxides, expressed as micrograms of chosen for any particular preparation should be such that:
sodium oxide per millilitre of the extraction solution using - the ingredients of the preparation in contact with the plastic
the following mass conversion factors: material are not significantly adsorbed on its surface and
- 1 Ilg of potassium oxide corresponds to 0.658 Ilg of sodium do not significantly migrate into or through the plastic,
oxide; - the plastic material does not reIease substances in quantities
- 1 Ilg of calcium oxide corresponds to 1.105 Ilg of sodium sufficient to affect the stability of the preparation or to
oxide. present a risk of toxicity.
Limits. For each container tested, the result is not greater than Using material or materials selected to satisfy these criteria, a
the value given in Table 3.2.1.-7. number of identical type samples of the container are made by
a well-defined pro ce dure and submitted to practica1 testing in
Table 3.2.1.-7. - Limit values in the test for surface hydrolytic conditions that reproduce those of the intended use, including,
resistance by flame atomic absorption spectrometry where appropriate, sterilisation. In order to confirm the
Maximum values for the
compatibility of the container and the contents and to ensure
conceníration of oxides, expressed that there are no changes detrimental to the quality of the
as sodium oxide (flg/mL) preparation, various tests are carried out such as verification of
FiIling volume (mL) Glass containers the absence of changes in physical characteristics, assessment
of any 10ss or gain through permeation, detection of pH
Types I and n changes, assessment of changes caused by light, chemical tests
Up to 1 5.00 and, where appropriate, biological tests.
Aboye 1 and up to 2 4.50 The method of manufacture is such as to ensure reproducibility
for subsequent bulk manufacture and the conditions of
Aboye 2 and up to 5 3.20 manufacture are chosen so as to preclude the possibility of
Aboye 5 and up to 10 2.50 contamination with other pI as tic materials or their ingredients.
The manufacturer of the product must ensure that containers
Aboye 10 and up to 20 2.00 made in production are similar in every respect to the type
Aboye 20 and up to 50 l.50 samples.
For the results of the testing on type samples to remain valid,
Aboye 50 and up to 100 1.20
it is important that:
Aboye 100 and up to 200 l.00 - there is no change in the composition of the material as
defined for the type samples,
Aboye 200 and up to 500 0.75
- there is no change in the manufacturing process as defined
Aboye 500 0.50 for the type samples, especially as regards the temperatures
to which the plastic material is exposed during conversion
or subsequent procedures such as sterilisation,
- scrap material is not used.
0112008:30202
Recycling of excess material of well-defined nature and
proportions may be permitted after appropriate validation.
3.2.2, PLASTIC CONTAINERS AND Subject to satisfactory testing for compatibility of each
CLOSURES FOR PHARMACEUTICAL different combination of container and contents, the materials
described in the Pharmacopoeia are recognised as being
USE suitable for the specific purposes indicated, as defined aboye.
A plastic container for pharmaceutical use is a plastic article
which contains or is intended to contain a pharmaceutical 01/2008:90003
product and is, or may be, in direct contact with it. The corrected 6.0
closure is a part of the container.
Plastic containers and closures for pharmaceutical use are 3,2.2.1. PLASTIC CONTAINERS FOR
made of materials in which may be'included certain additives;
these materials do not include in their composition any
AQUEOUS SOLUTIONS FOR INFUSION
substance that can be extracted by the contents in such DEFINITION
quantities as to alter the efficacy or the stability of the product Plastic containers for aqueous solutions for infusion are
or to present a risk of toxicity. manufactured from one or more polymers, if necessary
The most commonly used polymers are polyethylene (with with additives. The containers described in this section
and without additives), polypropy1ene, poly(vinyl chloride), are not necessarily suitable for emulsions. The polymers
poly(ethylene terephthalate) and poly(ethylene-vinyl acetate). most commonly used are polyethylene, polypropylene and
The nature and amount of the additives are determined by poly(vinyl chloride). The specifications of this text are to be
the type of the polymer, the process used to convert the read in conjunction with section 3.2.2. Plastic containers and
polymer into the container and the intended purpose of the closures for pharmaceutical use.
container. Additives may consist of antioxidants, stabilisers, The containers may be bags or botdes. They have a site
plasticisers, lubricants, colouring matter and impact modifiers. suitable for the attachment of an infusion set designed to
Antistatic agents and mouId-release agents may be used only ensure a secure connection. They may have a site that allows
for containers for preparations for oral use or for external an injection to be made at the time of use. They usually
use for which they are authorised. Acceptable additives are have a part that allows them to be suspended and which will
indicated in the type specification for each material described withstand the tension occurring during use. The containers
in the Pharmacopoeia. Other additives may be used provided must withstand the sterilisation conditions to which they will
they are approved in each case by the competent authority be submitted. The design of the container and the method of
responsible for the licensing for sale of the preparation. sterilisation chosen are such that aH parts of the containers

414 See the information section on general monographs (cover pages)


EUROPEAN PHARMACOPOEIA 8.0 3.2.3. Sterile plastic containers for human blood

that may be in contact with the infusion are sterilised. The 0112008:30203
container s are impermeable to micro-organisms after closure.
The containers are such that after filling they are resistant to 3.2.3. STERILE PLASTIC CONTAINERS
damage from accidental freezing which may occur during
transport of the final preparation. The containers are and FOR HUMAN BLOOD AND
remain sufficiently transparent to allow the appearance of the BLOOD COMPONENTS
contents to be examined at any time, unless otherwise justified
and authorised. Plastic containers for the collection, storage, processing and
administration of bIood and its components are manufactured
The empty containers display no defects that may lead to from one or more polymers, if necessary with additives.
leakage and the filled and closed containers show no leakage. The composition and the conditions of manufacture of
the containers are registe red by the appropriate competent
For satisfactory storage of sorne preparations, the container authorities in accordance with the relevant nationallegislation
has to be enclosed in a protective envelope. The initial and international agreements.
evaluation of storage has then to be carried out using the When the composition of the materials of the different parts
container enclosed in the envelope. of the container s correspond to the appropriate specifications,
their quality is controlled by the methods indicated in those
specifications (see 3.1. Materials usedfor the manufacture of
TESTS containers and subsections).
Solution S. Use solution S within 4 h of preparation. FiIl a Materials other than those described in the Pharmacopoeia
container to its nominal capacity with water R and close it, may be used provided that their composition is authorised by
if possible using the usual means of closure; otherwise close the competent authority and that the containers manufactured
using a sheet of pure aluminium. Heat in an autoclave so from them comply with the requirements prescribed for Sterile
that a temperature of 121 ± 2 oC is reached within 20 min Plastic Containers for Human Blood and Blood Components.
to 30 min and maintain at this temperature for 30 mino rf In normal conditions of use the materials do not releas e
heating at 121°C leads to deterioration of the container, heat monomers, or other substances, in amounts likely to be
at 100 oC for 2 h. harmful nor do they lead to any abnormal modifications of
Blanlc. Prepare a blank by heating water R in a borosilicate-glass the blood.
flask closed by a sheet of pure aluminium at the temperature The containers may contain anticoagulant solutions,
and for the time used for the preparation of solution S. depending on their intended use, and are supplied sterile.
Appearance of solution S. Solution S is clear (2.2.1) and Each container is fitted with attachments suitable for the
colourless (2.2.2, Method II). intended use. The container may be in the form of a single unit
or the collecting container may be connected by one or more
Acidity 01' alkalinity. To a volume of solution S corresponding tubes to one or more secondary containers to allow separation
to 4 per cent of the nominal capacity of the container add of the blood components to be effected within a closed system.
0.1 mL of phenolphthalein solution R. The solution is The outlets are of a shape and size allowing for adequate
colourless. Add 0.4 mL of 0.01 M sodium hydroxide. The connection ofthe container with the blood-giving equipment.
solution is pink. Add 0.8 mL of 0.01 M hydrochloric acid and The protective coverings on the blood-taking needle and on
0.1 mL of methyl red solution R. The solution is orange-red the appendages must be such as to ensure the maintenance
or red. of sterility. They must be easily removable but must be
Absorbance (2.2.25). Measure the absorbance of solution S tamper-proof.
from 230 nm to 360 nm, using the blank (see solution S) as the The capacity of the containers is related to the nominal
compensation liquido At these wavelengths, the absorbance is capacity prescribed by the national authorities and to the
not greater than 0.20. appropriate volume of anticoagulant solution. The nominal
Redudng substances. To 20.0 mL of solution S add 1 mL capacity is the volume ofblood to be collected in the container.
of dilute sulfuric acid R and 20.0 mL of 0.002 M potassium The containers are of a shape su eh that when filled they may
permanganate. Boil for 3 mino Cool immediately. Add 1 g be centrifuged.
of potassium iodide R and titrate immediately with 0.01 M The containers are fitted with a suitable device for suspending
sodium thiosulfate, using 0.25 mL of starch solution R as or fixing which do es not hinder the collection, storage,
indicator. Carry out a titration using 20.0 mL of the blank. processing or administration of the blood.
The difference between the titration volumes is not greater The containers are enclosed in sealed, protective envelopes.
than 1.5 mL.
CHARACTERS
Transparency. Fill a container previously used for the
preparation of solution S with a volume equal to the nominal The container is sufficiently transparent to allow adequate
capacity of the primary opalescent suspension (2.2.1) visual examination of its contents before and after the taking
of the blood and is sufficiently flexible to offer minimum
diluted 1 in 200 for a container made from polyethylene
resistance during filling and emptying under normal
or polypropylene and 1 in 400 for other containers. The
cloudiness of the suspension is perceptible when viewed conditions of use. The container contains not more than 5 mL
through the container and compared with a similar container of airo
filled with water R. TESTS
Solution SI' Fill the container with 100 mL of a sterile,
LABELLING pyrogen-free 9 gIL solution of sodium chloride R. Close the
container and heat it in an autoclave so that the contents are
The label accompanying a batch of empty containers includes maintained at llO oC for 30 mino
a statement of: If the container to be examined contains an anticoagulant
solution, first empty it, rinse the container with 250 mL of
- the name and address of the manufacturer, water for injections R at 20 ± 1 oC and discard the rinsings.
- a batch number which enables the history of the container Solution S2' Introduce into the container a volume of water
and of the plastic material of which it is manufactured to for injections R corresponding to the intended volume of
be traced. anticoagulant solution. Close the container and heat it in an

General Notices (1) apply to all monographs and other texts 415
3.2.3. Sterile plastic coníal.ners for human blood EUROPEAN PHARMACOPOEIA 8.0

autoclave so that the contents are maintained at 110 oC for Transpareney. Fill the empty container with a volume equal
30 mino After cooling, add sufficient water for injections R to to its nominal capacity of the primary opalescent suspension
fill the container to its nominal capacity. (2.2.1) diluted so as to have an absorbance (2.2.25) at 640 nm
of 0.37 to 0.43 (dilution factor about 1 in 16). The doudiness
If the container to be examined contains an anticoagulant of the suspension must be perceptible when viewed thraugh
solution, first empty it and rinse it as indicated aboye. the bag, as compared with a similar container filled with
Resistance to centrifugation. Introduce into the container a water R.
volume of water R, acidified by the addition of 1 mL of dilute Extractable matter. Tests are carried out by methods designed
hydrochloric acid R, sufficient to fill it to its nominal capacity. to simulate as far as possible the conditions of contact between
Envelop the container with absorbent paper impregnated with the container and its contents which occur in conditions of
a 1 in 5 dilution of bromophenol blue solution Rl or other use.
suitable indicator and then dried. Centrifuge at 5000 g for
The conditiol1s of contact and the tests to be carried out on
10 mino No leakage perceptible on the indicator paper and no
the eluates are prescribed, according to the nature of the
permanent distortion occur. constituent materials, in the particular requirements for each
Resistance to stretch. Introduce into the container a type of container.
volume of water R, acidified by the addition of 1 mL of Haemolytic effeets in buffered systems
dilute hydrochloric acid R, sufficient to fill it to its nominal
capacity. Suspend the container by the suspending device at Stock buffer solution. Dissolve 90.0 g of sodium chloride R,
the opposite end from the blood-taking tube and apply along 34.6 g of disodium hydrogen phosphate R and 2.43 g of sodium
the axis of this tube an immediate force of 20 N (2.05 kgf). dihydrogen phosphate R in water R and dilute to 1000 mL with
Maintain the traction for 5 s. Repeat the test with the force the same solvent.
applied to each of the parts for filling and emptying. No break Buffer solution Ao. To 30.0 mL of stock buffer solution add
and no deterioration occur. 10.0 mL of water R.
Leakage. Place the container which has been submitted to Buffer solution B o. To 30.0 mL of stock buffer solution add
the stretch test between two plates covered with absorbent 20.0 mL of water R.
paper impregnated with a 1 in 5 dilution of bromophenol Buffer solution Co. To 15.0 mL of stock buffer solution add
blue solution Rl or other suitable indicator and then dried. 85.0 mL of water R.
Progressively apply force to the plates to press the container
so that its internal pressure (i.e. the difference between the Introduce 1.4 mL of solution S2 into each of three centrifuge
applied pressure and atmospheric pressure) reaches 67 kPa tubes. To tube 1 add 0.1 mL ofbuffer solution Ao' to tube Ir
within 1 mino Maintain the pressure for 10 mino No signs of add 0.1 mL ofbuffer solution Bo and to tube III add 0.1 mL
leakage are detectable on the indicator paper or at any point of of buffer solution Co. To each tube add 0.02 mL of fresh,
attachment (seals, joints, ete.). heparinised human blood, mix well and warm on a water-bath
at 30 ± 1 oC for 40 mino Use blood collected less than
Vapour permeability. For a container containing an 3 h previously or blood collected into an anticoagulant
anticoagulant solution, fill with a volume of a 9 giL solution of citrate-phosphate-dextrose solution (CPD) less than 24 h
sodium chloride R equal to the volume ofblood for which the previously.
container is intended.
Prepare three solutions containing, respectively:
For an empty container, fill with the same mixture of 3.0 mL of buffer solution Aa and 12.0 mL of water R
anticoagulant solution and sodium chloride solution. Close (solution Al)'
the container, weigh it and store it at 5 ± 1 oC in an atmosphere
4.0 mL ofbuffer solution Bo and 11.0 mL of water R
with a relative humidity of (50 ± 5) per cent for 21 days. At (solution Bl ),
the end of this period the 10ss in mass is 110t greater than 1 per
cent. 4.75 mL ofbuffer solution Bo and 10.25 mL of water R
(solution C l ).
Emptying under pressure. Fill the container with a volume
of water R at 5 ± 1 oC equal to the nominal capacity. Attach To tubes 1, II and III add, respectively, 1.5 mL of solution Al'
a transfusion set without an intravenous cannula to one of 1.5 mL of solution Bl and 1.5 mL of solution C l . At the same
the connectors. Compress the container so as to maintain time and in the same manner, prepare three other tubes,
throughout the emptying an internal pressure (i.e the replacing solution S2 by water R. Centrifuge simultaneously
difference between the applied pressure and atmospheric the tubes to be examined and the control tubes at exactly
pressure) of 40 kPa. The container empties in less than 2 mino 2500 g in the same horizontal centrifuge for 5 mino After
centrifuging, measure the absorbances (2.2.25) of the liquids
Speed of filling. Attach the container by means of the at 540 11m using the stock buffer solution as compensation
blood-taking tube fitted with the needle to a reservoir liquido Calculate the haemolytic value as a percentage fram
containing a suitable solution having a viscosity equal to the express ion :
that of blood, such as a 335 giL solution of sucrose R at
37 oc. Maintain the internal pressure of the reservoir (i.e. Ae."p x 100
the difference between the applied pressure and atmospheric A lOO
pressure) at 9.3 kPa with the base ofthe reservoir and the
upper part of the container at the same leve!. The volume of absorbance of tube lII,
liquid which flows into the container in 8 min is not less than
.the nominal capacity of the container. absorbance of tube 1 or II or of the corresponding
control tubes.
Resistanee to temperature variations. Place the container
in a suitable chamber having an initial temperature of The solution in tube 1 gives a haemolytic value not greater
20-23 oc. Cool it rapidly in a deep-freeze to - 80 oC and than 10 per cent and the haemolytic value of the solution in
maintain it at this temperature for 24 h. Raise the temperature tube JI does not differ by more than 10 per cent from that of
to 50 oC and maintain for 12 h. Allow to cool to room the corresponding control tube.
temperature. The container complies with the tests for Sterility (2.6.1). The containers comply with the test for
resistance to centrifugation, resistance to stretch, leakage, sterility. Introduce aseptically into the container 100 mL of
vapour permeability emptying under pressure and speed of a sterile 9 giL solution of sodium chloride and shake the
filling prescribed aboye. container to ensure that the interna! surfaces have been

416 See the information section on general monographs (ca ver pages)
EUROPEAN PHARMACOPOEIA 8.0 3.2.4. Sterile coníainers of plasticised PVC for human blood

entirely wetted. Filter the contents of the container through a hydroxide. The solution is pink. Add 0.8 mL of 0.01 M
membrane filter and place the membrane in the appropriate hydrochloric acid and 0.1 mL of methyl red solution R. The
culture medium, as prescribed in the test for sterility. solution is orange-red or red.
Pyrogens (2.6.8). Solution SI complies with the test for Absorbance (2.2.25): maximum 0.30, determined between
pyrogens. Injeet 10 mL of the solution per kilogram of the wavelengths of 230 nm and 250 nm on solution S2; maximum
rabbit's mass. 0.10, determined between wavelengths of251 nm and 360 nm
Abnormal toxicity (2.6.9). Solution SI eomplies with the test on solution S2. Use the reference solution as the compensation
for abnormal toxicity. Inject 0.5 mL of the solution into each liquido
mouse. Oxidisable substances. Immediately after preparation of
solution S2 (see 3.2.3), transfer to a borosilicate-glass flask a
PACKAGING quantity corresponding to 8 per cent of the nominal capacity
The container s are packed in protective envelopes. of the container. At the same time, prepare a blank using an
On removal from its protective envelope the container shows equal volume of the freshly prepared reference solution in
another borosilicate-glass flask. To each solution add 20.0 mL
no leakage and no growth of micro-organisms. The protective
envelope is sufficiently robust to withstand normal handling. of 0.002 M potassium permanganate and 1 mL of dilute sulfuric
acid R. Allow to stand protected from light for 15 mino To
The protective envelope is sealed in such a manner that it each solution add 0.1 g of potassium iodide R. Allow to stand
cannot be opened and re-closed without leaving visible traces protected from light for 5 min and titrate immediately with
that the seal has been broken. 0.01 M sodium thiosulfate, using 0.25 mL of starch solution R
as indicator. The difference between the 2 titrations is not
LABELLING
more than 2.0 mL.
The labelling complíes with the relevant nationallegislation
Extractable di(2-ethylhexyl) phthalate. Extraction solvent,
and international agreements. The label states:
ethanol (96 per cent) R diluted with water R to have a
- the name and address of the manufacturer, relative density (2.2.5) of 0.9389 to 0.9395, measured with a
- a batch number which enables the history of the container pycnometer.
and of the plastic material of which it is manufactured to Stock solution. Dissolve 0.100 g of di(2-ethylhexyl) phthalate R
be traced. in the extraction solvent and dilute to 100.0 mL with the same
A part of the label is reserved for: solvent.
- the statement of the blood group, the reference number and Standard solutions. lnto 5 separate 100 mL volumetric flasks,
all other information required by nationallegislation or introduce respectively 1.0 mL, 2.0 mL, 5.0 mL, 10.0 mL
international agreements, and an empty space is provided and 20.0 mL of stock solution and dilute to 100.0 mL with
for the insertion of supplementary labelling. extraction solvent.
The label of the protective envelope or the label on the Measure the absorbances (2.2.25) of the standard solutions
container, visible through the envelope, states: at the absorption maximum at 272 nm, using the extraction
solvent as compensation liquid and plot a curve of absorbance
- the expiry date, against the concentration of di(2-ethylhexyl) phthalate.
- that, once withdrawn from its protective envelope, the Extraetíon procedure. Using the donor tubing and the needle
container must be used within 10 days. or adaptor, fill the empty container with a volume equal
The ink or other substance used to print the labels or the to half the nominal volume with the extraction solvent,
writing must not diffuse into the plastic material of the previously heated to 37 oC in a well-stoppered flask. Expel the
container and must remain legible up to the time of use. air completely from the container and seal the donor tube.
lmmerse the filled container in a horizontal position in a
water-bath maintained at 37 ± 1 oC for 60 ± 1 min without
shaking. Remove the container from the water-bath, invert
0112008:30204 it gently 10 times and transfer the contents to a glass flask.
Immediately measure the absorbance at the maximum at
3,2.4. EMPTY STERILE CONTAINERS 272 nm, using the extraction solvent as compensation liquido
Determine the concentration of di(2-ethylhexyl) phthalate in
OF PLASTICISED POLY(VINYL milligrams per 100 mL of extract from the calibration curve.
CHLORIDE) FOR HUMAN BLOOD The concentration does not exceed:
AND BLOOD COMPONENTS - 10 mg per 100 mL for containers of nominal volume greater
than 300 mL but not greater than 500 mL;
Unless otherwise authorised as described under Sterile plastic - 13 mg per 100 mL for containers of nominal volume greater
containers for human blood and blood components (3.2.3), than 150 mL but not greater than 300 mL;
the nature and composition of the material from which - 14 mg per 100 mL for containers of nominal volume up
the containers are made comply with the requirements for to 150 mL.
Materials based on plastieised poly(vinyl chloride) for containers
for human blood and blood components and for containers for Chlorides (2.4.4): maximum 0.4 ppm, determined with
aqueous solutions for intravenous infusion (3.1.1). solution S2.
Prepare the standard using a mixture of 1.2 mL of chloride
TESTS standard solution (5 ppm el) R and 13.8 mL of water R.
They comply with the tests prescribed for Sterile plastic Ammonium (2.4.1): maximum 2 ppm.
containers for human blood and blood components (3.2.3) and
Dilute 5 mL of solution S2 to 14 mL with water R.
with the following tests to detect extractable matter.
Residue on evaporation. Evaporate to dryness 100 mL
Reference solution. Heat water for injections R in a of solution S2 in a borosilicate-glass beaker of appropriate
borosilicate-glass flask in an autoclave at 110 oC for 30 mino capacity, previously heated to 105 oc. Evaporate to dryness in
Addity or alkalinity. To a volume of solution S2 the same conditions 100 mL of the reference solution (blank
corresponding to 4 per cent of the nominal capacity of the test). Dry to constant mass at 100-105 oc. The residue from
container add 0.1 mL of phenolphthalein solution R. The solution S2 weighs a maximum of 3 mg, allowing for the blank
solution remains eolourless. Add 0.4 mL of 0.01 M sodium test.

General Notices (1) apply to all monographs and other texts 417
3.2.5. Coníainers of plastidsed PVC with anticoagulant solution EUROPEAN PHARMACOPOEIA 8.0

PACKAGING 0112008:30206
See Sterile plastic containers for human blood and blood
components (3.2.3). 3.2.6. SETS FOR THE TRANSFUSION OF
BLOOD AND BLOOD COMPONENTS
LABELLING DEFINITION
See Sterile plastic containers for human blood and blood Sets for the transfusion of blood and blood components
components (3.2.3). consist principally of plastic tubing to which are fitted the
parts necessary to enable the set to be used for transfusion
in the appropriate manner. Sets indude a dosure-piercing
device, a blood filter, a drip chamber, a flow regulator, a Luer
connector and, usually, a site that allows an injection to be
made at the time of use. When the sets are to be used with
01/2008:30205
containers requiring an air-filter, this may be incorporated
in the dosure-piercing device or a separate air-inlet device
3.2.5. STERILE CONTAINERS may be used. The ehamber endosing the blood filter, the drip
chamber and the main tubing are transparent. The materials
OF PLASTICISED POLY(VINYL chosen and the design of the set are such as to ensure absence
CHLORIDE) FOR HUMAN BLOOD of haemolytic effects. The sets comply with current standard s
regardil1g dimensions and performance.
CONTAINING ANTICOAGULANT
AlI parts of the set that may be in contact with blood and
SOLUTION blood components are sterile and pyrogen-free. Each set is
presented in an individual package that maintains the sterility
Sterile plastic containers contail1ing an anticoagulant solution of the contents. The sets are not to be re-sterilised or re-used.
complying with the monographAnticoagulant and preservative Sets for the transfusion of blood and blood components
solutions for human blood (0209) are used for the collection, are manufactured in accordance with the rules of good
storage and administration of blood. Before filling they manufaeturing practice for medical devices al1d any relevant
comply with the description and characters givel1 under Empty national regulations.
sterile containers of plasticised poly(vinyl chloride) for human
blood and blood components (3.2.4). TESTS
Unless otherwise authorised as described under Sterile plastic Carry out the tests on sterilised sets.
containers for human blood and blood components (3.2.3), Solution S. Make a c10sed circulation system from 3 sets
the nature and composition of the material from which the and a 300 mL borosilicate-glass vessel. Fit to the vessel a
containers are made should comply with the requirements suitable thermostat device that maintains the temperature
prescribed for Materials based on plasticised poly(vinyl of the liquid in the vessel at 37 ± 1 oc. Circulate 250 mL of
chloride) for containers for human blood and blood components water for injections R through the system in the directiol1
and for containers for aqueous solutions for intravenous used for transfusion for 2 h at arate of 1 Llh (for example
infusion (3.1.1). using a peristaltic pump applied to as short a piece of suitable
silicone elastomer tubing as possible). Colleet the whole of
TESTS the solution and allow to cool.
Appearance oi solution. Solution S is dear (2.2.1) and
They comply with the tests prescribed for Sterile plastic
colourless (2.2.2, Method II).
containers for human blood and blood components (3.2.3)
and with the following tests to measure the volume of Addity 01' alkalil1ity. To 25 mL of solution S add 0.15 mL of
anticoagulant solution al1d to detect extractable matter. BRP indicator solution R. Not more than 0.5 mL of 0.01 M
sodium hydroxide is required to change the colour of the
Volume of anticoagulant solution. Empty the container, indicator to blue. To 25 mL of solution S add 0.2 mL of
collecting the anticoagulant solution in a graduated cylinder. methyl orange solution R. Not more than 0.5 mL of 0.01 M
The volume does not differ by more than ± 10 per cent from hydrochloric acid is required to reach the beginning of the
the stated volume. colour ehange of the indicator.
Spectrophotometric examination (2.2.25). Measure the Absorbance (2.2.25): maximum 0.30, determined between
absorbance of the anticoagulant solution from the container wavelengths of 230 nm and 250 nm on solution S; maximum
between 250 nm and 350 nm, using as the compensation liquid 0.15, determined between wavelengths of 251 nm and 360 nm
an anticoagulant solution of the same composition that has on solution S.
not been in contact with a plastic material. The absorbance at
the maximum at 280 nm is not greater than 0.5. Reducing substances. Carry out the test within 4 h of
preparation of solution S. To 20.0 mL of solution S add 1 mL
Extractable di(2-ethylhexyl) phthalate. Carefully remove of dilute sulfuric acid R and 20.0 mL of 0.002 M potassium
the anticoagulant solution by means of the flexible transfer permanganate. Boil for 3 min and cool immediately. Add
tube. Using a funnel fitted to the tube, completely fill the 1 g of potassium iodide R and titrate with 0.01 M sodium
container with water R, leave in contact for 1 min squeezing thiosulfate using 0.25 mL of starch solution R as indicator.
the container gently, then empty completely. Repeat the Carry out a blank test using 20 mL of water for injections R.
rinsing. The difference between the titration volumes is not greater
The container, so emptied and rinsed, complies with the test than 2.0 mL.
for extractable di(2-ethylhexyl) phthalate prescribed for Empty Ethylene oxide. Gas chromatography (2.2.28).
sterile plastic containers of plasticised poly(vinyl chloride) for Column:
human blood and blood components (3.2.4).
- material: stainless steel;
- size: 1= 1.5 m, 0 = 6.4 mm;
PACKAGING AND LABELLING
- stationary phase: silanised diatomaceous earth for gas
See Sterile plastic containers for human blood and blood chromatography R impregnated with macrogol1500 R (3 g
components (3.2.3). per 10 g).

418 See the information section on general monographs (cover pages)


EUROPEAN PHARMACOPOEIA 8.0 3.2.8. Sterile single-use plastic syringes

Carrier gas: helium for chromatography R. a tank of water at 20-23 oc. Apply progressive!y an excess
Flow rate: 20 mL/min. pressure of 100 kPa and maintain for 1 mino No air bubble
escapes from the set.
Temperature:
Transparency. Use as reference suspension the primary
- column: 40 oC;
opalescent suspension (2.2.1) diluted 1 in 8 for sets having
- injection port: 100 oC; tubing with an external di ame ter less than 5 mm and diluted
- detector: 150 oc. 1 in 16 for sets having tubing with an external diameter of
5 mm or greater. Circulate the reference suspension through
Detection: flame ionisatioll.
the set and compare with a set from the same batch filled
Verify the absence of peaks interfering with the ethylene with water R. The opalescence and presence of bubbles are
oxide peak by carrying out the test using an unsterilised set discernible.
or using the same chromatographic system with the following
modifications. Residue on evaporation. Evaporate 50.0 mL of solution S
to dryness on a water-bath and dry to constant mass in an
Column: oven at 100-105 oc. Carry out a blank test using 50.0 mL of
- size: 1= 3 m, 0 = 3.2 mm; water for injections R. The difference between the masses of
stationary phase: silanised diatomaceous earth the residues is not greater than 1.5 mg.
for gas chromatography R impregnated with Sterility (2.6.1). The sets comply with the test for sterility. If
triscyanoethoxypropane R (2 g per 10 g); the sets are stated to be sterile only internally, pass 50 mL of
- temperature: 60 oc. buffered sodium chloride-peptone solution pH 7.0 (2.6.12)
through the set and use to carry out the test by the membrane
Ethylene oxide solution. Prepare in a fume cupboard. Place filtration method.
50.0 mL of dimethylacetamide R in a 50 mL vial, stopper,
secure the stopper and weigh to the nearest 0.1 mg. Fill a If the sets are stated to be sterile both internally and externally,
50 mL polyethylene or polypropylene syringe with gaseous open the package with the necessary aseptic precautions and:
ethylene oxide R, allow the gas to remain in contact with the - for the direct inoculation method, place the set or its
syringe for about 3 min, empty the syringe and fin again with components in a suitable container containing a sufficient
50 mL of gaseous ethylene oxide R. Fit a hypodermic needle to quantity of the culture medium to ensure complete
the syringe and reduce the volume of gas in the syringe from immersion;
50 mL to 25 mL. Inject these 25 mL of ethylene oxide slowly - for the membrane filtration method, place the set or its
into the vial, shaking gently and avoiding contact between components in a suitable container containing a sufficient
the needle and the liquido Weigh the vial again: the increase quantity of buffered sodium chloride-peptone solution
in mass is 45 mg to 60 mg and is used to calculate the exact pH 7.0 (2.6.12) to allow total rinsing for 10 mino
concentration of the solution (about 1 giL). Pyrogens (2.6.8). Connect together 5 sets and pass through
Test. Weigh the set after removing the package. Cut the set into the assembly at a flow rate not exceeding 10 mL/min 250 mL
pieces of maximum dimension 1 cm and place the pieces in a of a sterile, pyrogen-free 9 giL solution of sodium chloride R.
250-500 mL vial containing 150 mL of dimethylacetamide R. Collect the solution aseptically in a pyrogen-free container.
Close the vial with a suitable stopper and secure the stopper. The solution complies with the test for pyrogens. Inject per
Place the vial in an oven at 70 ± 1 oC for 16 h. Remove 1 mL of kilogram of the rabbit's mass, 10 mL of the solution.
the hot gas from the vial and inject it onto the column. From
the calibration curve and the height of the peak obtained, LABELLING
calculate the mas s of ethylene oxide in the vial. The ¡abe! states, where applicable, that the set has been
Calibration curve. In a series of 7 vials of the same type sterilised using ethylene oxide.
as that used for the test and each containing 150 mL of
dimethylacetamide R, place respectively O mL, 0.05 mL,
01/2008:30208
0.10 mL, 0.20 mL, 0.50 mL, l.00 mL and 2.00 mL of the
ethylene oxide solution, i.e. about O ¡..tg, 50 ¡..tg, 100 ¡..tg, 200 ¡..tg,
500 ¡..tg, 1000 ¡..tg and 2000 ¡..tg of ethylene oxide. Stopper the 3.2.8. STERILE SINGLE- USE PLASTIC
vials, secure the stoppers and place the vials in an oven at SYRINGES
70 ± 1 oC for 16 h. Inject 1 mL of the hot gas from each vial
onto the column and draw a calibration curve from the heights DEFINITION
of the peaks and the mass of ethylene oxide in each flask. Sterile single-use plastic syringes are medical devices intended
Limit: if the label states that ethylene oxide has been used for for immediate use for the administration of injectable
sterilisation: preparations. They are supplied sterile and pyrogen-free
and are not to be re-sterilised or re- used. They consist of a
- ethylene oxide: maximum 10 ppm.
syringe barre! and a piston which may have an elastomer
Extraneous partides. Fill the set via the normal inlet with a sealing ring; they may be fitted with a needle which may be
0.1 giL solution of sodium laurilsulfate R, previously filtered non-detachable. Each syringe is presented with individual
through a sintered-glass filter (16) (2.1.2) and heated to 37 oc. protection for maintaining sterility.
Collect the liquid via the normal outlet. When examined The barre! of the syringe is sufficiently transparent to permit
under suitable conditions of visibility, the liquid is clear and dosages to be read without difficulty and allow air bubbles and
practically free from visible particles and filaments (it is foreign particles to be discerned.
assumed that particles and filaments with a diameter equal to
The plastics and e!astomer materials of which the barre! and
or greater than 50 ¡..tm are visible to the naked eye).
pisto n are made comply with the appropriate specification
Flow rateo Pass through a complete set with the flow regulator or with the requirements of the competent authority. The
fully open 50 mL of a solutíon having a viscosity of 3 mPa·s most commonly used materials are polypropylene and
(3 cP) (for example a 33 giL solution of macrogol 4000 R at polyethylene. The syringes comply with current standards
20 OC) under a static head of 1 m. The time required for regarding dimensions and performance.
passage of 50 mL of the solution is not greater than 90 s. Silicone oil (3.1.8) may be applied to the internal wall of the
Resistance to pressure. Make tight the extremities of the set barre! to assist in the smooth operation of the syringe but there
and any air-inlet device. Connect the set to a compressed remains no excess capable of contaminating the contents at
air outlet fitted with a pressure regulator. Immerse the set in the time of use. The inks, glues and adhesives for the marking

General Notices (1) apply to all monographs and other texts 419
3.2.8. Sterile single-use plastic syringes EUROPEAN PHARMACOPOEIA 8.0

on the syringe or on the package anO., where necessary, the Test, Weigh the syringe after removing the package, Cut the
assembly of the syringe anO. its package, do not migrate acrosssyringe into pieces of maximum dimension 1 cm and place
the walls, the pieces in a 250 mL to 500 mL vial containing 150 mL of
dimethylacetamide R. Close the vial with a suitable stopper
TESTS anO. secure the stopper. Place the vial in an oven at 70 ± 1 oC
Solution S, Prepare the solution in a manner that avoids for 16 h, Remove 1 mL of the hot gas from the vial anO. inject
contamination by foreign particles, Using a sufficient number it onto the column, From the calibration curve anO. the height
of syringes to produce 50 mL of 501ution, fill the syringes to of the peak obtained, calculate the mass of ethylene oxide in
their nominal volume with water for injections RanO. maintain the vial.
at 37°C for 24 h, Combine the contents of the syringes in a Limit: if the label states that ethylene oxide has been used for
suitable borosilicate-glass container. sterilisation:
Appearance of solution, Solution S is clear (2,2,1) anO. - ethylene oxide: maximum 10 ppm.
colourless (2.2,2, Method JI) anO. is practically free from Silicone oil, Calculate the internal surface area of a syringe in
foreign soliO. particles, square centimetres using the following expression:
Addity or alkalinity, To 20 mL of solution S add 0,1 mL
of bromothymol blue solution R1, Not more than 0.3 mL 2VV'7r,h
of 0,01 M sodium hydroxide or 0,01 M hydrochloric acid is
required to change the colour of the indicator, V nominal volume of the syringe, in cubic
centimetres;
Absorbance (2.2,25): maximum 0040, determined between
wavelengths of 220 nm anO. 360 nm on 501ution S, h height of the graduatiol1, in centimetres,
Ethylene oxide, Gas chromatography (2,2,28), Take a sufficient number of syringes to give an internal surface
are a of 100 cm 2 to 200 cm 2 , Aspirate into each syringe a
Column:
volume of methylene ehloride R equal to half the nominal
- material: stainless steel; volume and make up to the nominal vo1ume with air, Rinse
- size: 1= 1.5 m, 0 = 6.4 mm; the internal surface corresponding to the nominal volume with
- stationary phase: silanised diatomaceous earth for gas the solvent by inverting the syringe ten times in succession
chromatography R impregnated with macrogol1500 R (3 g with the needle fitting closed by a finger covered by a plastic
per 10 g), film inert to methylene chloride, Expel the extracts into a
tared dish anO. repeat the operation, Evaporate the combined
Carrier gas: helium for chromatography R,
extracts to dryness on a water-bath, Dryat 100-105 oC for
Flow rate: 20 mL/min, 1 h, The residue weighs not more than 0,25 mg per square
Temperature: centimetre of internal surface area,
- Column: 40 oC; Examine the residue by infrared absorption spectrophotometry
- Injection port: 100 oC; (2.2.24), It shows absorption bands typical of silicone oil at
805 cm- l, 1020 cm-l, 1095 cm- l, 1260 cm- 1 anO. 2960 cm-l.
- Detector: 150 oc,
Deteetion: flame ionisation, Redudng substances, To 20.0 mL of 501ution S add
2 mL of sulfuric acid RanO. 20,0 mL of 0,002 M potassium
Verify the absence of peaks interfering with the ethylene oxide permanganate. Boil for 3 min, Cool immediately, Add 1 g
peak, either by carrying out the test using an unsterilised of potassium iodide RanO. titrate immediately with 0,01 M
syringe or using the same chromatographic system with the sodium thiosulfate using 0,25 mL of stareh solution R as
following modifications: indicator. Carry out a blank titration using 20.0 mL of water
Column: for injections R. The difference between the titratiol1 volumes
- size: 1= 3 m, 0 = 3,2 mm; is not greater than 3,0 mL
- stationary phase: silanised diatomaceous earth Transparency, Fill a syringe with water R (blank) anO.
for gas chromatography R impregnated with fill another with a 1 in 10 dilution of primary opalescent
triscyanoethoxypropane R (2 g per 10 g); sllspension (2.2,1), Use primary opalescent suspension that
- tempera tu re: 60 oc, has been allowed to stand at 20 ± 2 oC for 24 h before use.
Compare with the naked eye in diffused light against a dark
Ethylene oxide solution, Prepare in a fume cupboard, Place background, The opalescence of the suspension is detectable
50.0 mL of dimethylacetamide R in a 50 mL vial, stopper, when compared with the blank.
secure the stopper anO. weigh to the nearest 0,1 mg, Fill a
50 mL polyethylene or polypropylene syringe with gaseous Sterility (2,6,1), Syringes stated to be sterile comply with the
ethylene oxide R, allow the gas to remain in contact with the test for sterility earried out as follows, Using aseptic technique,
syringe for about 3 min, empty the syringe anO. fill again with open the package, withdraw the syringe, separate the
50 mL of gaseous ethylene oxide R. Fit a hypodermic needle to components and place each in a suitable container containing
the syringe anO. reduce the volume of gas in the syringe from sufficient culture media to cover the part completely, Use both
50 mL to 25 mL Inject these 25 mL of ethylene oxide slowly the recommended media (2,6,1),
into the vial, shaking gently anO. avoiding contact between Syringes stated to be sterile only internally comply with the test
the needle anO. the liquid, Weigh the vial again: the increase for sterility carried out as follows, Use 50 mL of inoculation
in maS5 is 45 mg to 60 mg anO. i5 used to calculate the exact medium for each test syringe, Using aseptic technique, remove
concentration of the solution (about 1 giL), the needle protector and submerge the needle in the culture
Calibration curve, In a series of seven vials of the same medium, Flush the syringe five times by withdrawing the
type as that used for the test anO. each containing 150 mL plunger to its fullest extent.
of dimethylacetamide R, place respectively O mL, 0,05 mL, Pyrogens (2,6,8), Syringes with a nominal volume equal to or
0,10 mL, 0,20 mL, 0.50 mL, LOO mL and 2,00 mL ofthe greater than 15 mL comply with the test for pyrogens, Fill a
ethylene oxide solution, Le, about O flg, 50 [lg, 100 [lg, 200 Ilg, minimum of three syringes to their nominal volume with a
500 flg, 1000 Ilg anO. 2000 Ilg of ethylene oxide, Stopper the pyrogen-free 9 giL solution of sodium ehloride RanO. maintain
vials, secure the stoppers anO. place the vials in an oven at at a temperature of 37 oC for 2 h, Combine the solutions
70 ± 1 oC for 16 h, Inject 1 mL of the hot gas from each vial aseptically in a pyrogen-free container and carry out the test
onto the column and draw a calibration curve from the heights immediately, Inject per kilogram of the rabbit's mass 10 mL of
of the peaks anO. the mass of ethylene oxide in each flask the solution.

420 See ¡he information section on general monographs (ca ver pages)
EUROPEAN PHARMACOPOElA 8.0 3.2.9. Rubber dosul"es fOI" coniainers

LABELLING swelling may occur. They are homogeneous and practically


The label on the package states: free from flash and adventitious materials (for example, fibres,
foreign particles, waste rubber).
- the batch number;
Identificatíon of the type of rubber used for the closures is not
- a description of the syringe; within the scope of this specification. The identification tests
- that the syringe is for single-use only. given be/ow distinguish between closures made from rubber
The label on the outer package states: and those made from silicone elastomer and plastie materials
but do not differentiate all types of rubber. Other identity
- the method of sterilisation; tests may be carried out with the aim of detecting differences
- that the syringe is sterile or that it is sterile only internally; in a batch compared with the e/osures used for compatibility
- the identity of the manufacturer; testing. One or more of the following analytical methods
may be applied for this purpose: determination of relative
- that the syringe is not to be used if the packaging is density, determination of sulfated ash, determination of sulfur
damaged or the sterility protector is loose. content, thin-Iayer chromatography carried out on an extraet,
ultraviolet absorption spectrophotometry of an extract, infrared
absorption spectrophotometry of a pyrolysate or attenuated
total reflectance (ATR).

-
01/2014:30209
IDENTIFICATION
3.2.9. RUBBER CLOSURES FOR A. Infrared absorption spectrophotometry (2.2.24).
CONTAINERS FOR AQUEOUS Examine by attenuated total reflectan ce (ATR). The
PARENTERAL PREPARATIONS, spectrum obtained is identical to the spectrum obtained
with the type sample. rf necessary, cut the sample along an
FOR POWDERS AND FOR appropriate axis, examine the cut surface and compare the
FREEZE-DRIED POWDERS spectrum with that obtained with the type sample prepared
in the same way.
Rubber do sures for containers for aqueous parenteral lf direct ATR measurement on the surface is not feasible
preparations, for powders and for freeze-dried powders are (mainly rubber closures filled with carbon black), heat 1-2 g
made of materials obtained by vulcanisation (cross-linking), in a heat-resistant test-tube over an open flame to dry the
using appropriate additives, of macromolecular organic sample and continue heating until pyrolysate vapours are
substances (elastomers). The elastomers are produced from condensed near the top edge of the test -tube. Examine the
natural or synthetic substances by polymerisation. The choice pyrolysate of the sample by ATR and compare the spectrum
of the principal components and of the various additives with that obtained with the pyrolysate of the type sample.
(for example, vulcanisers, accelerators, stabilisers, pigments)
B. Total ash (2.4.16).
depends on the properties required for the finished artide.
The specifications do not apply to do sures made from silicone Determine the percentage content of total ash in the sample
elastomer (which are dealt with in chapter 3.1.9. Silicone to be examined and compare with the percentage content
elastomer for e/osures and tubing), to laminated dosures or of total ash in the type sample (A o)' The total ash content
to lacquered dosures. fans within the following ranges depending on the total ash
content of the type sample.
Rubber dosures may be dassified in 2 types: type 1 dosures
meet the strictest requirements and are preferred; type II Total ash in the type sample, Limit for total ash in tile
Ao (per cent) sample (per cent)
dosures have mechanical properties suitable for special
uses (for example, multiple piercing) and cannot meet Ao oS 5.0 (Ao - 0.75) to (Ao + 0.75)
requirements as severe as for type 1 closures because of their 5.0 < Ao oS 10 (A o - 1.0) to (A o + 1.0)
chemical composition.
Ao> 10 (Ao - 2.0) to (A o + 2.0)
The closures chosen for use with a particular preparation are
such that: In addition to the use of platinum and silica crucibles
- the components of the preparation in contact with the described in general chapter 2.4.16, porcelain crucibles may
closures are not adsorbed onto the surface of the do sures be used. The sample may be ignited using a microwave
and do not migrate into or through the closures to an oven instead of a muffle furnace.
extent sufficient to affect the preparation adversely;
TESTS
- the closures do not releas e substances in quantities
sufficient to affect the stability of the preparation or to The samples to be analysed may be washed and sterilised before
present a risk of toxicity. use.
The closures are compatible with the preparation for which Solution S. Place a number of un cut closures with a total
they are used throughout its period of validity. surface area of about 100 cm 2 in a suitable glass container,
cover with water R, boH for 5 min and rinse 5 times with cold
The manufacturer of the preparation must obtain from the water R. Place the washed closures in a wide-necked flask
supplier an assurance that the composition of the closure (type 1 glass, 3.2.1), add 200 mL of water R and weigh. Cover
does not vary and that it is identical to that of the closure the mouth of the flask with a borosilicate-glass beaker. Heat
used during compatibility testing. lf the supplier informs the in an autoclave so that a temperature of 121 ± 2 oC is reached
manufacturer of the preparation that changes have been made within 20-30 min and maintain at this temperature for 30 mino
to the composition, compatibility testing must be repeated, Cool to room temperature over about 30 mino Make up to
totally or partly, depending on the nature of the changes. the original mass with water R. Shake and decant the solution
The closures are washed and may be sterilised before use. immediately. Shake solution S before each test.
IÍusing a tightly closed flask (type 1 glass, 3.2.1) with an
CHARACTERS inert dosure instead of a wide- necked flask covered with a
Rubber closures are elastic. They are translucent or opaque borosilicate-glass beaker, it is 110t necessary to make up to the
and have no characteristic colour, the latter depending original mass.
on the additives used. They are practically insoluble in Blank solution. Prepare a blank solution in the same manner
tetrahydrofuran, in which, however, a considerable reversible using 200 mL of water R.

General Notices (1) apply to all monographs and other texts 421
3.2.9. Rubber dosures for coníainers EUROPEAN PHARMACOPOEIA 8.0

Appearance of solution S. For type l closures, solution S is Volatile sulfides. Place closures, cut if necessary, with a total
not more opalescent than reference suspension II (2.2.1) and sur fa ce area of 20 ± 2 cm 2 in a 100 mL conical flask and add
for type II closures, solution S is not more opalescent than 50 mL of a 20 giL solution of citric acid R. Place a piece of
reference suspension lII. Solution S is not more intensely lead acetate paper R over the mouth of the flask and maintain
coloured than reference solution GYs (2.2.2, Method II). the paper in position by placing over it an inverted weighing
Addity or alkalinity. To 20 mL of solution S add 0.1 mL bottle. Heat in an autoclave at 121 ± 2 oC for 30 mino Any black
of bromothymol blue solution R1. Not more than 0.3 mL of 5tain on the paper is not more intense than that of a standard,
0.01 M sodium hydroxide or 0.8 mL of 0.01 M hydrochloric treated in the same manner, prepared by mixing 50 mL of a
acid is required to change the colour of the indicator to blue 20 giL solution of citrie acid R and 5.0 mL of a freshly prepared
or yellow, respectively. 0.0308 giL solution of sodium sulfide R in water R.
Por the tests for penetrabi/ity, fragmentation and self-sealing,
Absorbance. Carry out the test within 5 h of preparation treat the e/osures as described for the preparation of so/ution S
of solution S. Filter solution S through a membrane filter and allow to dry.
(nominal pore size 0.45 flm), rejecting the first few millilitres
of filtrate. Measure the absorbance (2.2.25) of the filtrate at Penetrability. For closures intended to be pierced by a
wavelengths from 220-360 nm using the blank (see solution S) hypodermic needle, carry out the following test Fill 10
as compensation liquido At these wavelengths, the absorbance suitable vials to the nominal volume with water R, fit the
do es not exceed 0.2 for type l closures or 4.0 for type II closures to be examined and secure with a cap. Using for each
closures. lf necessary, dilute the filtrate before measurement closure a new, lubricated, 10ng-bevelO) (bevel angle 12 ± 20 )
of the absorbance and correct the result for the dilution. hypodermic needle with an external diameter of 0.8 mm,
pierce the closures with the needle perpendicular to the
Redudng substances. Carry out the test within 4 h of surface. The force required for piercing, determined with an
preparation of solution S. To 20.0 mL of solution S add accuracy of ± 0.25 N, is not greater than 10 N for each closure.
1 mL of dilute sulfuric acid R and 20.0 mL of 0.002 M
potassium permanganate. Boil for 3 mino Cool. Add 1 g Fragmentation. For closures intended to be pierced by a
of potassium iodide R and titrate immediately with 0.01 M hypodermic needle, carry out the following test. If the closures
sodium thiosulfate, using 0.25 mL of starch solution R as are to be used for aqueous preparations, introduce in 12
indicator. Carry out a titration using 20.0 mL of the blank. clean vials a volume of water R corresponding to the nominal
The difference between the titration volumes is not greater volume minus 4 mL, close the vials with the closures to be
than 3.0 mL for type l closures and 7.0 mL for type II closures. examined, secure with a cap and allow to stand for 16 h. lf the
closures are to be used with dry preparations, close 12 clean
Arnmonium (2.4.1, Method A): maximum 2 ppm. vials with the closures to be examined. Using a lubricated,
Dilute 5 mL of solution S to 14 mL with water R. long-bevel(J) (bevel angle 12 ± 2°) hypodermic needle with
an external diameter of 0.8 mm fitted to a clean syringe,
Extractable zinc: maximum of 5 flg of extractable Zn per inject into the viall mL of water R and remove 1 mL of air;
millilitre of solution S. carry out this operation 4 times for each closure, piercing the
Atomic absorption spectrometry (2.2.23, Method 1). closure each time at a different site. Use a new needle for each
closure and check that the needle is not blunted during the
Test so/ution. DiIute 10.0 mL of solution S to 100 mL with test. Pass the liquid in the vials through a filter with a pore
0.1 M hydrochloric acíd. size of 0.5 [lm. Count the fragments of rubber visible to the
Reference solutions. Prepare the reference solutions using naked eye. The total rtumber of fragments does not exceed 5.
zinc standard solution (10 ppm Zn) R diluted with 0.1 M This limit is based on the assumption that fragments with
hydrochloric acid. a diameter equal to or greater than 50 flm are visible to the
naked eye; in cases of doubt or dispute, the fragments are
Source:zinc hollow-cathode lampo examined with a microscope to verify their nature and size.
Wavelength: 213.9 nm. Self-sealing test. For closures intended to be used with
multidose containers, carry out the following test. Fill
Atomisation device: air-acetylene flameo 10 suitable vials to the nominal volume with water R, fit the
Extractable heavy metals (2.4.8): maximum 2 ppm. closures to be examined and secure with a cap. Using for each
closure a new hypodermic needle with an external diameter of
Solution S complies with test A. Prepare the reference solution 0.8 mm, pierce each closure 10 times, piercing the closure each
using lead standard solution (2 ppm Pb) R. time at a different site. Immerse the vials upright in a 1 giL
Residue onevaporation. Evaporate 50.0 mL of solution S to solution of methy/ene blue R and reduce the external pressure
dryness on a water-bath and dry at 100-105 oc. The residue by 27 kPa for 10 mino Restore atmospheric pressure and leave
weighs not more than 2.0 mg for type l closures and not more the vials immersed for 30 mino Rinse the outside of the vials.
than 4.0 mg for type JI closures. None of the vials contains any trace of coloured solution.

(1) See ISO 7864 "Sterile hypodermic needles for single use".

422 See the information sectíon on general monographs (caver pages)


EUROPEAN PHARMACOPOEIA 8.0

4. Reagents
4. Reagents ................................................................................. 425 4.1.3. Buffer solutions .............................................................. 540
4.1. Reagents, standard solutions, buffer solutions .............. 425 4.2. Volumetric analysis ........................................................... 545
4.1.1. Reagents .......................................................................... 425 4.2.1. Primary standards for volumetric solutions ............... 545
4.1.2. Standard solutions for limit tests ................................. 536 4.2.2. Volumetric solutions ...................................................... 546

General Notices (1) apply to all monographs and other texts 423
EUROPEAN PHARMACOPOEIA 8.0

424 See the informatian sectian an general managraphs (caver pages)


EUROPEAN PHARMACOPOEIA 8.0 4.1. L Reagents

0412013:40000 d§g: about 0.824.


nbD : about 1.382.
bp: about 103 oc.
4. REAGENTS
Acetaldehyde. C 2 Hp. (M, 44.1). 1000200. [75-07-0].
Additional information for reagents that can only be fully
Ethanal.
identified by a trademark or whose availability is limited may
be found in the Knowledge database on the EDQM website. Clear, colourless flammable liquid, miscible with water and
This information is given only to make it easier to obtain with ethanol (96 per cent).
such reagents and this does not suggest in any way that the d§g: about 0.788.
mentioned suppliers are especially recommended or certified by nbD : about 1.332.
the European Pharmacopoeia Commission or the Council of bp: about 21°C.
Europe. It is therefore acceptable to use reagents from another
so urce provided that they comply with the standards of the Acetaldehyde ammonia trimer trihydrate.
Pharmacopoeia. C 6H 1SN 3,3Hp. (M, 183.3). 1133500. [58052-80-5].
2,4,6-Trimethylhexahydro-l,3,5-triazine trihydrate.
mp: 95 oC to 97 oc.
04/2013:40100
Acetic add, anhydrous. C2 H 4 0 2 • (M, 60.1). 1000300.
4.1. REAGENTS, STANDARD [64-19-7].
Content: minimum 99.6 per cent m/m of C2H 4 0 2 •
SOLUTIONS, BUFFER SOLUTIONS
Colourless liquid or white or almost white, shining, fern-like
Where the name of a substance or a solution is followed by the crystals, miscible with or very soluble in water, in ethanol
letter R (the whole in italics), this indicates a reagent induded (96 per cent), in glycerol (85 per cent), and in most fatty and
in the following list. The specifications given for reagents do essential oils.
not necessarily guarantee their quality for use in medicines. d~g: 1.052 to 1.053.
Within the description of each reagent there is a 7-digit bp: 117 oC to 119 oc.
reference code in italics (for example, 1002501). This number,
A 100 giL solution is strongly acid (2.2.4).
which will remain unchanged for a given reagent during
subsequent revisions of the list, is used for identification A 5 giL solution neutralised with dilute ammonia R2 gives
purposes by the Secretariat, and users of the Pharmacopoeia reaction (b) of acetates (2.3.1).
may also find it useful, for example in the management of Freezing point (2.2.18): minimum 15.8 oc.
reagent stocks. The description may also indude a CAS Water (2.5.12): maximum 0.4 per cent. Ifthe water content
number (Chemical Abstract Service Registry Number) is more than 0.4 per cent it may be adjusted by adding the
recognisable by its typical format, for example 9002-93-1. calculated amount of acetic anhydride R.
Sorne of the reagents induded in the list are toxic and are to be Storage: protected from light.
handled in conformity with good quality controllaboratory
practice. Acetk acid, glaciaL C2HP2' (M, 60.1). 1000400. [64-19-7].
Reagents in aqueous solution are prepared using water R. See Acetic acid, glacial (0590).
Where a reagent solution is described using an expression such Acetic acid. 1000401.
as 'hydrochloric acid (lO giL HCl)', the solution is prepared by
an appropriate dilution with water R of a more concentrated Content: 290 giL to 310 giL ofC 2 HP2 6001).
reagent solution specified in this chapter. Reagent solutions DiIute 30 g of glacial acetic acid R to 100 mL with water R.
used in the limit tests for barium, calcium and sulfates are
Acetic add, dilute. 1000402.
prepared using distilled water R. Where the name of the
solvent is not stated, an aqueous solution is intended. Content: 115 giL to 125 giL of C2 H 4 02 (M, 60.1).
The reagents and reagent solutions are to be sto red in Dilute 12 g of glacial acetic acid R to 100 mL with water R.
well-dosed containers. The labelling should comply with the Acetic acid, dilute Rl. 1000403.
relevant nationallegislation and international agreements.
Content: 57.5 giL to 62.5 giL (M, 60.1).
Dilute 6 g of glacial acetic acid R to 100 mL with water R.
0112014:40101
Acetic anhydride. C4 H 6 0 3 • (M r 102.1). 1000500. [108-24-7].
Content: minimum 97.0 per cent m/m of C,¡H 6 03"
4.1.1. REAGENTS
Clear, colourless liquido
Acacia. 1000100. bp: 136 oC to 142 oc.
See Acacia (0307). Assay. Dissolve 2.00 g in 50.0 mL of 1 M sodium hydroxide
in a ground-glass-stoppered flask and boil under a reflux
Acada solution. 1000101.
condenser for 1 h. Titrate with 1 M hydrochloric acid, using
Dissolve 100 g of acacia R in 1000 mL of water R. Stir with 0.5 mL of phenolphthalein solution R as indicator. Calculate
a mechanical stirrer for 2 h. Centrifuge at about 2000 g for the number of millilitres of 1 M sodium hydroxide required
30 min to obtain a dear solution. for 1 g (n¡). Dissolve 2.00 g in 20 mL of cyclohexane R in
Storage: in polyethylene containers of about 250 mL a ground-glass-stoppered flask, coo1 in ice and add a cold
capacity at a temperature of O oC to - 20 oc. mixture of 10 mL of aniline R and 20 mL of cyclohexane R. Boil
the mixture under a re flux condenser for 1 h, add 50.0 mL of
Acebutolol hydrochloride. 1148900. [34381-68-5]. 1 M sodium hydroxide and shake vigorously. Titrate with 1 M
See Acebutolol hydrochloride (0871). hydrochloric acid, using 0.5 mL of phenolphthalein solution R
as indicator. Calculate the number of millilitres of 1 M sodium
Acetal. C6 H¡P2' (Mr 118.2). 1112300. [105-57-7].
hydroxide required for 1 g (n 2 ). Calculate the percentage of
Acetaldehyde diethyl aceta!. 1,1-Diethoxyethane.
C4 H 6 0 3 from the following expression:
Clear, colourless, volati1e liquid, miscible with water and with
ethanol (96 per cent).

General Notices (1) apply to all monographs and other texts 425
4.1,1. Reagents EUROPEAN PHARMACOPOEIA 8.0

Acetic anhydride solution Rl. 1000501. N-Acetyl-e-caprolactam. CSH 13 NO z' (Mr 155.2). 1102700.
Dissolve 25.0 mL of acetic anhydride R in anhydrous [1888-91-1]. N-Acetylhexane-6-1actam.
pyridine R and dilute to 100.0 mL with the same solvento Colourless liquid, miscible with anhydrous ethanol.
Storage: protected from light and airo d~g: about 1.100.
Acetic anhydride - sulfuric add solution. 1000502. niSo: about 1.489.
Carefully mix 5 mL of acetic anhydride R with 5 mL of bp: about 135 oc.
sulfuric acid R. Add dropwise and with cooling to 50 mL of Acetyl chloride. CZH}CIO. (M 78.5). 1000800. [75-36-5].
r
anhydrous ethanol R.
CIear, colourless Iiquid, flammable, decomposes in contact
Prepare immediately before use.
with water and with ethanol (96 per cent), miscible with
Acetone. 1000600. [67-64-1]. ethyIene chloride.
See Acetone (0872). d~g: about l.10.

Acetonitrile. C 2 H 3N. (M, 41.05). 1000700. [75-05-8]. Methyl


Distíllation range (2.2.11). Not less than 95 per cent distils
between 49 oC and 53 oc.
cyanide. Ethanenitrile.
Clear, colourless liquid, miscible with water, with acetone and Acetylcholine chloride. C 7H I6 CIN0 2• (Mr 181.7). 1001000.
with methanol. [60-31-1].
d~g: about 0.78. Crystalline powder, very soluble in cold water and in ethanol
niSo : about 1.344. (96 per cent). It decomposes in hot water and in alkalis.
A 100 giL solution is neutral to litmus papero Storage: at - 20 oc.
Distillation range (2.2.11). Not less than 95 per cent distils Acetyleugenol. C12HI403' (Mr 206.2). 1100700. [93-28-7].
between 80 oC and 82 oc. 2-Methoxy -4- (2-propenyl )phenylacetate.
Acetonitrile used in spectrophotometry complies with the Yellow coloured, oily liquid, practically insoluble in water,
following additional test. freely soluble in ethanol (96 per cent).
Minimum transmittance (2.2.25) using water R as niSo: about 1.521.
compensation liquid: 98 per cent from 255 nm to 420 nm.
bp: 281°C to 282 oc.
Acetonitrile for chromatography. 1000701. Acetyleugenol used in gas chromatography complies with the
See Acetonitrile R. following additional test.
Acetonitrile used in chromatography complies with the Assay. Gas chromatography (2.2.28) as prescribed in the
following additional tests. monograph Clove oil (1091).
Minimum transmittance (2.2.25) using water R as Test so/ution. The substance to be examined.
compensation liquid: 98 per cent from 240 nm.
Content: minimum 98.0 per cent, calculated by the
Content (2.2.28): mínimum 99.8 per cent. normalisation procedure.~
Acetonitrile RL 1000702. N-Acetylglucosarnine. CSH 1S N0 6 . (Mr 221.2). 1133600.
Complies with the requirements prescribed for acetonitrile R [7512-17-6]. 2-(Acetylamino)-2-deoxy-D-glucopyranose.
and with the following additional requirements. mp: about 202 oc.
Content: minimum 99.9 per cent.
Absorbance (2.2.25): maximum 0.10, determined at 200 nm Acetyl-ll-keto-~-boswemc acid. CJ 2 H 4 SOs' (Mr 512.7).
using water R as the compensation liquido 1167700. [67416-61-9]. 3a-(Acetyloxy)-11-oxours-12-en-24-
oic acid. (4~)-3a-(Acetyloxy)-11-oxours-12-en-23-oic acid.
Acetoxyvalerenic add. C 17 H 2P4' (Mr 292.4). 1165800. White or almost white powder, insoluble in water, soluble in
[81397 -67 -3]. (2E)-3- [(lRS,4S,7R,7aR)-1-(Acetyloxy)- acetone, in anhydrous ethanol and in methanol.
3,7 -dimethyl-2,4,5,6, 7, 7a -hexahydro-1H-inden-4-yl]-2-
mp: 271°C to 274 oc.
methylprop-2-enoic acid.
Colourless or pale yellow viscous oil. Acetyl-11-keto-f3-boswellic acid used in liquid chromatography
complies with the following additional test.
Absorbance (2.2.25). A solution in methanol R shows an
absorption maximum at about 216 nm. Assay. Liquid chromatography (2.2.29) as prescribed in the
monograph on Indian franlcincense (2310).
Acetylacetamide. C4 H 7 N0 2 . (Mr 101.l). 1102600. Content: minimum 90 per cent, calculated by the
[5977 -14-0]. 3-0xobutanamide. normalisation procedure.
mp: 53 oC to 56 oc.
N-Acetylneuraminic add. C ll H I9 N0 9 . (M; 309.3).1001100.
Acetylacetone. C SHP2' (Mr 100.1). 1000900. [123-54-6]. [131-48-6]. O-Sialic acid.
2,4- Pentanedione. White or almost white acicular crystals, soluble in water and
Colourless or slightly yellow, easily flammable liquid, freely in methanol, slightly soluble in anhydrous ethanol, practically
soluble in water, miscible with acetone, with ethanol (96 per insoluble in acetone.
cent) and with glacial acetic acid. [Ql~o: about - 36, determined on a 10 giL solution.
niSo: 1.452 to 1.453. mp: about 186 oC, with decomposition.
bp: 138 oC to 140 oc.
N-Acetyltryptophan. C 13 H I4N ZO}. (Mr 246.3). 1102800.
Acetylacetone reagent Rl. 1000901. [1218-34-4]. 2-Acetylamino-3-(indol-3-yl)propanoic acid.
To 100 mL of ammonium acetate solution R add 0.2 mL of White or almost white powder or colourless crystals, slightly
acetylacetone R. soluble in water. lt dissolves in dilute solutions of alkali
Acetylacetone reagent R2. 1000902. hydroxides.
Dissolve 0.2 mL of acetylacetone R, 3 mL of glacial acetic mp: about 205 oc.
acid R and 25 g of ammonium acetate R in water R and Assay. Liquid chromatography (2.2.29) as prescribed in the
dilute to 100 mL with the same solvent. monograph Tryptophan (1272).

426 See the information section on general monographs (cover pages)


EUROPEAN PHARMACOPOEIA 8.0 4.1. L Reagents

Test solution. Dissolve 10.0 mg in a mixture of 10 volumes al-Acid-glycoprotein si.lica gel for chiral separation.
of acetonitrile R and 90 volumes of water R and dilute to 1148700.
100.0 mL with the same mixture of solvents. A very finely divided silica gel for chromatography consisting
Content: minimum 99.0 per cent, calculated by the of spherical particles coated with al-acid glycoprotein. The
normalisation procedure. particle size is indicated after the name of the reagent in the
tests where it is used.
Acetyltyrosine ethyl estero C13HI7NO'l'H20. (Mr 269.3).
1001200. [36546-50-6]. N-Acetyl-L-tyrosine ethyl Acrylamide. C 3H sNO. (Mr 71.1). 1001500. [79-06-1].
ester monohydrate. Ethyl (S)-2-acetamido-3-(4- Propenamide.
hydroxyphenyl)propionate monohydrate.
Colourless or white flakes or a white or almost white,
White or almost white, crystalline powder suitable for the crystalline powder, very soluble in water and in methanol,
assay of chymotrypsin. freely soluble in anhydrous ethanol.
[al~o: + 21 to + 25, determined on a 10 giL soIution in ethanol mp: about 84 oc.
(96 per cent) R.
Ai~rn: 60 to 68, determined at 278 nm in ethanol (96 per 30 per cent acrylamide/bisacrylamide (29:1) solution.
cent) R. 1001501.

Acetyltyrosine ethyl ester, 0.2 M. 1001201. Prepare a solution containing 290 g of acrylamide R and
10 g of methylenebisacrylamide R per litre of water R. Filter.
Dissolve 0.54 g of acetyltyrosine ethyl ester R in ethanol
(96 per cent) R and dilute to 10.0 mL with the same solvent. 30 per cent acrylamide/bisacrylamide (36.5:1) solution.
1001502.
Add bIue 83. C4sH44N3Na07S2' (Mr 826). 1012200.
[6104-59-2]. Prepare a solution containing 292 g of acrylamide R and 8 g
Colour Index No. 42660. of methylenebisacrylamíde R per litre of water R. Filter.
Brilliant bIue. Coomassie brilliant blue R 250. Acrylic add. C3H 4 02' (Mr 72.l). 1133700. [79-10-7].
Brown powder insoluble in coId water, slightly soluble in Prop-2-enoic acid. Vinylformic acid.
boiling water and in anhydrous ethanol, soluble in sulfuric
Content: minimum 99 per cent.
acid, glacial acetic acid and in dilute solutions of alkali
hydroxides. It is stabilised with 0.02 per cent ofhydroquinone monomethyl
ether.
Acid biue 90. C47H4SN3Na07S2' (Mr 854). 1001300.
Corrosive liquid, miscible with water and ethanol (96 per
[6104-58-1].
cent). It polymerises readily in the presence of oxygen.
Colour Index No. 42655.
Sodium [4- [[ 4- [( 4-ethoxyphenyl)amino ]phenyl] [[ 4-( ethyl) (3- d~g: about 1.05.
sulfonatobenzyl)amino] phenyl] methylene] cyclo-hexa -2,5- n~o: about 1.421.
dien-l-ylidene] (ethyl)-(3-sulfonatobenzyl)ammonium. bp: about 141 oc.
A dark brown powder, with a violet sheen and sorne particles mp: 12 oC to 15 oc.
having a metallic lustre, soluble in water and in anhydrous
ethanol. Actein. C37Hs6011' (Mr 677). 1181500. [18642-44-9].
Ai~rn: greater than 500, determined at 577 nm in a 0.01 giL (23R,24R,25S,26S)-3~-(~- D-Xylopyranosyloxy)-
solution in buffer solution pH 7.0 and calculated with l6~,23:23,26:24,25-triepoxy-26-hydroxy-9,19-cyclolanosta11-
reference to the dried substance. 12~-yl acetate.
Loss on drying (2.2.32): maximum 5.0 per cent, determined
on 0.500 g by drying in an oven at 105 oc. Acteoside. C29H3601S' (Mr 624.6). 1145100.
[61276-17 -3]. 2-(3,4-Dihydroxyphenyl)ethyI
Acid bIue 92. C26H16N3Na301OS3' (Mr 696). 1001400. 3-0-( 6-deoxy-a -L- mannopyranosyl)-4-0- [(2E)- 3-
[3861-73-2]. (3,4-dihydroxyphenyl )prop-2 -enoyI]- ~- D-glucopyranoside.
Colour Index No. 13390. Verbascoside.
Coomassie blue. Anazolene sodium. Trisodium 8-hydroxy-4'- Light yellowish powder, freely soluble in water and in
(phenylamino) azonaphthalene-3 ,S' ,6-trisulfonate. methanol.
Dark bIue crystals, soluble in water, in acetone and in ethylene mp: about 140 oC, with decomposition.
glycol monoethylether, slightly soluble in ethanol (96 per
cent). Adamantane. ClOHl6" (Mr 136.2). 1181600. [281-23-2].
Tricyclo [3.3.1.1 3,7]decane.
Add bIue 92 solution. 1001401.
mp: about 270 oc.
Dissolve 0.5 g of acid blue 92 R in a mixture of 10 mL of
glacial acetic acid R, 45 mL of ethanol (96 per cent) R and Adenine. 1172800. [73-24-5].
45 mL of water R.
See Adenine (0800).
Add bIue 93. C37H27N3Na209S3' (Mr 800). 1134200.
[28983-56-4]. Adenosine. ClOHI3NS04' (Mr 267.2). 1001600. [58-61-7].
6- Amino-9-~- D-ribofuranosyl-9 H- purine.
Colour Index No. 42780.
Methyl blue. Poirrier bIue. White or almost white, crystalline powder, slightly soluble in
Mixture of triphenylrosaniline di- and trisulfonate and of water, practically insoluble in acetone and in ethanol (96 per
cent). It dissolves in dilute solutions of acids.
triphenylpararosaniline.
Dark bIue powder. mp: about 234 oc.
Colour change: pH 9.4 to pH 14.0. Adipi.c add. C6H lO 04' (Mr 146.1). 1095600. [124-04-9].
Add bIue 93 solution. 1134201. Prisms, freely soluble in methanol, soluble in acetone,
Dissolve 0.2 g of acid blue 93 R in water R and dilute to practically insoluble in light petroleum.
100 mL with the same solvent. mp: about 152 oc.

General Notices (1) apply to all monographs and other texts 427
4.1.1. Reagents EUROPEAN PHARMACOPOEIA 8.0

Adrenaline. C9 H 13 NO y (M, 183.2). 1155000. [51-43-4]. Agarose for electrophoresis. 1002000. [9012-36-6].
(IR) -1- (3,4-Dihydroxyphenyl)-2-(methylamino )ethanol. A neutral, linear polysaccharide, the main component of
4- [(lR)-1-hydroxy-2-(methylamino )ethyl]benzene-1,2-diol. which is derived from agar.
White or almost white powder, gradually becoming brown on White or almost white powder, practically insoluble in cold
exposure to light and air, very slightly soluble in water and water, very slightly soluble in hot water.
in ethanol (96 per cent), insoluble in acetone. It dissolves in
dilute solutions of mineral acids and alkali hydroxides. Agnuside. C Z2 HZPll' (Mr 466.4). 1162000.
mp: about 215 oc. [11027 -63-7]. (lRS,4aSR,5RS,7aRS)-5-Hydroxy-
7 -[[ (4-hydroxybenzoyl)oxy]methyl]-1,4a,5,7a-
Adrenalone hydrochloride. C9 H 12 CIN03" (Mr 217.7). tetrahydrocyclopenta [c ]pyran -1- yl ~- D-glucopyranoside.
1155100. [62-13-5]. 1-(3,4-Dihydroxyphenyl)-2- White or almost white crystals.
(methylamino )ethanone hydrochloride. 3',4'-Dihydroxy-2-
(methylamino )acetophenone hydrochloride. Alanine. 11 02900. [56-41-7].
Pale yellow crystals, freely soluble in water, soluble in ethanol See Alanine (0752).
(96 per cent).
~-Alanine. 1004500. [107-95-9].
mp: about 244 oc.
See 3-aminopropionic acid R.
Aesdn. 1001700. [6805-41-0].
Albumin, bovine. 1002300. [9048-46-8].
A mixture of related saponins obtained from the seeds of
Aeseulus hippoeastanum L. Bovine serum albumin containing about 96 per cent of protein.
Fine, almost white or slightly reddish or yellowish, amorphous White to light brownish-yellow powder.
powder. Water (2.5.12): maximum 3.0 per cent, determined on 0.800 g.
Chromatography. Thin-layer chromatography (2.2.27) as Alhumin, hovine Rl. 1183500. [9048-46-8].
prescribed in the monograph Senega root (0202): apply 20 ¡.tL
Bovine serum albumin containing about 96 per cent of protein.
of the solution; after spraying with anisaldehyde solution R
and heating, the chromatogram shows a principal band with White or light brownish-yellow powder.
an RF of about 0.4. Albumin, human. 1133800.
Aflatoxin Ej' C¡7H¡zÜ6' (Mr 312.3). 1166000. Human serum albumin containing not less than 96 per cent
[1162-65-8]. (6aR,9aS) -4-Methoxy- 2,3,6a,9a -tetrahydro- of albumin.
cyclopenta[c ]furo[3',2':4,5]furo[2,3-h] [1]benzopyran-1,11-
Albumin solution, human. 1002400. [9048-46-8].
dione.
See Human albumin so/utian (0255).
White or faint yellow crystals.
Agarosel cross-Hnked polyacrylamide. 1002200. Albumin solution, human Rl. 1002401.
Agarose trapped within a cross-linked polyacrylamide Dilute human albumin solution R with a 9 giL solution of
network; it is used for the separation of globular proteins with sodium ehloride R to a concentration of 1 giL of protein.
relative molecular masses of 2 x 104 to 35 X 104 . Adjust the pH to 3.5-4.5 with glacial aeetic acid R.

Agal'Ose-DEAE for ion-exchange chromatography. AlcohoL 1002500. [64-17-5].


1002100. [57407-08-6]. See Ethanol (96 per eent) R.
Cross-linked agarose substituted with diethylaminoethyl Alcohol (x per cent V/V). 1002502.
groups, presented as beads.
See Ethanol (x per cent V/V) R.
Agarose for chromatography. 1001800. [9012-36-6].
Alcohol, aldehyde-free. 1002501.
Swollen beads 60-140 ¡.tm in diameter presented as a 4 per
Mix 1200 mL of ethanol (96 per cent) R with 5 mL of a
cent suspension in water R.
400 giL solution of silver nitrate R and 10 mL of a cooled
Used in size-exclusion chromatography for the separation of 500 giL solution of potassium hydroxide R. Shake, allow to
proteins with relative molecular masses of 6 x 104 to 20 X 106 stand for a few days and filter. Distil the filtrate immediately
and of polysaccharides with relative molecular masses of before use.
3 x 10 3 to 5 X 10 6 .
Aldehyde dehydrogenase. 1103000.
Agarose for chromatography, cross-linked. 1001900.
[61970-08-9]. Enzyme obtained from baker's yeast which oxidises
acetaldehyde to acetic acid in the presence of
Prepared from agarose by reaction with 2,3-dibromopropanol nicotinamide-adenine dinucleotide, potassium salts
in strongly alkaline conditions. and thiols, at pH 8.0.
It occurs as swollen beads 60-140 ¡.tm in diameter and is
presented as a 4 per cent suspension in water R. Aldehyde dehydrogenase solution. 1103001.
Used in size-exclusion chromatography for the separation of Dissolve in water R a quantity of aldehyde dehydrogenase R,
proteins with relative molecular masses of 6 x 104 to 20 X 10 6 equivalent to 70 units and dilute to 10 mL with the same
and of polysaccharides with relative molecular masses of solvento This solution is stable for 8 h at 4 oc.
3 x 10 3 to 5 x 10 6.
Aldrin. C¡2HSC16' (M, 364.9). 1123100. [309-00-2].
Agarose for chromatography, cross-linked Rl. 1001901. bp: about 145 oc.
[65099-79-8]. mp: about 104 oc.
Prepared for agarose by reaction with 2,3-dibromopropanol in A suitable certified reference solution (10 ngl ¡.tL in
strongly alkaline conditions. cyclohexane) may be used.
It occurs as swollen beads 60-140 ¡.tm in diameter and is
presented as a 4 per cent suspension in water R. Aleuritic acid. C¡6H3zÜS' (M, 304.4). 1095700. [533-87-9].
(9RS,1 OSR)-9, 10, 16-Trihydroxyhexadecanoic acid.
Used in size-exclusion chromatography for the separation of
proteins with relative molecular masses of 7 x 104 to 40 x 10 6 White or almost white powder, greasy to the touch, soluble
and of polysaccharides with relative molecular masses of in methanol.
1 x 10 5 to 2 x 10 7 • mp: about 101 oc.

428 See the information section on general monographs (ca ver pages)
EUROPEAN PHARMACOPOEIA 8.0 4.1.1. Reagents

Alizarin S. C14H7Na07S,Hp. (Mr 360.3). 1002600. [130-22-3]. Ameridum-243 spiking solution. 1167500.
Schultz No. 1145. Contains 50 Bq/L 243 Am and a 134 giL solution of lanthanum
Colour Index No. 58005. chloride heptahydrate R in a 103 giL solution of hydrochloric
Sodium 1,2-dihydroxyanthraquinone-3-sulfonate acid R.
monohydrate. Sodium 3,4-dihydroxy-9, 10-dioxo-9, 10-
dihydroanthracene-2-sulfonate monohydrate. Amido black 1013. C22H14N6Nap9S2' (Mr 617). 1003100.
[1064-48-8].
Orange-yellow powder, freely soluble in water and in ethanol
(96 per cent). Schultz No. 299.
Colour Index No. 20470.
Alizarin S solution. 1002601. Disodium 5-amino-4-hydroxy-6- [( 4-nitrophenyl)azo]-3-
A 1 giL solution. (phenylazo )naphthalene-2, 7 -disulfonate.
Test for sensitivity. If alizarin S solution is used for the Dark-brown to black powder, sparingly soluble in water,
standardisation of 0.05 M barium perchlorate, it shows soluble in ethanol (96 per cent).
a colour change from yellow to orange-red when it is
tested according to the standardisation of 0.05 M barium Amido black lOE solution. 1003101.
perchlorate. A 5 giL solution of amido black lOB R in a mixture of
Colour change: pH 3.7 (yellow) to pH 5.2 (violet). 10 volumes of acetic acid R and 90 volumes of methanol R.

Alovudine. C lO H 13 FN 20,¡. (Mr 244.2). 1185400. [25526-93-6]. Aminoazobenzene. C I2 H ll N 3. (Mr 197.2). 1003200.
1- [(2R,45,5R)-4- Fluoro- 5- (hydroxymethyl)tetrahydrofuran- [60-09-3].
2-yl]-5-methylpyrimidine-2,4( 1H,3H)-dione. Colour Index No. 11000.
Fluorodeoxythymidine. 3'-Deoxy-3'-fluorothymidine. 4-(Phenylazo )aniline.
Content: mínimum 95 per cent. Brownish-yellow needles with a bluish tinge, slightly soluble
Colourless crystals. in water, freely soluble in ethanol (96 per cent).
mp: about 128 oc.
Aluminium. Al. (A r 26.98). 1118200. [7429-90-5].
White or almost white, malleable, flexible, bluish metal, 2-Aminobenzoic add. C7 H 7N0 2 • (Mr 137.1). 1003400.
available as bars, sheets, powder, strip s or wire. In moist air an [118-92-3]. Anthranilic add.
oxide film forms which protects the metal from corrosion. A white or pale-yellow, crystalline powder, sparingly soluble
Analytical grade. in cold water, freely soluble in hot water, in ethanol (96 per
cent) and in glycerol. Solutions in ethanol (96 per cent) or in
Aluminium chloride. AIC1 3,6Hp. (Mr 241.4). 1002700. ether and, particularly, in glycerol show a violet fluorescence.
[7784-13-6]. Aluminium chloride hexahydrate.
mp: about 145 oc.
Content: minimum 98.0 per cent of AICl3'6H 20.
White or slightly yellowish, crystalline powder, hygroscopic, 3-Aminobenzok add. C7 H 7N0 2. (Mr 137.1). 1147400.
freely soluble in water and in ethanol (96 per cent). [99-05-8].
5torage: in an airtight container. 'l\Thite ol' almost white crystals. An aqueous solution turns
brown on standing in airo
Aluminium chloride reagent. 1002702.
mp: about 174 oc.
Dissolve 2.0 g of aluminium chloride R in 100 mL of a 5 per
cent V/V solution of glacial acetic acid R in methanol R. Storage: in an airtight container, protected from light.

Aluminium chloride solution. 1002701. 4-Aminobenzoi.c add. C 7H 7N0 2. (Mr 137.1). 1003300.
[150-13-0].
Dissolve 65.0 g of aluminium chloride R in water R and
dilute to 100 mL with the same solvento Add 0.5 g of White or almost white, crystalline powder, slightly soluble
activated charcoal R, stir for 10 min, filter and add to the in water, freely soluble in ethanol (96 per cent), practically
filtrate, with continuous stirring, suffident of a 10 giL insoluble in light petroleum.
solution of sodium hydroxide R (about 60 mL) to adjust mp: about 187 oc.
the pH to about 1.5. Chromatography. Thin-layer chromatography (2.2.27) as
Aluminium nitrate. Al(N0 3 )3,9Hp. (Mr 375.1). 1002800. prescribed in the monograph Procaine hydrochloride (0050);
[7784-27-2]. Aluminium nitrate nonahydrate. the chromatogram shows only one principal spot.
Crystals, deliquescent, very soluble in water and ethanol Storage: protected from light.
(96 per cent), very slightly soluble in acetone. 4-Aminobenzok add solution. 1003301.
Storage: in an airtight container. Dissolve 1 g of 4-aminobenzoic acid R in a mixture of 18 mL
Aluminium oxide, anhydrous. 1002900. [1344-28-1]. of anhydrous acetic acid R, 20 mL of water R and 1 mL of
phosphoric acid R. Immediately before use, mix 2 volumes
Aluminium oxide, consisting of y-Al20v dehydrated and
of the solution with 3 volumes of acetone R.
activated by heat treatment.
Particle size: 75 flm to 150 flm. N-(4-Aminobenzoyl)-L-glutamic add. C 12 H 14 Nps'
(M, 266.3). 1141700. [4271-30-1]. ABGA. (25)-2-[(4-
Aluminium oxide, basic. 1118300. Aminobenzoyl)amino ]pentanedioic add.
A basic grade of anhydrous aluminium oxide R suitable for
White or almost white, crystalline powder.
column chromatography.
mp: about 175 oC, with decomposition.
pH (2.2.3). Shake 1 g with 10 mL of carbon dioxide-free
water R for 5 mino The pH of the suspension is 9 to 10. 4-Aminobutanoic add. C,¡H 9 N0 2 . (Mr 103.1). 1123200.
Aluminium oxide, neutral. 1118400. [56-12-2]. y-Aminobutyric acid. GABA.
See Aluminium oxide, hydrated (0311). Leaflets from methanol and ether, needles from water and
ethanol (96 per cent). Freely soluble in water, practically
Aluminium potassium sulfate. 1003000. [7784-24-9]. insoluble or slightly soluble in other solvents.
See Alum (0006). mp: about 202 oC (decreases on rapid heating).

General Notices (1) apply to al! monographs and other texts 429
4.1.1. Reagents EUROPEAN PHARMACOPOEIA 8.0

Aminobutanol. C 4 H ll NO. (Mr 89.1). 1003500. [5856-63-3]. Aminomethylalizarindiacetk add. C19H1SNOs,2H20.


2-Aminobutano!. (Mr 421.4). 1003900. [3952-78-1]. 2,2'-[(3,4-dihydroxy-
Oily liquid, miscible with water, soluble in ethanol (96 per anthraquinon -3-yl)methylenenitrilo] diacetic acid dihydrate.
cent). Alizarin complexone dihydrate.
Fine, pale brownish-yellow or orange-brown powder,
d~g : about 0.94.
practically insoluble in water, soluble in solutions of alkali
n~o : about 1.453. hydroxides.
bp: about 180 oc. mp: about 185 oc.
L055 on drying (2.2.32): maximum 10.0 per cent, determined
Aminochlorobenzophenone. C 13 H lO ClNO. (M, 231.7). on 1.000 g.
1003600. [719-59-5]. 2-Amino-5-chlorobenzophenone.
Aminomethylalizarindiacetic add reagent. 1003901.
Yellow, crystalline powder, practically insoluble in water, freely
soluble in acetone, soluble in ethanol (96 per cent). Solution A. Dissolve 0.36 g of cerous nitrate R in water R
and dilute to 50 mL with the same solvent.
mp: about 97 oc.
Solution B. Suspend 0.7 g of aminomethylalizarindiaeetic
Content: minimum 95.0 per cent. acid R in 50 mL of water R. Dissolve with the aid of about
Storage: protected from light. 0.25 mL of coneentrated ammonia R, add 0.25 mL of glacial
acetie acid R and dilute to 100 mL with water R.
4-Aminofolk aeid. C¡9H20NsOs' (M, 440.4). 1163700. Solution e. Dissolve 6 g of sodium acetate R in 50 mL of
[54-62-6]. (2S)-2- [[ 4- [[ (2,4-Diaminopteridin-6- water R, add 11.5 mL of glacial acetic acid R and dilute to
yl)methyl]amino ]benzoyl] amino ]pentanedioic aeid. 100 mL with water R.
N- [4- [[ (2,4-Diaminopteridin -6-yl)methyl] amino ]benzoyl]- L- To 33 mL of acetone R add 6.8 mL of solution C, 1.0 mL of
glutamic aeid. Aminopterine. solution B and 1.0 mL of solution A and dilute to 50 mL
Yellowish powder. with water R.
mp: about 230 oc. Test for sensitivity. To 1.0 mL of fluoride standard solution
(lO ppm F) R add 19.0 mL of water R and 5.0 mL of the
6-Aminohexanoic aeid. C6 H 13 N0 2 • (Mr 131.2). 1103100. aminomethylalizarindiacetic aeid reagent. After 20 min,
[60-32-2]. the solution assumes a blue colour.
Storage: use within 5 days.
Colourless crystals, freely soluble in water, sparingly soluble
in methanol, practically insoluble in anhydrous ethano!. Aminomethylalizarindiacetic add solution. 1003902.
mp: about 205 oc. Dissolve 0.192 g of aminomethylalizarindiaeetic acid R
in 6 mL of freshly prepared 1 M sodium hydroxide. Add
Aminohippuric aeid. C9 H lO NP3' (M, 194.2). 1003700. 750 mL of water R, 25 mL of succinate buffer solution
[61-78-9]. (4-Aminobenzamido)acetic aeid. pH 4.6 R and, dropwise, 0.5 M hydrochloric acid until the
White or almost white powder, sparingly soluble in water, colour changes from violet-red to yellow (pH 4.5 to 5). Add
soluble in ethanol (96 per cent). 100 mL of acetone R and dilute to 1000 mL with water R.
mp: about 200 oc. 4-Aminomethylbenzoic aeid. C SH 9N0 2 • (Mr 151.2).
1167800. [56-91-7].
Aminohippuric add reagent. 1003701.
Aminonitrobenzophenone. C13HlON203' (M, 242.2).
Dissolve 3 g of phthalic acid R and 0.3 g of aminohippuric 1004000. [1775-95-7]. 2-Amino-5-nitrobenzophenone.
acid R in ethanol (96 per cent) R and dilute to 100 mL with
Yellow, crystalline powder, practically insoluble in water,
the same solvent. soluble in tetrahydrofuran, slightly soluble in methano!.
Aminohydroxynaphthalenesulfonic add. C lO H 9 N0 4 S. mp: about 160 De.
(M, 239.3). 1112400. [116-63-2]. 4-Amino-3- Ai~m: 690 to 720, detennined at 233 nm using a 0.01 giL
hydroxynaphthalene-1-sulfonic aeid. solution in methanol R.
White or grey needles, turning pink on exposure to light, 6-AminopeniciHanic add. CSH¡2NP3S, (Mr 216.3). 1162100.
especially when moist, practically insoluble in water and in [551-16-6]. (2S,5R,6R)-6-Amino-3,3-dimethyl-7 -oxo-4-thia-
ethanol (96 per cent), soluble in solutions of alkali hydroxides 1-azabicyclo [3.2.0] heptane-2-carboxylic aeid.
and in hot solutions of sodium metabisulfite. Appearance: white or almost white powder.
Storage: protected from light. mp: about 205 oC, with decomposition.
Aminohydroxynaphthalenesulfonic aeid solution. Aminophenazone. C13HI7NP. (231.3). 1133900. [58-15-1].
1112401. 4- (Dimethylamino) -1 ,5-dimethyl-2-phenyl-1 ,2-dihydro-3H-
pyrazol-3-one.
Mix 5.0 g of anhydrous sodium sulfite R with
94.3 g of sodium hydrogensulfite R and 0.7 g of White or almost white, crystalline powder or colourless
aminohydroxynaphthalenesulfonic acid R. Dissolve 1.5 g of crystals, soluble in water, freely soluble in ethanol (96 per
the mixture in water R and dilute to 10.0 mL with the same cent).
solvent. Prepare the solution daily. mp: about 108 oc.

cis-Aminoindanol. CgH¡¡NO. (M, 149.2). 1168300. 2-Aminophenol. C6H7NO. (Mr 109.1). 1147500. [95-55-6].
[126456-43-7]. (lS,2R)-1-Amino-2,3-dihydro-lH-inden-2-0!. Pale yellowish-brown crystals which rapidly become brown,
(-) -cis-1- Aminoindan -2-0!. sparingly soluble in water, soluble in ethanol (96 per cent).
Content: minimum 98.0 per cent (sum of enantiomers, mp: about 172 oc.
determined by gas chromatography). Storage: in an airtight container, protected from Iight.
[al~o: - 69 to - 59, determined on a 2 giL solution in 3-Aminophenol. C6H7NO. (Mr 109.1). 1147600. [591-27-5].
chloroform R. Pale yellowish-brown crystals, sparingly soluble in water.
mp: 118 oC to 122 De. mp: about 122 oc.

430 See the information section on general monographs (caver pages)


EUROPEAN PHARMACOPOEIA 8.0 4.1.1. Reagents

4-AminophenoL C6H7NO. (M r 109.1). 1004300. [123-30-8]. Arnmonia, dUute Rl. 1004702.


Content: minimum 95 per cent. Content: 100 giL to 104 giL ofNH 3 (Mr 17.03).
White or slightly coloured, crystalline powder, becoming Dilute 41 g of concentrated ammonia R to 100 mL with
coloured on exposure to air and light, sparingly soluble in water R.
water, soluble in anhydrous ethanol. Ammonia, dilute R2. 1004703.
mp: about 186 oC, with decomposition. Content: 33 giL to 35 giL ofNH 3 (Mr 17.03).
Storage: protected from light. Dilute 14 g of concentrated ammonia R to 100 mL with
water R.
Arninopolyether. C¡SH36NP6' (Mr 376.5). 1112500.
[23978-09-8]. 4,7,13,16,21,24-hexaoxa-1,10- Amrnonia, dilute R3. 1004704.
diazabicyclo [8,8,8]hexacosane. Content: 1.6 giL to 1.8 giL ofNH 3 (Mr 17.03).
mp: 70 oC to 73 oc. Dilute 0.7 g of concentrated ammonia R to 100 mL with
water R.
3-ArninopropanoL C3H9NO. (Mr 75.1).1004400. [156-87-6].
3-Aminopropan-l-ol. Propanolamine. Arnrnonia, dilute R4. 1004706.
Clear, colourless, viscous liquido Content: 8.4 giL to 8.6 giL ofNH 3 (Mr 17.03).
d§g: about 0.99. Dilute 3.5 g of concentrated ammonia R to 100 mL with
water R.
ni?: about 1.461.
mp: about 11°C. Amrnonia, lead -free. 1004705.
Complies with the requirements prescribed for dilute
3-Arninopropionic add. C3H 7NO,. (Mr 89.1). 1004500. ammonia R1 with the following additional test: to 20 mL
[107-95-9]. ~-Alanine. ~ oflead-free ammonia, add 1 mL of lead-free potassium
Content: minimum 99 per cent. cyanide solution R, dilute to 50 mL with water R and add
0.10 mL of sodium sulfide solution R. The solution is not
White or almost white, crystalline powder, freely soluble in more intensely coloured than a reference solution prepared
water, slightly soluble in ethanol (96 per cent), practically without sodium sulfide.
insoluble in acetone.
mp: about 200 oC, with decomposition. Arnmonia, concentrated Rl. 1004800.
Content: minimum 32.0 per cent m/m ofNH 3 (Mr 17.03).
Aminopyrazolone. C¡¡H13NP. (M r 203.2). 1004600. A clear, colourless liquido
[83-07 -8]. 4-Amino-2,3-dimethyl-1-phenylpyrazolin -5-one.
d~g: 0.883 to 0.889.
Light-yellow needles or powder, sparingly soluble in water, Assay. Weigh accurately a ground-glass-stoppered flask
freely soluble in ethanol (96 per cent). containing 50.0 mL of 1 M hydrochloric acid. Introduce 2 mL
mp: about 108 oc. of the concentrated ammonia and weigh again. Titrate the
solution with 1 M sodium hydroxide, using 0.5 mL of methyl
Arninopyrazolone solution. 1004601. red mixed solution R as indicator.
A 1 giL solution in buffer solution pH 9.0 R. 1 mL of 1 M hydrochloric acid is equivalent to 17.03 mg of
NH y
3-Aminosalicylic add. C7H 7NO y (Mr 153.1). 1183600.
[570-23-0]. 3-Amino-2-hydroxybenzoic acid. Storage: protected from atmospheric carbon dioxide, at a
temperature below 20 oc.
mp: about 240 oc.
Slightly soluble in water. Ammouium acetate. C2 H 7N0 2 • (Mr 77.1). 1004900.
[631-61-8].
4-Arninosalicylic add. C7H 7NO y (Mr 153.1). 1183700. Colourless crystals, very deliquescent, very soluble in water
[65-49-6]. 4-Amino-2-hydroxybenzoic acid. and in ethanol (96 per cent).
White or almost white, bulky powder, slightly soluble in water, Storage: in an airtight container.
soluble in ethanol (96 per cent), in dilute nitric acid and in Ammonium acetate solution. 1004901.
sodium hydroxide. It darkens on exposure to air and light.
Dissolve 150 g of ammonium aeetate R in water R. Add
mp: 135 oC to 145 oc. 3 mL of glacial acetic acid R and dilute to 1000 mL with
Storage: at a temperature not exceeding 30 oC, in an airtight water R.
container, protected from light. Storage: use within 1 week.
Arnmonia, concentrated. 1004700. Arnmoniurn and ceriurn nitraíe. (NH'¡)2Ce(N03)6'
See Concentrated ammonia solution (0877). (M, 548.2). 1005000. [16774-21-3].
Orange-yellow, crystalline powder, or orange transparent
Amrnonia. 1004701. crystals, soluble in water.
Content: 170 giL to 180 giL of NH 3 (Mr 17.03).
Ammonium aud cerium sulfate. (NH4)4Ce(S04)4,2Hp.
Dilute 67 g of concentrated ammonia R to 100 mL with (Mr 633). 1005100. [10378-47-9].
water R. Orange-yeliow, crystalline powder or crystals, slowly soluble
d~g: 0.931 to 0.934. in water.
When used in the test for iron, ammonia R complies with (lR)- (-)- Ammonium lO-carnphorsulfonate. CJOHI9N04S.
the following additional requirement. Evaporate 5 mL of (Mr 249.3). 1103200.
ammonia to dryness on a water-bath, add 10 mL of water R,
Content: minimum 97.0 per cent of (lR)-( - )-ammonium
2 mL of a 200 giL solution of citrie acid R and 0.1 mL of
10-camphorsulfonate.
thioglycollie aeid R. Make alkaline by adding ammonia R
and dilute to 20 mL with water R. No pink colour develops. [Ql~o: - 18 ± 2, determined 011 a 50 giL solution.
Storage: protected from atmospheric carbon dioxide, at a Arnmoniurn carbamate. CH 6 NP2' (Mr 78.1). 1168400.
temperature below 20 oc. [1111-78-0]. Carbamic acid ammonium salto

General Notices (1) apply to all monographs and other texts 431
4.1,1. Reagents EUROPEAN PHARMACOPOEIA 8.0

Ammonium carbonate. 1005200. [506-87-6]. A Storage: in amber flasks at 37°C for 24 h.


mixture of varying proportions of ammonium hydrogen
carbonate (NH 4 HC0 3 , M, 79.1) and ammonium carbamate Ammonium molybdate reagent R2. 1005708.
(NH 2COONH,p M, 78.1). Dissolve 50 g of ammonium molybdate R in 600 mL of
White or almost white translucent mass, slowly soluble in water R. To 250 mL of cold water R add 150 mL of sulfuric
about 4 parts of water. It is decomposed by boiling water. acid R and cool. Mix the 2 solutions together. Storage: use
Ammonium carbonate liberates not less than 30 per cent m/m within 1 day.
ofNH 3 (Mr 17.03).
Ammonium molybdate solution. 1005702.
Assay. Dissolve 2.00 g in 25 mL of water R. Slowly add 50.0 mL
A 100 giL solution.
of 1 M hydrochloric add, titrate with 1 M sodium hydroxide,
using 0.1 mL of methyl orange solution R as indicator. Ammonium molybdate solution R2. 1005703.
1 mL of 1 M hydrochloric acid is equivalent to 17.03 mg of Dissolve 5.0 g of ammonium molybdate R with heating in
NH3' 30 mL of water R. Coo!, adjust the pH to 7.0 with dilute
Storage: at a temperature below 20 oc. ammonia R2 and dilute to 50 mL with water R.
Ammonium carbonate solution. 1005201. Ammoni.um molybdate solution R3. 1005704.
A 158 giL solution. Solution A. Dissolve 5 g of ammonium molybdate R in
Ammonium carbonate solution Rl. 1005202. 20 mL of water R with heating.
Dissolve 20 g of ammonium carbonate R in 20 mL of dilute Solution B. Mix 150 mL of ethanol (96 per cent) R with
ammonia R1 and dilute to 100 mL with water R. 150 mL of water R. Add with cooling 100 mL of sulfurie
acíd R.
Ammonium chloride. 1005300. [12125-02-9]. Immediately before use add 80 volumes of solution B to
See Ammonium ehloride (0007). 20 volumes of solution A.
Ammonium chloride solution. 1005301. Ammonium molybdate solution R4. 1005705.
A 107 giL solution. Dissolve 1.0 g of ammonium molybdate R in water R
Ammonium dtrate. C6H14N207' (M, 226.2). 1103300. and dilute to 40 mL with the same solvent. Add 3 mL of
[3012-65-5]. Diammonium hydrogen citrate. hydrochloric acid R and 5 mL of perchlorie acid R and dilute
to 100 mL with acetone R.
White or almost white, crystalline powder OI colourless
crystals, freely soluble in water, slightly soluble in ethanol Storage: protected from light; use within 1 month.
(96 per cent). Ammonium molybdate solution R5. 1005707.
pH (2.2.3) : about 4.3 for a 22.6 giL solution.
Dissolve 1.0 g of ammonium molybdate R in 40.0 mL of
Ammonium dihydrogen phosphate. (NH 4 )H 2P0 4. a 15 per cent V/V solution of sulfurie acid R. Prepare the
(Mr 115.0). 1005400. [7722-76-1]. Monobasic ammonium solution daily.
phosphate.
Ammonium molybdate soluHon R6. 1005709.
White or almost white, crystalline powder or colourless
crystals, freely soluble in water. Slowly add 10 mL of sulfuric acid R to about 40 mL of
water R. Mix and allow to cool. Dilute to 100 mL with
pH (2.2.3): about 4.2 for a 23 giL solution. water R and mix. Add 2.5 g of ammonium molybdate R and
Ammonium formateo CH sN0 2. (M, 63.1). 1112600. 1 g of cerium sulfate R, and shake for 15 min to dissolve.
[540-69-2].
Ammonium nitrate. NH 4N0 3 • (Mr 80.0). 1005800.
Deliquescent crystals or granules, very soluble in water, [6484-52-2].
soluble in ethanol (96 per cent).
White or almost white, crystalline powder or colourless
mp: 119 oC to 121°C. crystals, hygroscopic, very soluble in water, freeIy soluble in
Storage: in an airtight container. methanol, soluble in ethanol (96 per cent).
Ammonium hexafluorogermanate(IV). (NH4)2GeF6' Storage: in an airtight container.
(M, 222.7). 1134000. [16962-47-3].
Ammonium nitraíe Rl. 1005801.
White or almost white crystals, freely soluble in water.
Complies with the requirements prescribed for ammonium
Ammonium hydrogen carbonate. NH 4HC03' (M, 79.1). nitrate R with the following additional requirements.
1005500. [1066-33-7]. Acidity. The solution of the substance is slightly acid (2.2.4).
Content: minimum 99 per cent. Chlorides (2.4.4): maximum 100 ppm, determined on
Ammonium molybdate. (NH4)6MoP24,4Hp. (M, 1236). 0.50 g.
1005700. [12054-85-2]. Sulfates (2.4.13): maximum 150 ppm, determined on 1.0 g.
Colourless or slightly yellow or greenish crystals, soluble in Sulfated ash (2.4.14): maximum 0.05 per cent, determined
water, practically insoluble in ethanol (96 per cent). on 1.0 g.
Ammonium molybdate reagent. 1005701. Ammonium oxalate. C 2 H sNP4,Hp. (M, 142.1). 1005900.
Mix, in the given order, 1 volume of a 25 giL solution of [6009-70-7].
ammonium molybdate R, 1 volume of a 100 giL solution of Colourless crystals, soluble in water.
ascorbic aeid R and 1 volume of sulfurie acid R (294.5 giL
H 2S0 4 ). Add 2 volumes of water R. Ammonium oxalate solution. 1005901.
Storage: use within 1 day. A 40 giL solution.

Ammonium molybdate reagent Rl. 1005706. Ammonium persulfate. (NH4)2SPS' (M, 228.2). 1006000.
Mix 10 mL of a 60 giL solution of disodium arsenate R, [7727-54-0].
50 mL of ammonium molybdate solution R, 90 mL of dilute White or almost white, crystalline powder ar granular crystals,
sulfuric acid R and dilute to 200 mL in water R. freely soluble in water.

432 See the information section on general monographs (caver pages)


EUROPEAN PHARMACOPOEIA 8.0 4.1.1. Reagents

Ammonium phosphate. (NH4)2HP04' (Mr 132.1). 1006100. ~-Amyrin. C30 H so O. (M r 426.7). 1141800. [559-70-6].
[7783-28-0]. Diammonium hydrogen phosphate. Olean-12-en-3~-01.
White or almost white crystals or granules, hygroscopic, very White or almost white powder.
soluble in water, practically insoluble in ethanol (96 per cent). mp: 187 oC to 190 oc.
pH (2.2.3): about 8 for a 200 giL solution.
Storage: in an airtight container. Anethole. ClQH¡p. (Mr 148.2). 1006900. [4180-23-8].
1- Methoxy-4-(propen-1-yl)benzene.
Ammonium pyrrolidinedithiocarbamate. CSH 12 N 2S2' White or almost white, crystalline mass up to 20 oC to 21 oC,
(Mr 164.3). 1006200. [5108-96-3]. Ammonium liquid aboye 23 oC, practically insoluble in water, freely soluble
1- pyrrolidinyl-dithioformate. in anhydrous ethanol, soluble in ethyl acetate and in light
White or pale yellow, crystalline powder, sparingly soluble in petroleum.
water, very slightly soluble in ethanol (96 per cent). n~5 : about 1.56.
Storage: in a bottle containing a piece of ammonium carbonate bp: about 230 oc.
in a muslin bago
Anethole used in gas chromatography complies with the
Ammonium reineckate. NH 4[Cr(NCS)4(NH J )2],Hp. following additional test.
(M, 354.4). 1006300. [13573-16-5]. Ammonium Assay. Gas chromatography (2.2.28) as prescribed in the
diamine- tetrakis (isothiocyanato )chromate(III) monohydrate. monograph Anise oil (0804).
Red powder or crystals, sparingly soluble in cold water, soluble Test solution. The substance to be examined.
in hot water and in ethanol (96 per cent).
Content: minimum 99.0 per cent of trans-anethole (retention
Ammonium reineckate solution. 1006301. time: about 41 min), calculated by the normalisation
A 10 giL solution. Prepare immediately before use. procedure.

Ammonium sulfamate. NH 2 S0 3 NH 4 . (Mr 114.1). 1006400. AniHne. C6H7N. (Mr 93.1). 1007100. [62-53-3].
[7773-06-0]. Benzeneamine.
White or almost white, crystalline powder or colourless Colourless or slightly yellowish liquid, soluble in water,
crystals, hygroscopic, very soluble in water, slightly soluble in miscible with ethanol (96 per cent).
ethanol (96 per cent). d§g: about 1.02.
mp: about 130 oc. bp: 183 oC to 186 oc.
Storage: in an airtight container. Storage: protected from light.
Ammonium sulfate. (NH4)2S04' (M, 132.1). 1006500. AniHne hydrochloride. C 6 H sCIN. (Mr 129.6). 1147700.
[7783-20-2]. [142-04-1]. Benzenamine hydrochloride.
Colourless crystals or white or almost white granules, very Crystals.
soluble in water, practically insoluble in acetone and in ethanol It darkens on exposure to air and light.
(96 per cent).
mp: about 198 oc.
pH (2.2.3) : 4.5 to 6.0 for a 50 giL solution in carbon
dioxide-free water R. Storage: protected from lighí.
Sulfated ash (2.4.14): maximum 0.1 per cent. Content: minimum 97.0 per cent.

Ammonium sulfide solution. 1123300. Anion-exchange resino 1007200.


Saturate 120 mL of dilute ammonia R1 with hydrogen sulfide R Resin in chlorinated form containing quaternary ammonium
and add 80 mL of dilute ammonia Rl. Prepare immediately groups [CH 2N+(CH J )3] attached to a polymer lattice consisting
before use. of polystyrene cross-linked with 2 per cent of divinylbenzene.
It is available as spherical beads and the particle size is
Ammonium thiocyanate. NH 4SCN. (Mr 76.1). 1006700. specified in the monograph.
[1762-95-4].
Wash the resin with 1 M sodium hydroxide on a sintered-glass
Colourless crystals, deliquescent, very soluble in water, soluble filter (40) (2.1.2) until the washings are free from chloride,
in ethanol (96 per cent). then wash with water R until the washings are neutral.
Storage: in an airtight container. Suspend in freshly prepared ammonium-free water R and
protect from atmospheric cal"bon dioxide.
Ammonium thiocyanate solution. 1006701.
A 76 giL solution. Anion-exchange resin Rl. 1123400.
Ammonium vanadaíe. NH 4VO y (Mr 117.0). 1006800. Resin containing quaternary ammonium groups
[7803-55-6]. Ammonium trioxovanadate(V). [CH 2N+(CH 3)3] attached to a lattice consisting of methacrylate.
White or slightly yellowish, crystalline powder, slightly soluble Anion-exchange resin R2. 1141900.
in water, soluble in dilute ammonia R1. Conjugate of homogeneous 10 [lm hydrophilic polyether
Ammonium vanadate solution. 1006801. particles, and a quaternary ammonium salt, providing a
Dissolve 1.2 g of ammonium vanadate R in 95 mL of matrix suitable for strong anion-exchange chromatography
water R and dilute to 100 mL with sulfuric acid R. of proteins.

AmoxiciHin trihydrate. 1103400. Anion-exchange resin R3. 1180900.


See Amoxicillin trihydrate (0260). Resin with quaternary ammonium groups attached to a
lattice of ethylvinylbenzene crosslinked with 55 per cent of
a-Amylase. 1100800. l,4-a-D-glucane-glucanohydrolase divinylbenzene.
(EC 3.2.1.1).
White or light brown powder. Anion-exchange resin for chl'Omatography, strongly basi.c.
1112700.
a-Amylase solution. 1100801. Resin with quaternary amine groups attached to a lattice of
A solution of a-amylase R with an activity of 800 FAU/g. latex cross linked with divinylbenzene.

General Notices (1) apply to all monographs and other texts 433
4.1.1. Reagents EUROPEAN PHARMACOPOEIA 8.0

Anion-exchange resin for chromatography, strongly Antimony potassium tartrate. C4H4K07Sb,1/2HP.


basicRl. 1187400. (M, 333.9). 1007600. Potassium aqua[tartrato(4-)-OI,02,03]-
Non-porous resin agglomerated with a 100 nm alkyl antimoniate(III) hemihydrate.
quaternary ammonium functionalised latex. White or almost white, granular powder or colourless,
transparent crystals, soluble in water and in gIycerol, freely
Anion-exchange resin, strongly basic. 1026600. soluble in boiling water, practically insoluble in ethanol
Gel-type resin in hydroxide form containing quaternary (96 per cent). The aqueous solution is slightly acid.
ammonium groups [CH 2N+(CH 3 )3' type 1] attached to a
polymer lattice consisting of polystyrene cross-linked with Antimony trichloride. SbCl)" (Mr 228.1). 1007700.
8 per cent of divinylbenzene. [10025-91-9].
Brown transparent beads. Colourless crystals or a transparent crystalline mass,
hygroscopic, freely soluble in anhydrous ethanol. Antimony
Particle size: 0.2 mm to 1.0 mm. trichloride is hydrolysed by water.
Moisture cantent: about 50 per cent. Storage: in an airtight container, protected from moisture.
Total exehange eapacity: minimum 1.2 meq/mL.
Antimony trichloride solution. 1007701.
Anion-exchange resin, weak. 1146700. Rapidly wash 30 g of antimony trieh/oride R with two
Resin with diethylaminoethyl groups attached to a lattice quantities, each of 15 mL, of ethano/-free ehloroform R,
consisting of poly(methyl methacrylate). drain off the washings, and dissolve the washed crystals
immediately in 100 mL of ethanol-free eh/oroform R,
Anisaldehyde. CSHsÜ2' (Mr 136.1). 1007300. [123-11-5]. warming slightly.
4-Methoxybenzaldehyde.
Storage: over a few grams of anhydrous sodium sulfate R.
Oily liquid, very slightly soluble in water, miscible with
ethanol (96 per cent). Antimony trichloride solution Rl. 1007702.
bp: about 248 oc. So/ution A. Dissolve llO g of antimony trich/oride R in
Anisaldehyde used in gas chromatography camp/ies with the 400 mL of ethylene eh/oride R. Add 2 g of anhydrous
following additional test. aluminium oxide R, mix and filter through a sintered-glass
filter (40) (2.1.2). Dilute to 500.0 mL with ethy/ene
Assay. Gas chromatography (2.2.28) as prescribed in the eh/oride R and mix. The absorbance (2.2.25) of the solution,
monograph Anise oi/ (0804). determined at 500 nm in a 2 cm cell, is not greater than
Test solution. The substance to be examined. 0.07.
Content: minimum 99.0 per cent, calculated by the Solution B. In a fume cupboard, mix 100 mL offreshly
normalisation procedure. distilled acetyl eh/oride R and 400 mL of ethylene chloríde R.
Anisaldehyde solution. 1007301. Mix 90 mL of solution A and 10 mL of solution B.
Storage: in brown ground-glass-stoppered bottle for 7 days.
Mix in the following order, 0.5 mL of anísaldehyde R,
Discard any reagent in which colour develops.
10 mL of glacial acetic acid R, 85 mL of methanol R and
5 mL of sulfurie aeid R. Antithrombin IH. 1007800. [90170-80-2].
Anisaídehyde solution Rl. 1007302. Antithrombin III is purified from human plasma by heparin
agarose chromatography and should have a specific activity of
To 10 mL of anisa/dehyde R add 90 mL of ethano/ (96 per at least 6 IU/mg.
cent) R, mix, add 10 mL of sulfuric aeid R and mix again.
Antithrombin nI solution Rl. 1007801.
Anise ketone. C lO H 12 02' (Mr 164.2). 1174700. [122-84-9].
1-( 4-Methoxyphenyl)propan -2-one. Reconstitute antithrombin JIf Ras directed by the manu-
facturer and dilute with tris(hydroxymethyl)aminomethane
p-Anisidine. C7H9NO. (Mr 123.2). 1103500. [104-94-9]. sodium eh/oride buffer so/ution pH 7.4 R to 1 IU/mL.
4-Methoxyaniline.
Antithrombin In solution R2. 1007802.
White or almost white crystals, sparingly soluble in water,
Reconstitute antithrombin fJI R as directed by the manu-
soluble in anhydrous ethanol.
facturer and dilute with tris(hydroxymethyl)aminomethane
Content: minimum 97.0 per cent. sodium ch/oride buffer solution pH 7.4 R to 0.5 IU/mL.
Cautíon: skín írritant, sensítíser.
Antithrombin nI solution R3. 1007803.
Storage: protected from light, at O oC to 4 oc.
Reconstitute antithrombin JIf R as directed by the
On storage, p-anisidine tends to darken as a result of oxidation. manufacturer and dilute to 0.3 IU/mL with phosphate
A discoloured reagent can be reduced and decolorised in buffer so/ution pH 6.5 R.
the following way: dissolve 20 g of p-anisidine R in 500 mL
of water R at 75 oc. Add 1 g of sodium sulfite R and 10 g of Antithrombin In solution R4. 1007804.
activated eharcoa/ R and stir for 5 mino Filter, cool the filtrate Reconstitute antithrombin fJI R as directed by
to about O oC and allow to stand at this temperature for at least the manufacturer and dilute to 0.1 IU/mL with
4 h. Filter, wash the crystals with a small quantity of water R at tris(hydroxymethyl)aminomethane-EDTA buffer so/ution
about O oC and dry the crystals in vacuum over diphosphorus pH 8.4 R.
pentoxide R.
Apigenin. C 1sHlOOs' (Mr 270.2). 1095800. [520-36-5].
Anthracene. C 14H lO • (Mr 178.2). 1007400. [120-12-7]. 4',5,7 -Trihydroxyflavone.
White or almost white, crystalline powder, practically Light yellowish powder, practically insoluble in water,
insoluble in water, slightly soluble in chloroform. sparingly soluble in ethanol (96 per cent).
mp: about 218 oc. mp: about 310 oC, with decomposition.
Chromatography. Thin-layer chromatography (2.2.27) as
Anthrone. C 14H lO O. (M, 194.2). 1007500. [90-44-8].
prescribed in the monograph Roman ehamomi/e flower (0380):
9(10H)- Anthracenone.
apply 10 f.lL of a 0.25 giL solution in methanol R; the
Pale yellow, crystalline powder. chromatogram shows in the upper third a principal zone of
mp: about 155 oc. yellowish-green fluorescence.

434 See the informatiol1 seetion on general monographs (cover pages)


EUROPEAN PHARMACOPOEIA 8.0 4.1.1. Reagents

Apigenin 7-g1ucoside. C21HZaÜlO' (Mr 432.4). 1095900. Assay. Gas chromatography (2.2.28) as prescribed in the
[578-74-5]. Apigetrin. 7-(~-D-Glueopyranosyloxy)-5- monograph on Tea tree oil (1837).
hydroxy-2-( 4-hydroxyphenyl)-4H-l-benzopyran-4-one. Content: minimum 92 per cent, ealculated by the
Light yellowish powder, praetically insoluble in water, normalisation proeedure.
sparingly soluble in ethanol (96 per eent).
Arsenious trioxide. ASP3' (M, 197.8). 1008300. [1327-53-3].
mp: 198 oC to 201 oc.
Arsenious anhydride. Diarsenic trioxide.
Chromatography. Thin-Iayer ehromatography (2.2.27) as
Crystalline powder or a white or almost white mass, slightly
prescribed in the monograph Roman chamomile jlower (0380):
soluble in water, soluble in boiling water.
apply 10 I1L of a 0.25 giL solution in methanol R; the
ehromatogram shows in the middle third a principal zone of Arsenite solntion. 1008301.
yellowish fluoreseenee. Dissolve 0.50 g of arsenious trioxide R in 5 mL of diiute
Apigenin-7-glucoside used in liquid chromatography complies
sodium hydroxide solution R, add 2.0 g of sodium hydrogen
with the following additional test.
carbonate R and dilute to 100.0 mL with water R.
Assay. Liquid ehromatography (2.2.29) as prescribed in the
monograph Matricaria flower (0404). Ascorbic add. 1008400. [50-81-7].
Test solution. Dissolve 10.0 mg in methanol R and dilute to See Ascorbic acid (0253).
100.0 mL with the same solvent.
Ascorbic ¡¡cid solution. 1008401.
Content: minimum 95.0 per cent, ealculated by the
normalisation proeedure. Dissolve 50 mg in 0.5 mL of water R and dilute to 50 mL
with dimethylformamide R.
Aprotinin. 1007900. [9087-70-1].
Asiaticoside. C1sH78019' (Mr 959). 1123500.
See Aprotinin (0580). [16830-15-2]. 0-6-Deoxy-a-L-mannopyranosyl-
Arabinose. CSHlOOS' (Mr 150.1). 1008000. [87-72-9]. (104)-0-~- D-glueopyranosyl-( 106) -~- D-glucopyranosyl
L-( +)-Arabinose. 2a,3 ~,23- trihydroxy-4a -urs-12-en -28-oate.
White or almost white, erystalline powder, freely soluble in White or almost white powder, hygroseopic, soluble in
water. methanol, slightly soluble in anhydrous ethanol, insoluble in
[al~o: + 103 to + 105, determined on a 50 giL solution in
acetonitrile.
water R eontaining about 0.05 per eent of NH3' mp: about 232 oC, with deeomposition.
Water (2.5.12): 6.0 per eent.
Arachidylakohol. C 2o H 4p. (Mr 298.5). 1156300. [629-96-9].
1-Eicosanol. Asiaticoside used in liquid chromatography complies with the
following additional test.
mp: about 65 oc.
Assay. Liquid ehromatography (2.2.29) as preseribed in the
Cantent: minimum 96 per eent of C 2o H 42 0.
monograph Centella (1498).
Arimtin. C'2H160?' (Mr 272.3). 1008100. [497-76-7]. Content: minimum 97.0 per eent, calculated by the
Arbutoside. 4- Hydroxyphenyl-~- D-glueopyranoside. normalisation procedure.
Fine, white or almost white, shiny needles, freely soluble in : protected from humidity.
water, very soluble in hot water, soluble in ethanol (96 per
eent). Aspartic add. 1134100. [56-84-8].
Chromatography. Thin-Iayer chromatography (2.2.27) as See Aspartic acid (0797).
prescribed in the monograph Bearberry leaf (1054); the
L-Aspartyl-L-phenylalanine. C13H16N20s' (M, 280.3)0
ehromatogram shows only one principal spot. 10085000 [13433-09-5]. (S)-3-Amino-N-[(S)-I-earboxy-2-
Arginine. 1103600. [74-79-3]. phenylethyl] -suecinamic acid.
See Arginine (0806). White or almost white powder.
mp: about 210 oC, with deeomposition.
Argon. Ar. (A, 39.95).1008200. [7440-37-1].
Content: minimum 99.995 per eent V/V. Astragaloside IV. C41H6S0W (M, 785). 11782000
Carbon monoxide (2.5.25, Method 1): maximum 0.6 ppm V/V; [84687 -43-4]. (20R,24S)-20,24- Epoxy-16~,25-dihydroxy-
after passage of 10 L of argon R at a flow rate of 4 Llh, not 3~- (~- D-xylopyranosyloxy) -9, 19-cyclolanostan -6a -yl
more than 0.05 mL of 0.002 M sodium thiosulfate is required ~-D-glueopyranoside.
for the titration. Atropine sulfate. 1159000. [5908-99-6].
Argon Rl. Ar. (A r 39.95).1176000. [7440-37-1]. See Atropine sulfate (0068).
Content: minimum 99.99990 per cent V/V.
Aucubin. C 1s H 22 0 9 . (M, 346.3 ). 1145200. [479-98-1].
Argon far ehromatography. Ar. (A r 39.95). 1166200. [lS,4aR,5S,7aS)-5- Hydroxy-7 -(hydroxymethyl)-1,4a,5,7a-
[7440-37-1]. tetrahydroeyclopenta[c ]pyran -l-yl ~-D-glucopyranoside.
Content: mínimum 99.95 per cent V/V. Crystals, soluble in water, in ethanol (96 per cent) and in
methanol, practieally insoluble in light petroleum.
Aramadendrene. C 1S H24' (Mr 204.4).1139100. [489-39-4].
[al~o: about - 163.
(1R,2S,4R,8R, 11R) -3,3, 11-Trimethyl-7 -methylenetricyclo-
[6.3.0.02.4]undecane. mp: about 181 oc.
Clear, almost colourless liquido Azomethine H. C17H12NNaOSS2' (M, 445.4). 1008700.
d~o: about 0.911. [5941-07-1]. Sodium hydrogeno-4-hydroxy-5-(2-
nGo: about 1.497. hydroxybenzylideneamino)- 2, 7 -naphthalenedisulfonate.
[al~o: about + 12. Azomethine H solution. 1008701.
bp: about 263 oc. Dissolve 0.45 g of azomethine H R and 1 g of ascorbic acid R
Aromadendrene used in gas chromatography complies with with gen tI e heating in water R and dilute to 100 mL with
the following additional test. the same solvent.

General Notices (1) apply to all monographs and other texts 435
4.1.1. Reagents EUROPEAN PHARMACOPOEIA 8.0

BaicaHn. C2¡H¡Pll' (MI 446.4). 1179200. [21967-41-9]. Content: minimum 98.0 per cent.
5,6-Dihydroxy-4-oxo-2-phenyl-4H-l-benzopyran-7 -yl-~-D­ bp: about 261 oc.
glucopyranosiduronic acid. mp: about 39 oc.
Barbaloin. C21Hn09,HP. (Mr 436.4). 1008800.
Benzaldehyde. C7H60. (Mr 106.1). 1009600. [100-52-7].
[1415-73-2]. Aloin. 1,8- Dihydroxy-3-hydroxymethyl-1O-~-D­
glucopyranosyl-1 OH-anthracen -9-one. Colourless or slightly yellow liquid, slightly soluble in water,
Yellow to dark-yellow, crystalline powder, or yellow needles, miscible with ethanol (96 per cent).
darkening on exposure to air and light, sparingly soluble in d~g : about 1.05.
water and ín ethanol (96 per cent), soluble in acetone, in ntO : about 1.545.
ammonia and in solutions of alkali hydroxides. Distillation range (2.2.11). Not less than 95 per cent distils
Ai~rn: about 192 at 269 nm, about 226 at 296.5 nm, about between 177 oC and 180 oc.
259 at 354 nm, determined on a solution in methanol R and
Storage: protected from light.
calculated with reference to the anhydrous substance.
Chromatography. Thín-Iayer chromatography (2.2.27) as Benzene. C6H6' (Mr 78.1). 1009800. [71-43-2].
prescribed in the monograph Frangula bark (0025); the Clear, colourless, flammable liquid, practically insoluble in
chromatogram shows only one principal spot. water, miscible with ethanol (96 per cent).
Barbital. 1008900. [57-44-3]. bp: about 80 oC.
See Barbital (0170).
Benzene-l,2,4-triol. C6H60y (Mr 126.1). 1177500.
Barbital sodium. C S HJlN 2 NaO y (M, 206.2). 1009000. [533-73-3]. Hydroxyhydroquinone. Hydroxyquinol.
[144-02-5]. Sodium derivative of 5,5-diethyl-lH,3H,5H- Freely soluble in water, in ethanol (96 per cent) and in ethyl
pyrimídine-2,4,6-trione. acetate.
Content: mínimum 98.0 per cent. mp: about 140 oc.
A white or almost white, crystalline powder or colourless
crystals, freely soluble in water, slightly soluble in ethanol Benzethonium chloride. C27H42CIN02,Hp. (M, 466.1).
(96 per cent). 1009900. [121-54-0]. Benzyldimethyl[2-[2-[ 4-(1,1,3,3-
tetramethylbu tyl )phenoxy] ethoxy] ethyl] ammonium chloride
Barbituric add. C4H 4 Npy (Mr 128.1). 1009100. [67-52-7]. monohydrate.
lH,3H,5H-Pyrimidine-2,4,6-trione.
Fine, white or almost white powder or colourless crystals,
White or almost white powder, slightly soluble in water, freely soluble in water and in ethanol (96 per cent).
soluble in boiling water and in dilute acids.
mp: about 163 oc.
mp: about 253 oc.
Storage: protected from light.
Bari.um acetate. C4H 6Ba0 4. (MI 255.4). 1162700. [543-80-6].
Barium diacetate. Benzidine. C12H¡ZN2. (Mr 184.2). 1145300. [92-87-5].
Biphenyl-4,4' -diamine.
White or almost white powder, soluble in water.
d~g: 2.47. Content: minimum 95 per cent.
White or slightly yellowish or reddish powder, darkening on
Barium carbonate. BaC0 3. (M, 197.3). 1009200. [513-77-9]. exposure to air and light.
White or almost white powder or friable masses, practically mp: about 120 oc.
insoluble in water.
Storage: protected from light.
Barium chloride. BaCl2,2Hp. (M, 244.3). 1009300.
[10326-27-9]. Baríum dichloride. Benzil. C I4 H lO 0 2• (M, 210.2). 1117800. [134-81-6].
Colourless crystals, freely soluble in water, slightly soluble in Diphenylethanedione.
ethanol (96 per cent). Yellow, crystalline powder, practically insoluble in water,
soluble in ethanol (96 per cent), ethyl acetate and toluene.
Barium chloride solution Rl. 1009301.
mp: 95 oc.
A 61 giL solution.
Benzocaine. C9H¡IN0 2 • (Mr 165.2). 1123600. [94-09-7].
Barium chloride solution R2. 1009302.
See Benzocaine (0011).
A 36.5 giL solution.
Barium hydroxide. Ba(OH)2,8Hp. (M, 315.5). 1009400. Benzoic add. 1010100. [65-85-0].
[12230-71-6]. Barium dihydroxide. See Benzoic acid (0066).
Colourless crystals, soluble in water.
Benzoin. C¡4H¡P2' (MI 212.3). 1010200. [579-44-2].
Barium hydroxide solution. 1009401. 2-Hydroxy-1,2-dipheny!ethanone.
A 47.3 giL solution. Slightly yellowish crystals, very slightly soluble in water, freely
soluble in acetone, soluble in hot ethanol (96 per cent).
Barium nitrate. Ba(NOJ2' (Mr 261.3). 1163800.
[10022-31-8]. mp: about 137 oc.
Crystals or crystalline powder, freely soluble in water, very Benzophenone. C¡3HlOO. (Mr 182.2). 1010300. [119-61-9].
slightly soluble in ethanol (96 per cent) and in acetone. Diphenylmethanone.
mp: about 590 oc. Prismatic crystals, practically insoluble in water, freely soluble
Bari.um sulfate. 1009500. [7727-43-7]. in ethanol (96 per cent).
See Barium sulfate (0010). mp: about 48 oc.

Benzalacetone. ClOHlOO. (Mr 146.2). 1168500. [122-57-6]. 1,4-Benzoquinone. C 6 HP2' (M, 108.1).1118500. [106-51-4].
(3E)-4- phenylbut -3-en -2-one. Cyclohexa-2,5-diene-1,4-dione.
White or pale yellow mass. Content: minimum 98.0 per cent.

436 See the information section on general monographs (cover pages)


EUROPEAN PHARMACOPOEIA 8.0 4.1.1. Reagents

Benzoylarginíne ethyl ester hydrochloride. Yellow liquido


C¡SH 23 CINp3' (Mr 342.8). 1010500. [2645-08-1]. mp: 13 oC to 16 oc.
N-Benzoyl-L-arginine ethyl ester hydrochloride. Ethyl
(5) -2-benzamido- 5-guanidinovalerate hydrochloride. 4-Benzylpyridine. C 12 H ll N. (M, 169.2). 1181200.
White 01' almost white, crystalline powder, very soluble in [2116-65-6].
water and in anhydrous ethano!. Content: minimum 98.0 per cent.
[al~o: - 15 to - 18, determined on a 10 giL solution. Yellow liquido
mp: about 129 oc. mp: 72 oC to 78 oc.
Ai~rn: 310 to 340, determined at 227 nm using a 0.01 giL Benzyltrimethylammonium chloride. C¡oH¡6C1N. (Mr 185.7).
solution. 1155700. [56-93-9]. N,N,N- Trimethylphenylmethanaminium
chloride. N,N,N- Trimethylbenzenemethanaminium chloride.
Benzoyl chloride. C 7 H sCIO. (Mr 140.6). 1010400. [98-88-4].
White or almost white powder, soluble in water.
Colourless, lachrymatory liquid, decomposed by water and by
mp: about 230 oC, with decomposition.
ethanol (96 per cent).
d§g: about 1.21. Berberine chloride. C2oHIsCIN04,2Hp. (Mr 407.8). 1153400.
bp: about 197 oc. [5956-60-5]. 9,10-Dimethoxy-5,6-dihydrobenzo [g]-1,3-
benzodioxolo [5,6-a] quinolizinium chloride.
N- Benzoyl-L- prolyl-L-phenylalanyl-L-arginine Yellow crystals, slightly soluble in water, practically insoluble
4-nitroanilide acetate. C 3s H 42 Nps' (Mr 703). 1010600. in ethanol (96 per cent).
3-Benzoylpropionic add. CIOHIO03' (M, 178.2). 1171000. mp: 204 oC to 206 oc.
[2051-95-8]. 4-0xo-4-phenylbutanoic acid. Berberine chloride used in liquid chromatography complies
mp: about 118 oc. with the following additional test.
Assay. Liquid chromatography (2.2.29) as prescribed in the
2-Benzoylpyridine. C 12H gNO. (M, 183.2). 1134300. monograph Goldenseal rhizome (1831).
[91-02-1]. Phenyl(pyridin -2-yl)methanone.
Content: minimum 95 per cent, calculated by the
Colourless crystals, soluble in ethanol (96 per cent). normalisation procedure.
mp: about 43 oc.
Bergapten. C¡2HsÜ4' (Mr 216.2). 1103700. [484-20-8].
Benzyl alcohol. 1010700. [100-51-6]. 5-Methoxypsoralen.
See Benzyl alcohol (0256). Colourless crystals, practically insoluble in water, sparingly
soluble in ethanol (96 per cent) and slightly soluble in glacial
Benzyl benzoate. 1010800. [120-51-4]. acetic acid.
See Benzyl benzoate (0705). mp: about 188 oc.
Chromatography. Thin-Iayer chromatography (2.2.27) as
prescribed in the monograph Peru balsam (0754): apply 20 ¡.rL Betulin. C30Hso02' (Mr 442.7). 1011100. [473-98-3].
Lup-20(39)-ene-3~,28-diol.
of a 0.3 per cent V/V solution in ethyl aceta te R; after spraying
and heating, the chromatogram shows a principal band with White or almost white, crystalline powder.
an Rp of about 0.8. mp: 248 oC to 251 oc.
Benzyl dnnamate. C¡6H¡P2' (M, 238.3). 1010900. Bibenzyl. C¡4HI4' (Mr 182.3). 1011200. [103-29-7].
[103-41-3]. Benzy13-phenylprop-2-enoate. 1,2-Diphenylethane.
Colourless or yellowish crystals, practically insoluble in water, White or almost white, crystalline powder, pradically
soluble in ethanol (96 per cent). insoluble in water, very soluble in methylene chloride, freely
mp: about 39 oc. soluble in acetone, soluble in ethanol (96 per cent).
Chromatography. Thin-Iayer chromatography (2.2.27) as mp: 50 oC to 53 oc.
prescribed in the monograph Peru balsam (0754): apply

-
Biphenyl. Cl2HIO' (M, 154.2). 1168600. [92-52-4].
20 ¡.tL of a 3 giL solution in ethyl aceta te R; after spraying and mp: 68 oC to 70 oc.
heating, the chromatogram shows a principal band with an Rp
of about 0.6.
(-)-a-BisaboloL C 1sH 26 0. (M, 222.4). 1128800. [23089-26-1].
Benzyl cyanide. C S H 7 N. (Mr 117.2). 1171100. [140-29-4]. (25)-6-Methyl-2- [( 15)-4-methylcyclohex -3-enyl] hept -5-en-
Phenylacetonitrile. 2-0!. Levomenol.
Content: minimum 95.0 per cent. Colourless, viscous liquid with a slight, characteristic odour,
Clear, colourless or light yellow liquido practically insoluble in water, freely soluble in ethanol (96 per
cent), in methanol, in toluene, in fatty oils and in essential oils.
nbo : about 1.523.
bp: about 233 oc.
d§g: 0.925 to 0.935.
nbo: 1.492 to 1.500.
Benzyl ether. C14H140. (Mr 198.3). 1140900. [103-50-4]. [al~o: - 54.5 to - 58.0, determined on a 50 giL solution in
Dibenzyl ether. ethanol (96 per cent) R.
Clear, colourless liquid, practically insoluble in water, miscible (- )-a-Bisabolol used for gas eh ro m atography complies with
with acetone and with anhydrous ethano!. the following additional test.
d§g: about 1.043. Assay. Gas chromatography (2.2.28) as prescribed in the
nbo : about 1.562. monograph Matricaria oil (1836).
bp: about 296 oC, with decomposition. Test solution. A 4 giL solution in cyclohexane R.
Content: minimum 95.0 per cent, calculated by the
BenzylpenicHUn sodium. 1011000. [69-57-8].
normalisation procedure.
See Benzylpenicillin sodium (0114).
Bisbenzimide. C2sH27CI3N60,5H20. (Mr 624). 1103800.
2-Benzylpyridine. C 12 H 11 N. (Mr 169.2).1112900. [101-82-6]. [23491-44-3]. 4- [5- [5-( 4-Methylpiperazin-l-yl)benzimidazol-
Content: minimum 98.0 per cent. 2-yl]benzimidazol-2-yl] phenol trihydrochloride pentahydrate.

General Notices (1) apply to all monographs and other texts 437
4.1.1. Reagents EUROPEAN PHARMACOPOEIA 8.0

Bisbenzimide stock solution. 1103801. 130ric add solution, saturated, cold. 1011801.
Dissolve 5 mg of bisbenzimide R in water R and dilute to To 3 g of borie acid R add 50 mL of water R and shake for
100 mL with the same solvent. 10 mino Place the solution for 2 h in the refrigerator.
Storage: in the dark. Borneo!. ClOH1SO. (Mr 154.3). 1011900. [507-70-0].
Bisbenzimide wOl'king soh.l.tion. 1103802. endo-l, 7,7 -Trimethylbicyclo [2.2.1]heptan- 2-01.
Immediately before use, dilute 100 flL of bisbenzimide Colourless crystals, readily sublimes, practically insoluble
stock soluNon R to 100 mL with phosphate buffered saline in water, freely soluble in ethanol (96 per cent) and in !ight
pH 7.4 R. petroleum.
mp: about 208 oc.
Bismuth nitrate pentahydrate. Bi(N03)v5Hp. (Mr 485.1). Chromatography. Thin-layer chromatography (2.2.27), using
1165600. [10035-06-0]. si/iea gel G R as the coating substance. Apply to the plate
mp: about 30 oc. 10 flL of a 1 giL solution in toluene R. Develop over a path of
10 cm using chloroform R. Allow the plate to dry in air, spray
Bismuth subnitrate. 4BiN0 3 (OH)2,BiO(OH). (Mr 1462). with anisaldehyde solution R, using 10 mL for a plate 200 mm
1011500. [1304-85-4].
square, and heat at 100-105 oC for 10 mino The chromatogram
White or almost white püwder, practically insoluble in water. obtained shows only one principal spot.
Bismuth subnitrate Rl. 1011501. Bornyl acetate. C12H2002' (Mr 196.3). 1012000. [5655-61-8].
Content: 71.5 per cent to 74.0 per cent of bismuth (Bi), endo-1,7,7 - Trimethylbicyclo[2.2.1]hept -2-yl acetate.
and 14.5 per cent to 16.5 per cent of nitrate, calculated as Colourless crystals or a colourless liquid, very slightly soluble
nitro gen pentoxide (Nps)' in water, soluble in ethanol (96 per cent).
Bismuth subnitrate solutl.on. 1011502. mp: about 28 oc.
Dissolve 5 g of bismuth subnitrate R1 in a mixture of 8.4 mL Chromatography. Thin-layer chromatography (2.2.27), using
silica gel G R as the coating substance. Apply to the plate
of nitric acid R and 50 mL of water R and dHute to 250 mL
with water R. Filter if necessary. 10 flL of a 2 giL solution in toluene R. Develop over a path of
10 cm using ehloroform R. Allow the plate to dry in air, spray
AcidUy. To 10 mL add 0.05 mL of methyl orange solution R. with anisaldehyde so/ution R, using 10 mL for a plate 200 mm
5.0 mL to 6.25 mL of 1 M sodium hydroxide is required to square, and heat at 100-105 oC for 10 mino The chromatogram
change the colour of the indicator. obtained shows only one principal spot.
Bis-tris propane. C ll H 26 NP6' (Mr 282.3). 1185500. Boron tl'ichloride. BCl}' (Mr 117.2). 1112000. [10294-34-5].
[64431-96- 5]. 2,2' -(Propane-l ,3-diyldiimino) bis [2-
Colourless gas. Reacts violently with water. Available as
(hydroxymethyl)-1,3-propanediol.
solutions in suitable solvents (2-chloroethanol, methylene
Content: minimum 99.0 per cent. chloride, hexane, heptane, methanol).
Bimel. C2 H sNP2' (Mr 103.1). 1011600. [108-19-0]. ni? : about 1.420.
White or almost white crystals, hygroscopic, soluble in water, bp: about 12.6 oc.
sparingly soluble in ethanol (96 per cent). Caution: toxic and corrosive.
mp: 188 oC to 190 oC, with decomposition. Boron trichloride-methanol solutiol1. 1112001.
Storage: in an airtight container. A 120 giL solution of BCl3 in methanol R.
Biul'et reagent. 1011601. Storage: protected from light at - 20 oC, preferably in
sealed tubes.
Dissolve 1.5 g of capper sulfate R and 6.0 g of sodium
potassium tartrate R in 500 mL of water R. Add 300 mL of a 13oron trifluoride. BFy (Mr 67.8). 1012100. [7637-07-2].
carbonate-free 100 giL solution of sodium hydroxide R, dilute Colourless gas.
to 1000 mL with the same solution and mix.
Boron tl'ifiuoride-methanol soluHon. 1012101.
Blocking solution. 1122400. A 140 giL solution of boron trifluoride R in methanol R.
A 10 per cent V/V solution of acetic acid R.
Brilliant hiue. 1012200. [6104-59-2].
BIue dextran 2000. 1011700. [9049-32-5]. See acid blue 83 R.
Prepared from dextran having an average relative molecular
mass of 2 x 10 6 by introduction of a polycyclic chromophore Bromelains. 1012300. [37189-34-7].
that colours the substance blue. The degree of substitution is Concentrate of proteolytic enzymes derived from Ananas
0.017. comosus Merr.
It is freeze-dried and dissolves rapidly and completely in water Dull-yellow powder.
and aqueous saline solutions. Activity. 1 g liberates about 1.2 g of amino-nitrogen from a
Absorbance (2.2.25). A 1 giL solution in a phosphate buffer solution of gelatin R in 20 min at 45 oC and pH 4.5.
solution pH 7.0 R shows an absorption maximum at 280 nm. Bromelains solution. 1012301.
Boldine. C 19H 21 N0 4 . (Mr 327.3). 1118800. [476-70-0]. A 10 giL solution of bromelains R in a mixture of 1 volume
1,10-Dimethoxy-6aa -aporphine-2,9-diol. of phosphate buffer solution pH 5.5 R and 9 volumes of a
White or almost white crystalline powder, very slightly 9 giL solution of sodium chloride R.
soluble in water, soluble in ethanol (96 per cent) and in dilute Bromine. Br 2 • (Mr 159.8). 1012400. [7726-95-6].
solutions of acids. Brownish-red fuming liquid, slightly soluble in water, soluble
[a]g: about + 127, determined on a 1 giL solution in in ethanol (96 per cent).
anhydrous ethanol R. d~g: about 3.1.
mp: about 163 oc.
Bromine solution. 1012401.
Bode add. 1011800. [10043-35-3]. Dissolve 30 g of bromine R and 30 g of potassium bromide R
See Boric acid (0001). in water R and dilute to 100 mL with the same solvento

438 See the information section on general monographs (ca ver pages)
EUROPEAN PHARMACOPOEIA 8.0 4.1.1. Reagents

Bromine water. 1012402. Bromophenol bIue solution. 1012801.


Shake 3 mL of bromíne R with 100 mL of water R to Dissolve 0.1 g of bromophenol blue R in 1.5 mL of 0.1 M
saturation. sodium hydroxide and 20 mL of ethanol (96 per cent) R and
Storage: over an excess of bromine R, protected from light. dilute to 100 mL with water R.
Test for sensitivity. To 0.05 mL of the bromophenol blue
Bromine water Rl. 1012403.
solution add 20 mL of carbon dioxide-free water R and
Shake 0.5 mL of bromine R with 100 mL of water R. 0.05 mL of 0.1 M hydrochloric acid. The solution is yellow.
Storage: protected from light; use within 1 week. Not more than 0.1 mL of 0.1 M sodium hydroxide is
required to change the colour to bluish-violet.
Bromocresol green. C21H14Br40SS. (M, 698). 1012600.
Colour change: pH 2.8 (yellow) to pH 4.4 (bluish-violet).
[76-60-8]. 3',3",5',5"-Tetrabromo-m-cresol-sulfonphthalein.
4,4' -(3H-2,1-Benzoxathiol-3-ylidene)bis(2,6-dibromo-3-
Bromophenol biue solution Rl. 1012802.
methylphenol)-S,S-dioxide.
Dissolve 50 mg of bromophenol blue R with gentle heating
Brownish-white powder, slightly soluble in water, soluble
in 3.73 mL of 0.02 M sodium hydroxide and dilute to
in ethanol (96 per cent) and in dilute solutions of alkali
100 mL with water R.
hydroxides.
Bromocresol green-methyl red solution. 1012602. Bromophenol bIue solution R2. 1012803.
Dissolve 0.15 g of bromocresol green R and 0.1 g of methyl Dissolve with heating 0.2 g of bromophenol blue R in 3 mL
red R in 180 mL of anhydrous ethanol R and dilute to of 0.1 M sodium hydroxide and 10 mL of ethanol (96 per
200 mL with water R. cent) R. After solution is effected, allow to cool and dilute
to 100 mL with ethanol (96 per cent) R.
Bromocresol green solution. 1012601.
Dissolve 50 mg of bromocresol green R in 0.72 mL of 0.1 M Bromophos. C sH sBrCI 2 0 3 PS. (Mr 366.0). 1123700.
sodium hydroxide and 20 mL of ethanol (96 per cent) R and [2104-96-3].
dilute to 100 mL with water R. A suitable certified reference solution (lO ng/flL in iso-octane)
Test for sensitivity. To 0.2 mL of the bromocresol green maybe used.
solution add 100 mL of carbon dioxide-free water R.
The solution is blue. Not more than 0.2 mL of 0.02 M Bromophos-ethyl. CJOH12BrClzÜ3PS. (Mr 394.0). 1123800.
hydrochloric acid is required to change the colour to yellow. [4824-78-6].
Colour change: pH 3.6 (yellow) to pH 5.2 (blue). A suitable certified reference solution (lO ng/flL in iso-octane)
maybe used.
Bromocresol purple. C21H¡6Br20sS. (Mr 540.2). 1012700.
[115-40-2]. 3',3"-Dibromo-o-cresolsulfonphthalein. Bromothymol bIue. C27H2SBr20SS. (Mr 624). 1012900.
4,4' - (3H- 2,1-Benzoxathiol-3-ylidene )bis(2-bromo-6- [76- 59- 5]. 3',3"-Dibromothymolsulfonphthalein.
methylphenol)-S,S-dioxide. 4,4' -(3H-2, 1-Benzoxathiol-3-ylidene)bis(2-bromo-6-
isopropyl-3-methylphenol) S,S-dioxide.
Pinkish powder, practically insoluble in water, soluble
in ethanol (96 per cent) and in dilute solutions of alkali Reddish-pink or brownish powder, practically insoluble in
hydroxides. water, soluble in ethanol (96 per cent) and in dilute solutions
of alkali hydroxides.
Bromocresol purple solution. 101270l.
Dissolve 50 mg of bromocresol purple R in 0.92 mL of 0.1 M Bl'Omothymol bIue solution Rl. 1012901.
sodium hydroxide and 20 mL of ethanol (96 per cent) R and Dissolve 50 mg of bromothymol blue R in a mixture of 4 mL
dilute to 100 mL with water R. of 0.02 M sodium hydroxíde and 20 mL of ethanol (96 per
Test for sensitivity. To 0.2 mL of the bromocresol purple cent) R and dilute to 100 mL with water R.
SOlUtiOll add 100 mL of carbon dioxíde-free water R and Test for sensitivity. To 0.3 mL of bromothymol blue
0.05 mL of 0.02 M sodium hydroxide. The solution is solution R1 add 100 mL of carbon dioxide-free water R. The
bluish-violet. Not more than 0.2 mL of 0.02 M hydrochloric solution is yellow. Not more than 0.1 mL of 0.02 M sodium
acid is required to change the colour to yellow. hydroxide is required to change the colour to blue.
Colour change: pH 5.2 (yellow) to pH 6.8 (bluish-violet). Calour change: pH 5.8 (yellow) to pH 7.4 (bIne).
5-Bromo-2'-deoxyuridine. C9H¡¡BrNzÜs. (Mr 307.1).
Bl'Omothymol blue solution R2. 1012902.
1012500. [59-14-3]. 5-Bromo-l-(2-deoxy-p-d-erythro-
pentofuranosyl)-lH,3H-pyrimidine-2,4-dione. A 10 giL solution in dimethyiformamide R.
mp: about 194 oc. Bl'Omothymol blue solution R3. 1012903.
Chromatography. Thin-layer chromatography (2.2.27) as
Warm 0.1 g of bromothymol blue R with 3.2 mL of 0.05 M
prescribed in the monograph Idoxuridine (0669): apply 5 flL
sodium hydroxide and 5 mL of ethanol (90 per cent V/V) R.
of a 0.25 giL solution; the chromatogram shows only one
After solution is effected, dilute to 250 mL with ethanol
principal spot.
(90 per cent V/V) R.
Bromomethoxynaphthalene. CllH9BrO. (Mr 237.1).
1159100. [5111-65-9]. 2-Bromo-6-methoxynaphthalene. Bl'Omothymol blne solution R4. 1012904.
mp: about 109 oc. Dissolve 100 mg of bromothymol blue R in a mixture of
equal voIumes of ethanol (96 per cent) R and water R and
Bromophenol bine. C¡9HJOBrpsS. (Mr 670). 1012800. dilute to 100 mL with the same mixture of solvents. Filter
[115-39-9]. 3',3",5',5"-Tetrabromophenolsulfonphthalein. if necessary.
4,4' -(3H-2, 1-Benzoxathiol-3-ylidene)bis(2,6-dibromophenol)
S,S-dioxide. BRP indicator solution. 1013000.
Light orange-yellow powder, very slightly soluble in water, Dissolve 0.1 g of bromothymal blue R, 20 mg of methyl red R
slightly soluble in ethanol (96 per cent), freely soluble in and 0.2 g of phenolphthalein R in ethanol (96 per cent) R and
solutions of alkali hydroxides. dilute to 100 mL with the same solvento Filter.

General Notices (1) apply to all monographs and ather texts 439
4.1.1. Reagents EUROPEAN PHARMACOPOEIA 8.0

Brudne. C 23 H 26 NP4,2Hp. (Mr 430.5). 1013100. [357-57-3]. Butylboronic add. C 4 H¡¡B02' (M, 101.9). 1013700.
10,11-Dimethoxystrychnine. [4426-47-5].
Colourless crystals, slightly soluble in water, freely soluble in Content: mínimum 98 per cent.
ethanol (96 per cent). mp: 90 oC to 92 oc.
mp: about 178 oc.
tert-Butylhydroperoxide. C.¡H1002' (Mr 90.1). 1118000.
Butana!. C4H sO. (M, 72.1). 1134400. [123-72-8]. [75-91-2]. 1, 1- Dimethylethylhydroperoxide.
Butyraldehyde. Flammable liquid, soluble in organic solvents.
:
d;~ 0.806. d~g: 0.898.

n ~: 1.380.
nbo: 1.401.
bp: 35 oc.
bp: 75 oc.
Buty14-hydroxybenzoate. 1103900. [94-26-8].
Butane-l,4-diol. HO(CH2)40H. (M, 90.12). 1174800.
[110-63-4]. See Butyl parahydroxybenzoate R.
Butylhydroxytoluene. 1013800. [128- 37 -O].
ButanoL C 4HjQO. (M, 74.1).1013200. [71-36-3]. Butan-l-ol.
See Butylhydroxytoluene (0581).
CIear, colourless liquid, miscible with ethanol (96 per cent).
d~g: about 0.81. Butyl methacrylate. C S H¡P2' (M, 142.2). 1145400.
[97-88-1]. ButyI2-methylpropenoate.
bp: 116 oC to 119 oc.
Clear, colourless solution.
2-Buíanol Rl. C4 HjQO. (Mr 74.1). 1013301. [78-92-2]. d~o : about 0.894.
Butan-2-01. sec-Butyl alcohol. nbo : about 1.424.
Content: minimum 99.0 per cent. bp: about 163 oc.
Clear, colourless liquid, soluble in water, miscible with ethanol
tert-Butyl methyl ether. 1013900. [1634-04-4].
(96 per cent).
See 1,1-dimethylethyl methyl ether R.
d~g: about 0.81.
Distillation range (2.2.11). Not less than 95 per cent distils Butyl parahydroxybenzoate. 1103900. [94-26-8].
between 99 oC and 100 oc. See Butyl parahydroxybenzoate (0881).
Assay. Gas chromatography (2.2.28) as prescribed in the Butyric add. C 4 H sü 2. (Mr 88.1). 1014000. [107-92-6].
monograph Isopropyl alcohol (0970). Butanoic acid.
Butyl acetate. C 6 H 12 02' (Mr 116.2). 1013400. [123-86-4]. Content: minimum 99.0 per cent.
Clear, colourless liquid, flammable, slightly soluble in water, Oily liquid, miscible with water and with ethanol (96 per cent).
miscible with ethanol (96 per cent). d~g : about 0.96.
d~g : about 0.88. nbo : about 1.398.
nbD : about 1.395. bp: about 163 oc.
Distillation range (2.2.11). Not less than 95 per cent distils Butyrolactone. C4 H 6 0 2 • (Mr 86.1). 1104000. [96-48-0].
between 123 oC and 126 oc. Dihydro-2(3H)-furanone. y- Butyrolactone.
Oily liquid, miscible with water, soluble in methanol.
Butyl acetate Rl. 101340l.
nb5 : about 1.435.
Content: minimum 99.5 per cent, determined by gas
bp: about 204 oc.
chromatography.
Clear, colourless liquid, flammable, slightly soluble in Cadmium. Cd. (A r 112.4). 1014100. [7440-43-9].
water, miscible with ethanol (96 per cent). Silvery-white, lustrous metal, practically insoluble in water,
d~g : about 0.883. freely soluble in nitric acid and in hot hydrochloric acid.
nbo : about 1.395. Cadmium nitrate tetrahydrate. Cd(N0 3 )2,4Hp. (Mr 308.5).
Butanol: maximum 0.2 per cent, determined by gas 1174900. [10022-68-1].
chromatography. Hygroscopic orthorhombic crystals, very soluble in water,
n-Butylformate: maximum 0.1 per cent, determined by soluble in acetone and in ethanol (96 per cent).
gas chromatography. mp: about 59.5 oc.
n-Butyl propionate: maximum 0.1 per cent, determined by Caesium chloride. CsC!. (Mr 168.4). 1014200. [7647-17-8].
gas chromatography. White or almost white powder, very soluble in water, freely
Water: maximum 0.1 per cent. soluble in methanol, practically insoluble in acetone.
Butylamine. C 4H ll N. (Mr 73.1). 1013600. [109-73-9]. Caffeic add. C9HsÜ4' (Mr 180.2). 1014300. [331-39-5].
Butan -1-amine. (E)-3 -( 3,4-Díhydroxyphenyl)propenoic acid.
Distil and use within one month. White or almost white crystals OI plates, freely soluble in hot
Colourless liquid, miscible with water, with ethanol (96 per water and in ethanol (96 per cent), sparingly soluble in cold
cent). water.
mp: about 225 oC, with decomposition.
nbo: about 1.401.
Absorbance (2.2.25). A freshly prepared solution at pH 7.6
bp: about 78 oc.
shows 2 absorption maxima at 293 nm and 329 nm.
tert-Butylamine. 1100900. [75-64-9]. Caffeine. 1014400. [58-08-2].
See 1, 1-dimethylethylamine R. See Caffeine (0267).
Butylated hydroxytoluene. 1013800. [128-37-0]. Calcium carbonate. 1014500. [471-34-1].
See Butylhydroxytoluene R. See Calcium carbonate (0014).

440 See the information section on general monographs (caver pages)


EUROPEAN PHARMACOPOEIA 8.0 4.1.1. Reagents

Caldum carbonate Rl. 1014501. Test for sensitivity. Dissolve 50 mg of calconecarboxylic


Complies with the requirements prescribed for calcium acid triturate in a mixture of 2 mL of strong sodium
carbonate R with the following additional requirement. hydroxide solution R and 100 mL of water R. The solution
Chlorides (2.4.4): maximum 50 ppm. is blue but becomes violet on addition of 1 mL of a 10 giL
solution of magnesium sulfate R and 0.1 mL of a 1.5 giL
Cakium chloride. 1014600. [10035-04-8]. solution of calcium chloride R and turns pure blue on
See Calcium chloride (0015). addition of 0.15 mL of 0.01 M sodium edetate.

Caldum chloride solution. 1014601. Camphene. ClOH16' (Mr 136.2). 1139200. [79-92-5].
A 73.5 giL solutiol1. 2,2-Dimethyl-3-methylenebicyclo [2.2.1] heptane.
Camphene used in gas chromatography complies with the
Caldum chloride solution, 0.01 M. 1014602. following additional test.
Dissolve 0.147 g of calcium chloride R in water R and dilute Assay. Gas chromatography (2.2.28) as prescribed in the
to 100.0 mL with the same solvent. monograph Rosemary Oil (1846).
Calcium chloride solution, 0.02 M. 1014603. Content: minimum 90 per cent, calculated by the
Dissolve 2.94 g of calcium chloride R in 900 mL of water R, normalisation procedure.
adjust to pH 6.0 to 6.2 and dilute to 1000.0 mL with water R. Camphor. 1113000. [76-22-2].
5torage: at 2 oC to 8 oc.
See Camphor, racemic (0655).
Calcium chloride solution, 0.025 M. 1014604. Camphor used in gas chromatography complies with the
Dissolve 0.368 g of calcium chloride R in water R and dilute following addítional test.
to 100.0 mL with the same solvent. Assay. Gas chromatography (2.2.28) as prescribed in the
monograph Lavender oil (1338).
Calcium chloride Rl. CaC1 2,4Hp. (Mr 183.1). 1014700.
Calcium chloride tetrahydrate. Test solution. A 10 giL solution of the substance to be
examined in hexane R.
Iron: maximum 0.05 ppm.
Content: minimum 95.0 per cent, calculated by the
Cakium chloride, anhydrous. CaCl2. (Mr 111.0). 1014800. normalisation procedure.
[10043-52-4].
Content: minimum 98.0 per cent (dried substance). (15)-( + )-lO-Camphorsulfonk acid. ClQH1604S. (Mr 232.3).
1104100. [3144-16-9]. (l5AR)-( +)-2-0xo-1O-bornenesulfonic
White or almost white granules, deliquescent, very soluble in acid. [( 15)-7,7 -Dimethyl-2-oxobicyclo [2.2.1]heptan-l-
water, freely soluble in ethanol (96 per cent) and in methanol. yl]methanesulfonic acid. Reychler's acid.
Loss on drying (2.2.32): maximum 5.0 per cent, determined by
Prismatic crystals, hygroscopic, soluble in water.
drying in an oven at 200 oc.
Content: minimum 99.0 per cent of (5)-(+)-10-
5torage: in an airtight container, protected from moisture.
camphorsulfonic acid.
Calcium hydroxide. Ca(OH)2' (Mr 74.1). 1015000. [Clól~o: + 20 ± 1, determined on a 43 giL solution.
[l305-62-0]. Calcium dihydroxide. mp: about 194 oC, with decomposition.
White or almost white powder, almost completely soluble in ,l'l.A (2.2.41): 10.2 x 10 3 determined at 290.5 11m 011 a 1.0 giL
600 parts of water. solution.
Cakium hydroxide solution. 1015001.
Capric add. C 1o H 2aÜ2' (M, 172.3). 1142000. [334-48-5].
A freshly prepared saturated solution. Decanoic acid.
Cakium lactate. 1015100. [41372-22-9]. Crystalline solid, very slightly soluble in water, soluble in
See Calcium lacta te pentahydrate (0468). anhydrous ethanol.
bp: about 270 oc.
Caldum phosphate monobask monohydrate.
mp: about 31.4 oc.
CaH 4 0sP2,Hp. (Mr 252.1).1157200. [10031-30-8]. Calcium
tetrahydrogen bisphosphate monohydrate. Phosphoric acid Capric acid used in the assay of total fatty acids in Saw pa/metto
calcium salt (2:1) monohydrate. fruit (1848) complies with the following additional test.
White or almost white, crystalline powder, soluble in water. Assay Gas chromatography (2.2.28) as prescribed in the
monograph Saw palmetto fruit (1848).
Cakium sulfate. CaS04,1/2Hp. (Mr 145.1). 1015200.
Content: minimum 98 per cent, calculated by the
[10034-76-1]. Calcium sulfate hemihydrate.
normalisation procedure.
White or almost white powder, soluble in about 1500 parts of
water, practically insoluble in ethanol (96 per cent). When Capric alcohol. 1024700.
mixed with half its mass of water it rapidly solidifies to a hard See Decanol R.
and porous mass.
Caproic add. C6 H 12 02' (Mr 116.2). 1142100. [142-62-1J.
Caldum sulfate solution. 1015201. Hexanoic acid.
Shake 5 g of calcium sulfate R with 100 mL of water R for Oily liquid, sparingly soluble in water.
1 h and filter. d~o : about 0.926.
Cakonecarboxylic add. C21H14NP7S,3HzÜ· (Mr 492.5). n~o: about 1.417.
1015300. [3737-95-9]. 2-Hydroxy-l-(2-hydroxy-4-sulfo-1- bp: about 205 oc.
naphthylazo )naphthalene-3-carboxylic acid.
Caproic aeid used in the assay of total fatty acids in Saw
Brownish-black powder, slightly soluble in water, very slightly palmetto fruit (1848) complies with the following additional
soluble in acetone and in ethanol (96 per cent), sparingly test.
soluble in dilute solutions of sodium hydroxide.
Assay. Gas chromatography (2.2.28) as prescribed in the
Cakonecarboxylic acid triturate. 1015301. monograph Saw palmetto fruit (1848).
Mix 1 part of calconecarboxylic acid R with 99 parts of Content: minimum 98 per cel1t, calculated by the
sodium ehloride R. normalisatiol1 procedure.

General Notices (1) apply to all monographs and other texts 441
4.1.1. Reagents EUROPEAN PHARMACOPOEIA 8.0

E-Caprolactam. C6HllNO. (M r 113.2).1104200. [105-60-2). Do not use at a temperature higher than 400 oc.
Hexane-6-1actam.
Carbon for chromatography, graphitised Rl. 1153500.
Hygroscopic flakes, freely soluble in water, in anhydrous
ethanol and in methanol. Porous spherical carbon particles comprised of flat sheets of
hexagonally arranged carbon atoms.
mp: about 70 oc.
Particle size: 5 ¡..tm to 7 ¡..tm,
CapryHc acid. CSH¡602' (Mr 144.2). 1142200. [124-07-2). Pare volume: 0.7 cm 3 /g.
Octanoic acid.
Carbon monoxide. ca, (M, 28,01). 1016000. [630-08-0).
Slightly yellow, oily liquido
Content: minimum 99,97 per cent V/V,
d~o: about 0.910.
n6°: about 1.428. Carbon monoxide Rl. ca, (Mr 28,01), 1134600. [630-08-0).
bp: about 239.7 oc. Content: minimum 99 per cent V/V,
mp: about 16.7 oc. Carbon tetrachloride. CC14 , (Mr 153.8), 1016100. [56-23-5).
Caprylic acid used in the assay of total fatty acids in Saw Tetrachloromethane.
palmetto fruit (1848) complíes with the followíng additional Clear, colourless Iiquid, practically insoluble in water, miscible
test. with ethanol (96 per cent).
Assay. Gas chromatography (2.2.28) as prescribed in the d~g: 1.595 to 1.598.
monograph Saw palmetto fruít (1848).
bp: 76 oC to 77 oc.
Content: minimum 98 per cent, calculated by the
normalisation procedure. Carbophenothion. C ll H 16 Cl0 2 PS3' (Mr 342.9), 1016200,
[786-19-6), 0,0-Diethyl 5-[ [( 4-chlorophenyl)thio )methyl)-
Capsaicin. C¡SH27NOy (Mr 305.4). 1147900. [404-86-4). phosphorodithioate.
(E)-N-[(4-Hydroxy-3-methoxyphenyl)methyl)-8-methylnon-
Yellowish liquid, practically insoluble in water, miscible with
6-enamide.
organic solvents.
White or almost white, crystalline powder, practically
d~5 : about 1.27,
insoluble in water, freely soluble in anhydrous ethanol.
For the monograph Waal Fat (0134), a suitable certified
mp: about 65 oc.
reference solution (lO ng/¡..tL in iso-oetane) may be used,
Capsaícín used ín the assay in Capsicum (1859) complies with
the following additional test. Car-3-ene. ClOHl6' (M, 136,2). 1124000, [498-15-7).
Assay. Liquid chromatography (2.2.29) as prescribed in the 3,7,7 -Trimethylbicyclo[ 4,1,0)hept-3-ene. 4,7,7-Trimethyl-3-
monograph Capsicum (1859). norcarene.
Content: minimum 95.0 per cent, calculated by the Liquid with a pungent odour, slightly soluble in water, soluble
normalisation procedure. in organic solvents.
d~g: about 0,864,
Carbazole. C¡2H9N. (Mr 167.2). 1015400. [86-74-8).
Dibenzopyrrole.
n6°: 1.473 to 1.474,
[al~o: + 15 to + 17,
Crystals, practically insoluble in water, freely soluble in
acetone, slightly soluble in anhydrous ethanol. bp: 170 oC to 172 oc.
mp: about 245 oc. Car-3-ene used in gas chromatagraphy complies with the
following additianal test,
Carbomer. 1015500. [9007-20-9). Assay. Gas chromatography (2.2.28) as prescribed in the
A cross-linked polymer of acrylic acid; it contains a large monograph Nutmeg oil (1552).
proportion (56 per cent to 68 per cent) of carboxylic acid Content: minimum 95,0 per cent, calculated by the
(C0 2H) groups after drying at 80 oC for 1 h. Average relative normalisation procedure.
molecular mass about 3 x 10 6 •
pH (2.2.3): about 3 for a 10 giL suspension. Carminic add. C22 H 2 P13' (Mr 492.4), 1156700. [1260-17-9).
7-Q - D-Glucopyranosyl-3,5,6,8-tetrahydroxy -1-methyl-9, 10-
Carbon dioxide. 1015600. [124-38-9). dioxo-9,10-dihydroanthracene-2-carboxylic acid.
See Carbon dioxide (0375). Dark red powder, very slightly soluble in water, soluble in
dimethyl sulfoxide, very slightly soluble in ethanol (96 per
Carbon dioxide Rl. CO 2. (M, 44.01). 1015700. [124-38-9).
cent).
Content: minimum 99.995 per eent V/V.
Carbon monoxide: less than 5 ppm. Carob bean gum. 1104500,
Oxygen: less than 25 ppm. The ground endosperm of the fruit kernels of Ceratonia
siliqua L. Taub,
Nitric oxide: less than 1 ppm.
White or almost white powder containing 70 per cent to
Carbon dioxide R2. CO 2. (M, 44.01), 1134500, [124-38-9), 80 per cent of a water-soluble gum consisting mainly of
Content: minimum 99 per cent V/V, galactomannoglycone,

Carbon disulfide. CS 2 . (M, 76.1), 1015800, [75-15-0), Carvacrol. ClOH¡p. (Mr 150,2). 1016400, [499-75-2).
Colourless or yellowish, flammable liquid, practically insoluble 5-Isopropyl- 2-methylphenol.
in water, miscible with anhydrous ethanol. Brownish liquid, practically insoluble in water, very soluble in
d~g: about 1.26.
ethanol (96 per cent),
bp: 46 oC to 47 oc. d§g: about 0,975,
n6° : about 1.523.
Carbon for chromatography, graphitised. 1015900. bp: about 237 oc.
Carbon chains having a length greater than C 9 • Carvacrol used in gas chromatography complies with the
Particle size: 400 ¡..tm to 850 ¡..tm. fallowing additional test,
Relative densíty: 0,72, Assay. Gas chromatography (2.2,28) as prescribed in the
Surface area: 10 m 2/g, monograph Peppermint oil (0405),

442 See the informatian sectian on general monagraphs (cover pages)


EUROPEAN PHARMACOPOEIA 8.0 4.1.1. Reagents

Test salutian. Dissolve 0.1 g in about 10 mL of acetane R. CaryophyUene oxide. C¡SH 2p. (Mr 220.4).
Cantent: minimum 95.0 per cent, calculated by the 1149000. [1139-30-6]. (- )-~-Caryophyllene epoxide.
normalisation procedure. (lR,4R,6R, lOS) -4, 12, 12-Trimethyl-9-methylene-5-
oxatricyclo [8.2.0.04.6] dodecane.
Carveol. ClOH¡p. (Mr 152.2).1160400. [99-48-9]. p-Mentha- Colourless, fine crystals with lumps.
1(6),8-dien-2-01. 2-Methyl-5-( 1-methylethenyl)cyclohex-2-
mp: 62 oC to 63 oc.
erro!.
The substance contains a variable content of trans- and Caryaphyllene axide used in gas chramatagraphy complies with
cis-carveol. the fallawing additianal test.
Carveal used in gas chramatagraphy complies with the fallawing Assay. Gas chromatography (2.2.28) as prescribed in the
additiana/ test. monograph Turpentine ail, Pinus pinaster type (1627).
Assay. Gas chromatography (2.2.28) as prescribed in the Cantent: minimum 99.0 per cent, calculated by the
test for chromatographic profile in the monograph Caraway normalisation procedure.
ail (1817).
Casein. 1016600. [9000-71-9].
Cantent: minimum 97 per cent, calculated by the
normalisation procedure. Mixture of related phosphoproteins obtained from milk.
White or almost white, amorphous powder or granules, very
Carvone. C lO H¡40. (Mr 150.2). 1016500. [2244-16-8]. slightly soluble in water and in non-polar organic solvents. It
(+ )-p-Mentha-6,8-dien-2-one. (5S)-2-Methyl-5-(l- dissolves in concentrated hydrochloric acid giving a pale-violet
methylethenyl) -cyclohex -2-enone. solution. It forms salts with acids and bases. Its isoelectric
Liquid, practically insoluble in water, miscible with ethanol point is at about pH 4.7. Alkaline solutions are laevorotatory.
(96 per cent).
Castkin. C1 9 H¡SOs' (Mr 374.3). 1162200. [479-91-4].
d~g : about 0.965
5-Hydroxy-2- (3-hydroxy-4- methoxyphenyl)-3,6, 7-
n~o: about 1.500. trimethoxy-4H-1-benzopyran -4-one.
[al~o: about + 6l. Yellow crystals.
bp: about 230 oc.
Catalpol. C¡SHn0lO" (Mr 362.3). 1142300. [2415-24-9].
Carvane used in gas chramatagraphy camplies with the (1 aS, 1bS,2S,5aR,6S,6aS)-6- Hydroxy-l a -(hydroxymethyl)-
fallawing additianal test. 1a, 1b,2,5a,6,6a -hexahydrooxireno [4,5] cyclopenta [1,2-
Assay. Gas chromatography (2.2.28) as prescribed in the c]pyran-2-yl ~-D-glucopyranoside.
monograph Peppermint ail (0405) using the substance to be mp: 203 oC to 205 oc.
examined as the test solution.
Cantent: minimum 98.0 per cent, calculated by the Catechin. C1SH¡406,xH20. (Mr 290.3 for the anhydrous
normalisation procedure. substance). 1119000. [154-23-4]. (+)-(2R,3S)-2-(3,4-
Dihydroxyphenyl) -3,4-dihydro-2H-chromene-3,5,7 -trio!.
Carvone Rl. 1016501. Catechol. Cianidanol. Cyanidol.
Complies with the requirements prescribed for carvane R
with the following additional requirement. Cation-exchange resino 1016700.
Assay. Gas chromatography (2.2.28) as prescribed in the A resin in protonated form with sulfonic acid groups attached
test for chiral purity in the monograph Caraway ail (1817). to a polymer lattice consisting of polystyrene cross-Iinked with
8 per cent of divinylbenzene. It is available as beads and the
Cantent: minimum 98 per cent.
particle size is specified after the name of the reagent in the
(-)-Carvone. ClOH¡p. (Mr 150.2). 1160500. tests where it is used.
[6485-40-1]. (- )-p- Mentha-l (6),8-dien-2-one.
(5R)- 2-Methyl- 5-( 1-methylethenyl)cyclohex -2-enone.
Cation-exchange resin Rl. 1121900.

Liquid. A resin in protonated form with sulfonic acid groups attached


to a polymer lattice consisting of polystyrene cross-linked with
d§g: about 0.965. 4 per cent of divinylbenzene. It is available as beads and the
n~o : about 1.4988. particle size is specified after the name of the reagent in the
[al~o: about - 62. tests where it is used.
bp: about 230 oc. Cation-exchange resin, strong. 1156800.
Assay. Gas chromatography (2.2.28) as prescribed in the test Strong cation-exchange resin in protonated form with sulfonic
for chiral purity in the monograph Caraway ail (1817). acid groups attached to a polymer lattice consisting of
Cantent: minimum 99 per cent. polystyrene cross-linked with divinylbenzene. The particle
size is specified after the name of the reagent in the tests where
p-Caryophyllene. C¡SH 24 . (Mr 204.4). 1101000. it is used.
[87 -44-5]. (E)-(lR,9S)-4, 11,11-Trimethyl-8-methylene-
bicyclo[7.2.0]undec-4-ene. Cation-exchange resin (calcium form), strong. 1104600.
Oily liquid, practically insoluble in water, miscible with Resin in calcium form with sulfonic acid groups attached to
ethanol (96 per cent). a polymer lattice consisting of polystyrene cross-linked with
f3-Caryaphyllene used in gas chramatagraphy complies with 8 per cent of divinylbenzene. The particle size is specified after
the fallawing additianal test. the name of the reagent in the tests where it is used.
Assay. Gas chromatography (2.2.28) as prescribed in the Cation-exchange resin (sodium form), strong. 1176100.
monograph Clave ail (1091).
Resin in sodium form with sulfonic acid groups attached to
Test sa/utian. The substance to be examined. a polymer lattice consisting of polystyrene cross-linked with
Cantent: minimum 90.0 per cent, calculated by the divinylbenzene. The particle size is specified after the name of
normalisation procedure. the reagent in the tests where it is used.

General Natices (1) apply ta all managraphs and ather texts 443
4.1.1. Reagents EUROPEAN PHARMACOPOEIA 8.0

Cellulose for chromatography. 1016800. [9004-34-6]. Assay. Gas chromatography (2.2.28) as prescribed in the
Fine, white or almost white, homogeneous powder with an monograph Matricaria oil (1836).
average partide size less than 30 ¡.tm. Test solution: a 4 giL solution in cyclohexane R.
Preparation of a thin layer. Suspend 15 g in 100 mL of water R Content: minimum 95.0 per cent, calculated by the
ana homogenise in an electric mixer Íor 60 s. Coat carefully normalisation procedure.
deanea plates with a layer 0.1 mm thick using a spreading
device. Allow to dry in air. Charcoal, activated. 1017800. [64365-11-3].
See Activated charcoal (0313).
Cellulose for chromatography Rl. 1016900.
Microcrystalline cellulose. A fine, white or almost white Chloral hydrate. 1017900. [302-17-0].
homogeneous powder with an average particle size less than See Choral hydrate (0265).
30 11m.
Chloral hydrate solutkm. 1017901.
Preparatíon of a thín layer. Suspend 25 g in 90 mL of water R
and homogenise in an electric mixer for 60 s. Coat carefully A solution of 80 g in 20 mL of water R.
cleaned plates with a layer 0.1 mm thick using a spreading Chloramine. 1018000. [7080-50-4].
device. Allow to dry in airo See Tosylchloramide sodium (0381).
Cellulose for chromatography F254 • 1017000. Chloramine solution. 1018001.
Microcrystalline cellulose F1s4 . A fine, white or almost white, A 20 giL solution. Prepare immediately before use.
homogeneous powder with an average particle size less than
30 11m, containing a fluorescent indicator having an optimal Chloramine solution Rl. 1018002.
intensity at 254 nm. A 0.1 giL solution of chloramine R. Prepare immediately
Preparation of a thin layer. Suspend 25 g in 100 mL of before use.
water R and homogenise using an electric mixer for 60 s.
Coat carefully cleaned plates with a layer 0.1 mm thick using Chloramine solution R2. 1018003.
a spreading device. Allow to dry in air. A 0.2 giL solution. Prepare immediately before use.
Cerium sulfate. Ce(S04)1,4Hp. (Mr 404.3). 1017300. Chlordane. C lO H 6 Cls' 409.8). 1124100. [12789-03-6].
[10294-42-5]. Cerium(IV) sulfate tetrahydrate. Ceric sulfate. bp: about 175 oC
Yellow or orange-yellow, crystalline powder or crystals, very mp: about 106 oc.
slightly soluble in water, slowIy soluble in dilute acids. A suitable certifiea reference solution of technical grade
Cerous nitrate. Ce(N0 3)3,6Hp. (Mr 434.3). 1017400. (lO ng/l1L in iso-octane) may be used.
[10294-41-4]. Cerium trinitrate hexahydrate. Chlordiazepoxide. 1113200. [58-25-3].
Colourless or paje yellow, crystalline powder, freeIy soluble in
See Chlordíazepoxide (0656).
water and in ethanol (96 per cent).
Chlorfenvinphos. C12H14C1304P. 359.6). 1124200.
Cetostearyl alcohoL 1017500. [67762-27-0).
[470-90-6) .
See Cetostearyl alcohol (0702).
A suitable certified reference solution (lO ng/¡.tL in
Cetrimide. 1017600. [8044-71-1]. cyclohexane) may be used.
See Cetrimide (0378). Chloroacetanilide. CsHsCINO. (Mr 169.6). 1018100.
Cetyl alcohol. C 16 H 34o. (M, 242.4). 1160600. [36653-82-4]. [539-03-7]. 4' -Chloroacetanilide.
Hexadecan -1-01. Content: mínimum 95 per cent
Content: minimum 95.0 per cent Crystalline powder, practicany insoluble in water, soluble in
mp: about 48 oC ethanol (96 per cent).
mp: about 178 oc.
Cetylpyridinium chloride monohydrate. C1IH3SClN,HzÜ.
(M, 358.0). 1162800. [6004-24-6]. 1-Hexadecylpyridinium Chloroacetk add. C 2 H 3 CI0 2 • (M, 94.5). 1018200. [79-11-8].
chloride monohydrate. Colourless or white or almost white crystals, deliquescent,
White or almost white powder, freely soluble in water and in very soluble in water, soluble in ethanol (96 per cent).
ethanol (96 per cent). Storage: in an airtight container.
mp: 80 oC to 83 oC
Chloroaniline. C6 H 6 ClN. (Mr 127.6).1018300. [106-47-8].
Cetyitrimethylammonium bromide. C 19 H 42 BrN. 4-Chloroaniline.
(M, 3645). 1017700. [57-09-0]. Cetrimonium bromide. Crystals, soluble in hot water, freely soluble in ethanol (96 per
N- Hexadecyl-N,N,N- trimethylammonium bromide. cent).
White or almost white, crystalline powder, soluble in water, mp: about 71 oc.
freely soluble in ethanol (96 per cent).
mp: about 240 oC 4-Chlorobenzenesulfonamide. C 6H 6CIN0 2S. (M, 191.6).
1097400. [98-64-6].
Chamazulene. C¡,¡H w (M, 184.3). 1148000. [529-05-5]. White or almost white powder.
7 -Ethyl-1 ,4-dimethylazulene.
mp: about 145 oc.
Blue liquid, very slightly soluble in water, soluble in ethanol
(96 per cent), miscible with fatty oils, with essential oils and 2-Chlorobenzoic add. C7H sCI0 2 • (M, 156.6). 1139300.
with liquid paraffin, soluble with discolouration in phosphoric [118-91-2).
acid (85 per cent mlm) and sulfuric acid (50 per cent VIV). Soluble in water, slightly soluble in anhydrous ethanoL
Appearance of solution. 50 mg is soluble in 25 mL of hexane R. bp: about 285 oC
The blue solution is clear in a thin-Iayer obtained by tilting mp: about 140 oC
the test -tube.
Chamazulene used for gas chromatography complies with the Chlorobutanol. 1018400. [57-15-8].
followíng additional test. See Anhydrou5 chlorobutanol (0382).

444 See the information section on general monographs (caver pages)


EUROPEAN PHARMACOPOEIA 8,0 4.1. L Reagents

2-Chloro-2-deoxy-D-glucose. C 6 HIlClO s' (Mr 198,6), Content: minimum 99.8 per cent of CHC1 3, determined by gas
1134700, [14685-79-1], chromatography.
White or almost white crystalline, very hygroscopic powder, Chlorogenic add. C¡6H¡P9' (Mr 354.3). 1104700, [327-97-9].
soluble in water and in dimethyl sulfoxide, practically (lS,3R,4R,5R) -3 -[( 3,4-Dihydroxycinnamoyl)oxy]-1,4,5-
insoluble in ethanol (96 per cent), trihydroxycyclohexanecarboxylic acid.
2-Chloroethanol. C 2H sCIO. (Mr 80.5). 1097500. [107-07-3]. White 01' almost white, crystalline powder or needles, freely
Colourless liquid, soluble in ethanol (96 per cent). soluble in boiling water, in acetone and in ethanol (96 per
cent).
d~g: about 1.197.
[al~6: abont - 35.2.
n~o: about 1.442.
mp: about 208 oc.
bp: about 130 oc.
Ch ro m atography, Thin-Iayer chromatography (2.2.27) as
mp: about - 89 oc. prescribed on Identification A in the monograph Belladonna
leaf dry extract, standardised (1294); the chromatogram shows
2-Chloroethanol solution. 1097501.
only one principal zone.
Dissolve 125 mg of 2-chloroethanol R in 2-propanol R and
Chlorogenic acid used in liquid chramatography complies with
dilute to 50 mL with the same solvento Dilute 5 mL of the
the following additional test,
solution to 50 mL with 2-propanol R.
Assay, Liquid chromatography (2.2.29) as prescribed in the
Chloroethylarnine hydrochloride. C2H 7Cl 2N. (Mr 116,0). monograph Artichoke Leaf (1866).
1124300, [870-24-6]. 2-Chloroethanamine hydrochloride. Content: minimum 97.0 per cent.
mp: about 145 oc.
3-Chloro-2-methylaniline. C 7 H sCIN. (Mr 141.6). 1139400.
(2-Chloroethyl)diethylarnine hydrochloride. C 6H¡SC1 2 N. [87-60-5]. 6-Chloro-2-toluidine,
(Mr 172.1). 1018500, [869-24-9]. Not miscible with water, slightly soluble in anhydrous ethanol.
White or almost white, crystalline powder, very soluble in d~g : about 1.171.
water and in methanol, freely soluble in methylene chloride, n~o: about 1.587.
practically insoluble in hexane,
bp: about 115 oc.
mp: about 211 oc.
mp: abont 2 oc.
Chloroform. CHC1 3 • (Mr 11904). 1018600, [67-66-3]. 2-Chloro- N- (2,6-dimethylphenyl) acetamide. ClOH¡2CINO.
Trichloromethane. (Mr 197.7). 1168700. [1131-01-7].
Clear, colourless liquid, slightly soluble in water, miscible with
ethanol (96 per cent), 2-Chloroni.cotini.c acid. C 6 H 4 ClN0 2 • (Mr 157.6). 1157300,
[2942-59-8], 2-Chloropyridine-3-carboxylic acid,
d§g: 1.475 to 1.481.
White or almost white powder.
bp: about 60 oc.
mp: about 177 oc.
Ethanol: 004 per cent mlm to 1.0 per cent mlm.
Content: minimum 95 per cent.
Introduce 1.00 g (m g) into a ground-glass-stoppered f!ask.
Add 15.0 mL of nitrochromic reagent R, close the flask, shake 2-Chloro-4-nitroanHine. C 6 H sCIN 20 2 • (Mr 172.6). 1018800.
vigorously for 2 min and allow to stand for 15 mino Add [121-87-9].
100 mL of water R and 5 mL of a 200 giL solution of potassium Yellow, crystalline powder, freely soluble in methanol.
iodide R. After 2 min titrate with 0.1 M sodium thiosulfate, mp: about 107 oc.
using 1 mL of starch solution R as indicator, until a light
green colour is obtained (n¡ mL of 0.1 M sodium thiosulfate) , Starage: protected from light,
Carry out a blank assay (n 2 mL of 0.1 M sodium thiosulfate). 2-Chloro-5-nitrobenzoic add. C7H 4 CIN0 4 , (Mr 201.6).
Calculate the percentage of ethanol using the following 1183800. [2516-96-3].
expression: mp: 165 oC to 168 oc.
(n2 - nI) 0.115
Chlorophenol. C 6H 5CIO. (Mr 128,6). 1018900. [106-48-9].
m
4-Chlorophenol.
Chloroform, acidificd. 1018601. Colourless or almost colourless crystals, slightly soluble in
water, very soluble in ethanol (96 per cent) and in so!utions of
To 100 mL of chloroform R add 10 mL of hydrochloric
alkali hydroxides.
acid R. Shake, allow to stand and separate the 2 layers.
mp: about 42 oc.
Chloroform, ethanol-free. 1018602.
Chloroplatinic add. H 2Cl 6Pt,6HP, (Mr 517,9). 1019000.
Shake 200 mL of chloroform R with four quantities, each of [18497 -13-7]. Hydrogen hexachloroplatinate(IV) hexahydrate.
100 mL, of water R, Dry over 20 g of anhydraus sodium
Content: minimum 37.0 per cent mlm of platinum (A r 195.1).
sulfate R for 24 h. Distil the filtrate over 10 g of anhydrous
sodium sulfate R. Discard the first 20 mL of distillate. Brownish-red crystals or a crystalline mass, very soluble in
Prepare immediately before use. water, soluble in ethanol (96 per cent).
Assay. Ignite 0.200 g to constant mass at 900 ± 50 oC and
Chloroform stabilised with arnylene. CHCI 3 • (Mr 119.4). weigh the residue (platinnm).
1018700.
Storage: protected from light.
Clear, colourless liquid, slightly soluble in water, miscible with
ethanol (96 per cent), 3-Chloropropane-l,2-diol. CJH7CI02' (Mr 110.5), 1097600.
Water: maximum 0.05 per cent. [96-24-2].
Residue on evaporation: maximum 0.001 per cent. Colourless liquid, soluble in water and ethanol (96 per cent).
d~g : about 1.322.
Minimum transmittance (2.2.25) using water R as
compensation liquid: 50 per cent at 255 nm, 80 per cent at n~o: about 10480.
260 11m, 98 per cent at 300 nm, bp: about 213 oc.

General Natices (1) apply to all monographs and other texts 445
4.1.1. Reagents EUROPEAN PHARMACOPOEIA 8.0

5-Chloroquinolin-8-ol. C9H 6CINO. (Mr 179.6). 1156900. Chondroitinase Ae. 1163000.


[130-16-5]. 5-Chlorooxine. Pectin Iyase-like enzyme secreted by Flavobacterium
Sparingly soluble in coId dilute hydrochloric acid. heparinum. Available in vials containing 5-10 units. It cleaves
mp: about 123 oc. only glucuronate-containing disaccharides, e.g. chondroitin
Content: minimum 95.0 per cent. sulfate.

4-Chlororesordnol. C 6 H 5 Cl0 2 • (Me 144.6). Chromazurol S. C23H13CI2Na309S, (Me 605). 1019600.


1177700. [95-88-5]. 4-Chlorobenzene-1,3-diol. [1667-99-8].
1,3-Dihydroxy-4-chlorobenzene. Schultz No. 841.
mp: 106 oC to 108 oc. Colour Index No. 43825.
Trisodium 5- [( 3-carboxylato-5-methyl-4-oxocyclohexa -2,5-
5-Chlorosalicylic add. C 7H SCI03' (Me 172.6). 1019100. dien-l-ylidene )(2,6-dichloro-3-sulfonatophenyl)methyl]-2-
[321-14-2]. hydroxy-3-methylbenzoate.
White or almost white, crystalline powder, soluble in Brownish-black powder, soluble in water, slightly soluble in
methanol. ethanol (96 per cent).
mp: about 173 oc.
Chromic add deansing mixture. 1019700.
Chlorothiazide. C7H6ClNP4S2' (Mr 295.7). 1112100. A saturated solution of chromium trioxide R in sulfuric acid R.
[58-94-6]. 6-Chloro-2H-1,2,4-benzothiadiazine-7-
sulfonamide 1, l-dioxide. Chromic potassium sulfate. CrK(S04)2,12H 20. (Me 499.4).
Content: minimum 98.0 per cent. 1019800. [7788-99-0]. Chrome alum.
White or almost white, crystalline powder, very slightly Large, violet-red or black crystals, freely soluble in water,
soluble in water, sparingly soluble in acetone, slightly soluble practically insoluble in ethanol (96 per cent).
in ethanol (96 per cent). It dissolves in dilute solutions of
Chromium(HI) acetylacetonate. C 1s H 21 Cr0 6 . (Me 349.3).
alkali hydroxides.
1172900. [21679-31-2]. (OC-6-11)-Tris(2,4-pentanedionato-
ChlorotrimethylsHane. C 3H 9 CISi. (Me 108.6). 1019300. KO,KO')chromium.
[75-77-4].
Chromium(IH) trichloride hexahydrate.
CIear, colourless liquid, fuming in airo [Cr(H 20)¡CI 2]CI,2Hp. (M, 266.5). 1104800. [10060-12-5].
d§g: about 0.86. Dark green crystalline powder, hygroscopic.
nj'¡°: about 1.388. Storage: protected from humidity and oxidising agents.
bp: about 57 oc.
Chromium trioxide. Cr03' (M, 100.0).1019900. [1333-82-0].
Chlorpyriphos. C9 H ll C1 3N0 3PS. (M, 350.6). 1124400. Dark brownish-red needles or granules, deliquescent, very
[2921-88-2]. soluble in water.
bp: about 200 oc.
Storage: in an airtight glass container.
mp: 42 oC to 44 oc.
A suitable certified reference solution (lO ng/flL in Chromogenic substrate RL 1020000.
cyelohexane) may be used. Dissolve N-u -benzyloxycarbonyl-D-arginyl-L-
glycyl-L- arginine-4- nitroanilide dihydrochloride
Chlorpyriphos-methyl. C7H 7C1 3N0 3PS. (Mr 322.5). 1124500. in water R to give a 0.003 M solution. Dilute in
[5598-13-0]. tris(hydroxymethyl)aminomethane-EDTA buffer solution
mp: 45 oC to 47 oc. pH 8.4 R to 0.0005 M before use.
A suitable certified reference solution (lO ngl flL in
Chromogenic substrate R2. 1020100.
cyelohexane) may be used.
Dissolve D-phenylalanyl-L- pipecolyl-L-arginine-4-nitroanilide
Chlortetracydine hydrochloride. 1145500. dihydrochloride in water R to give a 0.003 M solution. Dilute
See Chlortetracycline hydrochloride (0173). before use in titrating in tris(hydroxymethyl)aminomethane-
EDTA buffer solution pH 8.4 R to give a 0.0005 M solution.
(5a)-Cholestane. C27 H 4S ' (M, 372.7).1167900. [481-21-0].
Slightly soluble in anhydrous ethanol. Chromogenic substrate R3. 1149100.
mp: about 81°C. Dissolve D-valyl-leucyl-lysyl-4-nitroanilide dihydrochloride in
water R to give a 0.003 M solution.
Cholesterol. 1019400. [57-88-5].
See Cholesterol (0993). Chromogenic substrate R4. 1163100.
Dissolve D-phenylalanyl-L-pipecolyl-L-arginine-4-nitroanilide
Choline chloride. CSH J4 ClNO. (Me 139.6). 1019500. dihydrochloride in water R to give a 0.008 M solution. Dilute
[67 c48-1]. (2-Hydroxyethyl)trimethylammonium chloride. to 0.0025 M with phosphate buffer solution pH 8.5 R before use.
Deliquescent crystals, very soluble in water and in ethanol
(96 per cent). Chromogenic substrate R5. 1163200.
Chromatography. Thin-layer chromatography (2.2.27) as Dissolve N- benzoyl-L-isoleucyl-L-glutamyl-glycyl-L-arginine-
prescribed in the monograph Suxamethonium chloride (0248): 4-nitroanilide hydrochloride in water R to give a 0.003 M
apply 5 flL of a 0.2 giL solution in methanol R; the solution.
chromatogram shows one principal spot. Chromotrope n B. C16H9N3Na201QS2' (Mr 513.4). 1020200.
Storage: in an airtight container. [548-80-1].
Chondroiti.nase ABe. 1162900. Schultz No. 67.
Pectin lyase-like enzyme secreted by Flavobacterium Colour Index No. 16575.
heparinum. Available in vials containing 5-10 units. It cleaves Disodium 4,5-dihydroxy-3-( 4-nitrophenylazo)naphthalene-
both glucuronate-containing disaccharides, e.g. chondroitin 2,7 -disulfonate.
sulfate, and iduronate-containing disaccharides, e.g. dermatan Reddish-brown powder, soluble in water giving a yellowish-red
sulfate. colour, practically insoluble in ethanol (96 per cent).

446 See the information section on general monographs (cover pages)


EUROPEAN PHARMACOPOEIA 8.0 4.1.1. Reagents

Chromotrope JI B solution. 1020201. Turpentine oil. Dissolve 1 g in 5 mL of ethanol (90 per


A 0.05 giL solution in sulfuric acid R. cent V/V) R. Add dropwise freshly prepared bromine water R.
Not more than 0.5 mL is required to give a yellow eolour
Chromotropic add, sodium salto CIOH6Na20SS2,2H20. lasting for 30 mino
(M r 400.3). 1020300. [5808-22-0].
Residue on evaporation: maximum 0.05 per cent.
Schultz No. 1136. To 10.0 mL add 25 mL of water R, evaporate on a water-bath
Disodium 4,5-dihydroxynaphthalene-2, 7 -disulfonate and dry the residue to constant mass at 100-105 oc.
dihydrate. Disodium 1,8-dihydroxynaphthalene-3,6-
disulfonate dihydrate. Cineole used in gas chromatography complies with the following
additional test.
A yellowish-white powder, soluble in water, practically
insoluble in ethanol (96 per cent). Assay. Gas chromatography (2.2.28) as prescribed in the
monograph Peppermint oil (0405).
Chromotropic add, sodium salí solution. 1020301. Test solution. The substance to be examined.
Dissolve 0.60 g of chromotropic acid, sodium salt R in Content: minimum 98.0 per eent, calculated by the
about 80 mL of water R and dilute to 100 mL with the same normalisation procedure.
solvento Use this solution within 24 h.
1,4-Cineole. ClOH¡p. (M, 154.3). 1142500. [470-67-7].
Chromotropic acid-sulfuric add solution. 1020302. 1- Methyl-4-( 1-methylethyl)-7 -oxabicyclo[2.2. l]heptane.
Dissolve 5 mg of chromotropic acid, sodium salt R in 10 mL 1-Isopropyl-4-methyl-7 -oxabicyelo[2.2. l]heptane.
of a mixture of 9 mL of sulfuric acid R and 4 mL of water R. Colourless liquido
Chrysanthemin. C 21 H 21 CIO Il . (Mr 485.8). 1134800. d~o: about 0.900.
[7084-24-4]. Cyanidin 3-0-glucoside chloride. nbo : about 1.445.
Kuromanin chloride. 2-(3,4- Dihydroxyphenyl)-3-(~- D- bp: about 173 oc.
glucopyranosyl)oxy-5, 7-dihydroxy-1-benzopyrylium chloride.
Reddish-brown crystalline powder, soluble in water and in Cinnamamide. C9H9NO. (Mr 147.2). 1154800. [621-79-4].
ethanol (96 per cent). (E)-3- Phenylprop-2-enamide.
Absorbance (2.2.25). A 0.01 giL solution in a mixture White or almost white powder.
of 1 volume of hydrochloric acid R and 999 volumes of mp: about 149 oc.
methanol R shows an absorption maximum at 528 nm.
trans-Cinnamic acid. C9 HP2' (M, 148.2). 1159200.
a-Chymotrypsin for peptide mapping. 1142400. [140-10-3]. trans-3-Phenylacrylic aeid. (2E)-3-Phenylprop-
a -Chymotrypsin of high purity, treated to eliminate tryptic 2-enoic aeid.
activity. Colourless crystals, very slightly soluble in water, freely soluble
in ethanol (96 per cent).
Cimifugin. C¡6H¡sÜ6' (M, 306.3). 1181700. [37921-38-3].
mp: 133 oc.
(25)-7 -(Hydroxymethyl)-2-(1-hydroxy-l-methylethyl)-4-
methoxy-2,3-dihydro- 5H-furo [3,2-g] [l]benzopyran -5-one. Cinnamic aldehyde. C9H BO. (M, 132.2). 1020700. [104-55-2].
3-Phenylpropenal.
Cinchonidine. C¡9H22NP. (MI 294.4). 1020400. [485-71-2].
(R) -( Quinol-4-yl) [(25,4S,5R) - 5-vinylquinuclidin -2- Yellowish 01' greenish-yellow, oily liquid, slightly soluble in
yl]methanol. water, very soluble in ethanol (96 per cent).
White or almost white, crystalline powder, very slightly nbo : about 1.620.
soluble in water and in light petroleum, soluble in ethanol Storage: protected from light.
(96 per cent).
trans-Cinnamic aldehyde. C 9HgÜ. (MI 132.2). 1124600.
[a];o: - 105 to - 110, determined on a 50 giL solution in [14371-10-9]. (E)-3-Phenylprop-2-enal.
ethanol (96 per cent) R.
trans-Cinnamic aldehyde used in gas chromatography complies
mp: about 208 oC, with decomposition. with the following additional test.
5torage: protected from Iight. Assay. Gas chromatography (2.2.28) as prescribed in the
Cinchonine. C¡9H22NP. (Mr 294.4). 1020500. [1l8-1O-5]. monograph Cassia oil (1496).
(S)- (Quinol-4-yl) [(2R,4S,5R)-5-vinylquinuclidin-2- Content: minimum 99.0 per cent, calculated by the
yl]methanol. normalisation procedure.
White or almost white, crystalline powder, very slightly Cinnamyl acetate. CllH1202' (M, 176.2). 1124700.
soluble in water, sparingly soluble in ethanol (96 per cent) [103-54-8]. 3-Phenylprop-2-en-1-yl acetate.
and in methanol.
nbo : about 1.542.
[a];o: + 225 to + 230, determined on a 50 giL solution in
bp: about 262 oc.
ethanol (96 per cent) R.
mp: about 263 oc. Cinnamyl acetate used in gas chromatography complies with
the following additional test.
Storage: protected from light.
Assay. Gas chromatography (2.2.28) as prescribed in the
Cineole. CIOH¡SO. (MI 154.3). 1020600. [470-82-6]. monograph Cassia oil (1496).
1,8-Cineole. Eucalyptol. 1,8-Epoxy-p-menthane. Content: minimum 99.0 per cent, calculated by the
Colourless liquid, practically insoluble in water, miscible with normalisation procedure.
anhydrous ethanol.
CUral. ClOH¡60. (MI 152.2). 1020800. [5392-40-5]. Mixture of
d~g: 0.922 to 0.927. (2E)- and (2Z)-3,7 -Dimethylocta-2,6-dienal.
nbo: 1.456 to 1.459. Light yellow liquid, practically insoluble in water, miscible
°
Freezing point (2.2.18): oC to 1 oc. with ethanol (96 per cent) and with propylene glycol.
Distillation range (2.2.11): 174 oC to 177 oc. Chromatography. Thin-layer chromatography (2.2.27), using
Phenol. Shake 1 g with 20 mL of water R. Allow to separate si/ica gel GF254 R as the coating substance: apply to the plate
and add to 10 mL of the aqueous layer 0.1 mL of ferric chloride 10 I1L of a 1 giL solution in toluene R. Develop over a path of
solution R1. No violet colour develops. 15 cm using a mixture of 15 volumes of ethyl acetate R and

General Notices (1) apply to all monographs and other texts 447
4.1. L Reagents EUROPEAN PHARMACOPOEIA 8.0

85 volumes of toluene R. Allow the plate to dry in air and Citropten. C¡lHlQ04' (M, 206.2). 1021300. [487-06-9].
examine in ultraviolet light at 254 nm. The chromatogram Limettin. 5,7-Dimethoxy-2H-1-benzopyran-2-one.
shows only one principal spot. Needle-shaped crystals, practically insoluble in water and
Citral used in gas chromatography complies with the fo/lowing in light petroleum, freely soluble in acetone and in ethanol
additional test. (96 per cent).
Assay. Gas chromatography (2.2.28) as prescribed in the mp: about 145 oc.
monograph Citrone/la oil (1609). Chromatography. Thín-Iayer chromatography (2.2.27), using
Content of citral (neral + geranial) : minimum 95.0 per cent, si/ica gel GF254 R as the coating substance: apply to the plate
calculated by the normalisation procedure. 10 [1L of a 1 giL solution in toluene R. Develop over a path of
15 cm using a mixture of 15 volumes of ethyl acetate R and
Citrated rabbit plasma. 1020900. 85 volumes of toluene R. Allow the plate to dry in air and
Collect blood by intracardiac puncture from a rabbit kept examine in ultraviolet light at 254 nm. The chromatogram
fasting for 12 h, using a plastic syringe with a No. 1 needle obtained shows only one principal spot.
containing a suitable volume of 38 giL solution of sodium
citrate R so that the final volume ratio of citrate solution to Clobetasol propionate. C2sH32ClFOs' (Mr 467.0). 1097700.
blood is 1: 9. Separate the plasma by centrifugation at 1500 g [25122-46-7J. 21-Chloro-9-fiuoro-I1 ~,l7 -dihydroxy-16~­
to 1800 g at 15 oC to 20 oC for 30 mino methylpregna-1,4-diene-3,20-dione 17 -propionate.
Storage: at O oC to 6 oC; use within 4 h of collection. White or almost white crystalline powder, insoluble in water,
soluble in ethanol (96 per cent) and in acetone.
Citric add. 1021000. [5949-29-1]. [aJ~o: about + 104 (in dioxan).
See Citric acid monohydrate (0456). mp: about 196 oc.
When used in the test for iron, it complies with the following
additional requirement. Coagulation factor V solution. 1021400.
Dissolve 0.5 g in 10 mL of water R, add 0.1 mL of thioglycollic Coagulation factor V solution may be prepared by the
acid R, mix and make alkaline with ammonia R. Dilute to following method or by any other method which excludes
20 mL with water R. No pink colour appears in the solution. factor VIII.
Citric add, anhydrous. 1021200. [77-92-9J. Prepare the factor V reagent from fresh oxalated bovine
plasma, by fractionation at 4 oC with a saturated solution of
See Anhydrous citric acid (0455). ammonium sulfate R prepared at 4 oc. Separate the fraction
CitroneUal. ClQH¡p. (Mr 154.3). 1113300. [106-23-0]. which precipitates between 38 per cent and 50 per cent
3,7 -Dimethyl-6-octenal. of saturation, which contains factor V without significant
Very slightly soluble in water, soluble in ethanol (96 per cent). contamination with factor VIII. Remove the ammonium
sulfate by dialysis and dilute the solution with a 9 giL solution
d§g: 0.848 to 0.856. of sodium chloride R to give a solution containing between
n~o : about 1.446. 10 per cent and 20 per cent of the quantity offactor V present
Citrone/lal used in gas chromatography complies with the in fresh human normal plasma.
following additional test. Assay offactor V. Prepare two dilutions of the preparation
Assay. Gas chromatography (2.2.28) as prescribed in the of factor V in imidazole buffer solution pH 7.3 R containing
monograph Citronella oil (1609). 1 volume of the preparation in 10 volumes and in 20 volumes
Content: minimum 95.0 per cent, calculated by the of the buffer solution respectively. Test each dilution as
normalisation procedure. follows: mix 0.1 mL of plasma substrate deficient in factor
V R, 0.1 mL of the solution to be examined, 0.1 mL of
Citronellol. C lO H 2p. (M, 156.3). 1134900. [106-22-9]. thromboplastin R and 0.1 mL of a 3.5 giL solution of calcium
3,7 -Dimethyloct-6-en-l-ol. chloride R and measure the coagulation times, i.e. the interval
Clear, colourless liquid, practically insoluble in water, miscible between the moment at which the calcium chloride solution is
with ethanol (96 per cent). added and the first indication of the formation of fibrin, which
d;g: 0.857. may be observed visually or by means of a suitable apparatus.
In the same manner, determine the coagulation time (in
n ~ : 1.456.
duplicate) of four dilutions of human normal plasma in
bp: 220 oC to 222 oc. imidazole buffer solution pH 7.3 R, containing respectively,
Citrone/lol used in gas chromatography complies with the 1 volume in 10 (equivalent to 100 per cent offactor V),
fo/lowing additional test. 1 volume in 50 (20 per cent), 1 volume in 100 (10 per cent),
Assay. Gas chromatography (2.2.28) as prescribed in the and 1 volume in 1000 (l per cent). Using two-way logarithmic
monograph Citrone/la oil (1609). paper plot the average coagulation times for each dilution of
Content: minimum 95.0 per cent, calculated by the human plasma against the equivalent percentage of factor V
normalisation procedure. and read the percentage of factor V for the two dilutions of the
factor V solution by interpolation. The mean of the two results
Storage: in an airtight container, protected from light.
gives the percentage of factor V in the solution to be examined.
Citronellyl acetate. C¡2H2zÜZ' (Mr 198.3). 1135000. Storage: in the frozen state at a temperature not higher than
[150-84-5]. 3,7-Dimethyl-6-octen-1-yl acetate. - 20 oc.
:
d;~ 0.890.
Cobalt chIoride. CoC1 2,6Hp. (Me 237.9). 1021600.
n ~: l.443. [7791-13-1].
bp: 229 oc. Red, crystalline powder or deep-red crystals, very soluble in
Citronellyl acetate used in gas chromatography complies with water, soluble in ethanol (96 per cent).
the following additional test.
Cobalt nitrate. Co(N0 3 )2,6Hp. (Mr 291.0). 1021700.
Assay. Gas chromatography (2.2.28) as prescribed in the [10026-22-9].
monograph Citrone/la oil (1609).
Small garnet-red crystals, very soluble in water.
Content: minimum 97.0 per cent, calculated by the
normalisation procedure. Codeine. 1021800. [6059-47-8].
Storage: in an airtight container, protected from light. See Codeine (0076).

448 See the information section on general monographs (cover pages)


EUROPEAN PHARMACOPOEIA 8.0 4.1.1. Reagents

Codeine phosphate. 1021900. [52-28-8]. the temperature below 20 oC, add dropwise with continuous
See Codeine phosphate hemihydrate (0074). shaking 30 mL of strong sodium hydroxide solution R.
Filter through a sintered-glass filter (40) (2.1.2), wash with
Congo red. C32H22N6Nap6S2' (Mr 697). 1022000. [573-58-0]. water R until the filtrate is clear and take up the precipitate
Schultz No. 360. with 200 mL of concentrated ammonia R. Filter through a
Colour Index No. 22120. sintered-glass fiIter (2.1.2) and repeat the filtration to reduce
Disodium (biphenyl-4,4' -diyl-bis-2,2' -azo )bis( 1-amino- the residue to a minimum.
nap hthalene-4-sulfonate).
Cortisone. C21 H 2SOs' (Mr 360.4). 1175000. [53-06-5].
Brownish-red powder, soluble in water.
Content: minimum 95.0 per cent.
Congo red papel'. 1022002. mp: 223-228 oc.
Immerse strip s of filter paper for a few minutes in congo
red solution R. Allow to dry. Cortisone acetate. 1097800. [50-04-4].
See Cortisone acetate (0321).
Congo red solution. 1022001.
Dissolve 0.1 g of congo red R in a mixture of 20 mL of Coumaphos. CI4HI6CIOsPS. (M, 362.8). 1124800. [56-72-4].
ethanol (96 per cent) R and water R and dilute to 100 mL mp: 91°C to 92 oc.
with water R. A suitable certified reference solution (lO ng/flL in iso-octane)
Test for sensitivity. To 0.2 mL of the congo red solution add may be used.
100 mL of carbon dioxide-free water R and 0.3 mL of 0.1 M
hydrochloric acid. The solution is blue. Not more than o-CoumarÍC acid. C9HP3" (Mr 164.2). 1157400. [614-60-8].
0.3 mL of 0.1 M sodium hydroxide is required to change (E)-2-Hydroxycinnamic acid. (2E)-3-(2-Hydroxyphenyl)prop-
the colour to pink. 2-enoic acid.
Colour change: pH 3.0 (blue) to pH 5.0 (pink). White or almost white powder.
mp: about 217 oc.
Coomassie bIue. 1001400. [3861-73-2].
See acid blue 92 R. p-Coumaric acid. C9HPJ' (M, 164.2). 1157500. [7400-08-0].
4-Hydroxycinnamic acid. 3-(4-Hydroxyphenyl)-prop-2-enoic
Coomassie bIue solution. 1001401. acid.
See acid blue 92 solution R. White or almost white needles, practically insoluble in water,
Coomassie staining solution. 1012201. soluble in acetone and in methanol.
A 1.25 giL solution of acid blue 83 R in a mixture consisting of mp: 214 oC to 217 oc.
1 volume of glacial acetic acid R, 4 volumes of methanol R and p-Coumaric acid used in the assay in Nettle leaf (1897) complies
5 volumes of water R. Filter. with the following additional tests.
Coomassie staining solution Rl. 1173000. Loss on drying (2.2.32): maximum 5.0 per cent, determined
Dissolve 0.275 g of acid blue 83 R in 200 mL of methanol R. on 0.200 g by drying in an oven at 105 oC for 2 h.
Stir until complete dissolution of the crystals (for about 2 h). Assay. Liquid chromatography (2.2.29) as prescribed in the
Add 750 mL of water R and 50 mL of glacial acetic acid R. Stir monograph Nettle leaf (1897).
overnight (for at least 16 h); frIter. Content: minimum 95 per cent, calculated by the
normalisation procedure.
Copper. Cu. (A r 63.55). 1022100. [7440-50-8].
Cleaned foil, turnings, wire or powder of the pure metal of Coumarin. C9H602' (Me 146.1). 1124900. [91-64-5].
electrolytic grade. 2H-Chromen-2-one. 2H-1- Benzopyran-2-one.
Copper acetate. C4H6CU04,Hp. (Mr 199.7). 1022200. Colourless, crystalline powder or orthorhombic or rectangular
[142-71-2]. crystals, very soluble in boiling water, soluble in ethanol
(96 per cent). It dissolves in solutions of alkali hydroxides.
Blue-green crystals or powder, freely soluble in boiling water,
soluble in water and in ethanol (96 per cent), slightly soluble mp: 68 oC to 70 oc.
in glycerol (85 per cent). Coumarin used in gas chromatography complies with the
following additional test.
Copper edetate solution. 1022300.
Assay. Gas chromatography (2.2.28) as prescribed in the
To 2 mL of a 20 giL solution of copper acetate R add 2 mL of monograph Cassia oi! (1496).
0.1 M sodium edetate and dilute to 50 mL with water R.
Content: minimum 98.0 per cent, calculated by the
Copper nitrate. CU(NO J)2,3Hp. (M, 24l.6). 1022400. normalisation procedure.
[10031-43-3]. Chloride dinitrate trihydrate.
Dark blue crystals, hygroscopic, very soluble in water giving a Cresol. C 7 Hp. (Me 108.1). 1022700. [95-48-7]. o-Cresol.
strongly acid reaction, freely soluble in ethanol (96 per cent) 2-Methylphenol.
and in dilute nitric acid. Crystals or a super-cooled ¡¡quid becoming dark on exposure
Storage: in an airtight container. to light and air, miscible with anhydrous ethanol, soluble
in about 50 parts of water and soluble in solutions of alkali
Copper sulfate. CuS0 4,5Hp. (Mr 249.7). 1022500. hydroxides.
[7758-99-8]. d~g: about 1.05.
Blue powder or deep-blue crystals, slowly efflorescent, very n~o: 1.540 to 1.550.
soluble in water, slightly soluble in ethanol (96 per cent).
bp: about 190 oc.
Copper sulfate solution. 1022501. Freezing point (2.2.18): minimum 30.5 oc.
A 125 giL solution. Residue on evaporation: maximum 0.1 per cent m/m,
Copper tetrammine, ammoniacal solution of. 1022600. determined by evaporating on a water-bath and drying in an
oven at 100-105 oc.
Dissolve 34.5 g of copper sulfate R in 100 mL of water R and,
whilst stirring, add dropwise concentrated ammonia R until Storage: protected from light, moisture and oxygen.
the precipitate which forms dissolves completely. Keeping Distil before use.

General Notices (1) apply to all monographs and other texts 449
4.1.1. Reagents EUROPEAN PHARMACOPOEIA 8.0

m-Creso!. 1177100. [108-39-4]. a) To 25.0 mL add 3 g of potassium iodide R. Add 25 mL of a


See metacresol (2077). 25 per cent m/m solution of sulfurie acid R with precaution
and in small quantities. Titrate with 0.1 M sodium thiosulfate
p-Cresol. C 7H sO. (Mr 108.1). 1153100. [106-44-5]. using 0.5 mL of starch solution R, added towards the end of
4-Methylphenol. the titration, as indicator.
Colourless or white OI almost white crystals or crystalline 24.5 mL to 25.5 mL of 0.1 M sodium thiosulfate is used in
mass. the titration.
d~g : about 1.02. b) Dilute 10.0 mL to 100.0 mL with water R and mix. To
bp: about 202 oc. 10.0 mL ofthe solution, add 25.0 mL ofO.1 M hydrochloric acid
and heat for 1 h on a water-bath. Cool, adjust with water R to
m-Cresol purpk C2JH¡sÜsS, (Mr 382.44). 1121700. the initial volume and titrate with 0.1 M sodium hydroxide,
[2303-01-7]. m-Cresolsulfonphthalein. using 0.1 mL of phenolphthalein solution R1 as indicator.
Olive-green, crystalline powder, slightly soluble in water,
5.7 mL to 6.3 mL of 0.1 M sodium hydroxide is used in the
soluble in ethanol (96 per cent), in glacial acetic acid and in
titration.
methanol.
c) Dilute 10.0 mL to 100.0 mL with water R and mix. Titrate
m-Cresol purple solution. 1121701. 10.0 mL of the solution with 0.1 M hydrochloric acid, using
Dissolve 0.1 g of m-cresal purple R in 13 mL of 0.01 M 0.1 mL of phenolphthalein solution R1 as indicator.
sodium hydroxide, dilute to 100 mL with water R and mix. 6.0 mL to 7.5 mL of 0.1 M hydrochloríc acid is used in the
Colour change: pH l.2 (red) to pH 2.8 (yellow); pH 7.4 titration.
(yellow) to pH 9.0 (purple).
Cupriethylenediamine hydroxide solution. 3008700.
Cresol red. C21H180sS. (M, 382.4). 1022800. [1733-12-6]. [14552-35-3].
Cresolsulfonphthalein. 4,4' -(3H-2,1- Benzoxathiol-3- The molar ratio of ethylenediamine to copper is 2.00 ± 0.04.
ylidene)bis-(2-methylphenol) S,S-dioxide. This solution i5 commercially available.
A reddish-brown crystalline powder, slightly soluble in water,
soluble in ethanol (96 per cent) and in dilute solutions of Cupri-tartaric soludon. 1023300.
alkali hydroxides. Solution A. Dissolve 34.6 g of copper sulfate R in water R and
dilute to 500 mL with the same solvent.
Cresol red §olution. 1022801.
Solution B. Dissolve 173 g of sodium potassium tartrate R
Dissolve 0.1 g of cresol red R in a mixture of 2.65 mL of and 50 g of sodium hydroxide R in 400 mL of water R. Heat
0.1 M sodium hydraxide and 20 mL of ethanol (96 per
to boiling, allow to cool and dilute to 500 mL with earbon
cent) R and dilute to 100 mL with water R.
dioxide-free water R.
Test for sensitivity. A mixture of 0.1 mL of the cresol
Mix equal volumes of the 2 solutions immediately before use.
red solution and 100 mL of carbon dioxide-free water R
to which 0.15 mL of 0.02 M sodium hydroxide has been Cupri-tartar:ic solution R2. 1023302.
added is purple-red. Not more than 0.15 mL of 0.02 M Add 1 mL of a solution containing 5 giL of copper sulfate R and
hydrochloric acid is required to change the colour to yellow. 10 giL of potassium tartrate R to 50 mL of sodium carbonate
Colour ehange: pH 7.0 (yellow) to pH 8.6 (red). so/ution R1. Prepare immediately before use.
Crystal violeto CZSHJoClNJ' (Me 408.0). 1022900. [548-62-9]. Cupri-tartaric solutl.on R3. 1023303.
Schultz No. 78. Prepare a solution containing 10 giL of copper sulfate R and
Colour Index No. 42555. 20 giL of sadium tartrate R. To 1.0 mL of the solution add
Hexamethyl-pararosanilinium chloride. 50 mL of sodium carbonate solution R2. Prepare immediately
Dark-green powder or crystals, soluble in water and in ethanol before use.
(96 per cent). Cupri-tartaric solutlon R4. 1023304.
Crystal violet solution. 1022901. Solution A. 150 giL copper sulfate .R.
Dissolve 0.5 g of erystal violet R in anhydrous acetic acid R Solution B. Dissolve 2.5 g of anhydrous sodium carbonate R,
and dilute to 100 mL with the same solvent. 2.5 g of sodium potassium tartrate R, 2.0 g of sodium hydrogen
Test for sensitivity. To 50 mL of anhydrous aeetie acid R carbonate R, and 20.0 g of anhydrous sodium sulfate R in
add 0.1 mL of the crystal violet solution. On addition of water R and dilute to 100 mL with the same solvent.
0.1 mL of 0.1 M perchloric acid the bluish-purple solution Mix 1 part of solution A with 25 parts of solution B
turns bluish-green. immediately before use.
Cupric chloride. CuC1 2 ,2Hp. (Mr 170.5). 1023000. Curcumin. C21 H 2aÜ6' (Mr 368.4). 1023500. [458-37-7]. 1,7-
[10125-13-0]. Cupric chloride dihydrate. Bis( 4-hydroxy- 3-methoxyphenyl)hepta-l ,6-diene-3,5-dione.
Greenish-blue powder or crystals, deliquescent in moist air, Orange-brown, crystalline powder, practically insoluble in
efflorescent in dry air, freely soluble in water, in ethanol water, soluble in glacial acetic acid.
(96 per cent) and in methanol, sparingly soluble in acetone. mp: about 183 oc.
Storage: in an airtight container.
Curcuminoids. 1183900.
Cupri-citric solution. 1023100. A mixture of curcumin (C 21 H 20 0 6 ; Me 368.4),
Dissolve 25 g of copper sulfate R, 50 g of citrie acid R and 144 g demethoxycurcumin (C 20H¡SOS; M r 338.4) and
of anhydrous sodium carbonate R in water R and dilute to bis-demethoxycurcumin (C¡9H¡604; M r 308.3).
1000 mL with the same solvent.
Cyanoaceti.c add. C 3H 3 N0 2 • (Mr 85.1). 1097900. [372-09-8].
Cupri-dtric solution Rl. 1023200. White or yellowish-white, hygroscopic crystals, very soluble in
Dissolve 25 g of copper sulfate R, 50 g of eitrie aeid R and 144 g water.
of anhydrous sadium carbonate R in water R and dilute to Storage: in an airtight container.
1000 mL with the same solvent.
Adjust the solution so that it complies with the following Cyanocobalamin. 1023600. [68-19-9].
requirements. See Cyanocobalamin (0547).

450 See the information section on general monographs (cover pages)


EUROPEAN PHARMACOPOEIA 8.0 4.1.1. Reagents

Cyanogen bromide solution. 1023700. [506-68-3]. d§g: about 0.998.


Add dropwise, with cooling 0.1 M ammonium thiocyanate to n5°: about 1.4648.
bromine water R until the yellow colour disappears. Prepare bp: about 130 oc.
immediately before use.
Cyhalothrin. C23H¡9ClF3N03" (Mr 449.9). 1125000.
Cyanoguanidine. C 2H 4N 4. (Mr 84.1). 1023800. [461-58-5]. [91465-08-6].
Dicyandiamide. l-Cyanoguanidine.
bp: 187 oC to 190 oc.
White or almost white, crystalline powder, sparingly soluble
mp: about 49 oc.
in water and in ethanol (96 per cent), practically insoluble
in methylene chloride. A suitable certified reference solution (10 ng/ fiL in
mp: about 210 oc. cyclohexane) may be used.

u-Cydodextrin. C 36 H 6aÜ30. (Mr 972). 1176200. p-Cymene. C lO H)4" (Mr 134.2). 1113400. [99-87-6].
[10016-20-3]. Cyclohexakis-(1-+4)-( u- D-glucopyranosyl). 1-Isopropyl-4-methylbenzene.
Cyclomaltohexaose. Alfadex. Colourless liquid, practically insoluble in water, soluble in
ethanol (96 per cent).
~-Cydodextrin. 1184000. [7585-39-9].
d§g: about 0.858.
See Betadex (1070).
n5° : about 1.4895.
~-Cydodextrin for chiral chromatography, modifled. bp: 175 oC to 178 oc.
1154600. p-Cymene used in gas chromatagraphy complies with the
30 per cent of 2,3-di-O-ethyl-6-0-tert-butyldimethyl- following additional test.
silyl-~-cyclodextrin dissolved in poly(dimethyl)(85)(diphen-
Assay. Gas chromatography (2.2.28) as prescribed in the
yl)(15)siloxane R. monograph Peppermint oil (0405).
~-Cydodextrin for chiral chromatography, modifled Rl. Test solution. The substance to be examined.
1160700. Content: minimum 96.0 per cent, calculated by the
30 per cent of 2,3-di-O-acetyl-6-0-tert-butylsilyl-~-cyclo­ normalisation procedure.
dextrin dissolved in poly(dimethyl)(85)( diphenyl)(15 )silox-
ane R. Cynarin. C2sH24012. (Mr 516.4). 1159300. [30964-13-7].
(la,3a,4((,5~)-1,3- Bis[ [3-(3,4-Dihydroxyphenyl)-1-oxo-2-
Cydohexane. C6H¡2. (Mr 84.2). 1023900. [110-82-7]. propenyl] oxy]-4,5-dihydroxycyclohexanecarboxylic acid.
Clear, colourless, flammable liquid, practically insoluble in White or almost white amorphous mass, odourless.
water, miscible with organic solvents.
d§g: about 0.78. Cypermethrin. C22H¡9ClzN03' (Mr 416.3). 1125100.
[52315-07-8].
bp: about 80.5 oc.
bp: 170 oC to 195 oc.
Cyclohexane used in spectrophotometry complies with the
mp: 60 oC to 80 oc.
following additional test.
Minimum transmittance (2.2.25) using water R as A suitable certified reference solution (10 ng/fiL in
compensation liquid: 45 per cent at 220 nm, 70 per cent at cyclohexane) may be used.
235 nm, 90 per cent at 240 nm, 98 per cent at 250 nm. L-Cysteine. C3H 7N0 2 S. (Mr 121.1). 1024200. [52-90-4].
Cyelohexane Rl. 1023901. Powder, freely soluble in water, in ethanol (96 per cent) and in
Complies with the requirements prescribed for acetic acid, practically insoluble in acetone.
cyclohexane R with the following additional requirement. Cysteine hydrochloride. 1024300. [7048-04-6].
The fluorescence, measured at 460 nm, under illumination See Cysteine hydrachloride monohydrate (0895).
with an excitant light beam at 365 nm, is not more intense
than that of a solution containing 0.002 ppm of quinine R L-Cystine. C6H¡2NP4S2. (Mr 240.3). 1024400. [56-89-3].
in 0.05 M sulfuric acid. White or almost white, crystalline powder, practically
Cydohexylamine. C 6HlJN. (Mr 99.2).1024000. [108-91-8]. insoluble in water and in ethanol (96 per cent). It dissolves in
Cyclohexanamine. dilute solutions of alkali hydroxides.
Colourless liquid, soluble in water, miscible with usual organic [Ql~o: - 218 to - 224, determined in 1 M hydrochloric acid.
solvents. mp: 250 oC, with decomposition.
n5° : about 1.460. Cytosine. C4 H sNp. (Mr 111.1).1160800. [71-30-7].
bp: 134 oC to 135 oc.
Content: minimum 95.0 per cent.
Cydohexylenedinitrilotetra-acetic add. C¡4H22N20S,H20. Daidzein. C¡SH1004. (M¡ 254.2). 1178400. [486-66-8].
(Mr 364.4). 1024100. trans-Cyclohexylene-1,2-dinitrilo-
7 -Hydroxy-3-( 4-hydroxyphenyl) -4H-l-benzopyran-4-one.
N,N,N;N'-tetra-acetic acid.
White or almost white, crystalline powder. Dairlzin. C 21 H 2aÜ9' (Mr 416.4). 1178300. [552-66-9].
mp: about 204 oc. Daidzein -7 -O-glucoside. 7-(~- D-Glucopyranosyloxy)-3-( 4-
hydroxyphenyl)-4H-1-benzopyran-4-one.
Cydohexylmethanol. C7 H¡p. (Mr 114.2). 1135200.
[100-49-2]. Cyclohexylcarbinol. Dantron. C¡4HS04. (Mr 240.2). 1024500. [117-10-2].
1,8-Dihydroxyanthraquinone. 1,8-Dihydroxyanthracene-9,1 0-
Liquid with a slight odour of camphor, soluble in ethanol
dione.
(96 per cent).
Crystalline orange powder, practically insoluble in water,
ni;': about 1.464. slightly soluble in ethanol (96 per cent), soluble in solutiol1s of
bp: about 185 oc. alkali hydroxides.
3-Cydohexylpropionic add. C9H¡602. (Mr 156.2). 1119200. mp: about 195 oc.
[701-97-3]. Dantron used in the sesquiterpenic acids assay in Valerian
Clear liquido root (0453) complies with the fallowing additional tests.

General Natices (1) apply to all monographs and other texts 451
4. L 1. Reagents EUROPEAN PHARMACOPOEIA 8.0

Ai~m: 355 to 375, determined at 500 nm in 1 M potassium Demedocydine hydrochloride. 1145600.


hydroxide. See Demeclocycline hydrochloride (0176).
Assay. Liquid chromatography (2.2.29) as prescribed in the
monograph Valerian Root (0453) at the concentration of the DemethyIflumazenil. C 14 H 1Z FNP3' (Mr 289.3). 1149300.
reference solution. [79089-72-8]. Ethyl 8-fluoro-6-oxo-5,6-dihydro-4H-
imidazo [1,5-a] [1 ,4]benzodiazepine-3-carboxylate.
Content: minimum 95 per cent, calculated by the
normalisation procedure. Colourless needles, soluble in dimethyl sulfoxide and in hot
methanol.
o,p'-DDD. C 14 H IO C14 • (Mr 320.0). 1125200. [53-19-0]. mp: about 288 oc.
1-(2-Chlorophenyl)-1-( 4-chlorophenyl)-2,2-dichloroethane.
A suitable certified reference solution (lO ngl¡.tL in 2-Deoxy-D-ribose. CSH1004' (Mr 134.1). 1163900.
cyclohexane) maybe used. [533-67 -5]. Thyminose. 2-Deoxy- D-erythro-pentose.
p,p'-DDD. C14 H IO C1 4 • (Mr 320.0). 1125300. [72-54-8]. 2'-Deoxyuridine. C9 H 12 NP5' (Me 228.2). 1024800.
1,1-Bis( 4-chlorophenyl)-2,2-dichloroethane. [951-78-0]. 1-(2- Deoxy-~-d-erythro-pentofuranosyl)-lH,3H­
bp: about 193 oc. pyrimidine-2,4-dione.
mp: about 109 oc. mp: about 165 oc.
A suitable certified reference solution (10 ngl¡.tL in Chromatography. Thin-Iayer chromatography (2.2.27) as
cyclohexane) may be used. prescribed in the monograph Idoxuridine (0669): apply 5 flL
of a 0.25 giL solution; the chromatogram shows only one
o,p'-DDE. C14 H sC14 • (Mr 318.0). 1125400. [3424-82-6]. principal spot.
1- (2-Chlorophenyl)-1-( 4-chlorophenyl)-2,2-dichloroethylene.
A suitable certified reference solution (lO ngl ¡.tL in 4-Deoxypyridoxine hydrochloride. C SH 1Z NO zCl.
cyclohexane) may be used. (Me 189.6). 1175500. [148-51-6]. 5-(Hydroxymethyl)-2,4-
dimethylpyridin -3-01.
p,p'-DDE. C 14 H sC1 4 • (Mr 318.0). 1125500. [72-55-9].
1,1-Bis( 4-chlorophenyl)-2,2-dichloroethylene. Desmethylmisonidazole. C6 H 9NP4' (Me 187.2). 1185600.
bp: 316 oC to 317 oc. [13551-92-3]. (2RS)-3-(2- Nitro-lH-imidazol-l-yl)propane-
l,2-diol.
mp: 88 oC to 89 oc.
Content: minimum 95 per cent.
A suitable certified reference solution (lO ngl¡.tL in
cyclohexane) may be used. Yellow powder.
o,p'-DDT. C 14 H 9 CIs' (Mr 354.5). 1125600. [789-02-6]. Destainíng solution. 1012202.
1-(2-Chlorophenyl)-1-( 4-chlorophenyl)-2,2,2-trichloroethane. A mixture consisting of 1 volume of glacial acetic acid R,
A suitable certified reference solution (lO ngl¡.tL in 4 volumes of methanol R and 5 volumes of water R.
cyclohexane) may be used.
Deuterated acetic add. C/H 4 02' (M, 64.1). 1101100.
p,p'-DDT. C I4 H 9 CIs. (Me 354.5). 1125700. [50-29-3]. [1186-52-3]. Tetradeuteroacetic acid. Acetic-d3 acid-d.
1,1-Bis( 4-chlorophenyl) -2,2,2-trichloroethane. Degree of deuteration: minimum 99.7 per cent.
bp: about 260 oc. d~g : about 1.12.
mp: 108 oC to 109 oc. n~o : about 1.368.
A suitable certified reference solution (10 ngl ¡.tL in bp: about 115 oc.
cyclohexane) may be used.
mp: about 16 oc.
Decanal. CIOHzoO. (Me 156.3). 1149200. [112-31-2]. Decyl
aldehyde. Deuterated acetone. C/H60. (Mr 64.1). 1024900. [666-52-4].
Acetone-dG• (2H 6)-Acetone.
Oily, colourless liquid, practically insoluble in water.
Decanal used in gas chromatography complies with the Degree of deuteration: minimum 99.5 percent.
following additional test. Clear, colourless liquid, miscible with water, with
Assay. Gas chromatography (2.2.28) as prescribed in the dimethylformamide, with anhydrous ethanol and with
monograph Sweet orange oil (1811). methanol.
Content: minimum 97 per cent, calculated by the d~g: about 0.87.
normalisation procedure. n~o: about 1.357.

Decane. ClOHn' (Me 142.3). 1024600. [124-18-5]. bp: about 55 oc.


Colourless liquid, practically insoluble in water. Water and deuterium oxide. Not more than 0.1 per cent.
n~o: about 1.411. Deuterated acetonitrile. C/H3N. (M, 44.1). 1173100.
bp: about 174 oc. [2206-26-0].
Decano!. CIOHzp. (Mr 158.3). 1024700. [112-30-1]. Degree of deuteration: minimum 99.8 per cent.
Decan -1-01. Clear, colourless liquid, miscible with water, with acetone and
Viscous liquid, solidifying at about 6 oC, practically insoluble with methanol.
in water, soluble in ethanol (96 per cent). d~g: about 0.78.
n~o : about l.436. n~o : about 1.344.
bp: about 230 oc.
Deuterated ~hloroform. C2HCl y (Mr 120.4). 1025000.
Deltamethrin. C22H19BrzN03' (Mr 505.2). 1125800. [865-49-6]. (-H)-Chloroform. Chloroform-d.
[52918-63-5]. Degree of deuteration: minimum 99.7 per cent.
bp: about 300 oc. Clear, colourless liquid, practically insoluble in water, miscible
mp: about 98 oc. with acetone and with ethanol (96 per cent). 1t may be
A suitable certified reference solution (10 ng/¡.tL in stabilised over silver foil.
cyclohexane) may be used. d~g: about 1. 51.

452 See the information section on general monographs (caver pages)


EUROPEAN PHARMACOPOEIA 8.0 4.1.1. Reagents

n~O : about 1.445. 3,3' -Diaminobenzidine tetrahydrochloride.


bp: about 60 oc. C¡2H¡SC14N4, 2HzÜ. (Mr 396.1). 1098000. [7411-49-6].
3,3',4,4' -Biphenyl-tetramine.
Water and deuterium oxide: maximum 0.05 per cent.
Almost white or slightly pink powder, soluble in water.
Deuterated dimethyl sulfoxide. C/H 6 0S. (Mr 84.2). mp: about 280 oC, with decomposition.
1025100. [2206-27-1]. (2H 6)-Dimethyl sulfoxide. Dimethyl
sulfoxide-d6 • Diammonium 2,2'-azinobis( 3-ethylbenzothiazoline-6-
Degree of deuteration: minimum 99.8 per cent. sulfonate). C¡SH24N606S4' (Mr 548.7). 1153000. [30931-67-0].
ABTS. Diammonium 2,2'-( diazanediylidene)bis [3-ethyl-2,3-
Very hygroscopic liquid, practically colourless, viscous, soluble
dihydrobenzothiazole-6-sulfonate ].
in water, in acetone and in anhydrous ethanol.
Chromogenic substrate suitable for use in ELISA procedures.
d~g: about 1.l8.
Green tablets, freely soluble in water.
mp: about 20 oc.
pH (2.2.3): 4.2 to 5.8 for a 0.1 giL solution.
Water and deuterium oxide: maximum 0.1 per cent.
Storage: in an airtight container. Diaíomaceous earth. 1025900. [91053-39-3].
White or almost white, fine granular powder, made up of
Deuterated methanol. C2 H 4 0. (Mr 36.1). 1025200.
siliceous frustules of fossil diatoms or of debris of fossil
[811-98-3]. (2H)-Methanol. Methanol-d.
diatoms, practically insoluble in water and in ethanol (96 per
Degree of deuteration: minimum 99.8 per cent. cent).
CIear, colourless liquid miscible with water, with ethanol The substance may be identified by microscopic examination
(96 per cent) and with methylene chloride. with a magnification of x 500.
d~g : about 0.888.
Diatomaceous earth for gas chromatography. 1026000.
n~o: about 1.326.
White or almost white, fine granular powder, made up of
bp: 65.4 oc. siliceous frustules of fossil diatoms or of debris of fossil
Deuterated sodium trimethylsilylpropionate. diatoms, practically insoluble in water and in ethanol (96 per
C6 H/H 4Na0 2Si. (Mr 172.3). 1179100. [24493-21-8]. Sodium cent). The substance may be identified by microscopic
3 -(trimethylsilyl) (2,2,3,3 -2H 4 )propionate. TSP -d4 • examination with a magnification of x 500. The substance
is purified by treating with hydrochloric acid R and washing
Degree oj deuteration: minimum 98 per cent. with water R.
White or almost white powder. Particle size: maximum 5 per cent is retained on a sieve
Deuterium chloride. 2HCl. (Mr 37.47). 1178800. [7698-05-7]. No. 180. Maximum 10 per cent passes a sieve No. 125.
Deuterated hydrochloric acid. Diatomaceous earth for gas chromatography Rl. 1026100.
Gas. White or almost white, fine granular powder, made up of
Degree oj deuteration: mínimum 99 per cent. siliceous frustules of fossil diatoms or of debris of fossil
Caution: toxico diatoms, practically insoluble in water and in ethanol (96 per
cent). The substance may be identified by microscopic
Deuterium chloride solution. 1178801. examination with a magnification of x 500. The substance
Dilute 1 mL of deuterium chloride R (38 per cent mlm) is purified by treating with hydrochloric acid R and washing
with 5 mL of deuterium oxide R. with water R.
Particle size. Maximum 5 per cent is retained on a sieve
Deuterium oxide. 2Hp. (Mr 20.03). 1025300. [7789-20-0].
No. 250. Maximum 10 per cent passes a sieve No. 180.
Deuterated water.
Degree of deuteration: minimum 99.7 per cent. Diatomaceous earth for gas chromatography R2. 1026200.
d~g : about 1.11. White or almost white, fine granular powder with a specific
n~o : about 1.328. surface are a of about 0.5 m 2 /g, made up of siliceous frustules
of fossil diatoms or of debris of fossil diatoms, practically
bp: about 101°C. insoluble in water and in ethanol (96 per cent). The substance
Deuterium oxide Rl. 2H,O. (M 20.03). 1025301. may be identified by microscopic examination with a
[7789-20-0]. Deuterated ~ater. r magnification of x 500. The substance is purified by treating
with hydrochloric aeid R and washing with water R.
Degree of deuteration: minimum 99.95 per cent.
Particle size. Maximum 5 per cent is retained on a sieve
Developer solution. 1122500. No. 180. Maximum 10 per cent passes a sieve No. 125.
Dilute 2.5 mL of a 20 giL solution of citrie acid R and 0.27 mL Diatomaceous earth for gas chromatography, silanised.
ofjormaldehyde R to 500.0 mL with water R. 1026300.
Dextran for chromatography, cross-linked R2. 1025500. Diatomaceous earth jor gas chromatography R silanised with
Bead-form dextran with a fraction range suitable for the dimethyldichlorosilane or other suitable silanising agents.
separation of peptides and proteins with relative molecular Diatomaceous earth Cor gas chromatography, silanised Rl.
masses of 15 x 10 2 to 30 X 10 3• When dry, the beads have a
1026400.
diameter of 20-80 fIm.
Prepared from crushed pink firebrick and silanised with
Dexíran for chromatography, cross-linked R3. 1025600. dimethyldichlorosilane or other suitable silanising agents. The
Bead-form dextran with a fraction range suitable for the substance is purified by treating with hydrochloric acid R and
separation of peptides and proteins with relative molecular washing with water R.
masses of 4 x 103 to 15 X 104 . When dry, the beads have a Diazinon. C 12 H 21 N Z0 3 PS. (Mr 304.3).1125900. [333-41-5].
diameter of 40-120 fIm.
bp: about 306 oc.
Dextrose. 1025700. [50-99-7]. A suitable certified reference solution (10 ngl [lL in iso-octane)
See glucose R. maybe used.

General Notices (1) apply to all monographs and other texts 453
4.1.1. Reagents EUROPEAN PHARMACOPOEIA 8.0

Diazobenzenesulfonic add solution Rl. 1026500. Dichlorobenzene. C 6H 4 Cl 2 • (Mr 147.0).1027100. [95-50-1].


Dissolve 0.9 g of sulfanilic acid R in a mixture of 30 mL of 1,2-Dichlorobenzene.
dilute hydrochloric acid R and 70 mL of water R. To 3 mL of Colourless, oily liquid, practically insoluble in water, soluble
the solution add 3 mL of a 50 giL solution of sodium nitrite R. in anhydrous ethanol.
Cool in an ice-bath for 5 min, add 12 mL of the sodium nitrite d~g : about l.31.
solution and cool again. Dilute to 100 mL with water R and
bp: about 180 oc.
keep the reagent in an ice-bath. Prepare extemporaneously
but allow to stand for 15 min before use. 2,4-Dichlorobenzoic add. C7 H 4 C1 2 0 2. (Mr 191.0). 1185700.
[50-84-0].
DibutyJ.amine. C SH 19 N. (Mr 129.3). 1126000. [111-92-2].
N-Butylbutan-l-amine. Faintly beige powder.
Colourless liquido mp: about 160 oc.
n5°: about 1.417. 2,3- Dichloro-5,6-dicyanobenzoquinone. CsCl 2N 202'
bp: about 159 oc. (Mr 227.0). 1153600. [84-58-2]. 4,5-Dichloro-3,6-dioxo-
cyc!ohexa -1 A-diene-l ,2-dicarbonitrile.
Dibutylammonium phosphate for ion-pairing. 1168800.
Yellow or orange crystals, soluble in dioxan and in acetic acid,
A colourless solution of 10 per cent to 15 per cent V/V slightly soluble in methylene chloride. It decomposes in water.
of di-n-butylamine and 12 per cent to 17 per cent V/V of mp: about 214 oc.
phosphoric acid in water, suitable for ion-pairing in liquid
chromatography. Storage: at a temperature of 2 oC to 8 oc.

Dibutyl etller. CSH 1S O. (Mr 130.2). 1026700. [142-96-1]. (S)-3,5- Dichloro-2,6-dihydroxy-N- [( l-ethylpyrroHdin-
2-yl) methyl] benzamide hydrobromide. C14H19BrClzN P3'
Colourless, flammable liquid, practically insoluble in water, (Mr 414.1). 1142600. [113310-88-6].
miscible with anhydrous ethano!.
White or almost white, crystalline powder.
d~g: about 0.77.
[a l~2 : + 11.4, determined on a 15.0 giL solution in anhydrous
n5°: about 1.399. ethanol R.
Do not distil if the dibutyl ether does not comply with the test mp: about 212 oc.
for peroxides.
Peroxides. Place 8 mL of potassium iodide and starch solution R Dichlorofluorescein. C20HlQC1 20 s' (Mr 401.2).
in a 12 mL ground-glass-stoppered cylinder about 1.5 cm in 1027200. [76-54-0]. 2,7 -Dichlorofluorescein.
diameter. Fill completely with the substance to be examined, 2- (2,7 -Dichloro-6-hydroxy-3-oxo-3H-xanthen -9-yl) benzoic
shake vigorously and allow to stand protected from light for acid.
30 mino No colour is produced. Yellowish-brown or yellow-orange powder, slightly soluble
The name and concentration of any added stabiliser are stated in water, freely soluble in ethanol (96 per cent) and in dilute
on the labe!' solutions of alkali hydroxides giving a solution showing a
yellowish-green fluorescence.
Dibutyl phthalate. C 16 H 22 0 4 • (Mr 278.3). 1026800. [84-74-2].
Dibutyl benzene-l,2-dicarboxylate. 2,6-DichlorophenoL C 6 H 4 CI 2 0. (Mr 163.0). 1177600.
Clear, colourless or faintly coloured, oily liquid, very slightly [87-65-0].
soluble in water, miscible with acetone and with ethanol mp: 64 oC to 66 oc.
(96 per cent).
Di.chlorophenolindophenol, sodium salto
d~g: 1.043 to 1.048. C12H6CI2NNa02,2Hp. (Mr 326.1). 1027300. [620-45-1].
n5°: 1.490 to 1.495. The sodium derivative of 2,6-dichloro-N-(4-hydroxy-
phenyl) -1 04- benzoquinone monoimil1e dihydrate.
Dicarboxidine hydrochloride. C2oH26C12N206' (Mr 461.3).
Dark-green powder, freely soluble in water and in anhydrous
1026900. [56455-90-4]. 4A'-[(4A'-Diaminobiphenyl-3,3'-
ethanol. The aqueous solution is dark blue; when acidified
diyl)dioxy]dibutanoic acid dihydrochloride.
it becomes pink.
Dichlofenthion. ClOH13ClP3PS, (Mr 315.2). 1126100.
Dichlorophenolindophenol standard solution. 1027301.
[97-17-6].
Dissolve 50.0 mg of dichlorophenolindophenol, sodium
A suitable certified reference solution (lO ngl flL in
salt R in 100.0 mL of water R and filter.
cyelohexane) may be used.
Assay. Dissolve 20.0 mg of ascorbic acid R in 10 mL of a
Dichloroacetic add. C 2H 2ClP2' (Mr 128.9). 1027000. freshly prepared 200 giL solution of metaphosphoric acid R .
[79-43-6]. and dilute to 250.0 mL with water R. Titrate 5.0 mL rapidly
Colourless liquid, miscible with water and ethanol (96 per with the dichloro-phenolindophenol standard solution,
cent). added from a microburette graduated in 0.01 mL, until the
pink colour persists for 10 s, the titration occupying not
d~g: about 1.566.
more than 2 mino Dilute the dichlorophenolindophenol
n5°: about 1.466. solution with water R to make 1 mL of the solution
bp: about 193 oc. equivalent to 0.1 mg of ascorbic acid (C6HsÜ6)'
Storage: use within 3 days.
Dichloroacetic add solution. 1027001.
Standardise immediately before use.
Dilute 67 mL of dichloroacetic acid R to 300 mL with
water R and neutralise to blue litmus paper R using 5,7-Dichloroquinolin-8-ol. C 9 H 5 C1 2NO. (Mr 214.1).
ammonia R. Coo!, add 33 mL of dichloroacetic acid R and 1157000. [773-76-2]. 5,7-Dichlorooxine.
dilute to 600 mL with water R.
Yellow, crystalline powder, soluble in acetone, slightly soluble
3,5-DichloroaniHne. C 6H sCI 2N. (Mr 162.0). 1177800. in ethanol (96 per cent).
[626-43-7]. 3,5-dichlorophenylamine. mp: about 179 oc.
mp: 46 oC to 52 oc. Content: minimum 95.0 per cent.

454 See the information section on general monographs (cover pages)


EUROPEAN PHARMACOPOEIA 8.0 4.1.1. Reagents

Dichloroquinonechlorimide. C6H 2CI 3NO. (M, 210.4). Reference solutions. Dissolve 0.50 g of ethanolamine R in
1027400. [101-38-2]. 2,6-Dichloro-N-chloro-l,4- acetone R and dilute to 10.0 mL with the same solvent. To
benzoquinone mono-imine. 0.5 mL, 1.0 mL and 2.0 mL of this solution, add 1.0 mL of
Pale yellow or greenish-yellow crystalline powder, practically the internal standard solution and dilute to 10.0 mL with
insoluble in water, soluble in ethanol (96 per cent) and in acetone R.
dilute alkaline solutions. Column:
mp: about 66 oc. - size: 1 = 1 m, 0 = 4 mm;
- stationary phase: diphenylphenylene oxide polymer R
Dichlorvos. C 4 H 7C1 2 0 4P. (M, 221). 1101200. [62-73-7].
(180-250 flm).
2,2-Dichlorovinyl dimethyl phosphate.
Carrier gas: nitrogen for chromatography R.
Colourless or brownish-yellow liquid, soluble in water,
miscible with most organic solvents. Flow rate: 40 mLlmin.
n55 : about 1.452. Temperature:
Time Temperature
Dicydohexyl. C 12 H 22 • (Me 166.3). 1135300. [92-51-3]. (min) (OC)
Bicyclohexyl. o..,. 3 125
Column
:
d;~ about 0.864.
3 ..,. 17.6 125 ..,. 300
bp: about 227 oc.
mp: about 4 oc. Injection port 250

Detector 280
Dicyelohexylamine. C 12 H n N. (M, 181.3). 1027500.
[101-83-7]. N,N- Dicyclohexylamine. Detectiol1: flame- ionisation.
Colourless liquid, sparingly soluble in water, miscible with the Injectiol1: 1.0 flL.
usual organic solvents.
Limit:
n50: about 1.484. - ethanolamine: maximum 1.0 per cent.
bp: about 256 oc.
Freezing point (2.2.18) : O oC to 1 oc. Diethoxytetrahydrofuran. CSH¡60y (M, 160.2). 1027900.
[3320-90-9]. 2,5-Diethoxytetrahydrofuran. A mixture of the
Dicyclohexylurea. C¡3H24NP. (M, 224.4). 1027600. cis and trans isomers.
[2387 -23-7]. 1,3-Dicyclohexylurea. Clear, colourless or slightly yellowish liquid, practically
White or almost white, crystalline powder. insoluble in water, soluble in ethanol (96 per cent) and in most
mp: about 232 oc. other organic solvents.
d~g : about 0.98.
Didocosahexaenoin. C47H6S0S' (Me 713.0). 1142700.
[88315-12-2]. Diglyceride of docosahexaenoic acid (C22:6).
n50: about l.418.
Glycerol didocosahexaenoate. (all-Z)-Docosahexaenoic acid, Diethylamine. C"¡H ll N. (M, 73.1). 1028000. [109-89-7].
diester with propane-1,2,3-triol. Clear, colourless, flammable liquid, strongly alkaline, miscible
Didodecy13,3'-thiodipropionate. C30H,sÜ4S, (Me 514.8). with water and with ethanol (96 per cent).
1027700. [123-28-4]. d~g : about 0.71.
White or almost white, crystalline powder, practically bp: about 55 oc.
insoluble in water, freely soluble in acetone and in light Diethylamine Rl. C4 H¡¡N. (Me 73.1).1028001. [109-89-7].
petroleum, slightly soluble in ethanol (96 per cent). N- Ethylethanamine.
mp: about 39 oc. Content: minimum 99.5 per cent.
Dieldrin. C 12 H sCI 6 0. (Me 380.9). 1126200. [60-57-1]. Clear, colourless, flammable liquid, strongly alkaline,
bp: about 385 oc. miscible with water and with ethanol (96 per cent).
mp: about 176 oc. d~g: about 0.71.
A suitable certified reference solution (lO ng/flL in bp: about 55 oc.
cyclohexane) may be used. Diethylaminoethyldextran. 1028200.
Diethanolamine. C4H ll N0 2 • (Me 105.1). 1027800. Anion -exchange resin presented as the hydrochloride.
[111-42-2]. 2,2'-Iminobisethanol. Powder forming gel s with water.
Viscous, clear, slightly yellow liquid or deliquescent crystals N,N-Diethylaniline. ClOH15N. (M, 149.2). 1028400. [91-66-7].
melting at about 28 oC, very soluble in water, in acetone and
d~g: about 0.938.
in methanol.
bp: about 217 oc.
d~g: about 1.09.
mp: about - 38 oc.
pH (2.2.3): 10.0 to 11.5 for a 50 giL solution.
Diethanolamine used in the test for alkaline phosphatase Diethylene glycoL C4HlOOy (Me 106.1).1028300. [111-46-6].
complies with the following additional test. 2,2' -Oxydiethanol.
Ethanolamine. Gas chromatography (2.2.28). Content: minimum 99.5 per cent mlm.
Internal standard solution. Dissolve 1.00 g of Clear, colourless liquid, hygroscopic, miscible with water, with
3-aminopropanol R in acetone R and dilute to 10.0 mL with acetone and with ethanol (96 per cent).
the same solvento d~g: about l.1l8.
Test solution (a). Dissolve 5.00 g of the substance to be n5°: about 1.447.
examined in acetone R and dilute to 10.0 mL with the same bp: 244 oC to 246 oc.
solvent. Storage: in an airtight container.
Test solution (b). Dissolve 5.00 g of the substance to be
examined in acetone R, add 1.0 mL of the internal standard N,N- Diethylethane-l ,2-diamine. 1028500. [100- 36-7].
solution and dilute to 10.0 mL with the same solvento See N,N-diethylethylenediamine R.

General Notices (1) apply to all monographs and other texts 455
4.1.1. Reagents EUROPEAN PHARMACOPOEIA 8.0

N,N-Diethylethylenediamine. C6H¡6N2' (Mr 116.2). 1028500. Content calculated by the normalisation procedure:
[100-36-7]. - major camponent (trans-dihydrocarvone): minimum 70 per
Content: minimum 98.0 per cent. cent;
Slightly oily liquid, colourless or slightly yellow, strong odour - sum of cis- and trans-dihydrocarvone: minimum 98 per
of ammonia, irritant to the skin, eyes and mucous membranes. cent.
d~g: 0.827. 2,5-Dihydroxybenzoic acid. C7H604' (Mr 154.1). 1148200.
bp: 145 oC to 147 oc. [490-79-9]. Gentisic acid.
Water (2.5.12): maximum 1.0 per cent, determined on 0.500 g. Light yellow crystals.
mp: about 200 oc.
Di(2-ethylhexyl) phthalate. C Z4H J sÜ4' (Mr 390.5). 1028100.
Di(2-ethylhexyl) benzene-1 ,2-dicarboxylate. 5,7-Dihydroxy-4-methykoumarin. C lO H s04' (Mr 192.2).
Colourless, oily liquid, practically insoluble in water, soluble 1149400. [2107-76-8]. 5,7-Dihydroxy-4-methyl-2H-l-
in organic solvents. benzopyran -2-one.
d~g: about 0.98. Light yellowish powder, practically insoluble in water,
nbo : about 1.486. sparingly soluble in ethanol (96 per cent).
mp: 295 oC to 303 oc.
Viscasity (2.2.9): about 80 mPa·s.
Dihydroxynaphthalene. 1029000. [132-86-5].
Diethylphenylenediamine sulfate. ClQH¡SNP4S, (Mr 262.3).
1028600. [6283-63-2]. N,N'- Diethyl-p-phenylenediamine See 1,3-dihydroxynaphthalene R.
sulfate. N,N'- Diethylbenzene-1,4-diamine sulfate. 1,3-Dihydroxynaphthalene. ClOHsÜ2' (Mr 160.2). 1029000.
White or slightly yellow powder, soluble in water. [132-86-5]. Naphthalene-1,3-diol.
mp: about 185 oC, with decomposition. Crystalline, gene rally brownish-violet powder, freely soluble
Storage: protected from light. in water and in ethanol (96 per cent).
mp: about 125 oc.
Diethylphenylenediamine sulfate solution. 1028601.
To 250 mL of water R add 2 mL of sulfuric acid R and 2,7-Dihydroxynaphthalene. ClQHP2' (M, 160.2). 1029100.
25 mL of 0.02 M sodium edetate. Dissolve in this solution [582-17 -2]. Naphthalene-2,7 -diol.
1.1 g of diethylphenylenediamine sulfate R and dilute to Needles, soluble in water and in ethanol (96 per cent).
1000 mL with water R. mp: about 190 oc.
Do not use if the solution is not colourless.
2,7-Dihydroxynaphthalene solution. 1029101.
Storage: protected from light and heat for 1 month. Dissolve 10 mg of 2,7-dihydroxynaphthalene R in 100 mL
Diflubenzuron. C¡4H9CIF2N202' (Mr 310.7). 1180000. of sulfuric acid R and allow to stand until decolorised.
[35367 -38-5]. 1-( 4-Chlorophenyl)-3-(2,6-difluoro- Storage: use within 2 days.
benzoyl)urea.
5,7-Diiodoquinoli.n-8-ol. C9H sI 2 NO. (Mr 397.0). 1157100.
Colourless or white 01' almost white crystals, practically [83-73-8]. 5,7-Diiodooxine.
insoluble in water, freely soluble in dimethyl sulfoxide, slightly
soluble in acetone. Yellowish-brown powder, sparingly soluble in acetone and in
ethanol (96 per cent).
mp: 230 to 232 oc.
Content: minimum 95.0 per cent.
DigHonin. CS6H92029' (Mr 1229). 1028700. [11024-24-1]. Di-isobutylketone. CyH¡p. (M 142.2).1029200. [108-83-8].
r
3~- [O-~-D-Glucopyranosyl-(l ~3)-O-~-D-galactopyranosyl-
(1 ~2)-O- [~-D-xylopyranosyl-(l ~3) ]-O-~-D-galactopyranosyl­ Clear, colourless liquid, slightly soluble in water, miscible with
(1 ~4) -O-~- D-galactopyranosyloxy ]-(25R)-5a -spirostan- most organic solvents.
2a,15~-diol. nbo: about 1.414
Crystals, practically insoluble in water, sparingly soluble in bp: about 168 oc.
anhydrous ethanol, slightly soluble in ethanol (96 per cent).
Di-isopropyl ether. C6 H¡40. (Mr 102.2). 1029300. [108-20-3].
Digitoxin. 1028800. [71-63-6]. CIear, colourless liquid, very slightly soluble in water, miscible
See Digitoxin (0078). with ethanol (96 per cent).
d~g: 0.723 to 0.728.
Dihydrocapsaicin. C¡SH 29 N03' (M, 307.4). 1148100. bp: 67 oC to 69 oc.
[19408-84-5]. N- [( 4- Hydroxy-3-methoxyphenyl)methyl]-8-
methylnonanamide. Do not distil if the di-isopropyl ether does not comply with the
test for peroxides.
White or almost white, crystalline powder, practically
insoluble in cold water, freely soluble in anhydrous ethanol. Peroxides. Place 8 mL of potassium iodíde and starch solution R
in a 12 mL ground-glass-stoppered cylinder about 1.5 cm in
10,11-Dihydrocarbamazepine. C¡SH¡4Np. diameter. Fill completely with the substance to be examined,
(Mr 238.3). 1028900. [3564-73-6]. 10,1l-Dihydro- shake vigorously and allow to stand protected from light for
5H-dibenzo [b,j] azepine- 5 -carboxamide. 30 mino No colour is produced.
mp: 205 oC to 210 oc. The name and concentration of any added stabiliser are stated
on the label.
Dihydrocarvone. ClQH¡60. (Mr 152.2). 1160900.
Storage: protected from light.
[7764-50-3]. p-Menth-8-en-2-one. 2-Methyl-5-(l-
methylethenyl)cyclohexanone. N,N~Diisopropylethylenediamine. CSH 20N 2. (Mr 144.3).
Dihydrocarvone used in gas chromatography complies with the 1140600. [4013-94-9]. N,N"-Bis(l-methylethyl)-1,2-
following additional test. ethanediamine.
Assay. Gas chromatography (2.2.28) as prescribed in the Colourless or yellowish, corrosive, flammable, hygroscopic
test for chromatographic profile in the monograph Caraway liquido
oil (1817). d§g: about 0.798.

456 See the informatíon section on general monographs (caver pages)


EUROPEAN PHARMACOPOEIA 8.0 4. L 1. Reagents

n~?: about 1.429. Storage: protected from light; use within 4 weeks.
bp: about 170 oc.
Dimethylaminobenzaldehyde solution R8. 1029805.
4,4'-Dimethoxybenzophenone. C1SH140y (Mr 242.3). Dissolve 0.25 g of dimethylaminobenzaldehyde R in a
1126300. [90-96-0]. Bis(4-methoxyphenyl)methanone. mixture of 5 g of phosphoric acid R, 45 g of water R and 50 g
White or almost white powder, practically insoluble in water of anhydrous acetic acid R. Prepare immediately before use.
and slightly soluble in ethanol (96 per cent).
4-Dimethylaminodnnamaldehyde. CllH13NO.
mp: about 142 oc. (M¡ 175.2). 1029900. [6203-18-5]. 3-(4-Dimethylamino-
Dimethoxypropane. CSH 12 0 2 • (Mr 104.1). 1105200. phenyl)prop-2-enal.
[77 -7 6-9]. 2,2-Dimethoxypropane. Orange or orange-brown crystals or powder. Sensitive to light.
Colourless liquid, decomposing on exposure to moist air or mp: about 138 oc.
water.
d~g: about 0.847.
4- Dimethylaminocinnamaldehyde solution. 1029901.
nir about 1.378. Dissolve 2 g of 4-dimethylaminocinnamaldehyde R in a
mixture of 100 mL of hydrochloric acid R1 and 100 mL of
bp: about 83 oc. anhydrous ethanol R. Dilute the solution to four times its
Dimethylacetamide. C4 H 9 NO. (M¡ 87.1). 1029700. volume with anhydrous ethanol R immediately before use.
[127-19-5]. N,N-Dimethylacetamide.
2- (Dimethylamino )ethyl methacrylate. CSH 1S N0 2 •
Content: minimum 99.5 per cent. (M, 157.2). 1147200. [2867-47-2]. 2-(Dimethylamino)ethyl
Colourless liquid, miscible with water and with many organic 2-methylpropenoate.
solvents. d~o: about 0.930.
d~g : about 0.94. bp: about 187 oc.
n~o: about 1.437.
bp: about 165 oc. Dimethylaminonaphthalenesulfonyl chloride.
C 12 H 12 CIN0 2 S. (Mr 269.8). 1030000. [605-65-2].
Dimethylamine. C2 H 7 N. (Mr 45.08). 1168900. [124-40-3]. 5-Dimethyl-amino-1-naphthaIenesulfonyl chloride.
N-Methylmethanamine. Yellow, crystalline powder, slightly soluble in water, soluble
Colourless, flammable gas. in methanol.
bp: about 7 oc. mp: about 70 oc.
mp: about - 92.2 oc.
3-Dimethylaminophenol. CSHllNO. (M, 137.2). 1156500.
Dimethylamine solution. 1168901. [99-07 -O]. 3-(Dimethylamino )phenol.
A 400 giL solution. Grey powder, slightly soluble in water.
Clear, colourless solution. mp: about 80 oc.
Density: about 0.89.
2- (Dimethylamino )thioacetamide hydrochloride.
bp: about 54 oc. C4 H ll ClN 2S. (M, 154.7). 1181800. [27366-72-9].
mp: about - 37 oc.
Dimethylaniline. 1030100. [121-69-7].
Dimethylaminobenzaldehyde. C9HllNO. (Mr 149.2). See N,N-Dimethylaniline R.
1029800. [100-10-7]. 4-Dimethylaminobenzaldehyde.
White or yellowish-white crystals, soluble in ethanol (96 per N,N-Dimethylaniline. CSH 11 N. (M¡ 121.2). 1030100.
cent) and in dilute acids. [121-69-7].
mp: about 74 oc. CIear, oily liquid, almost colourless when freshly distilled,
darkening on storage to reddish-brown, practically insoluble
Dimethylaminobenzaldehyde solution Rl. 1029801. in water, freely soluble in ethanol (96 per cent).
Dissolve 0.2 g of dimethylaminobenzaldehyde R in 20 mL n~o: about 1.558.
of ethanol (96 per cent) R and add 0.5 mL of hydrochloric
acid R. Shake the solution with activated charcoal R and Distillation range (2.2.11). Not less than 95 per cent distils
filter. The colour of the reagent is less intense than that of between 192 oC and 194 oc.
iodine solution R3. Prepare immediately before use. 2,3-Dimethylaniline. CS H 11 N. (Mr 121.2). 1105300.
Dimethylaminobenzaldehyde solution R2. 1029802. [87-59-2]. 2,3-Xylidine.
Dissolve 0.2 g of dimethylaminobenzaldehyde R, without Yellowish liquid, sparingly soluble in water, soluble in ethanol
heating, in a mixture of 4.5 mL of water R and 5.5 mL of (96 per cent).
hydrochloric acid R. Prepare immediately before use. d~g: 0.993 to 0.995.
Dimethylaminobenzaldehyde solution R6. 1029803. n~o: about 1.569.

Dissolve 0.125 g of dimethylaminobenzaldehyde R in a bp: about 224 oc.


cooled mixture of 35 mL of water R and 65 mL of sulfuric
2,6-Dimethyianiline. CSHllN. (Mr 121.2). 1030200.
acid R. Add 0.1 mL of a 50 giL solution ofjerric chloride R.
[87-62-7]. 2,6-Xylidine.
Before use allow to stand for 24 h, protected from light.
Storage: when stored at room temperature, use within Colourless liquid, sparingly soluble in water, soluble in ethanol
1 week; when stored in a refrigerator use within several (96 per cent).
months. d~g : about 0.98.

Dimethylaminobenzaldehyde solution R7. 1029804. 2,6-Dimethylaniline hydrochloride. C sH 12 CIN. (Mr 157.6).


Dissolve 1.0 g of dimethylaminobenzaldehyde R in 50 mL 1169000. [21436-98-6]. 2,6-Dimethylbenzenamide
of hydrochloric acid R and add 50 mL of ethanol (96 per hydrochloride. 2,6-Xylidine hydrochloride.
cent) R. Content: minimum 98.0 per cent.

General Notices (1) apply to all monographs and other texts 457
4.1.1. Reagents EUROPEAN PHARMACOPOEIA 8.0

2,4-Dimethyl-6-tert-butylphenol. C 12H¡p. (M, 178.3). Dimethylglyoxime. C 4 H gNP1' (M, 116.1). 1030400.


1126500. [1879-09-0]. [95-45-4]. 2,3-Butanedione dioxime.
White or almost white, erystalline powder or eolourless
Dimethyl carbonate. C3H603' (M, 90.1). 1119300. erystals, praetically insoluble in eold water, very slightly
[616-38-6]. Carbonie acid dimethyl estero soluble in boiling water, soluble in ethanol (96 per eent).
Liquid, insoluble in water, miscible with ethanol (96 per eent). mp: about 240 oC, with deeomposition.
dl 7 : 1.065. Sulfated ash (2.4.14): maximum 0.05 per eent.
n~o: 1.368.
1,3-Dimethyl-2-imidazolidinone. CSHIONzO. (M, 114.2).
bp: about 90 oc. 1135400. [80-73-9]. N,N'-Dimethylethylene urea.
1,3-Dimethyl-2-imidazolidone.
Dimethyl-~-cydodextrin. CS6H9S03S' (M, 1331). 1169100.
[51166-71-3]. Heptakis(2,6-di-0-methyl)eyclomaltoheptaose. n~o: 1.4720.
Cycloheptakis-( 1-74)-(2,6-di -0-methyl-a -D-glueopyranosyl). bp: about 224 oc.
2A,2 B,2 e,2 D,2 E,2 F,2 G,6 A ,6 B,6 c,6 D,6 E,6 F,6 G - Tetradeea -0-methyl- ~_
eyclodextrin. N,N-Dimethyloctylamine. CIOH23N. (M, 157.3). 1030500.
[7378-99-6]. Octyldimethylamine.
White or almost white powder.
Colourless liquido
DimethyIdecylamine. C¡2H27N. (M, 185.4). 1113500. d~g: about 0.765.
[1120-24-7]. N,N-dimethyldeeylamine. n~o : about 1.424.
Content: minimum 98.0 per eent mlm. bp: about 195 oc.
bp: about 234 oc.
2,5-DimethylphenoL CSHIOO. (M, 122.2). 1162300.
l,l-Dimethylethylamine. C4 H ll N. (M, 73.1). 1100900. [95-87-4]. p-Xylenol.
[75-64-9]. 2-Amino-2-methylpropane. tert-Butylamine. White or almost white erystals.
Liquid, miscible with ethanol (96 per eent). 2,6-DimethylphenoL CSHIOO. (M, 122.2). 1030600.
d~g : about 0.694. [576-26-1].
n~o: about 1.378. Colourless needles, slightly soluble in water, very soluble in
bp: about 46 oc. ethanol (96 per eent).
bp: about 203 oc.
1,1-Dimethylethyl methyl ether. CSH¡p. (M, 88.1). 1013900. mp: 46 oC to 48 oc.
[1634-04-4]. 2-Methoxy-2-methylpropane. tert- Butyl methyl
ether. 3,4-Dimethylphenol. CSHIOO. (M, 122.2). 1098100.
Colourless, clear, flammable liquido [95-65-8].
n~o: about 1.376.
White or almost white erystals, slightly soluble in water, freeIy
soluble in ethanol (96 per eent).
Minimum transmittance (2.2.25) using water R as
bp: about 226 oc.
eompensation liquid: 50 per eent at 240 nm, 80 per eent at
255 nm, 98 per cent at 280 nm. mp: 25 oC to 27 oc.

l,l-Dimethylethyl methyl ether Rl. CSH¡p. (Mr 88.1). N,N-Dimethyl-L-phenylalanine. C ll H¡SN0 2. (M, 193.2).
1126400. [1634-04-4]. 2-Methoxy-2-methylpropane. 1164000. [17469-89-5]. (2S)-2-(Dimethylamino)-3-
tert-Butyl methyl ether. phenylpropanoic acid.
mp: about 226 oc.
Content: minimum 99.5 per eent.
d~g: about 0.741. Dimethylpiperazine. C 6 H 14N1. (M, 114.2). 1030700.
n~o: about 1.369.
[106-58-1]. 1,4-Dimethylpiperazine.
A eolourless liquid, miscible with water and with ethanol
bp: about 55 oc.
(96 per eent).
Dimethylformamide. C3H7NO. (Mr 73.1). 1030300. d~g : about 0.85.
[68-12-2]. n~o : about 1.446.
Clear, colourless neutralliquid, miscible with water and with bp: about 131 oc.
ethanol (96 per eent).
DimethyIstearamide. C20 H 4 ¡NO. (M, 311.6). 1030800.
d~g: 0.949 to 0.952.
N,N- Dimethylstearamide.
bp: about 153 oc. White or almost white solid mass, soluble in many organic
Water (2.5.12): maximum 0.1 per eent. solvents, including aeetone.
mp: about 51 oc.
Dimethylformamide diethylacetal. C7H¡7N02' (M, 147.2).
1113600. [1188-33-6]. N, N-Dimethylformamide diethylaeetal. Dimethylstearylamide. 1030800.
n~o : about 1.40. See dimethylstearamide R.
bp: 128 oC to 130 oc.
Dimethyl sulfone. C 2H 6 0 1S. (M, 94.1). 1030900. [67-71-0].
N,N-Dimethylformamide dimethylaeetal. CSH 13 N0 1. White or almost white, erystalline powder, freeIy soluble in
(M, 119.2).1140700. [4637-24-5]. l,l-Dimethoxytrimethyl- water, soluble in acetone and ethanol (96 per cent).
amine. mp: 108 oC to 110 oc.
Clear, eolourless liquido
Dimethyl sulfoxide. 1029500. [67-68-5].
d~g: about 0.896. See Dimethyl sulfoxide (0763).
n~o: about 1.396. Dimethyl sulfoxide used in spectrophotometry complies with
bp: about 103 oc. the following additional test.

458 See the information section on general monographs (ca ver pages)
EUROPEAN PHARMACOPOEIA 8.0 4,1.1. Reagents

Mínimum transmíttance (2.2.25) using water R as Dinitrophenylhydrazine-hydrochloric solution.


compensation liquid: 10 per cent at 262 nm, 35 per cent at 1031502.
270 nm, 70 per cent at 290 nm, 98 per cent at 340 nm and Dissolve by heating 0.50 g of dinitrophenylhydrazine R in
at higher wavelengths. dilute hydrochloric acid R and dilute to 100 mL with the
Dimethyl sulfoxide RL 102950l. same solvent. Allow to coo1 and filter. Prepare immediately
before use.
Content: minimum 99.7 per cent, determined by gas
chromatography. Dinitrophenylhydrazine-suUuric add solution. 1031503.
Dimeticone, 1105400. [9006-65-9]. Dissolve 1.5 g of dinitrophenylhydrazine R in 50 mL of
a 20 per cent V/V soIution of sulfuric acid R. Prepare
See Dimeticone (0138).
immediately before use.
Dimidium bromide, C20H1SBrNy (Me 380.3). 1031100.
[518-67 -2]. 3,8-Diamino-5-methyl-6-phenylphen- Dinonyl phthalate. C 26 H 4P4' (Me 418.6). 1031600.
[28553-12-0].
anthridinium bromide.
Dark-red crystals, slightly soluble in water at 20 oC, sparinglyColourless to pale yellow, viscous liquido
soluble in water at 60 oC and in ethanol (96 per cent). d~g : 0.97 to 0.98.
nbO: 1.482 to 1.489.
Dimidium bromide-sulfan blue mixed solution,
1031101. Acidity. Shake 5.0 g with 25 mL of water R for 1 mino Allow
to stand, filter the separated aqueous layer and add 0.1 mL of
Dissolve separately 0.5 g of dimidium bromide R and 0.25 g
phenolphthalein solution R. Not more than 0.3 mL of 0.1 M
of sulfan blue R in 30 mL of a hot mixture of 1 volume of sodium hydroxide is required to change the colour of the
anhydrous ethanol R and 9 volumes of water R, stir, mix the solution (0.05 per cent, calculated as phthalic acid).
two solutions, and dilute to 250 mL with the same mixture
of solvents. Mix 20 mL of this solution with 20 mL of a Water (2.5.12): maximum 0.1 per cent.
14.0 per cent V/V solution of sulfuric acid R previously Dioctadecyl disulfide. C36H74S2' (Mr 571.1). 1031700.
diluted with about 250 mL of water R and dilute to 500 mL [2500-88-1].
with water R.
White or almost white powder, practically insoluble in water.
Storage: protected from light.
mp: 53 oC to 58 oc.
Dinitrobenzene, C6H4NzÜ4' (Me 168.1). 1031200. [99-65-0].
1,3-Dinitrobenzene. 2,2' -Di( octadecyloxy) -5,5'-spirobi( 1,3,2-dioxaphosphorin-
ane). C 41 H sP6P2' (M¡ 733). 1031800.
Yellowish crystalline powder or crystals, practically insoluble
in water, slightly soluble in ethanol (96 per cent). White or almost white, waxy solid, practically insoluble in
water, soluble in hydrocarbons.
mp: about 90 oc.
mp: 40 oC to 70 oc.
Dinitrobenzene solution, 103120l.
DioctadecyI3,3'-thiodipropionate. C42HsP4S, (Me 683).
A 10 giL solution in ethanol (96 per cent) R.
1031900. [693-36-7].
Dinitrobenzoic add. C7H4NzÜ6' (M, 212.1). 1031300. White or almost white, crystalline powder, practically
[99- 34- 3]. 3,5-Dinitrobenzoic acid. insoluble in water, freeIy soluble in methylene chloride,
Almost colourless crystals, slightly soluble in water, very sparingly soluble in acetone, in ethanol (96 per cent) and in
soluble in ethanol (96 per cent). light petroleum.
mp: about 206 oc. mp: 58 oC to 67 oc.
Dinitrobenzoic add solution. 1031301. Dioxan. C4HsÜ2' (Me 88.1). 1032000. [123-91-1]. l,4-Dioxan.
A 20 giL solution in ethanol (96 per cent) R. CIear, colourless liquid, miscible with water and with most
organic solvents.
Dinitrobenzoyl chloride. C7H3ClNzÜs' (Mr 230.6). 1031400.
[99-33-2]. 3,5-Dinitrobenzoyl chloride. d~g : about 1.03.
Translucent, yellow or greenish-yellow powder or yellowish Freezing-point (2.2.18): 9 oC to 11 oc.
crystals, soluble in acetone and in toluene. Water (2.5.12): maximum 0.5 per cent.
mp: about 68 oc. Do not distil if the dioxan does not comply with the test for
Suitability test. To 1 mL of anhydrous ethanol R and 0.1 g peroxides.
of dinitrobenzoyl chloride R add 0.05 mL of dilute sulfuric Peroxides. Place 8 mL of potassium iodide and starch solution R
acid R and boil under a reflux condenser for 30 mino After in a 12 mL ground-glass-stoppered cylinder about 1.5 cm in
evaporation on a water-bath add 5 mL of heptane R to the diameter. Fill completely with the substance to be examined,
residue and heat to boiling. Filter the hot solution. Wash the shake vigorously and allow to stand in the dark for 30 mino
crystals formed on cooling to room temperature with a small No colour is produced.
quantity of heptane R and dry in a desiccator. The crystals Dioxan used for liquid scintillation is of a suitable analytical
melt (2.2.14) at 94 oC to 95 oc. grade.
Dinitrophenylhydrazine. C 6 H 6NP4' (Me 198.1). 1031500. Dioxan solution. 1032002.
[119-26-6]. 2,4-Dinitrophenylhydrazine.
Dilute 50.0 mL of dioxan stock solution R to 100.0 mL with
Reddish-orange crystals, very slightly soluble in water, slightly water R. (0.5 mg/mL of dioxan).
soluble in ethanol (96 per cent).
mp: about 203 oC (instantaneous method). Dioxan solution Rl. 1032003.
Dilute 10.0 mL of dioxan solution R to 50.0 mL with
Dinitrophenylhydrazine- aceto- hydrochlork solution. water R. (0.1 mg/mL of dioxan).
1031501.
Dissolve 0.2 g of dinitrophenylhydrazine R in 20 mL of Dioxan stock solution. 1032001.
methanol R and add 80 mL of a mixture of equal volumes of Dissolve 1.00 g of dioxan R in water R and dilute to
acetic acid R and hydrochloric acid R1. Prepare immediately 100.0 mL with the same solvent. Dilute 5.0 mL of this
before use. soIution to 50.0 mL with water R (1.0 mg/mL).

General Notices (1) apply lo all monographs and other texts 459
401.1. Reagents EUROPEAN PHARMACOPOEIA 8.0

Diphenylamine. C 12 H ll N. (Mr 169.2). 1032100. [122-39-4]. 2,2-Diphenylglycine. C 1j H 13 N0 2. (Mr 227.26). 1174300.


White or almost white crystals, slightly soluble in water, [3060-50-2]. Amino(diphenyl)acetic acid.
soluble in ethanol (96 per cent). 1,2-Diphenylhydrazine. CJ2HJ2N2. (Mr 184.3). 1140800.
mp: about 55 oc. [122-66-7]. Hydrazobenzene. 1,2-Diphenyldiazane.
Storage: protected from light. Orange powder.
Diphenylamine solution. 1032101. mp: about 125 oc.
A 1 giL solution in sulfuric acid R. Diphenylmethanol. C13H120. (Mr 184.2). 1145700. [91-01-0].
Storage: protected from light. Benzhydrol.
Diphenylamine solution Rl. 1032102. White or almost white, crystalline powder.
A 10 giL solution in sulfurie aeid R. The solution is mp: about 66 oc.
colourless. Diphenyloxazole. C1sHllNO. (Mr 221.3). 1032700. [92-71-7].
Diphenylamine solution R2. 1032103. 2,5-Diphenyloxazole.
Dissolve 1 g of diphenylamine R in 100 mL of glacial aeetie White or almost white powder, practically insoluble in water,
acid R and add 2.75 mL of sulfuric aeid R. Use immediately. soluble in methanol, sparingly soluble in dioxan and in glacial
acetic acid.
Diphenylanthracene. C 26 H J8 . (Mr 330.4). 1032200. mp: about 70 oc.
[1499-10-1]. 9,10- Diphenylanthracene.
Ai~rn: about 1260 determined at 305 nm in methanol R.
Yellowish or yellow, crystalline powder, practically insoluble
in water. Diphenyloxazole used for liquid scintillation is of a suitable
mp: about 248 oc.
analytical grade.
Diphenylphenylene oxide polymer. 1032800.
Diphenylbenzidine. C24H20N2' (Mr 336.4). 1032300.
2,6-Diphenyl-p-phenylene oxide polymer.
[531-91-9]. N,N'- Diphenylbenzidine. N,N'-Diphenylbiphenyl-
4,4' -diamine. White or almost white, porous beads. The size range of the
White or faintly grey, crystalline powder, practically insoluble beads is specified after the name of the reagent in the tests
in water, slightly soluble in acetone and in ethanol (96 per where it is used.
cent). Diphosphorus pentoxide. pps' (Mr 141.9). 1032900.
mp: about 248 oc. [1314-56-3]. Phosphorus pentoxide. Phosphoric anhydride.
Nitrates. Dissolve 8 mg in a cooled mixture of 5 mL of water R White or almost white powder, amorphou5, deliquescent. It i5
and 45 mL of nitrogen-free sulfurie aeid R. The solution is hydrated by water with the evolution of heat.
colourless or very pale blue. Storage: in an airtight container.
Sulfated ash (2.4.14): maximum 0.1 per cent.
Dipotassium hydrogen phosphateo K2HP0 4 • (Mr 174.2).
Storage: protected from light.
1033000. [7758-11-4].
Diphenylboric add aminoethyl estero C14H 16BNO. White or almost white, crystalline powder, hygroscopic, very
(Mr 225.1). 1032400. [524-95-8]. soluble in water, slightly soluble in ethanol (96 per cent).
White or slightly yellow, crystalline powder, practically Storage: in an airtight container.
insoluble in water, soluble in ethanol (96 per cent).
mp: about 193 oc. Dipotassium hydrogen phosphate trihydrateo
K2HP0 4,3H 20. (Mr 228.2).1157600. [16788-57-1].
Diphenylcarbazide. C J3 H J4 Np. (Mr 242.3). 1032500. Colourless or white or almost white powder or crystals, freeIy
[140-22-7]. 1,5-Diphenylcarbonodihydrazide. soluble in water.
White or almost white, crystalline powder which gradually
becomes pink on exposure to air, very slightly soluble in water, Dipotassium sulfate. K2 S0 4 . (Mr 174.3). 1033100.
soluble in acetone, in ethanol (96 per cent) and in glacial [7778-80-5].
acetic acid. Colourless crystals, soluble in water.
mp: about 170 oc. 2,2'-Dipyridylamine. C lO H 9N3' (Mr 171.2). 1157700.
Sulfated ash (2.4.14): maximum 0.1 per cent. [1202-34- 2]. N-(Pyridin -2-yl)pyridin-2-amine.
Storage: protected from light. mp: about 95 oc.
Diphenykarbazide solution. 1032501. Disodium arsenate. Na 2HAs0 4,7H 20. (Mr 312.0). 1102500.
Dissolve 0.2 g of diphenylearbazíde R in 10 mL of glacial [10048-95-0]. Disodium hydrogen arsenate heptahydrate.
acetic acid R and dilute to 100 mL with anhydrous Dibasic sodium arsenate.
ethanol R. Prepare immediately before use. Crystals, efflorescent in warm air, freely soluble in water,
Diphenylcarbazone. C13H12N'IO. (Mr 240.3). 1032600. soluble in glycerol, slightly soluble in ethanol (96 per cent).
[538-62-5]. 1,5-Diphenylcarbazone. The aqueous solution is alcaline to litmus.
Orange-yellow, crystalline powder, practically insoluble in d~g: about 1.87.
water, freely soluble in ethanol (96 per cent). mp: about 57 oC when rapidly heated.
mp: about 157 oC, with decomposition. Disodium bicinchoninate. C2oHlON2Naz04'
Diphenylcarbazone mercuric reagent. 1032601. (M, 388.3). 1126600. [979-88-4]. Disodium
2,2' -biquinoline-4-4' -dicarboxylate.
Solution A. Dissolve 0.1 g of diphenylcarbazone R in
anhydrous ethanol R and dilute to 50 mL with the same Disodium hydrogen citrate. C6H6NaP7,l1/2H20. (Mr 263.1).
solvent. 1033200. [144-33-2]. Sodium acid citrate. Disodium hydrogen
Solutíon B. Dissolve 1 g of mercuric chloride R in anhydrous 2-hydroxypropane-1 ,2,3-tricarboxylate sesquihydrate.
ethanol R and dilute to 50 mL with the same solvent. White or almost white powder, soluble in less than 2 parts of
Mix equal volumes of the two solutions. water, practically insoluble in ethanol (96 per cent).

460 See the information section on general monographs (caver pages)


EUROPEAN PHARMACOPOEIA 8.0 4.1.1. Reagents

Disodium hydrogen phosphate. 1033300. [10039-32-4]. water R and 10 mL of a 200 giL solution of hydroxylamine
See Disodium phosphate dodecahydrate (0118). hydrochloride R. Titrate with the dithizone solution; after
each addition, shake the mixture twenty times and towards
Disodium hydrogen phosphate solution. 1033301. the end of the titration allow to separate and discard
A 90 giL solution. the chloroform layer. Titrate until a bluish-green colour
is obtained. Calculate the equivalent in micrograms of
Disodium hydrogen phosphate, anhydrous. Na2HP0 4 • mercury per millilitre of the dithizone solution from the
(Mr 142.0). 1033400. [7558-79-4]. expression 201V, where Vis the volume in millilitres of the
Disodium hydrogen phosphate dihydrate. 1033500. dithizone solution used in the titration.
[10028-24-7]. Dithizone Rl. C13H12N4S, (M, 256.3). 1105500. [60-10-6].
See Disodium phosphate dihydrate (0602). 1,5-Diphenylthiocarbazone.
Disodium tetraborate. 1033600. [1303-96-4]. Content: minimum 98.0 per cent.
See Borax (0013). Bluish-black, brownish-black or black powder, practically
insoluble in water, soluble in ethanol (96 per cent).
Borate solution. 1033601.
Storage: protected from light.
Dissolve 9.55 g of disodium tetraborate R in sulfuric acid R,
heating on a water-bath, and dilute to 1 L with the same Divanadium pentoxide. V 20 S' (M, 181.9). 1034000.
acid. [1314-62-1]. Vanadic anhydride.
Content: minimum 98.5 per cent.
Ditalimphos. C 12 H 14 N0 4 PS. (M, 299.3). 1126700.
[5131-24-8]. O,O-Diethyl (1,3-dihydro-1,3-dioxo-2H- Yellow-brown Ol" rust-brown powder, slightly soluble in water,
isoindol-2-yl)phosphonothioate. soluble in strong mineral acids and in solutions of alkali
Very slightly soluble in water, in ethyl acetate and in anhydrous hydroxides with formation of salts.
ethano!. Appearance oi solution. Heat 1 g for 30 min with 10 mL of
A suitable certified reference solution may be used. sulfuric acid R. Allow to cool and dilute to 10 mL with the
same acid. The solution is clear (2.2.1).
S,S'-Dithiobis(2-nitrobenzoic acid). C14HsNzÜsS2' Sensitivity to hydrogen peroxide. Dilute 1.0 mL of the solution
(Mr 396.4). 1097300. [69-78-3]. 3-Carboxy-4- prepared for the test for appearance of solution cautiously to
nitrophenyldisulfide. Ellman's reagent. DTNB. 50.0 mL with water R. To 0.5 mL ofthe solution add 0.1 mL
Yellow powder sparingly soluble in ethanol (96 per cent). of a solution of hydrogen peroxide R (0.1 giL of H 2 0z). The
mp: about 242 oc. solution has a distinct orange colour compared with a blank
prepared from 0.5 mL of the solution to be examined and
DithioL C 7H sS2 • (Me 156.3). 1033800. [496-74-2]. 0.1 mL of water R. After the addition of 0.4 mL of hydrogen
Toluene-3,4-dithio!. 4-Methylbenzene-1,2-dithiol. peroxide solution (0.1 giL H 20 2), the orange solution becomes
White or almost white crystals, hygroscopic, soluble in orange-yellow.
methanol and in solutions of alkali hydroxides. Loss on ignition: maximum 1.0 per cent, determined on 1.00 g
mp: about 30 oc. at 700 ± 50 oc.
Storage: in an airtight container. Assay. Dissolve 0.200 g with heating in 20 mL of a 70 per
cent m/m solution of sulfuric acid R. Add 100 mL of water R
Dithiol reagent. 1033801. and 0.02 M potassium permanganate until a reddish colour is
To 1 g of dithiol R add 2 mL of thioglycollic acid R and dilute obtained. Decolorise the excess of potassium permanganate
to 250 mL with a 20 giL solution of sodium hydroxide R. by the addition of a 30 giL solution of sodium nitrite R. Add
Prepare immediate!y before use. 5 g of urea R and 80 mL of a 70 per cent m/m solution of
Dithiothreitol. C 4H lO 0 2S2. (M, 154.2). 1098200. sulfuric acid R. Coo!. Using 0.1 mL of ferro in R as indicator,
[27565-41-9]. threo-l,4- Dimercaptobutane-2,3-diol. titrate the solution immediately with 0.1 M ferrous sulfate
until a greenish-red colour is obtained.
Slightly hygroscopic needles, freely soluble in water, in acetone
and in anhydrous ethano!. 1 mL of 0.1 M ferrous sulfate is equivalent to 9.095 mg ofV 20 S '
Storage: in an airtight container. Divanadium pentoxide solution in sulfuric add.
1034001.
Dithizone. C 13 H 12 N 4S. (Me 256.3). 1033900. [60-10-6].
1,5-Diphenylthiocarbazone. Dissolve 0.2 g of divanadium pentoxide R in 4 mL of
sulfuric acid R and dilute to 100 mL with water R.
A bluish-black, brownish-black or black powder, practically
insoluble in water, soluble in ethanol (96 per cent). Docosahexaenoic acid methyl estero C 2J H 3P2' (Me 342.5).
Storage: protected from light. 1142800. [301-01-9]. DHA methyl estero Cervonic acid
methyl ester. (all- Z)- Docosa -4,7,10,13,16,19-hexaenoic acid
Dithizone solution. 1033901. methyl estero
A 0.5 giL solution in chloroform R. Prepare immediate!y Content: minimum 90.0 per cent, determined by gas
before use. chromatography.
Dithizone solution R2. 1033903. Docusate sodium. 1034100. [577-11-7].
Dissolve 40.0 mg of dithizone R in chloroform R and dilute See Docusate sodium (1418).
to 1000.0 mL with the same solvent. Dilute 30.0 mL of the
solution to 100.0 mL with chloroform R. Dodecyltrimethylammonium bromide. C 1s H 34 BrN.
Assay. Dissolve a quantity of mercuric chloride R equivalent (M, 308.4). 1135500. [1119-94-4]. N,N,N- Trimethyldodecan-
to 0.1354 g of HgC12 in a mixture of equal volumes of di/u te 1-aminium bromide.
sulfuric acid R and water R and dilute to 100.0 mL with the White or almost white crystals.
same mixture of solvents. Dilute 2.0 mL of this solution mp: about 246 oc.
to 100.0 mL with a mixture of equal volumes of di/u te
sulfuric acid R and water R. (This solution contains 20 ppm D-Dopa. C9 H 11 N0 4. (M, 197.2). 1164100. [5796-17-8].
of Hg). Transfer 1.0 mL of the solution to a separating (2R) - 2 -Amino-3 -( 3 ,4-dihydroxyphenyl)propanoic acid.
funnel and add 50 mL of dilute sulfuric acid R, 140 mL of 3-Hydroxy-D-tyrosine. 3,4-Dihydroxy-D-phenylaJanine.

General Notices (1) apply to all monographs and other texts 461
4.1.1. Reagents EUROPEAN PHARMACOPOEIA 8.0

[o: 1~o: + 9.5 to + 11.5, determined on a 10 giL solution in 1 M p- Endosulfan. C9H6Cl603S, (M, 406.9). 1126900.
hydrochloric acid. [33213-65-9].
mp: about 277 oc. bp: about 390 oc.
mp: about 207 oc.
Dotriacontane. C 32 H 66 . (M, 450.9). 1034200. [544-85-4].
A suitable certified reference solution (10 ngl flL in
n- Dotriacontane.
cyclohexane) may be used.
White or almost white plates, practically insoluble in water,
sparingly soluble in hexane. Endrin. C j2 H SCI60. (M, 380.9). 1127000. [72-20-8].
mp: about 69 oc. A suitable certified reference solution (10 ng/flL in
cyclohexane) may be used.
Impurities. Not more than 0.1 per cent of impurities with
the same t R value as a-tocopherol acetate, determined by the Erucamide. C22 H 43 NO. (M, 337.6). 1034500. [112-84-5].
gas chromatographic method prescribed in the monograph (Z)- Docos-13-enoamide.
(X- Tocopherol acetate (0439).
Yellowish or white powder or granules, practically insoluble
Doxycydine. 1145800. in water, very soluble in methylene chloride, soluble in
anhydrous ethanol.
See Doxycycline monohydrate (0820).
mp: about 70 oc.
Echinacoside. C3sH46020' (M, 786.5). 1159400. [82854-37-3]. Erythritol. 1113800. [149-32-6].
~-( 3',4' -Dihydroxyphenyl)-ethyl-O-a -L- rhamnopyranosyl
(1-¿3)-O-~-D- [~- D-glucopyranosyl( 1-¿6) ]-( 4-0-caffeoyl)-
See Erythritol (1803).
glucopyranoside. Esculetin. C9H604' (Mr 178.1). 1185800. [305-01-1].
Pale yellow powder, odourless. 6,7 -Dihydroxy-2H-l-benzopyran-2-one. Aesculetin.

Edotreotide. C6SH92Nj4018S2' (M, 1422). 1182400. Esculin. ClsH1609,11/2Hp. (M, 367.3). 1119400. [531-75-9].
[204318-14-9]. N- [[ 4,7,10-Tris( carboxymethyl)-1,4,7,10- 6- (~- D-Glucopyranosyloxy)-7 -hydroxy- 2H-chromen -2-one.
tetraazacyclododecan-l-yl] acetyl]-D-phenylalanyl-L-cysteinyl- White or almost white powder or colourless crystals, sparingly
L-tyrosyl-D-tryptophyl-L-lysyl-L-threonyl- N - [( 1R,2R) - 2- soluble in water and in ethanol (96 per cent), freely soluble in
hydroxy-1- (hydroxymethyl)propyl]-L-cysteinamide cyclic hot water and in hot ethanol (96 per cent).
(H 7)-disulfide. DOTATOC. DOTA -[Tyr 3 ]-octreotide. Chromatography (2.2.27). Thin-layer chromatography (2.2.27)
White or almost white powder. as prescribed in the monograph Eleutherococcus (1419); the
Content: minimum 95.0 per cent. chromatogram shows only one principal spot.

Electrolyte reagent for the micro determination of water. Estradiol. C 1sH 2P2' (M, 272.4). 1135600. [50-28-2].
1113700.
Estra-1,3,5( 10)-triene-3,17~-diol. ~- Estradiol.
Prisms stable in air, practically insoluble in water, freely
Commercially available anhydrous reagent or a combination
soluble in ethanol (96 per cent), soluble in acetone and in
of anhydrous reagents for the coulometric titration of water,
dioxane, sparingly soluble in vegetable oils.
containing suitable organic bases, sulfur dioxide and iodide
dissolved in a suitable solvent. mp: 173 oC to 179 oc.

Elementary standard solution for atomic spectrometry 17a-EstradioL ClsH2402' (M, 272.4). 1034600. [57-91-0].
(LOOO giL). 5004000. White or almost white, crystalline powder or colourless
This solution is prepared, generally in acid conditions, from crystals.
the element or a salt of the element whose minimum content mp: 220 oC to 223 oc.
is not less than 99.0 per cent. The quantity per litre of solution
Estragole. ClOHIP. (M, 148.2). 1034700. [140-67-0].
is greater than 0.995 g throughout the guaranteed period, as
1-Methoxy-4-prop-2-enylbenzene.
long as the vial has not been opened. The starting material
(element or salt) and the characteristics of the final solvent Liquid, miscible with ethanol (96 per cent).
(nature and acidity, etc.) are mentioned on the label. n6°: about 1.52.
bp: about 216 oc.
Emetine dihydrochloride. 1034300. [316-42-7].
Es trago le used in gas chromatography complies with the
See Emetine hydrochloride pentahydrate (0081). following test.
Emodin. C1sH100S' (Mr 270.2). 1034400. [518-82-1]. Assay. Gas chromatography (2.2.28) as prescribed in the
1,3,8-Trihydroxy -6-methylanthraquinone. monograph Anise oil (0804).
Orange-red needles, practically insoluble in water, soluble in Test solution. The substance to be examined.
ethanol (96 per cent) and in solutions of alkali hydroxides. Content: minimum 98.0 per cent, calculated by the
Chromatography. Thin-Iayer chromatography (2.2.27) normalisation procedure.
as prescribed in the monograph Rhubarb (0291); the Ethanol. 1034800. [64-17-5].
chromatogram shows only one principal spot.
See Ethanol, anhydrous R.
Endoprotease LysC. 1173200. Ethanol, anhydrous. 1034800. [64-17-5].
Microbial extracellular proteolytic enzyme secreted by See Ethanol, anhydrous (1318).
Achromobacter lyticus. A lyophilised powder, free of salts.
Ethanol Rl. 103480l.
a-Endosulfan. C9H 6CI60 3S. (Mr 406.9). 1126800. [959-98-8].
Complies with the requirements prescribed for the
bp: about 200 oc. monograph Ethanol, anhydrous (1318) with the following
mp: about 108 oc. additional requirement.
A suitable certified reference solution (10 ngl flL in Methanol. Gas chromatography (2.2.28).
cyclohexane) may be used. Test solution. The substance to be examined.

462 See the information section on general monographs (ca ver pages)
EUROPEAN PHARMACOPOEIA 8.0 4.1.1. Reagents

Reference solution. Dilute 0.50 mL of anhydrous methanol R Storage: in an airtight container, protected from light, at a
to 100.0 mL with the substance to be examined. Dilute temperature not exceeding 15 oc.
LO mL of this solution to 100.0 mL with the substance to
be examined. Ether, peroxide-free. 1035100.
Column: See Anaesthetic ether (0367).
- material: glass; Ethion. C9H2P4P2S4' (M, 384.5). 1127100. [563-12-2].
- size: l '" 2 m, 0 '" 2 mm; mp: - 24 oC to - 25 oc.
- stationary phase: ethylvinylbenzene-divinylbenzene A suitable certified reference solution (lO ng/¡.tL in
copolymer R (75-100 flm). cyclohexane) may be used.
Carrier gas: nitrogen for chromatography R.
Ethoxychrysoidine hydrochloride. C¡4H17ClNp. (M, 292.8).
Flow rate: 30 mLlmin. 1035200. [2313-87-3]. 4-[( 4-Ethoxyphenyl)diazenyl]phenyl-
Temperature: ene-1,3-diamine hydrochloride.
- column: 130 oC; Reddish powder, soluble in ethanol (96 per cent).
- injection port: 150 oC;
Ethoxychrysoidine solution. 1035201.
- detector: 200 oc.
A 1 giL solution in ethanol (96 per cent) R.
Detection: flame- ionisation.
Test for sensitivity. To a mixture of 5 mL of dilute
Injection: 1 flL of the test solution and 1 flL of the reference hydroehloríe acid R and 0.05 mL of the ethoxy-chrysoidine
solution, alternately, three times. solution add 0.05 mL of 0.0167 M bromide-bromate. The
After each chromatography, heat the column to 230 oC colour changes from red to light yellow within 2 mino
for 8 mino Integrate the methanol peak. Calculate
the percentage methanol content from the following Ethyl acetate. C4HsÜ2' (M, 88.1). 1035300. [141-78-6].
expression: Clear, colourIess liquid, soluble in water, miscible with ethanol
a xb (96 per cent).
c-b d§g: 0.901 to 0.904.
bp: 76 oC to 78 oc.
a percentage V/V content of methanol in the Ethyl acetate, treaíed. 1035301.
reference solution,
Disperse 200 g of sulfamic acid R in ethyl acetate R and
b area of the methanol peak in the chromatogram make up to 1000 mL with the same solvent. Stir the
obtained with the test solution, suspension obtained for three days and filter through a
e area of the methanol peak in the chromatogram filter papero
obtained with the reference solution. Storage: use within 1 month.
Limit:
Ethyl acrylate. C S H S0 2. (Mr 100.1). 1035400. [140-88-5].
- methanol: maximum 0.005 per cent V/V. Ethyl prop-2-enoate.
Ethanol (96 per cent). 1002500. [64-17-5]. Colourless liquido
See Ethanol (96 per cent) (1317). d~g : about 0.924.
n~o : about 1.406.
Ethanol (x per cenl V/V). 1002502.
bp: about 99 oc.
Mix appropriate volumes of water R and ethanol (96 per
cent) R, allowing for the effects of warming and volume mp: about - 71°C.
contraction inherent to the preparation of such a mixture, 4-[(Ethylamino)methyl]pyridine. CSH¡2N2' (M, 136.2).
to obtain a solution whose final content of ethanol 1101300. [33403-97-3].
corresponds to the value of X.
Pale yellow liquido
Ethanolamine. C2H 7 NO. (M, 61.1). 1034900. [141-43-5]. d~g: about 0.98.
2-Aminoethanol. n~o: about 1.516.
Clear, colourless, viscous, hygroscopic liquid, miscible with bp: about 98 oc.
water and with methanol.
d§g: about 1.04. Ethylbenzene. CSHlO" (M, 106.2). 1035800. [100-41-4].
n~o : about 1.454. Content: minimum 99.5 per cent m/m, determined by gas
mp: about 11°C. chromatography.
Storage: in an airtight container. Clear, colourless liquid, practically insoluble in water, soluble
in acetone, and in ethanol (96 per cent).
Ether. C 4 HlQO. (M, 74.1). 1035000. [60-29-7]. d~g: about 0.87.
Clear, colourless, volatile and very mobile liquid, very n~o : about 1.496.
flammable, hygroscopic, soluble in water, miscible with
bp: about 135 oc.
ethanol (96 per cent).
d~g: 0.713 to 0.715. Ethyl benzoate. C 9H¡aÜ2' (M, 150.2). 1135700. [93-89-0].
bp: 34 oC to 35 oc. A clear, colourless, refractive liquid, practically insoluble
Do not distil if the ether does not comply with the test for in water, miscible with ethanol (96 per cent) and with light
peroxides. petroleum.
Peroxides. Place 8 mL of potassium iodide and starch solution R d 245 : about 1.050.
in a 12 mL ground-glass-stoppered cylinder about 1.5 cm in n ~ : about 1.506.
d¡ameter. Fill completely with the substance to be examined, bp: 211 oC to 213 oc.
shake vigorously and allow to stand in the dark for 30 mino
No colour is produced. EthyI5-bromovalerate. C7H13Br02' (Mr 209.1). 1142900.
The name and concentration of any added stabilisers are [14660-52-7]. Ethy15-bromopentanoate.
stated on the labe!' Clear, colourless liquido

General Notiees (1) apply to all monographs and other texts 463
4.1.1. Reagents EUROPEAN PHARMACOPOEIA 8.0

d~g: about 1.321. Clear, co1ourless liquid, miscible with water, with acetone and
bp: 104 oC to 109 oc. with ethano1 (96 per cent).
d~g: about 0.97.
Ethyl cyanoacetate. C SH 7N0 2 • (Mr 113.1). 1035500.
n~o : about 1.403.
[105-56-6].
bp: about 125 oc.
Colourless or pale yellow liquid, slightly soluble in water,
miscible with ethanol (96 per cent). Ethylene oxide. C2H 4 0. (M, 44.05). 1036400. [75-21-8].
bp: 205 oC to 209 oC, with decomposition. Oxirane.
Colourless, flammable gas, very soluble in water and in
Ethylene chloride. C 2H 4 C12. (Mr 99.0). 1036000. [107-06-2]. anhydrous ethanol.
1,2-Dichloroethane.
Liquefaction point: about 12 oc.
Clear, colourless liquid, soluble in about 120 parts of water
and in 2 parts of ethanol (96 per cent). Ethylene oxide solution. 1036402.
d~g : about 1.25. Weigh a quantity of coo1 ethylene oxide stock solution R
Distillation range (2.2.11). Not less than 95 per cent distils equivaIent to 2.5 mg of ethylene oxide into a co01 flask and
between 82 oC and 84 oc. dilute to 50.0 g with macrogol 200 R1. Mix well and dilute
2.5 g of this solution to 25.0 mL with macrogol 200 R1
Ethylenediamine. C2H sN 2• (Mr 60.1). 1036500. [107-15-3]. (5 [1g of ethylene oxide per gram of solution). Prepare
Ethane-l,2-diamine. immediately before use.
Clear, colourless, fuming liquid, strongly alkaline, miscible The solution can be prepared using commercially availab1e
with water and with ethanol (96 per cent). reagents instead of ethylene oxide stock solution R, making
bp: about 116 oc. appropriate dilutions.

Ethylene bis [3,3-di( 3- tert- butyl-4-hydroxyphenyl) buty- Ethylene oxide solution Rl. 1036403.
rate]. 1035900. [32509-66-3]. Dilute 1.0 mL of cooled ethylene oxide stock solution R
See ethylene bis[3,3-di(3-(1,l-dimethylethyl)-4-
(check the exact volume by weighing) to 50.0 mL with
macrogol 200 Rl. Mix well and dilute 2.5 g of this solution
hydroxyphenyl)butyrate} R.
to 25.0 mL with macrogol200 R1. Calculate the exact
Ethylene bis [3,3-di(3-( 1,1-dimethylethyl)-4- amount of ethylene oxide in parts per million from the
hydroxyphenyl)butyrate]. CSOH660S' (Mr 795). 1035900. volume determined by weighing and taking the relative
[32509-66-3]. Ethylene bis[3,3-di(3-tert-butyl-4- density of macrogol200 R1 as 1.127. Prepare immediately
hydroxyphenyl)butyrate ]. before use.
Crystalline powder, practically insoluble in water and in light The solution can be prepared using commercially available
petroleum, very soluble in acetone and in methanol. reagents instead of ethylene oxide stock solution R, making
mp: about 165 oc. appropriate dilutions.
Ethylene oxide solution R2. 1036404.
(Ethylenedinitrilo)tetra-acetic add. ClOH16N20s'
(M, 292.2). 1105800. [60-00-4]. N,N'-1,2-Ethanediylbis[N- Weigh 1.00 g of cold ethylene oxide stock solution R
(carboxymethyl)glycine]. Edetic acid. (equivalent to 2.5 mg of ethylene oxide) into a cold flask
containing 40.0 g of cold m acrogo 1200 R1. Mix and
White or almost white crystalline powder, very slightly soluble
determine the exact mass and dilute to a calculated mass
in water.
to obtain a solution containing 50 [1g of ethylene oxide per
mp: about 250 oC, with decomposition. gram of solution. Weigh 10.00 g into a flask containing
Ethylene glycol. C2H 60 2. (Mr 62.1). 1036100. [107-21-1]. about 30 mL of water R, mix and dilute to 50.0 mL with
Ethane-1,2-diol. water R (lO [1g/mL of ethylene oxide). Prepare immediately
before use.
Content: minimum 99.0 per cent.
The solution can be prepared using commercially available
Colourless, slightly viscous liquid, hygroscopic, miscible with reagents instead of ethylene oxide stock solution R, making
water and with ethanol (96 per cent). appropriate dilutions.
d~g: 1.113 to 1.115.
Ethylene oxide solution R3. 1036405.
n~o : about 1.432.
Dilute 10.0 mL of ethylene oxide solution R2 to 50.0 mL with
bp: about 198 oc.
water R (2 [1g/mL of ethylene oxide). Prepare immediately
mp: about - 12 oc. befare use.
AcidUy. To 10 mL add 20 mL of water R and 1 mL of
phenolphthalein solution R. Not more than 0.15 mL of 0.02 M Ethylene oxide solution R4. 1036407.
sodium hydroxide is required to change the colour of the Dilute 1.0 mL of ethylene oxide stock solution R1 to
indicator to pink. 100.0 mL with water R. Dilute 1.0 mL of this solution to
25.0 mL with water R.
Water (2.5.12): maximum 0.2 per cent
Ethylene oxide stock solution. 1036401.
Ethylene glycol monoethyl ether. C4H lO ü 2 • (Mr 90.1).
1036200. [110-80-5]. 2-Ethoxyethanol. All operations carried out in the preparation of these
solutions must be conducted in a fume cupboard. The
Content: minimum 99.0 per cent. operator must protect both hands and face by wearing
CIear, colourless liquid, miscible with water, with acetone and polyethylene protective g/oves and an appropriate face mask.
with ethanol (96 per cent). Store all solutions in an airtight container in a refrigerator
d~g: about 0.93. at 4 oC to 8 oc. Carry out all determinations three times.
n~5 : about 1.406. 1nto a dry, clean test-tube, cooled in a mixture of 1 part of
bp: about 135 oc. sodium chloride R and 3 parts of crushed ice, introduce a
slow current of ethylene oxide R gas, allowing condensation
Ethylene glycol monomethyl ether. C3HsÜ2' (M, 76.1). onto the inner wall of the test -tube. Using a glass
1036300. [109-86-4]. 2-Methoxyethanol. syringe, previously cooled to - 10 oC, inject about 300 [1L
Content: minimum 99.0 per cent. (corresponding to about 0.25 g) ofliquid ethylene oxide R

464 See the information section on general monographs (cover pages)


EUROPEAN PHARMACOPOEIA 8.0 4.1.1. Reagents

into 50 mL of macrogol 200 R1. Determine the absorbed EthyI4-hydroxybenzoate. 1035700. [120-47-8].
quantity of ethylene oxide by weighing before and after See Ethyl parahydroxybenzaate R.
absorption (Meo)' Dilute to 100.0 mL with m acrogo 1200 Rl.
Mix well before use. N-Ethylmaleimide. C6H 7N0 2 • (MI 125.1). 1036700.
Assay. To 10 mL of a 500 giL suspension of magnesium [128-53-0]. 1-Ethyl-1H-pyrrole-2,5-dione.
chloride R in anhydrous ethanol R add 20.0 mL of 0.1 M Colourless crystals, sparingly soluble in water, freely soluble
alcoholic hydrochloric acid in a flask. Stopper and shake to in ethanol (96 per cent).
obtain a saturated solution and allow to stand overnight to mp: 41 oC to 45 oc.
equilibrate. Weigh 5.00 g of ethylene oxide stock solution Storage: at a temperature of 2 oC to 8 oc.
(2.5 giL) into the flask and allow to stand for 30 mino Titrate
with 0.1 M alcoholic potassium hydroxide determining the Ethyl methanesulfonate. C3Hs03S, (M¡ 124.2). 1179300.
end-point potentiometrically (2.2.20). [62-50-0].
Carry out a blank titration, replacing the substance to be Clear, colourless liquido
examined with the same quantity of macrogol 200 Rl. Cantent: minimum 99.0 per cent.
Ethylene oxide content in milligrams per gram is given by: Density: about 1.206 g/cm 3 (20 OC).
(Vo - V1 ) x f x 4.404 n5°: about 1.418.
m bp: about 213 oc.

volumes of 0.1 M alcoholic patassium Ethyl methyl ketone. 1054100. [78-93-3].


hydroxide used respectively for the blank See methyl ethyl ketone R.
titration and the assay,
2-Ethyl-2-methylsuccinic acid. C7H¡zÜ4' (Mr 160.2).
f factor of the alcoholic potassium hydroxide 1036800. [631-31-2]. 2-Ethyl-2-methylbutanedioic acid.
solution,
mp: 104 oC to 107 oc.
m mass of the sample taken (g).
Ethyl parahydroxybenzoate. 1035700. [120-47-8].
Ethylene oxide stock solution Rl. 1036406. See Ethyl parahydraxybenzaate (0900).
A 50 giL solution of ethylene oxide R in methanol R.
2-Ethylpyridine. C7H9N. (Mr 107.2). 1133400. [100-7l-0].
Either use a commercially available reagent or prepare the
Colourless or brownish liquido
solution corresponding to the aforementioned composition.
d~g: about 0.939.
Ethylene oxide stock solution R2. 1036408. n5° : about 1.496.
A 50 giL solution of ethylene oxide R in methylene bp: about 149 oc.
chloride R.
Either use a commercially available reagent or prepare the Ethylvinylbenzene-divinylbenzene copolymer. 1036900.
solution corresponding to the aforementioned composition. Porous, rigid, cross-linked polymer beads. Several grades are
available with different sizes of bead. The size range of the
Ethyl formateo C3H602' (MI 74.1). 1035600. [109-94-4]. Ethyl beads is specified after the name of the reagent in the tests
methanoate. where it is used.
CIear, colourless, flammable liquid, freely soluble in water,
miscible with ethanol (96 per cent). Ethylvinylbenzene-divinylbenzene copolymel' RL 1036901.
d~g: about 0.919. Porous, rigid, cross-linked polymer beads, with a nominal
n5° : about 1.36. specific surface area of 500 m 2 /g to 600 m 2 /g and having pores
with a mean diameter of 7.5 nm. Several grades are available
bp: about 54 oc. with different sizes of beads. The size range of the beads is
2-Ethylhexane-l,3-diol. CSH¡sÜ2' (M¡ 146.2). 1105900. specified after the name of the reagent in the tests where it
[94-96-2]. is used.
Slightly oily liquid, soluble in anhydrous ethanol, 2-propanol, Eugenol. C lO H¡zÜ2' (Me 164.2). 1037000. [97-53-0].
propylene glycol and castor oil. 4-Allyl-2-methoxyphenol.
d~g: about 0.942. Colourless or pale yellow, oily liquid, darkening on exposure
n5°: about 1.451. to air and light and becoming more viscous, practically
bp: about 244 oc. insoluble in water, miscible with ethanol (96 per cent) and
with fatty and essential oils.
2-Ethylhexanoic aeid. CSH¡P2' (M, 144.2). 1036600. d~g: about 1.07.
[149-57 -5].
bp: about 250 oc.
Colourless liquido
Eugenol used in gas chramatagraphy complies with the
d~g: about 0.91. follawing additional test.
n50: about 1.425. Assay. Gas chromatography (2.2.28) as prescribed in the
Related substances. Gas chromatography (2.2.28). monograph Clove oil (1091).
Injectian: 1 ¡.tL of the test solution. Test solutian. The substance to be examined.
Test solutian: suspend 0.2 g of the 2-ethylhexanoic acid in Cantent: minimum 98.0 per cent, calculated by the
5 mL of water R, add 3 mL of dilute hydrochlaric acid R and normalisation procedure.
5 mL of hexane R, shake for 1 min, allow the layers to separate Storage: protected from light.
and use the upper layer. Carry out the chromatographic
procedure as prescribed in the test for 2-ethylhexanoic acid in Euglobulins, bovine. 1037100.
the monograph on Amaxicillin sodium (0577). Use fresh bovine blood collected into an anticoagulant
Limit: the sum of the area of any peaks, apart from the solution (for example, sodium citrate solution). Discard any
principal peak and the peak due to the solvent, is not greater haemolysed blood. Centrifuge at 1500-1800 g at 15-20 oC to
than 2.5 per cent of the are a of the principal peak. obtain a supernatant plasma poor in platelets.

General Natices (1) apply ta all monagraphs and other texts 465
4.1.1. Reagents EUROPEAN PHARMACOPOEIA 8.0

To 1 L of bovine plasma add 75 g of barium sulfate R and in a water-bath at 37 oC introduce 0.1 mL of a solution of a
shake for 30 mino Centrifuge at not less than 1500-1800 g at reference preparation of streptokinase containing 10 IU of
15-20 oC and draw off the clear supernatant. Add 10 mL of a streptokinase activity per millilitre and 0.1 mL of a solution of
0.2 mg/mL solution of aprotinin R and shake to ensure mixing. human thrombin R containing 20 IU/mL. Add rapidly 1 mL
In a container with a minimum capacity of 30 L in a chamber of a solution containing 10 mg of human euglobulins per
at 4 oC introduce 25 L of distilled water R at 4 oC and add about millilitre. A firm clot forms in less than 10 S. Note the time
500 g of solid carbon dioxide. Immediately add, while stirring, that elapses between the addition of the solution of human
the supernatant obtained from the plasma. A white precipitate euglobulins and the lysis of the clot. The lysis time does not
is formed. Allow to settle at 4 oC for 10-15 h. Remove the clear exceed 15 mino
supernatant solution by siphoning. Collect the precipitate by Storage: in an airtight container at 4 oC; use within 1 year.
centrifuging at 4 oc. Suspend the precipitate by dispersing
mechanically in 500 mL of distilled water R at 4 oC, shake Factor VH-deficient plasma. 1185900.
for 5 min and collect the precipitate by centrifuging at 4 oc. Plasma that is deficient in factor VII.
Disperse the precipitate mechanically in 60 mL of a solution
containing 9 giL of sodium chloríde R and 0.9 giL sodium Factor Xa, bovine, coagulation. 1037300. [9002-05-5].
citrate R and adjust to pH 7.2-7.4 by adding a 10 giL solution An enzyme which converts prothrombin to thrombin. The
of sodium hydroxide R. Filter through a sintered glass filter semi-purified preparation is obtained from liquid bovine
(2.1.2); to facilitate the dissolution of the precipitate crush the plasma and it may be prepared by activation of the zymogen
particles of the precipitate with a suitable instrument. Wash factor X with a suitable activator such as Russell's viper venom.
the filter and the instrument with 40 mL of the chloride-citrate Storage: freeze-dried preparation at - 20 oC and frozen
solution described aboye and dilute to 100 mL with the same solution at a temperature lower than - 20 oc.
solution. Freeze-dry the solution. The yields are generally 6 g
to 8 g of euglobulins per litre of bovine plasma. Factor Xa solution, bovine. 1037301.
Test for suitability. For this test, prepare the solutions using Reconstitute as directed by the manufacturer and dilute
phosphate buffer solution pH 7.4 R containing 30 giL of bovine with tris(hydroxymethyl)aminomethane sodium chloride
albumin R. buffer solutíon pH 7.4 R.
lnto a test-tube 8 mm in diameter placed in a water-bath at Any change in the absorbance of the solution, measured at
37 oC introduce 0.2 mL of a solution of a reference preparation 405 nm (2.2.25) against trís(hydroxymethyl)aminomethane
of urokinase containing 100 IU ImL and 0.1 mL of a solution of sodium chloríde buffer solutíon pH 7.4 R and from which
human thrombin R containing 20 IU/mL. Add rapidly 0.5 mL the blank absorbance has been substracted, is not more
of a solution containing 10 mg of bovine euglobulins per than 0.20 per minute.
millilitre. A firm clot forms in less than 10 s. Note the time
Factor Xa solution, bovine Rl. 1037302.
that elapses between the addition of the solution of bovine
euglobulins and the lysis of the clot. The lysis time does not Reconstitute as directed by the manufacturer and dilute to
exceed 15 mino 1.4 nkat/mL with tris(hydroxymethyl)aminomethane-EDTA
buffer so/utíon pH 8.4 R.
Storage: protected from moisture at 4 oC; use within 1 year.
(E,E)-Farnesol. C 1s H 26 0. (Mr 222.4). 1161000. [106-28-5].
Euglobulins, human. 1037200.
trans, trans- Farnesol. (2E,6E) - 3, 7 ,11-Trimethyldodeca -2,6,1 0-
For the preparation, use fresh human blood collected into an trien -1-01.
anticoagulant solution (for example sodium citrate solution)
or human blood for transfusion that has been collected in Fast biue 13 salto C14H12C12N402' (M, 339.2). 1037400.
plastic blood bags and which has just reached its expiry date. [84633-94-3].
Discard any haemolysed blood. Centrifuge at 1500-1800 g Schultz No. 490.
at 15 oC to obtain a supernatant plasma poor in platelets. Colour Index No. 37235.
Iso-group plasmas may be mixed. 3,3' -Dimethoxy(biphenyl)-4,4' -bisdiazonium dichloride.
To 1 L of the plasma add 75 g of baríum sulfate R and shake Dark green powder, soluble in water. It is stabilised by
for 30 mino Centrifuge at not less than 15 000 g at 15 oC and addition of zinc chloride.
draw off the clear supernatant. Add 10 mL of a solution
of aprotinin R containing 0.2 mg/mL and shake to ensure Storage: in an airtight container, at a temperature between
mixing. In a container with a minimum capacity of 30 L in a 2 oC and 8 oc.
chamber at 4 oC introduce 25 L of distilled water R at 4 oC Fast red 13 salto C17H13NP9S2' (Mr 467.4). 1037500.
and add about 500 g of solid carbon dioxide. Immediately [49735-71-9].
add while stirring the supernatant obtained from the plasma.
Schultz No. 155.
A white precipitate is formed. Allow to settle at 4 oC for
10-15 h. Remove the clear supernatant solution by siphoning. Colour Index No. 37125.
Collect the precipitate by centrifuging at 4 oc. Suspend the 2-Methoxy-4-nitrobenzenediazonium hydrogen
precipitate by dispersing mechanically in 500 mL of distilled naphthalene-1,5-disulfonate.
water R at 4 oC, shake for 5 min and collect the precipitate by Orange-yellow powder, soluble in water, slightly soluble in
centrifuging at 4 oc. Disperse the precipitate mechanically in ethanol (96 per cent).
60 mL of a solution containing 9 giL of sodium chloride R and Storage: in an airtight container, protected from light, at 2 oC
0.9 giL of sodium citrate R, and adjust the pH to 7.2-7.4 by to 8 oc.
adding a 10 giL solution of sodium hydroxide R. Filter through
a sintered-glass filter (2.1.2); to facilitate the dissolution of the Fenchlorphos. C sH sCI 3 0 3 PS. (Mr 321.5). 1127200.
precipitate crush the particles of the precipitate with a suitable [299-84-3].
instrument. Wash the filter and the instrument with 40 mL mp: about 35 oc.
of the chloride-citrate solution described aboye and dilute to A suitable certified reference solution (lO ng/[lL in
100 mL with the same solution. Freeze-dry the solution. The cyclohexane) may be used.
yields are generally 6 g to 8 g of euglobulins per litre ofhuman
plasma. Fenchone. ClOH1P, (M, 152.2). 1037600. [7787-20-4].
Test for suitability. For this test, prepare the solutions using (lR)-1,3,3- Trimethylbicyclo[2.2.1]heptan-2-one.
phosphate buffer solution pH 7.2 R containing 30 giL of Oily liquid, miscible with ethanol (96 per cent), practically
bovine albumin R. lnto a test-tube 8 mm in diameter placed insoluble in water.

466 See the informatíon section on general monographs (ca ver pages)
EUROPEAN PHARMACOPOEIA 8.0 4. lo 1. Reagents

n~O: about 1.46. Ferric sulfate. Fe 2(S04)3,xHp. 1037900. [10028-22-5].


bP15mm: 192 oC to 194 oc. lron(III) trisulfate hydrated.
Fenchone used in gas chromatography complies with the Yellowish-white powder, very hygroscopic, decomposes in air,
following test. slightly soluble in water and in ethanol (96 per cent).
Assay. Gas chromatography (2.2.28) as prescribed in the Storage: in an airtight container, protected from light.
monograph Bitter fennel (0824). Ferrie sulfate solution. 1037901.
Test solution. The substance to be examined. Dissolve 50 g of ferric sulfate R in an excess of water R,
Content: minimum 98.0 per cent, calculated by the add 200 mL of sulfuric acid R and dilute to 1000 mL with
normalisation procedure. water R.

Fenvalerate. Czs H 22 CINO y (M, 419.9). 1127300. Ferric sulfate pentahydrate. Fe 2(S04)3,5Hp. (M, 489.9).
[51630-58-1]. 1153700. [142906-29-4].
bp: about 300 oc. White or yellowish powder.
A suitable certified reference solution (lO ng/flL in Ferroeyphene. CZ6H16FeN6' (M, 468.3). 1038000.
cyclohexane) may be used. [14768-11-7]. Dicyanobis(1,10-phenanthroline)iron(II).
Ferric ammonium sulfate. FeNH 4 (S04)2,12H 2 0. (M, 482.2). Violet-bronze, crystalline powder, practically insoluble in
1037700. [7783-83-7]. Ammonium iron di sulfate water and in ethanol (96 per cent).
dodecahydrate. Storage: protected from Iight and moisture.
Pale-violet crystals, efflorescent, very soluble in water, Ferroin. 1038100. [14634-91-4].
practically insoluble in ethanol (96 per cent).
Dissolve 0.7 g of ferrous sulfate R and 1.76 g of phenanthroline
Ferric ammonium sulfate solution R2. 1037702. hydrochloride R in 70 mL of water R and dilute to 100 mL
A 100 giL solution. If necessary filter before use. with the same solvent.
Test for sensítivity. To 50 mL of dilute sulfuric acid R add
Ferric ammonium sulfate solution R5. 1037704. 0.15 mL of osmium tetroxide solution R and 0.1 mL ofthe
Shake 30.0 g offerric ammonium sulfate R with 40 mL ferroin. After the addition of 0.1 mL of 0.1 M ammonium and
of nítric acid R and dilute to 100 mL with water R. If the cerium nitrate the colour changes from red to light bIue.
solution is turbid, centrifuge or filter it.
Ferrous ammonium sulfate. Fe(NH 4MS04)2,6H 20.
Storage: protected from light. (M¡ 392.2). 1038200. [7783-85-9]. Diammonium iron
disulfate hexahydrate.
Ferric ammonium sulfate solution R6. 1037705.
PaIe bluish-green crystals or granules, freely soluble in water,
Dissolve 20 g of ferric ammonium sulfate R in 75 mL of practically insoluble in ethanol (96 per cent).
water R, add 10 mL of a 2.8 per cent V/V solution of sulfuric
acid R and dilute to 100 mL with water R. Storage: protected from light.
Fermus sulfate. 1038300. [7782-63-0].
Ferrk chloride. FeCl 3,6Hp. (M, 270.3). 1037800.
[10025- 77 -1]. lron trichloride hexahydrate. See Ferrous sulfate heptahydrate (0083).
Yellowish-orange or brownish crystalline masses, deliquescent, Ferrous sulfate solution R2. 1038301.
very soluble in water, soluble in ethanol (96 per cent). On
Dissolve 0.45 g of ferrous sulfate R in 50 mL of 0.1 M
exposure to light, ferric chloride and its solutions are partIy
hydrochloric acid and dilute to 100 mL with carbon
reduced. dioxide-free water R. Prepare immediately before use.
Storage: in an airtight container.
Ferulic add. ClOHIO04' (M, 194.2). 1149500.
Ferric ehloride solution Rl. 1037801. [1135-24-6]. 4-Hydroxy-3-methoxycinnamic acid.
A 105 giL solution. 3-( 4- Hydroxy-3-methoxyphenyl)propenoic acid.
Faint yellow powder, freely soluble in methanol.
Ferric ehloride solution R2. 1037802.
mp: 172.9 oC to 173.9 oc.
A 13 giL solution.
Ferulic acid used in the assay of eleutherosides in
Ferrie chloride solution R3. 1037803. Eleutherococcus (1419) complies with the following additional
test.
Dissolve 2.0 g of ferric chloride R in anhydrous ethanol R
and dilute to 100.0 mL with the same solvento Assay. Liquid chromatography (2.2.29) as prescribed in the
monograph Eleutherococcus (1419).
Ferric chloride-ferricyanide-arsenite reagent. 1037805. Content: minimum 99 per cent, calculated by the
Immediately before use mix 10 mL of a 27 giL solution normalisation procedure.
of ferric chloride R in dilute hydrochloric acid R, 7 mL of
Fibrin bIue. 1101400.
potassium ferricyanide solution R, 3 mL of water R and
10 mL of sodium arsenite solution R. Mix 1.5 g of fibrin with 30 mL of a 5 giL solution of indigo
carmine R in 1 per cent V/V dilute hydrochloric acid R.
Ferric chloride-sulfamic add reagent. 1037804. Heat the mixture to 80 oC and maintain at this temperature
A solution containing 10 giL ofjerric chloride R and 16 giL whilst stirring for about 30 mino Allow to cool. Filter.
of sulfamic acid R. Wash extensively by resuspension in 1 per cent V/V dilute
hydrochloric acid R and mixing for about 30 min; filter. Repeat
Ferric nitraíe. Fe(N0 3 )3,9Hp. (M¡ 404). 1106100. the washing operation three times. Dry at 50 oc. Grind.
[7782-61-8].
Fibrin congo red. 1038400.
Content: minimum 99.0 per cent m/m ofFe(N0 3 )3,9Hp.
Take 1.5 g of fibrin and leave overnight in 50 mL of a 20 giL
Light-purple crystals or crystalline mass, very soluble in water. solution of congo red R in ethanol (90 per cent V/V) R. Filter,
Free acid: not more than 0.3 per cent (as HN0 3 ). rinse the fibrin with water R and store under ether R.

General Notices (1) apply to all monographs and other texts 467
4. L 1. Reagents EUROPEAN PHARMACOPOEIA 8.0

Fibrinogen. 1038500. [9001-32-5]. DL-6-Fluorodopa hydrochloride. C9 H ll ClFN0 4 •


See Human fibrinogen, freeze-dried (0024). (Mr 251.6). 1169200. (2RS)-2-Amino-3-(2-fluoro-
4,5-dihydroxyphenyl)propanoic acid hydrochloride.
Fixing solution. 1122600. 2-Fluoro- 5-hydroxy- DL- tyrosine hydrochloride.
To 250 mi of methanol R, add 0.27 mi of formaldehyde R and White or almost white powder.
dilute to 500.0 mi with water R.
6-Fluorolevodopa hydroehloride. C 9H 11 CIFN0 4 • (M, 251.6).
Fixing solution for isoelectric focusing in polyacrylamide 1169300. [144334-59-8]. (2S)-2-Amino-3-(2-fluoro-
geL 1138700. 4,5-dihydroxyphenyl)propanoic acid hydrochloride.
2-Fluoro- 5-hydroxy- L-tyrosine hydrochloride.
A solution containing 35 g of sulfosalicylic acid R and 100 g of
trichloroacetíc acid R per litre of water R. Colourless or almost colourless solid, soluble in water.
Fluoromisonidazole. C6HsFl'''¡30y (Mr 189.1). 1186000.
Flufenamic add. C14HlOF3NOz. (M, 281.2). 1106200.
[13551-89-8]. (2RS)-1- Fluoro-3-(2-nitro-lH-imidazol-l-
[530-78-9]. 2- [[3-(Trifluoromethyl)phenyl] amino ]benzoic
yl)propan-2-ol. FMISO.
acid.
Content: minimum 95 per cent.
Pale yellow, crystalline powder or needles, practically insoluble
in water, freely soluble in ethanol (96 per cent). Yellow crystals.
mp: 132 oC to 135 oc. 1- Fluoro- 2-nitro-4- (trifluoromethyl) benzene. C7H3F 4N02'
(M r 209.1). 1038900. [367-86-2].
Flumazenil. 1149600. [78755-81-4]. mp: about 197 oc.
See Flumazenil (1326).
Folie acid. 1039000. [75708-92-8].
Flunitrazepam. 1153800. [1622-62-4].

-
See Folie acid (0067).
See Flunitrazepam (0717).
Formaldehyde. 1039100. [50-00-0].
See Formaldehyde solution R.
Fhwrene. C13HlO' (M, 166.2). 1127400. [86-73-7].
Diphenylenemethane. Formaldehyde solution. 1039101.
White or almost white crystals, freely soluble in anhydrous See Formaldehyde solution (35 per eent) (0826).
acetic acid, soluble in hot ethanol (96 per cent). Formamide. CH 3 NO. (Mr 45.0). 1039200. [75-12-7].
mp: 113 oC to 115 oc. Clear, colourless, oily liquid, hygroscopic, miscible with water
and with ethanol (96 per cent). It is hydrolysed by water.
(9-Fluorenyl)methyl chloroformate. C¡SH ll Cl0 2.
(M, 258.7). 1180100. [28920-43-6]. Fluoren-9-ylmethyl d§g: about 1.134.
chloromethanoate. bp: about 210 oc.
mp: about 63 oc. Content: minimum 99.5 per cent.
Storage: in an airtight container.
Fluorescamine. C17HlQ04' (M, 278.3). 1135800. [38183-12-9].
4- Phenylspiro [furan -2( 3H), l' (3' H) - isobenzofuran ]-3,3' - Formamide Rl. 1039202.
dione. Comphes with the requirements prescribed for formamide R
mp: 154 oC to 155 oc. with the following additional requirement.
Water (2.5.12): maximum 0.1 per cent determined with an
Fluoreseein. C20 H¡zÜs' (M, 332.3). 1106300. [2321-07-5]. equal volume of anhydrous methanol R.
3',6'-Dihydroxyspiro[isobenzofurane-l (3H),9' - [9H]xanthen]-
3-one. Formamide, treated. 1039201.
Orange-red powder, practically insoluble in water, soluble in Disperse 1.0 g of sulfamic acid R in 20.0 mi of formamide R
warm ethanol (96 per cent), soluble in alkaline solutions. In containing 5 per cent VIV of water R.
solution, fluorescein displays a green fluorescence. Formic add, anhydrouso CHzÜ2' (Mr 46.03). 1039300.
mp: about 315 oc. [64-18-6].
Fluorescein-conjugated rabies antiserum. 1038700. Content: minimum 98.0 per cent mlm.
Colourless liquid, corrosive, miscible with water and with
Immunoglobulin fraction with a high rabies antibody titre,
ethanol (96 per cent).
prepared from the sera of suitable animals that have been
immunised with inactivated rabies virus; the immunoglobulin d§g : about 1.22.
is conjugated with fluorescein isothiocyanate. Assay. Weigh accurately a conical flask containing 10 mL of
water R, quickly add about 1 mL of the acid and weigh again.
2-Fluoro-2-deoxy-D-glucose. C6HllFOs' (M, 182.2). Add 50 mL of water R and titrate with 1 1V1 sodium hydroxide,
1113900. [86783-82-6]. using 0.5 mL of phenolphthalein solution R as indicator.
White or almost white crystalline powder. 1 mL of 1 M sodium hydroxíde is equivalent to 46.03 mg of
mp: 174 oC to 176 oc. CHzÜ2'

2-Fluoro-2-deoxy-D-mannose. C6HllFOS' (M, 182.1). Fructose. 1106400. [57-48-7].


1172100. [38440-79-8]. See Fructose (0188).
Colourless semi -solid. Fuchsin, basic. 1039400. [632-99-5].
Fluorodinitrobenzene. C6H3FNzÜ4' (M, 186.1). 1038800. A mixture of rosaniline hydrochloride (C 2o H 2o CIN 3; M, 337.9;
[70-34-8]. 1-Fluoro-2,4-dinitrobenzene. Colour Index No. 42510; Schultz No. 780) and para-rosaniline
hydrochloride (C19H1SCINJ; M, 323.8; Colour Index
Pale yellow liquid or crystals, soluble in propylene glycol. No. 42500; Schultz No. 779).
mp: about 29 oc. lf necessary, purify in the following manner. Dissolve 1 g in
Content: minimum 99.0 per cent, determined by gas 250 mL of dilute hydrochloric acid R. Allow to stand for 2 h
chromatography. at room temperature, filter and neutralise with dilute sodium

468 See the information seetion on general monographs (caver pages)


EUROPEAN PHARMACOPOEIA 8.0 4.1.1. Reagents

hydroxide solution R and add 1 mL to 2 mL in excess. FiIter It loses its water of crystallisation at 120 oc.
the precipitate through a sintered -glass fiIter (40) (2.1.2) mp: about 260 oC, with decomposition.
and wash with water R. Dissolve the precipitate in 70 mL of
Chromatography. Thin-Iayer chromatography (2.2.27) as
methanol R, previously heated to boiling, and add 300 mL of
prescribed in the monograph Bearberry lea! (1054); the
water R at 80 oc. Allow to cool to room temperature, fiIter
chromatogram shows only one principal spot.
and dry the crystals in vacuo.
Crystals with a greenish-bronze sheen, soluble in water and in GaUium (68 Ga) chloride solution. 68GaCI3" (Mr 174.3).
ethanol (96 per cent). 1182500.
Storage: protected from light. Solution containing gallium-68 in the form of gallium chloride
in dilute hydrochloric acid R.
Fuchsin solution, decolorised. 1039401. Content: 90 per cent to 110 per cent of the declared gallium-68
Dissolve 0.1 g of basic fuchsin R in 60 mL of water R. Add radioactivity at the date and time stated on the label.
a solution containing 1 g of anhydrous sodium sulfite R or Gastric juice, artificial. 1039900.
2 g of sodium sulfite R in 10 mL of water R. Slowly and with
Dissolve 2.0 g of sodium chloride R and 3.2 g of pepsin
continuous shaking add 2 mL of hydrochloric acid R. Dilute
powder R in water R. Add 80 mL of 1 M hydrochloric acid and
to 100 mL with water R. Allow to stand protected from
dilute to 1000 mL with water R.
light for at least 12 h, decolorise with activated charcoal R
and filter. lf the solution becomes cloudy, fiIter before use. GC concentrical column. 1135100.
lf on standing the solution becomes violet, decolorise again A commercially available system consisting of 2 concentrically
by adding activated charcoal R. arranged tubes. The outer tube is packed with molecular
Test for sensitivity. To 1.0 mL add 1.0 mL of water R and sieves and the inner tube is packed with a porous polymer
0.1 mL of aldehyde-free alcohol R. Add 0.2 mL of a solution mixture. The main application is the separation of gases.
containing 0.1 giL offormaldehyde (CH 2 0, M, 30.0). A
pale-pink colour develops within 5 mino Gelatin. 1040000. [9000-70-8].
See Gelatín (0330).
Storage: protected from light.
Gelatin, hydrolysed. 1040100.
Fuchsin solution, decolorised Rl. 1039402.
Dissolve 50 g of gelatin R in 1000 mL of water R. Autoclave in
To 1 g of basic fuchsin R add 100 mL of water R. Heat to saturated steam at 121 oC for 90 min and freeze dry.
50 oC and allow to cool with occasional shaking. Allow to
stand for 48 h, shake and fiIter. To 4 mL of the filtrate add Geraniol. CIOH¡SO. (Mr 154.2). 1135900. [106-24-1].
6 mL of hydrochloric acid R, mix and dilute to 100 mL with (E)-3, 7 -Dimethylocta-2,6-dien-1-01.
water R. Allow to stand for at least 1 h before use. Oily liquid, slight odour of rose, practically insoluble in water,
miscible with ethanol (96 per cent).
Fucose. C6 H 12 0s' (Mr 164.2). 1039500. [6696-41-9]. Geraniol used in gas chromatography complies with the
6-Deoxy-L-galactose. following additional test.
White or almost white powder, soluble in water and in ethanol Assay. Gas chromatography (2.2.28) as prescribed in the
(96 per cent). monograph Citronella oil (1609).
[al~o: about - 76, determined on a 90 giL solution 24 h after Content: minimum 98.5 per cent, calculated by the
dissolution. normalisation procedure.
mp: about 140 oc. Storage: in an airtight container, protected from light

Fumaric add. C 4 HP4' (Mr 116.1). 1153200. [110-17-8]. Geranyl acetate. C¡2H2P2' (Mr 196.3).1106500. [105-87-3].
(E)- Butenedioic acid. (E)-3,7 -Dimethylocta-2,6-dien-1-yl acetate.
White or almost white crystals, slightly soluble in water, Colourless or slightly yellow liquid, slight odour of rose and
soluble in ethanol (96 per cent), slightly soluble in acetone. lavender.
Geranyl acetate used in gas chromatography camplies with the
mp: about 300 oc.
following additíonal test.
Furfural. CSH 4 0 2. (Mr 96.1). 1039600. [98-01-1]. Assay. Gas chromatography (2.2.28) as prescribed in the
2-Furaldehyde. 2-Furanecarbaldehyde. monograph Bitter-orange-flower oil (1175).
Clear, colourless to brownish-yellow, oily liquid, miscible in Test solution. The substance to be examined.
11 parts of water, miscible with ethanol (96 per cent). Content: minimum 98.0 per cent, calculated by the
d~g: 1.155 to 1.161. 110rmalisation procedure.
Distillation range (2.2.11). Not less than 95 per cent distils Ginsenoside Rbl. C s4 H 9P23,3Hp. (M¡ 1163). 1127500.
between 159 oC and 163 oc. [41753-43-9]. (20S)-3~- Di-D-glucopyranosyl-20-di-D-
Storage: in a dark place. glucopyranosylprotopanaxadiol. (20S)-3~-[(2-0-~-D­
Glucopyranosy! -~- D-glucopyranosyl)oxy]-20- [( 6-0-~- D-
Galactose. C6H¡P6' (Mr 180.2). 1039700. [59-23-4]. glucopyranosyl-~-D-g!ucopyranosyl)oxyl-5a-dammar-
D-( +)-Galactose. 24-en-12~-0l. (20S)-3~- [(2-0-~-D-Glucopyranosyl-~- D-
glucopyranosyl)oxy]-20- [( 6-0-~- D-glucopyranosy! -~- D-
White or almost white, crystalline powder, freely soluble in glucopyranosyl)oxy ]-4,4,8,14-tetramethyl-18-nor-5a -cholest-
water. 24-en-12~-0l.
[al~o: + 79 to + 81, determined on a 100 giL solution in A colourless solid, soluble in water, in anhydrous ethanol and
water R containing about 0.05 per cent of NH y in methanol.
Gallic acid. C 7HPs,Hp. (M¡ 188.1). 1039800. [5995-86-8]. [al~o: + 11.3 determined on a 10 giL so!ution in methanol R.
3,4,5-Trihydroxybenzoic acid monohydrate. mp: about 199 oc.
Crystalline powder or long needles, colourless or slightly Water (2.5.12): maximum 6.8 per cent.
yellow, soluble in water, freely soluble in hot water, in ethanol Assay. Liquid chromatography (2.2.29) as prescribed in the
(96 per cent) and in glycerol. monograph Ginseng (1523).

General Nofices (1) apply ta all monographs and other texts 469
4.1.1. Reagents EUROPEAN PHARMACOPOEIA 8.0

Test solution. Dissolve 3.0 mg, accurately weighted, of Shows mutarotation: [al~4: + 11.7 -7 + 36.3.
ginsenoside Rbl in 10 mL of methanol R. Assay. Dissolve 0.150 g in 50 mL of anhydrous methanol R
Content: minimum 95.0 per cent, calculated by the while stirring under nitrogen. Titrate with 0.1 M
normalisation procedure. tetrabutylammonium hydroxide, protecting the solution from
atmospheric carbon dioxide throughout solubilisation and
Ginsenoside Re. C 4s H szÜ1S. (Mr 947.2). 1157800. titration. Determine the end-point potentiometrically (2.2.20).
[52286-59-6]. (3~,6a,12~) - 20-(~- D-GIucopyranosyloxy)-
3, 12-dihydroxydammar-24-en -6-yI 2-0- (6-deoxy-a -L- 1 mL of 0.1 M tetrabutylammonium hydroxide is equivalent to
mannopyranosyl) -~- D-glucopyranoside. 19.41 mg of C 6H lO 07"
Colourless solid, soluble in water, in ethanol (96 per cent) and Glutamic add. 1040400. [56-86-0].
in methanol. See Glutamic acid (0750).
Ginsenoside Rf. C42H720l4'2HzÜ. (Mr 837). 1127700.
Glutamyl endopeptidase for peptide mapping. 1173300.
[52286-58-5]. (20S)-6-0- [~-D-Glucopyranosyl-(l-72)-~-D­
[13 70 10-42- 5]. Endoproteinase Glu -C of high purity from
glycopyranoside ]-dammar- 24-ene-3~,6a, l2~,20- tetrol.
Staphylococcus aureus strain V8 (EC 3.4.21.19).
A colourless solid, soluble in water, in anhydrous ethanol and
in methanol. L-y-Glutamyl-L-cysteine. CSH14N20SS. (Mr 250.3). 1157900.
[al~o: + 12.8 determined on a 10 giL solution in methanol R. [636-58-8].
mp: about 198 oc. Glutaraldehyde. CSHsÜ2. (Mr 100.1). 1098300. [111-30-8].
Ginsenoside Rgl. C42H720l4'2HzÜ. (Mr 837). Oily liquid, soluble in water.
1127600. [22427-39-0]. (20S)-6~-D-Glucopyranosyl­ n~5 : about 1.434.
D-glucopyranosylprotopanaxatriol. (20S)-6a,20- Bis(~­ bp: about 188 oc.
D-glucopyranosyloxy)-5a-dammar-24-ene-3~,12~-diol.
(20S)-6u,20- Bis(~- D-glucopyranosyloxy)-4,4,8, 14- Glutaric acid. CSHsÜ4. (Mr 132.1). 1149700. [110-94-1].
tetramethyI-18- nor-5u -cholest -24-ene-3~, 12~-diol. Pentanedioic acid.
A colourless solid, soluble in water, in anhydrous ethanol and White or almost white, crystalline powder.
in methanol.
[al~o: + 31.2 determined on a 10 giL solution in methanol R. L-Glutathione,oxidised. C2oH32N6012S2. (Mr 612.6). 1158000.
[27025-41-8]. Bis(L-y-glutamyl-L-cysteinylglycine) disulfide.
mp: 188 oC to 191 oc.
Water (2.5.12): maximum 4.8 per cent. Glycerol. 1040500. [56-81-5].
Assay. Liquid chromatography (2.2.29) as prescribed in the See Glycerol (0496).
monograph Ginseng (1523).
Glycerol Rl. 1040501.
Test solution. Dissolve 3.0 mg, accurately weighted, of
ginsenoside Rgl in 10 mL of methanol R. Complies with the requirements prescribed for the
monograph Glycerol (0496) and free fram diethylene glycol
Content: minimum 95.0 per cent, calculated by the
when examined as prescribed in the test for impurity A and
normalisation procedure.
related substances in that monograph.
Ginsenoside Rg2. C42HnOl3" (Mr 785). 1182600.
Glycerol (85 per cent). 1040600.
[52286-7 4-5]. 3~,l2~,20- Trihydroxydammar-24-en-6a-yl
2-0-(6-deoxy-a-L-mannopyranosyl)-~-D-glucopyranoside. See Glycerol (85 per cent) (0497).

Gitoxin. C 41 H 6 P14. (Mr 781). 1040200. [4562-36-1]. Glycerol (85 per cent) Rl. 1040601.
Glycoside of Digitalis purpurea L. 3~-(0-2,6-Dideoxy- Complies with the requirements prescribed for the
~-d -ribo- hexopyranosyl-( 1-74) -0- 2,6-dideoxy-~-d- monograph Glycerol 85 per cent (0497) and free from
ribo- hexopyranosyl-( 1-74)- 2,6-dideoxy-~-d-ribo­ diethylene glycol when examined as prescribed in the test
hexopyranosyloxy) -14, 16~-dihydroxy-5~, 14~-card -20(22)- for impurity A and related substances in that monograph.
enolide.
A white or almost white, crystalline powder, practically Glyceroll-decanoate. C I3 H 26 0 4 . (Mr 246.3). 1169400.
insoluble in water and in most common organic solvents, [2277-23-8]. (2RS)-2,3-Dihydroxypropyl decano ate.
soluble in pyridine. a -Monocaprin. 1-Monodecanoyl-rac-glycerol.
[al~o: + 20 to + 24, determined on a 5 giL solution in a mixture Content: about 99 per cent.
of equal volumes of chloroform R and methanol R. Glyceroll-octanoate. C ll H 2P4. (M, 218.3). 1169500.
Chromatography. Thin-layer chromatography (2.2.27) as [502-54-5]. (2RS)-2,3-Dihydroxypropyl octanoate.
prescribed in the monograph Digitalis leaf (0117); the a -Monocaprylin. 1- Monooctanoyl-rac-glycerol.
chromatogram shows only one principal spot. Cantent: about 99 per cent.
Glucosamine hydrochloride. C6 H 14 CINO s. (Mr 215.6).
Glycidol. C3H602. (Mr 74.1). 1127800. [556-52-5].
1040300. [66-84-2]. D-Glucosamine hydrochloride.
Slightly viscous liquid, miscible with water.
Crystals, soluble in water.
d~o: about 1.115.
[al~o: + 100, decreasing to + 47.5 after 30 min, determined
on a 100 giL solution. n~o : about 1.432.

Glucose. 1025700. [50-99-7]. Glycine. 1040700. [56-40-6].


See Anhydrous glucase (0177). See Glycine (0614).

D-Glucuronic acid. C 6 H lO 07" (Mr 194.1). 1119700. Glycollic acid. C2H 4 0 y (Mr 76.0). 1040800. [79-14-1].
[6556-12-3]. 2-Hydroxyacetic acid.
Content: minimum 96.0 per cent, calculated with reference to Crystals, soluble in water, in acetone, in ethanol (96 per cent)
the substance dried in vacuo (2.2.32). and in methanol.
Soluble in water and in ethanol (96 per cent). mp: about 80 oc.

470 See the information section on general monographs (caver pages)


EUROPEAN PHARMACOPOEIA 8.0 4.1.1. Reagents

Glycyrrhetic acid. C 30 H 4P4' (Mr 470.7). 1040900. Guaiazulene. C1sH 1S ' (Mr 198.3). 1041500. [489-84-9].
[471-53-4]. Glycyrrhetinic aeid. 12,13-Didehydro-3~­ 1,4-Dimethyl-7 -isopropylazulene.
hydroxy-l1-oxo-olean-30-oic aeid. Dark-blue crystals or blue liquid, very slightly soluble in water,
A mixture of a- and ~-glycyrrhetic aeids in whieh the ~-isomer miseible with fatty and essential oils and with liquid paraffin,
is predominant. sparingly soluble in ethanol (96 per cent), soluble in 500 giL
sulfuric aeid and 80 per cent m/m phosphoric aeid, giving a
White or yellowish-brown powder, practically insoluble in colourless solution.
water, soluble in anhydrous ethanol and in glaeial acetic aeid.
mp: about 30 oc.
[Q;l~o: + 145 to + 155, determined on a 10.0 giL solution in Storage: protected from light and airo
anhydrous ethanol R.
Chromatography. Thin-Iayer chromatography (2.2.27) using Guanidine hydrochloride. CHsNJHCl. (Mr 95.5). 1098500.
silica gel GF254 Ras the coating substance; prepare the slurry
[50-01-1].
using a 0.25 per cent V/V solution of phosphoric acid R. Crystalline powder, freely soluble in water and in ethanol
Apply to the plate 5 flL of a 5 giL solution of the glycyrrhetie (96 per cent).
aeid in a mixture of equal volumes of chloroform R and Guanine. CsHsNsO. (Mr 15l.l). 1041600. [73-40-5].
methanol R. Develop over a path of 10 cm using a mixture 2-Amino-l, 7-dihydro-6H-purin -6-one.
of 5 volumes of methanol R and 95 volumes of chloroform R.
Examine the chromatogram in ultraviolet light at 254 nm. The Amorphous white or almost white powder, practically
chromatogram shows a dark spot (R p about 0.3) corresponding insoluble in water, slightly soluble in ethanol (96 per cent).
to ~-glycyrrhetie aeid and a smaller spot (R p about 0.5) It dissolves in ammonia and in dilute solutions of alkali
corresponding to a-glycyrrhetie aeid. Spray with anisaldehyde hydroxides.
solution R and heat at 100-105 oC for 10 mino Both spots are Haemoglobin. 1041700. [9008-02-0].
coloured bluish-violet. Between them a smaller bluish-violet Nitrogen: 15 per cent to 16 per cent.
spot may be presento
Iron: 0.2 per cent to 0.3 per cent.
18a-Glycyrrhetinic aeid. C30H4604' (Mr 470.7). 1127900. L05s on drying (2.2.32): maximum 2 per cent.
[1449-05-4]. (20~)-3~- Hydroxy-ll-oxo-18a-olean-12-en-29- Sulfated ash (2.4.14): maximum 1.5 per cent.
oie aeid.
Haemoglobin solution. 1041701.
White or almost white powder, practically insoluble in water,
Transfer 2 g of haemoglobin R to a 250 mL beaker and add
soluble in anhydrous ethanol, sparingly soluble in methylene
75 mL of dilute hydrochloric acid R2. Stir until solution is
chloride.
complete. Adjust the pH to 1.6 ± 0.1 using 1 M hydrochloric
Glyoxalhydroxyanil. C 14 H 12 NP2' (Mr 240.3). 1041000. acid. Transfer to a 100 mL flask with the aid of dilute
[1149-16-2]. Glyoxal bis(2-hydroxyanil). hydrochloric acid R2. Add 25 mg of thiomersal R. Prepare
daily, sto re at 5 ± 3 oC and readjust to pH 1.6 before use.
White or almost white crystals, soluble in hot ethanol (96 per Storage: at 2 oC to 8 oc.
cent).
mp: about 200 oc. Harpagoside. C24H30011' (Mr 494.5). 1098600.
White or almost white, crystalline powder, very hygroscopic,
Glyoxal solution. 1098400. [107-22-2]. soluble in water and in ethanol (96 per cent).
Contains about 40 per cent (m/m) glyoxal. mp: 117 oC to 121 oc.
Storage: in an airtight container.
Assay. In a ground-glass stoppered flask place 1.000 g of
glyoxal solution, 20 mL of a 70 giL solution of hydroxylamine Hederacoside C. CS9H96026' (Mr 1221). 1158100.
hydrochloride R and 50 mL of water R. Allow to stand for [14216-03-6]. 0-6-Deoxy-a-L-mannopyranosyl-(l-74)-
30 min and add 1 mL of methyl red mixed solution R and 0- ~- D-glucopyranosyl-( 1-76)-~- D-glucopyranosyl
titrate with 1 M sodium hydroxide until the colour changes (4R)-3~- [[2-0( -6-deoxy-a-L-mannopyranosyl)-a-L-
from red to green. Carry out a blank titration. arabinopyranosyl] oxy]-23-hydroxyolean -12-en -28-oate.
1 mL of 1 M sodium hydroxide is equivalent to 29.02 mg of Colourless crystals or white or almost white powder.
glyoxal (C 2 H 20 2). mp: about 220 oc.
Hederacoside C used in liquid chromatography complies with
Gonadotrophin, chorionic. 1041100. [9002-61-3].
the following additional test.
See Chorionic gonadotrophin (0498). Assay. Liquid chromatography (2.2.29) as prescribed in the
monograph Ivy leaf (2148).
Gonadotrophin, serum. 1041200.
Test solution. Dissolve 5.0 mg of hederacoside C in 5.0 mL
See Equine serum gonadotrophin for veterinary use (0719). of methanol R.
Content: minimum 95 per cent, calculated by the
Guaiacol. C7 H s02' (Mr 124.1). 1148300. [90-05-1].
normalisation procedure.
2-Methoxyphenol. 1-Hydroxy-2-methoxybenzene.
Crystalline mass or colourless or yellowish liquid, hygroscopic, Hederagenin. C30 H 4sÜ4' (Mr 472.7). 1184100.
slightly soluble in water, very soluble in methylene chloride, [465-99-6]. Astrantiagenin E. Caulosapogenin.
freely soluble in ethanol (96 per cent). 3~,23- Dihydroxy-4a-olean-12-en-28-oic acid.

bp: about 205 oc. a-Hederin. C41H66012' (Mr 751.0).1158200. [27013-91-8].


(+ )-( 4R)-3~- [[2-0-( 6-Deoxy-a-L-mannopyranosyI)-a-L-
mp: about 28 oc.
arabinopyranosyl] oxy]-23-hydroxyolean -l2-en -28-oie aeid.
Guaiacum resino 1041400. White or almost white powder.
Resin obtained from the heartwood of Guaiacum officinale L. mp: about 256 oc.
and Guaiacum sanctum L. HeHum for chromatography. He. (A r 4.003). 1041800.
Reddish-brown or greenish-brown, hard, glassy fragments; [7440-59-7].
fracture shiny. Content: minimum 99.995 per cent V/VofHe.

General Notices (1) apply to all monographs and other texts 471
4.1.1. Reagents EUROPEAN PHARMACOPOEIA 8.0

Heparin. 1041900. [9041-08-1]. Hexacosane. C26 H s4 ' (Mr 366.7). 1042200. [630-01-3].
See Heparin sodium (0333). Colourless or white or almost white flakes.
mp: about 57 oc.
Heptachlor. C lO H sC1 7 • (Mr 373.3). 1128000. [76-44-8].
bp: about 135 oc. Hexadimethrine bromide. (C13H30Br2Nz)l1' 1042300.
mp: about 95 oc. [28728-55-4]. 1,5-Dimethyl-1,5-diazaundecamethylene
polymethobromide. Poly( 1,1,5,5-tetramethyl-1 ,5-azonia-
A suitable certified reference solution (lO ng/flL in undecamethylene dibromide).
cyclohexane) may be used.
White or almost white, amorphous powder, hygroscopic,
Heptachlor epoxide. ClQH SCI,o. (Mr 389.3). 1128100. soluble in water.
[1024-57-3]. 5torage: in an airtight container.
bp: about 200 oc.
2,2',2",6,6',6"- Hexa( 1, l-dimethylethyl) -4,4',4"- [( 2,4,6-
mp: about 160 oc. trimethyl-l ,3,5-benzenetriyl)trismethylene] triphenol.
A suitable certified reference solution (10 ng/flL in C s4 H 7sÜ3' (Mr 775). 1042100. 2,2',2",6,6',6"-
cyclohexane) may be used. Hexa -tert-butyl-4,4',4" - [(2,4,6-trimethyl-1,3,5-
benzenetriyl)trismethylene] trip henol.
Heptafluorobutyric add. C4 HF,02' (Mr 214.0). 1162400.
[375-22-4]. HFBA. Crystalline powder, practically insoluble in water, soluble in
acetone, slightly soluble in ethanol (96 per cent).
Clear, colourless liquido Corrosive.
mp: about 244 oc.
d~g: about 1.645.
n~o : about 1.300. 1,1,1,3,3,3-Hexafluoropropan-2-ol. C3H2F60. (Mr 168.0).
1136000. [920-66-1].
bp: about 120 oc.
Content: minimum 99.0 per cent, determined by gas
Content: minimum 99.5 per cent.
chromatography.
Heptafluoro- N- methyl- N- (trimethylsilyl) butanamide. Clear, colourless liquid, miscible with water and with
CS H i2 F7NOSi. (Mr 299.3). 1139500. [53296-64-3]. anhydrous ethanol.
2,2,3,3,4,4,4-Heptafluoro- N- methyl- N-( trimethylsilyl)-
butyramide.
:
d;~ about 1.596.
bp: about 59 oc.
Clear, colourless liquid, flammable.
n~o: about 1.351.
Hexamethyldisilazane. C6Hi9NSiz. (Mr 161.4). 1042400.
[999-97-3].
bp: about 148 oc.
Clear, colourless liquido
Heptane. C7Hi6' (M, 100.2). 1042000. [142-82-5]. d~g: about 0.78.
Colourless, flammable liquid, practically insoluble in water, n~o : about 1.408.
miscible with anhydrous ethanol. bp: about 125 oc.
d~g: 0.683 to 0.686. 5torage: in an airtight container.
n~o: 1.387 to 1.388.
Hexamethylenetetramine. C6H i2 N 4 • (M, 140.2). 1042500.
Distillation range (2.2.11). Not less than 95 per cent distils [100-97-0]. Hexamine. 1,3,5,7-Tetraazatricyclo[3.3.1.1 3•7]-
between 97 oC and 98 oc.
decane.
Hesperidin. C2s H 3PiS' (M, 611). 1139000. [520-26-3]. Colourless, crystalline powder, very soluble in water.
(5)-7 - [[ 6-0-( 6-Deoxy-a- L-mannopyranosyl)-~- D-
glucopyranosyl] oxy]-5-hydroxy-2-( 3-hydroxy-4- Hexane. C 6 H[4" (Mr 86.2). 1042600. [110-54-3].
methoxyphenyl)-2,3-dihydro-4H-1-benzopyran -4-one. Colourless, flammable liquid, practically insoluble in water,
Hygroscopic powder, slightly soluble in water and in methanol. miscible with anhydrous ethanol.
mp: 258 oC to 262 oc. d~g: 0.659 to 0.663.
n~o: 1.375 to 1.376.
Hexachlorobenzene. C 6 C16 . (M, 284.8). 1128200. [118-74-1]. Distillation range (2.2.11). Not less than 95 per cent distils
bp: about 332 oc. between 67 oC and 69 oc.
mp: about 230 oc. Hexane used in spectrophotometry complies with the following
A suitable certified reference solution (lO ng/flL in additional test.
cyclohexane) may be used. Minimum transmittance (2.2.25) using water R as
compensation liquid: 97 per cent from 260 nm to 420 nm.
a-Hexachlorocydohexane. C6H6Cl6' (Mr 290.8). 1128300.
[319-84-6]. Hexylamine. C6 H JS N. (Mr 101.2). 1042700. [111-26-2].
bp: about 288 oc. Hexan -l-amine.
mp: about 158 oc. Colourless liquid, slightly soluble in water, soluble in ethanol
(96 per cent).
A suitable certified reference solution (10 ngl flL in
cyclohexane) may be used. d~g: about 0.766.
n~o: about l.418.
p- Hexachlorocydohexane. C6H 6C16. (M, 290.8). 1128400. bp: 127 oC to 131 oc.
[319-85-7].
A suitable certified reference solution (10 ngl flL in Histamine dihydrochloride. 1042800. [56-92-8].
cyclohexane) may be used. See Histamine dihydrochloride (0143).
ó-Hexachlorocydohexane. C6 H 6C1 6 . (Mr 290.8). 1128500. Histamine solution. 104290 l.
[319-86-8]. A 9 giL solution of sodium chloride R containing 0.1 flg
A suitable certified reference solution (lO ng/flL in per millilitre of histamine base (as the phosphate or
cyclohexane) may be used. dihydrochloride) .

472 See the information section on general monographs (ca ver pages)
EUROPEAN PHARMACOPOEIA 8.0 4.1.1. Reagents

Histidine monohydroehloride. C6H lO CINP2,Hp. Hydrazine sulfate. H(;NP4S, (M, 130.1). 1043400.
(Mr 209.6). 1043000. [123333-71-1]. (RS)-2-Amino-3- [10034-93-2].
(imidazol-4-yl)propionic acid hydrochloride monohydrate. Colourless crystals, sparingly soluble in cold water, soluble
Crystalline powder or colourless crystals, soluble in water. in hot water (50 oC) and freely soluble in boiling water,
mp: about 250 oC, with decomposition. practically insoluble in ethanol (96 per cent).
Chromatography. Thin-Iayer chromatography Arsenic (2.4.2, Method A) : maximum 1 ppm, determined on
(2.2.27) as prescribed in the monograph Histamine l.0 g.
dihydrochloride (0143); the chromatogram shows only one Sulfated ash (2.4.14): maximum 0.1 per cent.
principal spot. Hydriodic add. HI. (Mr 127.9). 1098900. [10034-85-2].
Holmium oxide. HOP3" (Mr 377.9). 1043100. [12055-62-8]. Prepare by distilling hydriodic acid over red phosphorus,
Diholmium trioxide. passing carbon dioxide R or nitrogen R through the apparatus
Yellowish powder, practically insoluble in water. during the distillation. Use the colourless or almost colourless,
constant-boiling mixture (55 per cent to 58 per cent of HI)
Holmium perchlorate solution. 1043101. distilling between 126 oC and 127 oc.
A 40 giL solution of holmium oxide R in a solution of Place the acid in small, amber, glass-stoppered bottles
perchloric acid R containing 141 giL ofHCI0 4. previously flushed with carbon dioxide R or nitrogen R, seal
with paraffin.
DL-Homocysteine. C4H 9 N0 2S. (M r 135.2). 1136100. Storage: in a dark place.
[454-29-5]. (2RS)-2- Amino-4-sulfanylbutanoic acid.
White or almost white, crystalline powder. Hydrobromic add, 30 per eent. 1098700. [10035-10-6].
mp: about 232 oc. A 30 per cent solution of hydrobromic acid in glacial acetic
acid R.
L- Homocysteine thiolactone hydrochloride. Degas with caution the contents before opening.
C4H sCINOS. (Mr 153.6). 1136200. [31828-68-9].
(3S)- 3 -Aminodihydrothiophen -2( 3H)-one hydrochloride.
Hydrobromic add, dUute. 1098701.
Place 5.0 mL of 30 per cent hydrobromic acid R in amber
White or almost white, crystalline powder.
vials equipped with polyethylene stoppers. Seal under
mp: about 202 oc. argon R and stofe in the dark. Add 5.0 mL of glacial acetic
acid R immediately before use. Shake.
HonokioL C1S H J s0 2 • (Mr 266.3). 1182700. [35354-74-6].
3',5-Di(prop-2-enyl)biphenyl-2,4' -diol. 3',5-Diallyl-2,4'- Storage: in the dark.
dihydroxybiphenyl. 3',5-Di-2-propenyl- [1, l' -biphenyl]-2,4'- Hydrobromk add, 47 per eent. 1118900.
diol.
A 47 per cent m/m solution of hydrobromic acid.
Human tissue factor solution. 1186100. Hydrobromic acid, diluie Rl. 1118901.
Solution containing human tissue factor, which may be Contains 7,9 giL of HBr.
produced by recombinant DNA technology, combined with
Dissolve 16.81 g of 47 per cent hydrobromic acid R in
phospholipids and calcium buffers. Suitable stabilisers may
water R and dilute to 1000 mL with the same solvent.
be added.
Hydroehlork add. 1043500. [7647-01-0].
Hyaluronidase diluent. 1043300.
See Concentrated hydrochloric acid (0002).
Mix 100 mL of phosphate buffer solution pH 6.4 R with 100 mL
of water R. Dissolve 0.140 g of hydrolysed gelatin R in the 2 M Hydrochloric adJ. 3001700.
solution at 37 oc. Dilute 206.0 g of hydrochloric acid R to 1000.0 mL with
Storage: use within 2 h. water R.

Hydrastine hydroehloride. C21H2ZCIN06' (Mr 419.9). 3 M Hydrochloric add. 3001600.


1154000. [5936-28-7]. (3S)-6,7-Dimethoxy-3-[(5R)-6- Dilute 309.0 g of hydrochloric acid R to 1000.0 mL with
methyl- 5,6, 7,8-tetrahydro-1,3-dioxolo [4,5-g] isoquinolin- 5- water R.
yl]isobenzofuran-1 (3H)-one hydrochloride.
6 M Hydrochloric acid. 3001500.
White or almost white powder, hygroscopic, very soluble in
Dilute 618.0 g of hydrochloric acid R to 1000.0 mL with
water and in ethanol (96 per cent).
water R.
[a]:;: about + 127.
Hydrochloric add RL 1043501.
mp: about 116 oc.
Contains 250 giL of HCl.
Hydrastine hydrochloride used in liquid chromatography
complies with the following additional test. Dilute 70 g of hydrochloric acid R to 100 mL with water R.
Assay. Liquid chromatography (2.2.29) as prescribed in the Hydroehloric add, brominated. 1043507.
monograph Goldenseal rhizome (1831). To 1 mL of bromine solution R add 100 mL of hydrochloric
Content: minimum 98 per cent, calculated by the acid R.
normalisation procedure.
Hydrochloric add, diluie. 1043503.
Hydrazine. H 4N z. (Mr 32.05). 1136300. [302-01-2]. Diazane. Contains 73 giL of HCl.
Slightly oily liquid, colourless, with a strong odour of Dilute 20 g of hydrochloric acid R to 100 mL with water R.
ammonia, miscible with water. Dilute solutions in water are
Hydrochloric add, dUute, heavy metal-free. 1043509.
commercially available.
Complies with the requirements prescribed for dilute
n ~ : about 1.470. hydrochloric acid R with the following maximum contents
bp: about 113 oc. of heavy metals.
mp: about 1.5 oc. As: 0.005 ppm.
Caution: toxic and corrosive. Cd: 0.003 ppm.

General Notices (1) apply to all monographs and other texts 473
4.1.1. Reagents EUROPEAN PHARMACOPOEIA 8.0

Cu: 0.003 ppm. Hydrogen for chromatography. H 2• (Mr 2.016). 1043700.


Fe: 0.05 ppm. [1333-74-0].
Hg: 0.005 ppm. Content: minimum 99.95 per cent V/V.
Ni: 0.004 ppm. Hydrogen peroxide solution, dilute. 1043800. [7722-84-1].
Pb: 0.001 ppm. See Hydrogen peroxide solution (3 per cent) (0395).
Zn: 0.005 ppm.
Hydrogen peroxide solution, strong. 1043900. [7722-84-1].
Hydrochloric add, dilute Rl. 1043504. See Hydrogen peroxide solution (30 per cent) (0396).
Contains 0.37 giL of HCl.
Hydrogen sulfide. H 2S. (Mr 34.08). 1044000. [7783-06-4].
Dilute 1.0 mL of dilute hydrochloric acid R to 200.0 mL
Gas, slightly soluble in water.
with water R.
Hyrlrogen sulfide solution. 1136400.
Hydrochlork add, dHute R2. 1043505.
A recently prepared solution of hydrogen sulfide R in
Dilute 30 mL of 1 M hydrochloric acid to 1000 mL with
water R. The saturated solution contains about 0.4 per cent
water R; adjust to pH 1.6 ± 0.1.
to 0.5 per cent of H 2S at 20 oc.
Hydl'Ochloric add, ethanolic. 1043506.
Hydrogen sulfide Rl. H 2S. (Mr 34.08). 1106600. [7783-06-4].
Dilute 5.0 mL of 1 M hydrochloric acid to 500.0 mL with Content: minimum 99.7 per cent V/V.
ethanol (96 per cent) R.
Hydroquinone. C6H602' (Mr 110.1). 1044100. [123-31-9].
Hydrochloric add, heavy metal-free. 1043510. Benzene-l A-diol.
Complies with the requirements prescribed for hydrochloric Fine, colourless or white 01' almost white needles, darkening
acid R with the following maximum contents of heavy on exposure to air and light, soluble in water and in ethanol
metals. (96 per cent). .
As: 0.005 ppm. mp: about 173 oc.
Cd: 0.003 ppm. Storage: protected from light and airo
Cu: 0.003 ppm.
Hydroquinone solution. 1044101.
Fe: 0.05 ppm.
Dissolve 0.5 g of hydroquinone R in water R, add 20 ¡..tL of
Hg: 0.005 ppm.
sulfuric acid R and dilute to 50 mL with water R.
Ni: 0.004 ppm.
Pb: 0.001 ppm. 2-Hydroxybenzimidazole. C7H6NP. (Mr 134.1). 1169600.
[615-16-7]. lH-benzimidazol-2-ol.
Zn: 0.005 ppm.
4-Hydroxybenzohyrlrazide. C7H sNP2' (Mr 152.2). 1145900.
Hydl'Ochloric add, lead-free. 1043508.
[5351-23-5]. p-Hydroxybenzohydrazide.
Complies with the requirements prescribed for hydrochloric
acid R with the following additional requirement. 4-Hydroxybenzoic add. C7H603' (Mr 138.1). 1106700.
Lead: maximum 20 ppb. [99-96-7].
Atomic emission spectrometry (2.2.22, Method 1). Crystals, slightly soluble in water, very soluble in ethanol
(96 per cent), soluble in acetone.
Test so/ution. In a quartz crucible evaporate 200 g of the
mp: 214 oC to 215 oc.
acid to be examined almost to dryness. Take up the residue
in 5 mL of nitric acid prepared by sub-boiling distillation of 4-Hydroxycoumarin. C9H603' (M, 162.2). 1169700.
nitric acid R and evaporate to dryness. Take up the residue [1076-38-6]. 4-Hydroxy-2H-l-benzopyran-2-one.
in 5 mL of nitric acid prepared by sub-boiling distillation White or almost white powder, freely soluble in methanol.
of nitric acid R.
Content: minimum 98.0 per cent.
Reference solutions. Prepare the reference solutions using
lead standard solution (0.1 ppm Pb) R diluted with nitric 6-Hydroxydopa. C9HIlNOS' (Mr 213.2). 1169800.
acid prepared by sub-boiling distillation of nitric acid R. [21373-30-8]. (2RS)-2-Amino-3-(2A,5-trihydroxyphenyl)-
Wavelength: 220.35 nm. propanoic acid. 2,5-Dihydroxy-DL-tyrosine.
mp: about 257 oc.
Hydrochloric add, methanolic. 1043511.
Dilute 4.0 mL of hydrochloric acid R to 1000.0 mL with 2- [4- (2- Hydroxyethyl)piperazin-l-yl] ethanesulfonic add.
methanol R2. CSH1SNP4S, (M, 238.3). 1106800. [7365-45-9]. HEPES.
White or almost white powder.
Hydrocortisone acetate. 1098800. [50-03-3].
mp: about 236 oC, with decomposition
See Hydrocortisone aceta te (0334).
4-Hydroxyisophthalk add. C SH 60 S' (Mr 182.1). 1106900.
Hydrofluoric add. HE (M¡ 20.01). 1043600. [7664-39-3]. [636-46-4]. 4-Hydroxybenzene-l ,3-dicarboxylic acid.
Content: minimum 40.0 per cent mimo Needles or platelets, very slightly soluble in water, freely
Clear, colourless liquido soluble in ethanol (96 per cent).
Loss on ignition: not more than 0.05 per cent m/m; evaporate mp: about 314 oC, with decomposition.
the hydrofluoric acid in a platinum crucible and gently ignite
the residue to constant mass. Hydroxylamine hydrochloride. NH 4 CIO. (Mr 69.5).
1044300. [5470-11-1].
Assay. Weigh accurately a glass-stoppered flask containing
50.0 mL of 1 M sodium hydroxide. Introduce 2 g of the V/hite or almost white, crystalline powder, very soluble in
hydrofluoric acid and weigh again. Titrate the solution with water, soluble in ethanol (96 per cent).
0.5 M sulfuric acid, using 0.5 mL of phenolphthalein solution R Hydroxylamine hydrochloride soh.l.tion R2. 1044304.
as indicator.
Dissolve 2.5 g of hydroxylamine hydrochloride R in 4.5 mL
1 mL of 1 M sodium hydroxide is equivalent to 20.01 mg ofHE of hot water R and add 40 mL of ethanol (96 per cent) R
Storage: in a polyethylene container. and 0.4 mL of bromophenol blue solution R2.~ Add 0.5 M

474 See the information section on general monographs (cover pages)


EUROPEAN PHARMACOPOEIA 8.0 4.1.1. Reagents

alcoholic potassium hydroxide until a greenish-yellow Hypericin. C30H160S' (Mr 504.4). 1149800.
colour is obtained. Dilute to 50.0 mL with ethanol (96 per [548-04-9]. 1,3,4,6,8,13-Hexahydroxy-lO,ll-
cent) R. dimethylphenanthro [1, 1O,9,8-opqra )perylene-7, 14-dione.
Content: minimum 85 per cent.
Hydroxylamine solution, alcoholic. 104430l.
Dissolve 3.5 g of hydroxylamine hydrochloride R in 95 mL Hyperoside. C21 H 2aÜ12' (Mr 464.4). 1045000.
of ethanol (60 per cent V/V) R, add 0.5 mL of a 2 giL 2- (3,4-Dihydroxyphenyl) -3- ~- D-galactopyranosyloxy-
solution of methyl orange R in ethanol (60 per cent V/V) R 5,7 -dihydroxychromen -4-one.
and sufficient 0.5 M potassium hydroxide in alcohol (60 per Faint yellow needles, soluble in methanol.
cent V/V) to give apure yellow colour. Dilute to 100 mL
with ethanol (60 per cent V/V) R. mp: about 240 oC, with decomposition.
Absorbance (2.2.25). A solution in methanol R shows
Hydroxylamine solution, alkaline. 1044302. 2 absorption maxima at 259 nm and at 364 nm.
Immediately befo re use, mix equal volumes of a 139 giL
solution of hydroxylamine hydrochloride R and a 150 giL Hypophosphorous reagent. 1045200.
solution of sodium hydroxide R. Dissolve with the aid of gentle heat, 10 g of sodium
hypophosphite R in 20 mL of water R and dilute to 100 mL
Hydroxylamine solution, alkaline Rl. 1044303. with hydrochloric acid R. Allow to settle and decant or fiIter
Solution A. Dissolve 12.5 g of hydroxylamine hydrochloride R through glass wool.
in methanol R and dilute to 100 mL with the same solvento
Hypoxanthine. C SH 4Np. (Mr 136.1). 1045300. [68-94-0].
Solution B. Dissolve 12.5 g of sodium hydroxide R in
lH-Purin-6-one.
methanol R and dilute to 100 mL with the same solvento
Mix equal volumes of solution A and solution B White or almost white, crystalline powder, very slightly soluble
immediately befo re use. in water, sparingly soluble in boiling water, soluble in dilute
acids and in dilute alkali hydroxide solutions, decomposes
HydroxymethyIfurfuraL C6H603' (Mr 126.1). 1044400. without melting at about 150 oc.
[67-47-0].5-Hydroxymethylfurfural. Chromatography. Thin-Iayer chromatography (2.2.27) as
Acicular crystals, freely soluble in water, in acetone and in prescribed in the monograph Mercaptopurine (0096); the
ethanol (96 per cent). chromatogram shows only one principal spot.
mp: about 32 oc. Imidazole. C 3H 4N 2 • (Mr 68.1). 1045400. [288-32-4].
Hydroxynaphthol blue, sodium salto C2oHllN2Na3011S3" White or almost white, crystalline powder, soluble in water
(Mr 620). 1044500. [63451-35-4]. Trisodium and in ethanol (96 per cent).
2,2' -dihydroxy-l, l' -azonaphthalene-3',4,6' -trisulfonate. mp: about 90 oc.
2- Hydroxypropylbetadex for chromatography R. 1146000. IminodibenzyL C 14H 13 N. (M r 195.3).1045500. [494-19-9].
Betacyclodextrin modified by the bonding of (R) or (RS) 10,11-Dihydrodibenz lb,)] azepine.
propylene oxide groups on the hydroxyl groups.
Pale yellow, crystalline powder, practically insoluble in water,
Hydroxypropyl-~-cydodextrin. 1128600. [94035-02-6]. freely soluble in acetone.
See Hydroxypropylbetadex (1804). mp: about 106 oc.
pH (2.2.3): 5.0 to 7.5 for a 20 giL solution. Imperatorin. C16H1404' (Mr 270.3). 1180200. [482-44-0].
Hydroxyquinoline. C9H7NO. (Mr 145.2). 1044600. 9- [(3-Methylbut-2-enyl)oxy]-7H-furo[3,2-g] [l]benzopyran-
[148-24-3]. 8-Hydroxyquinoline. Quinolin-8-01. 7-one.
White or slightly yellowish, crystalline powder, slightly soluble 2-Indanamine hydrochloride. C9H 12 CIN. (Mr 169.7).
in water, freely soluble in acetone, in ethanol (96 per cent) 1175800. [2338-18-3]. 2-Aminoindane hydrochloride.
and in dilute mineral acids. 2,3-Dihydro-1H-inden -2-amine hydrochloride.
mp: about 75 oc.
Sulfated ash (2.4.14): maximum 0.05 per cent. Indigo carmine. C16HsN2NazÜSS2' (Mr 466.3). 1045600.
[860-22-0].
12-Hydroxysteark add. ClsH3603' (M, 300.5). 1099000. Schultz No. 1309.
[106-14-9]. 12-Hydroxyoctadecanoic acid.
Colour Index No. 73015.
White or almost white powder. 3,3' -Dioxo-2,2' -bisindolylidene-5,5' -disulfonate disodium.
mp: 71°C to 74 oc. E 132.
It usually contains sodium chloride.
5-HydroxyuradL C4 H 4NP3" (Mr 128.1). 1044700.
[496-76-4]. Isobarbituric acid. Pyrimidine-2,4,5-trio!. Blue or violet-blue powder or blue granules with a coppery
lustre, sparingly soluble in water, practically insoluble in
White or almost white, crystalline powder.
ethanol (96 per cent). It is precipitated from an aqueous
mp: about 310°C, with decomposition. solution by sodium chloride.
Chromatography. Thin-layer chromatography (2.2.27)
as prescribed in the monograph Fluorouracil (0611); the Indigo carmine solution. 1045601.
chromatogram shows a principal spot with an Rp of about 0.3. To a mixture of 10 mL of hydrochloric acid R and 990 mL
Storage: in an airtight container. of 200 giL nitrogen-free sulfuric acid R add 0.2 g of indigo
carmine R.
Hyoscine hydrobromide. 1044800. [6533-68-2]. The solutíon complies with the following test: add 10 mL
See Hyoscine hydrobromide (0106). to a solution of 1.0 mg of potassium nitrate R in 10 mL of
water R, rapidly add 20 mL of nitrogenlree sulfuric acid R
Hyoscyamine sulfate. 1044900. [620-61-1]. and heat to boiling. The blue colour is discharged within
See Hyoscyamine sulfate (0501). 1 mino

General Notices (1) apply to all monographs and other texts 475
4.1.1. Reagents EUROPEAN PHARMACOPOEIA 8.0

Indigo carmine solution Rl. 1045602. Storage: pratected fram light.


Dissolve 4 g of indigo earmine R in about 900 mL of water R
Iodine chloride. lCl. (Mr 162.4). 1143000. [7790-99-0].
added in several portions. Add 2 mL of sulfuric acid R and
dilute to 1000 mL with water R. Black crystals, soluble in water, in acetic acid and in ethanol
(96 per cent).
Assay. Place in alOa mL conica! flask with a wide neck
10.0 mL of nitrate standard solution (lOO ppm NO) R, bp: about 97.4 oc.
10 mL of water R, 0.05 mL of the indigo carmine solution R1,
Iodine chloride solution. 1143001.
and then in a single addition, but with caution, 30 mL of
sulfuric acid R. Titrate the solution immediately, using the Dissolve 1.4 g of iodine ehloride R in glacial acetic acid R
índigo carmine solution R1, until a stable blue colour is and dilute to 100 mL with the same acid.
obtained. Storage: protected from light.
The number of millilitres used, n, is equivalent to 1 mg of
Iodine pentoxide, recrystaUised. Ips' (Mr 333.8). 1046000.
N03' [12029-98-0]. Di-iodine pentoxide. lodic anhydride.
Indometadn. 1101500. [53-86-1]. Content: minimum 99.5 per cent.
See Indometacin (0092). White or almost white, crystalline powder, or white or
greyish-white granules, hygrascopic, very soluble in water
Inosine. CJOH12N40S' (M, 268.2). 1169900. [58-63-9].
9-~- D- Ribofuranosylhypoxanthine. 9-~- D- Ribofuranosyl-l ,9-
forming HI03'
dihydro-6H-purin-6-one. Stability on heating. Dissolve 2 g, previously heated for 1 h at
mp: 222 oC to 226 oc. 200 oC, in 50 mL of water R. A colourless solution is obtained.
Assay. Dissolve 0.100 g in 50 mL of water R, add 3 g of
myo- Inositol. 1161100. potassium iodide R and 10 mL of dilute hydrochloric acid R.
See myo-Inositol (1805). Titrate the liberated iodine with 0.1 M sodium thiosulfate,
using 1 mL of starch solution R as indicator.
Iodine. 1045800. [7553-56-2].
1 rnL of 0.1 M sodium thiosulfate is equivalent to 2.782 mg
See Iodine (0031).
of 12°5'
Iodine solution Rl. 1045801. Storage: in an airtight container, protected fram light.
To 10.0 mL of 0.05 M iodine add 0.6 g of potassium iodide R
and dilute to 100.0 mL with water R. Prepare immediately Iodoacetamide. C2 H 4 INO. (Mr 185.0). 1186200. [144-48-9].
before use. 2-Iodoacetamide.
Slightly yellow, crystalline powder, soluble in water.
Iodine solution R2. 1045802. mp: about 92 oc.
To 10.0 l11L of 0.05 M íodine add 0.6 g of potassíum iodide R
and dilute to 1000.0 mL with water R. Prepare immediately Iodoacetic add. C2 H 3I0 2 • (Mr 185.9). 1107000. [64-69-7].
before use. Colourless or white or almost white crystals, soluble in water
Iodine solution R3. 1045803. and in ethanol (96 per cent).
Dilute 2.0 mL of iodíne solution R1 to 100.0 l11L with mp: 82 oC to 83 oc.
water R. Prepare immediately before use. 2-Iodobenzoic adJ. C 7 H sI02' (M, 248.0). 1046100.
Iodine solution R4. 1045806. [88-67-5].
Dissolve 14 g of iodine R in 100 mL of a 400 giL solution of White or slightly yellow, crystalline powder, slightly soluble in
potassium íodide R, add 1 l11L of dilute hydrochloric acid R water, soluble in ethanol (96 per cent).
and dilute to 1000 mL with water R. mp: about 160 oc.
Storage: protected from light. Chromatography. Thin-layer chromatography (2.2.27), using
cellulose for chromatography f2S4 R as the coating substance:
Iodi.ne solution, akoholic. 1045804.
apply to the p!ate 20 flL of a solution of the 2-iodobenzoic
A 10 giL solution in ethanol (96 per cent) R. acid, prepared by dissolving 40 mg in 4 mL of 0.1 M sodium
Storage: protected fram light. hydroxide and diluting to 10 mL with water R. Develop over
a path of about 12 cm using as the mobile phase the upper
Iodine solution, chloroformic. 1045805. layer obtained by shaking together 20 volumes of water R,
A 5 giL solution in chloroform R. 40 volumes of glacial acetic acid R and 40 volumes of toluene R.
Storage: protected from light. Allow the plate to dry in air and examine in ultraviolet light at
254 nm. The chromatogram shows only one principal spot.
Iorline-123 and ruthenium-l 06 spiking solution. 1166700.
Prepare immediately before use. Mix 3.5 mL of an 18.5 kBq/mL 3-Iodobenzylammonium chloride. C7H 9CIIN. (Mr 269.5).
solution of ruthenium-106 in the form of ruthenium 1168000. [3718-88- 5]. 1-(3-lodophenyl)methanamine
trichloride in a mixture of equal volumes of glacial acetic hydrochloride. 1-(3-Iodophenyl)methanaminium chloride.
acíd R and water R with 200 flL of a 75 kBq/mL solution of m- Iodobenzylamine hydrochloride.
iodine-123 in the form of sodium iodide in water R. White or almost white crystals.
Iodine bromide. IBr. (Mr 206.8). 1045900. [7789-33-5]. mp: 188 oC to 190 oc.
Bluish-black or brownish-black crystals, freely soluble in Iodoethane. C 2H sI. (M, 155.9). 1099100. [75-03-6].
water, in ethanol (96 per cent) and in glacial acetic acid. Colourless or slightly yellowish liquid, darkening 011 exposure
bp: about 116 oc. to air and light, miscible with ethanol (96 per cent) and most
mp: about 40 oc. organic solvents.
Storage: protected from light. d§g: about 1.95.
Iodine bromide solution. 1045901. nj'¡°: about 1.513.
Dissolve 20 g of iodine bromide R in glacial acetic acid R bp: about 72 oc.
and dilute to 1000 rnL with the same solvent Storage: in an airtight container.

476 See the informatíon section on general monographs (cover pages)


EUROPEAN PHARMACOPOEIA 8.0 4.1.1. Reagents

2-Iodohippuric add. C9HsIN0 3 ,2Hp. (Me 341.1). 1046200. Preparation of a column. Unless otherwise prescribed, use a
[147-58-0]. 2-(2-Iodobenzamido)acetic acid. tube with a fused-in sintered glass disc having a length of
White or almost white, crystalline powder, sparingly soluble 400 mm, an internal diameter of 20 mm and a filling height
in water. of about 200 mm. Introduce the resin, mixing it with water R
mp: about 170 oc. and pouring the slurry into the tube, ensuring that no air
bubbles are trapped between the particles. When in use, the
Water (2.5.12): 9 per cent to 13 per cent, determined on liquid must not be allowed to fall below the surface of the
1.000 g. resino If the res in is in its protonated form, wash with water R
Chromatography. Thin-Iayer chromatography (2.2.27), using until50 mL requires not more than 0.05 mL of 0.1 M sodium
cellulose for chromatography F254 R as the coating substance: hydroxide for neutralisation, using 0.1 mL of methyl orange
apply to the plate 20 ¡.tL of a solution of the 2-iodohippuric solution R as indicator.
acid, preparedby dissolving 40 mg in 4 mL of 0.1 M sodium If the resin is in its sodium form or if it requires regeneration,
hydroxide and diluting to 10 mL with water R. Develop over pass about 100 mL of a mixture of equal volumes of
a path of about 12 cm using as the mobile phase the upper hydrochloric acid Rl and water R slowly through the column
layer obtained by shaking together 20 volumes of water R, and then wash with water R as described aboye.
40 volumes of glacial acetic acid R and 40 volumes of toluene R.
Allow the plate to dry in air and examine in ultraviolet light at lrisnorentin. C2oH1SOS' (Mr 386.4).1186300. [41743-73-1].
254 nm. The chromatogram shows only one principal spot. 9-Methoxy-7 -(3,4,5-trimethoxyphenyl)-8H-1,3-dioxolo-
[4,5-g] [1]benzopyran-8-one.
Iodoplatinate reagent. 1046300.
To 3 mL of a 100 giL solution of chloroplatinic acid R add !ron. Fe. (A e 55.85). 1046600. [7439-89-6].
97 mL of water R and 100 mL of a 60 giL solution of potassium Grey powder or wire, soluble in dilute mineral acids.
iodide R.
Iron salicylate solution. 1046700.
Storage: protected from light.
Dissolve 0.1 g of ferric ammonium sulfate R in a mixture of
Iodoplatinate reagent Rl. 1172200. 2 mL of dilute sulfuric acid R and 48 mL of water R and dilute
Mix 2.5 mL of a 50 giL solution of chloroplatinic acid R, to 100 mL with water R. Add 50 mL of a 11.5 giL solution of
22.5 mL of a 100 giL solution of potassium iodide R and 50 mL sodium salicylate R, 10 mL of dilute acetic acid R, 80 mL of
of water R. a 136 giL solution of sodium acetate R and dilute to 500 mL
with water R. The solution should be recently prepared.
Storage: protected from light, at a temperature of 2-8 oc.
Storage: in an airtight container, protected from light.
Iodosulfurous reagent. 1046400.
Isatin. C sH sN0 2. (Me 147.1). 1046800. [91-56-5].
The apparatus, which must be kept closed and dry during the
Indoline-2,3-dione.
preparation, consists of a 3000 mL to 4000 mL round-bottomed
flask with three inlets for a stirrer and a thermometer and Small, yellowish-red crystals, slightly soluble in water, soluble
fitted with a drying tube. To 700 mL of anhydrous pyridine R in hot water and in ethanol (96 per cent), soluble in solutions
and 700 mL of ethylene glyeol monomethyl ether R add, with of alkali hydroxides giving a violet colour becoming yellow
constant stirring, 220 g of finely powdered iodine R, previously on standing.
dried over diphosphorus pentoxide R. Continue stirring until mp: about 200 oC, with partial sublimation.
the iodine has completely dissolved (about 30 min). Cool to Sulfated ash (2.4.14): maximum 0.2 per cent.
- 10 oC, and add quickly, still stirring, 190 g of sulfur dioxide R.
Do not allow the temperature to exceed 30 oc. Coo!. Isatin reagent. 1046801.
Assay. Add about 20 mL of anhydrous methanol R to a titration Dissolve 6 mg of ferric sulfate R in 8 mL of water R and add
vessel and titrate to the end-point with the iodosulfurous cautiously 50 mL of sulfuric acid R. Add 6 mg of isatin R
reagent (2.5.12). Introduce in an appropriate form a suitable and stir until dissolved.
amount of water R, accurately weighed, and repeat the The reagent should be pale yellow, but not orange or red.
determination of water. Calculate the water equivalent in Isoarnyl alcohoL C SH 120. (Me 88.1). 1046900. [123-51-3].
milligrams per millilitre of iodosulfurous reagent. 3-Methylbutan-1-01.
The minimum water equivalent is 3.5 mg of water per millilitre Colourless liquid, slightly soluble in water, miscible with
of reagent. ethanol (96 per cent).
Work protected from humidity. Standardise immediately bp: about 130 oc.
before use.
Storage: in a dry container. Isoarnyl benzoate. C12H1602' (M, 192.3). 1164200. [94-46-2].
Isopentyl benzoate. 3-Methylbutyl benzoate.
5-Iodouradl. C4 H)N 20 2 • (Me 238.0).1046500. [696-07-1]. n~o : about 1.494.
5-Iodo-lH,3H-pyrimidine-2,4-dione.
bp: about 261°C.
mp: about 276 oC, with decomposition.
Colourless or pale yellow liquido
Chromatography. Thin-Iayer chromatography (2.2.27) as
prescribed in the monograph Idoxuridine (0669): apply 5 ¡.tL Isoandrosterone. C 19 H 3oÜ2' (Mr 290.4).1107100. [481-29-8].
of a 0.25 giL solution; the chromatogram obtained shows only Epiandrosterone. 3~- Hydroxy-5a-androstan-17 -one.
one principal spot. White or almost white powder, practically insoluble in water,
soluble in organic solvents.
Ion -exdusion resin for chromatography. 1131000.
A res in with sulfonic acid groups attached to a polymer lattice [al~o: + 88, determined on 20 giL solution in methanol R.
consisting of polystyrene cross-linked with divinylbenzene. mp: 172 oC to 174 oc.
LlA (2.2.41): 14.24 x 10 3, determined at 304 nm on a 1.25 giL
Ion-exchange resin, strongly addic. 1085400. solution.
Resin in protonated form with sulfonic acid groups attached to
a lattice consisting of polystyrene cross-linked with 8 per cent N-Isobutyldodecatetraenarnide. C 16H 2S NO. (Mr 247.4).
of divinylbenzene. It is available as spherical beads; unless 1159500. [866602-52-0]. (2E,4E,8Z,lOEZ)-N-2-
otherwise prescribed, the particle size is 0.3 mm to 1.2 mm. (Methylpropyl)dodeca -2,4,8,10-tetraenamide.
Capacity. 4.5 mmol to 5 mmol per gram, with a water content White or almost white or non -coloured crystals.
of 50 per cent to 60 per cent. mp: about 70 oc.

General Notices (1) apply to al! monographs and other texts 477
4.1.1. Reagents EUROPEAN PHARMACOPOEIA 8.0

N-Isobutyldodecatetraenamide solution. 1159501. Isopropyl methanesulfonate. C 4 H IO 0 3 S. (M, 138.2). 1179400.


A solution of N-isobutyldodecatetraenamide R, exactly [926-06-7]. 1- methylethyl methanesulfonate.
weighed, in methanol R at a concentration of about Clear, colourless liquido
10 mg/mL. Content: minimum 99.0 per cent.
Isodrin. C 12 H 8 CI 6 • (M, 364.9). 1128700. [465-73-6]. Density: about 1.129 g/cm 3 (20 OC).
1,2,3,4,10,10-Hexachloro-l,4,4a,5,8,8a -hexahydro-endo,endo- n~o: 1.418-1.421.
1,4:5,8-dimethanonaphthalene. bp: abont 82 oC at 6 mm Hg.
Practically insoluble in water, soluble in common organic
solvents such as acetone. Isopropyl myristate. 1047200. [110-27-0].
A suitable certifled reference solution may be used. See Isopropyl myristate (0725).

Isoleudne. 1185000. [73-32-5]. 4-IsopropylphenoL C 9 H¡p. (M, 136.2). 1047300. [99-89-8].


See Isoleucine (0770). Content: minimum 98 per cent.
bp: about 212 oc.
Isomalt. C 12 H 2Pll' (Mr 344.3). 1164300. [64519-82-0].
mp: 59 oC to 61°C.
Mixture of 6-0-a-D-glucopyranosyl-D-glucitol and of
l-O-a -D-glucopyranosyl-D-mannito!. Isopulegol. CIOH¡SO. (M, 154.2). 1139600. [89-79-2].
White or almost white powder or granules, freely soluble in (-)-Isopulegol. (lR,2S,5R)-2-Isopropenyl-5-methyl-
water. cyclohexanol.
d~o: about 0.911.
Isomaltitol. C12H24011' (M, 344.3). 1161200. [534-73-6].
n~o: about 1.472.
6-0-a -D-Glucopyranosyl-D-glucitol.
bp: about 91°C.
White or almost white powder, freely soluble in water.
Isopulegol used in gas chromatography camplies with the
IsomenthoL C IO H 20 0. (M, 156.3). 1047000. [23283-97-8]. following additional test.
(+ )-Isomenthol: (lS,2R,5R)-2-isopropyl-5-methylcyclo- Assay. Gas chromatography (2.2.28) as prescribed in the
hexanol. (±)-Isomenthol: a mixture of equal parts of monograph Mint oil, partly dementholised (1838).
(1 S,2R,5R)- and (lR,2S,5S)- 2-isopropyl-5-methylcyclohexanol.
Content: minimum 99 per cent, calculated by the
Colourless crystals, practically insoluble in water, very soluble normalisation procedure.
in ethanol (96 per cent).
[al~o: (+ )-Isomenthol: about + 24, determined on a 100 giL Isoquercitroside. C 21 H 2aܡ2' (M, 464.4). 1136500.
solution in ethanol (96 per cent) R. [21637 -25-2]. Isoquercitrin. 2-(3,4- Dihydroxyphenyl)-3-(~-D­
glucofuranosyloxy)-5,7 -dihydroxy-4H -1- benzopyran -4-one.
bp: (+ )-Isomenthol: about 218 oc. (±)-Isomenthol: about
218 oc.
3,3',4',5,7 -Pentahydroxyflavone- 3 -glucoside.

mp: (+)- Isomenthol: about 80 oc. (±)- Isomenthol: about 53 oc. Isosilibinin. C2SH2201O' (M, 482.4). 1149900. [72581-71-6].
3,5,7 -Trihydroxy- 2- [2-( 4- hydroxy-3-methoxyphenyl)- 3-
(+)-Isomenthone. CIOH¡p. (M, 154.2). 1047100. hydroxymethyl-2,3 -dihydro-l,4-benzodioxin-6-yl] chroman-
(IR) -cis-p- Menthan-3 -one. (lR)-cis- 2-Isopropyl-5- 4-one.
methylcyclohexanone.
White to yellowish powder, practically insoluble in water,
Contains variable amounts of menthone. A colourless liquid, soluble in acetone and in methanol.
very slightly soluble in water, soluble in ethanol (96 per cent).
d~g : about 0.904. Kaolin, Hght. 1047400. [1332-58-7].
n~o: about 1.453. A purifled native hydrated aluminium silicate. It contains a
suitable dispersing agent.
[al~o: about + 93.2.
Light, white or almost white powder free from gritty particles,
Isomenthone used in gas chromatography complies with the unctuous to the touch, practically insoluble in water and in
following additional test.
mineral acids.
Assay. Gas chromatography (2.2.28) as prescribed in the
Coarse particles: maximum 0.5 per cent.
monograph Peppermint oil (0405).
Place 5.0 g in a ground-glass-stoppered cylinder about
Test solution. The substance to be examined.
160 mm long and 35 mm in diameter and add 60 mI. of a
Content: minimum 80.0 per cent, calculated by the 10 giL solution of sodium pyrophosphate R. Shake vigorously
normalisation procedure. and allow to stand for 5 mino Using a pipette, remove 50 mL
of the Iiquid from a point about 5 cm below the surface. To
Isomethyleugenol. C¡¡H¡P2' (M, 178.2).1181900. [93-16-3].
the remaining liquid add 50 mI. of water R, shake, allow to
1,2-Dimethoxy-4-prop-1-enylbenzene.
stand for 5 min and remove 50 mL as before. Repeat the
Isomethyleugenol used in gas chromatography complies with operatio11S until a total of 400 mL has been removed. Transfer
the following additional test. the remaining suspension to an evaporating dish. Evaporate
Assay. Gas chromatography (2.2.28) as prescribed in the to dryness on a water-bath and dry the residue to constant
monograph Niaouli oil, cineole type (2468). mass at 100-105 oc. The residue weighs not more than 25 mg.
Content: minimum 97.0 per cent, calculated by the Fine particles. Disperse 5.0 g in 250 mL of water R by shaking
normalisation procedure. vigorously for 2 mino Immediately pour into a glass cylinder
50 mm in diameter and, using a pipette, transfer 20 mL to
Isopropylamine. C3 H gN. (M, 59.1). 1119800. [75-31-0]. a glass dish, evaporate to dryness on a water-bath and dry
Propan-2-amine. to constant mas s at 100-105 oc. Allow the remainder of the
Colourless, highly volatile, flammable liquido suspension to stand at 20 oC for 4 h and, using a pipette with
n~o: about 1.374. its tip exactly 5 cm below the surface, withdraw a further
bp: 32 oC to 34 oc. 20 mL without disturbing the sediment, place in a glass dish,
evaporate to dryness 011 a water-bath and dry to constant mass
Isopropyl iodide. C 3H 7I. (M, 170.0). 1166600. [75-30-9]. at 100-105 oc. The mass of the second residue is not less than
2-Iodopropane. 70 per ce11t of that of the first residue.

478 See the ínformation section on general monographs (caver pages)


EUROPEAN PHARMACOPOEIA 8.0 4.1.1. Reagents

H-Keto-~-bo§wellic add. CJOH4604' (Mr 470.7). 1167600. Lactic acid. 1047800. [50-21-5].
[17019-92-0]. 3a- Hydroxy-l1-oxours-12-en-24-oic add. See Lactie acid (0458).
(4~) -3a -Hydroxy-11-oxours-12-en -23-oic acid.
White or almost white powder, insoluble in water, soluble in Lactic reagent. 1047801.
acetone, in anhydrous ethanol and in methanol. Solution A. To 60 mL of lactie acid R add 45 mL of
mp: 195 oC to 197 0e. previously filtered lactie acid R saturated without heating
with Sudan red G R; as lactic acid saturates slowly without
11-Keto-f3-boswellie acid used in liquid ehromatography
heating, an excess of colorant is always necessary.
complies with the following additional test
Solution B. Prepare 10 mL of a saturated solution of
Assay. Liquid chromatography (2.2.29) as prescribed in the
aniline R. Filter.
monograph Indian frankincense (2310).
Solution C. Dissolve 75 mg of potassium iodide R in water
Content: minimum 90 per cent, calculated by the
and dilute to 70 mL with the same solvent. Add 10 mL of
normalisation procedure.
ethanol (96 per cent) R and 0.1 g of iodine R. Shake.
Kieselguhr for chromatography. 1047500. Mix solutions A and B. Add solution e.
White or yellowish-white, light powder, practically insoluble
in water, in dilute acids and in organic solvents. Lacíobionic acid. C12H22012' (Mr 358.3). 1101600. [96-82-2].
White or almost white, crystalline powder, freely soluble in
Filtration rateo Use a chromatography column 0.25 m long and
water, practically insoluble in ethanol (96 per cent).
10 mm in internal diameter with a sintered-glass (100) plate
and two marks at 0.10 m and 0.20 m aboye the plateo Place mp: about 115 0e.
sufficient of the substance to be examined in the column to Lactose. 1047900. [5989-81-1].
reach the first mark and fill to the second mark with water R.
When the first drops begin to flow from the column, fill to See Lactose (0187).
the second mark again with water R and measure the time ~-Lactose. C12H22011' (Mr 342.3). 1150100. [5965-66-2].
required for the first 5 mL to flow from the column. The flow ~- D-Lactose.
rate is not less than 1 mL/min.
White or slightly yellowish powder.
Appearance of the eluate. The eluate obtained in the test for Content: minimum 99 per cent.
filtration rate is colourless (2.2.2, Method I).
a-D-Lactose: not greater than 35 per cent.
Acidity or alkalinity. To 1.00 g add 10 mL of water R, shake
vigorously and allow to stand for 5 mino Filter the suspension Assay. Gas chromatography (22.28) : use the normalisation
on a filter previously washed with hot water R until the procedure.
washings are neutral. To 2.0 mL of the filtrate add 0.05 mL Column:
of methyl red solution R; the solution is yellow. To 2.0 mL of - size: 1= 30 m, 0 = 0.25 mm;
the filtrate add 0.05 mL of phenolphthalein solution R1 ; the - stationary phase: poly{(cyanopropyl)(phenyl)] [dimeth-
solution is at most slightly pink. yl]siloxane R (film thickness 1 flm).
Water-soluble substances. Place 10.0 g in a chromatography Carrier gas: helium for chromatography R.
column 0.25 m long and 10 mm in internal diameter and elute
with water R. Collect the first 20 mL of eluate, evaporate to Temperature:
dryness and dry the residue at 100 oC to 105 0e. The residue Time Temperature
weighs 110t more than 10 mg. (min) CC)
Iron (2.4.9): maximum 200 ppm. Column 0-32.5 20 -7 280
To 0.50 g add 10 mL of a mixture of equal volumes of Injection port 250
hydrochloric acid Rl and water R, shake vigorously, allow to
Detector 250
stand for 5 min and filter. 1.0 mL of the filtrate compEes with
the test for iron.
Detection: flame ionisation.
Loss on ignition: maximum 0.5 per cent. During heating to
Injection: an appropriate derivatised sample.
red heat (600 ± 50 OC) the substance does not become brown
or black. a-Lactose monohydrate. C12H22011'H20. (Mr 360.3).
1150000. [5989-81-1]. a-D-Lactose monohydrate.
Kieselguhr G. 1047600.
White or almost white powder.
Consists of kieselguhr treated with hydrochloric acid and
calcined, to which is added about 15 per cent of calcium Content: minimum 97 per cent.
sulfate hemihydrate. f3-D-Lactose: less than 3 per cent.
A fine greyish-white powder; the grey colour becomes more Assay. Gas chromatography (2.2.28): use the normalisation
pronounced on triturating with water. The average particle procedure.
size is 10-40 flm. Column:
Calcium sulfate contento Determine by the method prescribed - size: 1 = 30 m, (2) = 0.25 mm;
for silica gel G R. - stationary phase: poly(dimethyl)siloxane R (film thickness
pH (2.2.3). Shake 1 g with 10 mL of carbon dioxide-free 1 flm).
water R for 5 mino The pH of the suspension is 7 to 8. Carrier gas: helium for chromatography R.
Chromatographie separation. Thin-layer chromatography Temperature:
(2.227). Prepare plates using a slurry of the kieselguhr G
with a 2.7 giL solution of sodium acetate R. Apply 5 ¡.tL of a Time Temperature
solution containing 0.1 giL oflactose, sucrose, glucose and (min) (oC)
fructose in pyridine R. Develop over a path of 14 cm using a Column o 12.5 230 -7 280
mixture of 12 volumes of water R, 23 volumes of 2-propanol R Injection port 250
and 65 volumes of ethyl acetate R. The migration time of
the solvent is about 40 mino Dry, spray onto the plate about Detector 280
10 mL of anisaldehyde solution R and heat for 5-10 min at
100-105 oc. The chromatogram shows four well-defined spots Detection: flame ionisation.
without tailing and well separated from each other. 1njection: an appropriate derivatised sample.

General Notices (1) apply to all monographs and other texts 479
4.1.1. Reagents EUROPEAN PHARMACOPOEIA 8.0

Lanatoside C. C49H760Z0' (M, 985). 1163300. [17575-22-3]. Test solution. The substance to be examined.
3~- [(~-D-Glucopyranosyl-(1-+4)-3-0-acetyl-2,6- Content: minimum 93.0 per cent, calculated by the
dideoxy-~- D-ribo- hexopyranosyl-( 1-+4)-2,6-dideoxy- normalisatiol1 procedure.
~- D-ribo- hexopyranosyl- (1-+4)-2,6-dideoxy- ~- D-ribo-
hexopyranosyl)oxy ]-12~, 14-dihydroxy-5~-card -20(22)- Lead aeetate. C4 H 6 0 4 Pb,3Hp. (M, 379.3). 1048100.
enolide. [6080-56-4]. Lead di-acetate.
Long, flat prisms obtained after recrystallisation in ethanol Colourless crystals, efflorescent, freely soluble in water, soluble
(96 per cent), freely soluble in pyridine and in dioxane. in ethanol (96 peY cent).
Lanthanum chloride heptahydrate. LaCI J ,7Hp. (M, 371.4). Lead acetate cotton. 1048101.
1167200. Immerse absorbent cotton in a mixture of 1 volume
White Ol' almost white powder or colourless crystals, freely of di/ute aeetie acid R and 10 volumes of lead aeetate
soluble in water. solution R. Drain off the excess of liquid, without squeezing
the cotton, by placing it 011 severallayers of filter papero
Lanthanum chloride solution. 1114001. Allow to dry in airo
To 58.65 g of lanthanum trioxide R slowly add 100 mL of Storage: in an airtight container.
hydroehlorie aeid R, Heat to boiling. Allow to cool and dilute
to 1000.0 mL with water R, Lead acetate papero 1048102.
Lanthanmn nitraíe. La(NO J )3,6Hp. (M, 433.0). 1048000. Immerse filter paper weighing about 80 g/m Z in a mixture
[10277-43-7]. Lal1thanum trinitrate hexahydrate. of 1 volume of di/ute aeetie acid R and 10 volumes of lead
aceta te solution R. After drying, cut the paper into strip s
Colourless crystals, deliquescent, freely soluble in water.
15 mm by 40 mm.
Storage: in an airtight container.
Lead acetate solution. 1048103.
Lanthanum nitrate solution. 1048001.
A 95 giL solution in earbon dioxide-free water R.
A 50 giL solution.
Lead dioxide. Pb0 2 • (Mr 239.2). 1048200. [1309-60-0].
Lanthanum trioxide. LazO), (M, 325.8). 1114000.
[1312-81-8]. Dark brown powder, evolving oxygen when heated, practically
insoluble in water, soluble in hydrochloric acid with evolution
An almost white, amorphous powder, practically insoluble in of chlorine, soluble in dilute nitric acid in the presence of
water R. It dissolves in dilute solutions of mineral acids and hydrogen peroxide, oxalic acid or other reducing agents,
absorbs atmospheric carbon dioxide. soluble in hot, concentrated alkali hydroxide solutions.
Calcium: maximum 5 ppm.
Lead nitrate. Pb(N0 3)2' (M, 331.2). 1048300. [10099-74-8].
Laude acid. C 12 H 24 0Z' (Mr 200.3). 1143100. [143-07-7]. Lead dinitrate.
Dodecanoic acid.
White or almost white, crystalline powder or colourless
White OI almost white, crystalline powder, practically crystals, freely soluble in water.
insoluble in water, freely soluble in ethanol (96 per cent).
mp: about 44 oc. Lead nitrate solution. 1048301.
Laurie aeid used in the assay of total fatty aeids in Saw pa/metto A 33 giL solution.
fruit (1848) complies with the following additiona/ test. Lead subacetate solution. 1048400. [1335-32-6]. Basic lead
Assay. Gas chromatography (2.2.28) as prescribed in the acetate solution.
monograph Saw palmetto fruít (1848). Content: 16.7 per cent m/m to 17.4 per ceut m/m of Pb
Content: minimum 98 per cent, calculated by the (A r 207.2) in a form corresponding approximately to the
normalisation procedure. formula C8H¡401OPby
Lauryl alcohoL C12 H 26 0. (Mr 186.3). 1119900. [112-53-8]. Dissolve 40.0 g of lead aeetate R in 90 mL of earbon dioxide-free
Dodecan-1-01. water R. Adjust the pH to 7.5 with strong sodium hydroxide
d~g : about 0.820.
solution R. Centrifuge and use the clear colourless supernatant
solution.
mp: 24 oC to 27 oc.
The SOIUtiOll remains clear when stored in a well-closed
Content: minimum 98.0 per cent, determined by gas container.
chromatography.
Leiocarposide. C27H340l6' (M, 614.5). 1150200.
LavanduloL ClOH¡p. (M, 154.2). 1114100. [498-16-8].
[71953-77 -O]. 2-(~-D-Glucopyranosyloxy)benzyl
(R)-5- Methyl-2-(l-methylethenyl)-4-hexen-1-01.
3 - (~- D-gl ucopyranosyloxy) -6-hydroxy-2-methoxybenzoate.
Oily liquid with a characteristic odour. 2- [[ [3-( ~- D-Glucopyranosyloxy)-6-hydroxy-2-
Lavandulol used in gas ehromatography complies with the methoxybenzoyl] oxy] methyl] p henyl- ~ -D-glucopyranoside.
following additional test. White or almost white powder, soluble in water, freely soluble
Assay. Gas chromatography (2.2.28) as prescribed in the in methanol, slightly soluble in ethanol (96 per cent).
monograph Lavender oil (1338). mp: 190 oC to 193 oc.
Test solution. The substance to be examined.
Lernon oil. 1101700.
Content: minimum 90.0 per cent, calculated by the
normalisation procedure. See Lemon oil (0620).

Lavandulyl acetate. C 12 H 20 0 2. (M, 196.3). 1114200. Leucine. 1048500. [61-90-5].


[25905-14-0]. 2-Isopropenyl-5-methylhex-4-en-1-yl acetate. See Leueine (0771).
Colourless liquid with a characteristic odour.
Levodopa. 1170000. [59-92-7].
Lavandulyl aeetate used in gas eh ro m atography complies with
See Levodopa (0038).
the following additional test.
Assay. Gas chromatography (2.2.28) as prescribed in the (Z)-Ligustilide. C 1Z H¡Pz. (Mr 190.2).1180300. [81944-09-4].
monograph Lavender oil (1338). (3Z)-3- Butylidene-1,3,4,5-tetrahydroisobenzofuran -l-one.

480 See the information section on general monographs (cover pages)


EUROPEAN PHARMACOPOEIA 8.0 4.1.1. Reagents

Limonene. CIOHl6' (Mr 136.2). 1048600. [5989-27-5]. Linolenic acid. ClsH3002' (Mr 278.4). 1143300. [463-40-1].
D- Límonene. (+ )-p- Mentha-I,8-diene. (R)-4- Isopropenyl-1- (9Z, 12Z, 15Z)-Octadeca-9,12,15-trienoie acid. a-Linolenie
methylcyclohex -l-ene. acid.
Colourless liquid, practieally insoluble in water, soluble in Colourless liquid, practically insoluble in water, soluble in
ethanol (96 per cent). organie solvents.
d~g : about 0.84. d¡o: about 0.915.
n~o: 1.471 to 1.474. nbo:about 1.480.
[al~o: about + 124. Linolenic acid used in the assay of total fatty acids in Saw
bp: 175 oC to 177 oc. palmetto fruit (1848) complíes with the following additional
test.
Limonene used in gas chromatography complies with the
following additional test. Assay. Gas chromatography (2.2.28) as prescribed in the
monograph Saw palmetto fruit (1848).
Assay. Gas chromatography (2.2.28) as prescribed in the
monograph Peppermint oil (0405). Content: minimum 98 per cent, calculated by the
normalisation procedure.
Test solution. The substance to be examined.
Content: minimum 99.0 per cent, calculated by the Linolenyl alcohol. C 1s H 3p. (Mr 264.4). 1156200.
normalisation procedure. [24149-05-1]. (9Z, 12Z,15Z)-Octadeca-9,12,15-trien -1-01.
a -Linolenyl alcohol.
Linalol. CIOHI80. (Mr 154.2). 1048700. [78-70-6].
Content: minimum 96 per cenl.
(RS)-3,7 - Dimethylocta -1 ,6-dien -3-0l.
Mixture of two stereoisomers (licareol and coriandrol). Linoleyl alcohoL C 1s H 3p. (Mr 266.5). 1155900. [506-43-4].
Líquid, practieally insoluble in water. (9Z, 12Z) -Octadeca -9, 12-dien-l-ol.

d~g : about 0.860. Relative density: 0.830.


n~o : about 1.462. Content: minimum 85 per cent.
bp: about 200 oc. Linsidomine hydrochloride. C6HllCIN402' (Mr 206.6).
LinaZol used in gas chromatography eomplies with the following 1171200. [16142-27-1]. 3-(Morpholin-4-yl)sydnonimine
test. hydrochloride. 3-(Morpholin-4-yl)-1,2,3-oxadiazol-3-ium-5-
Assay. Gas chromatography (2.2.28) as prescribed in the aminide hydrochloride.
monograph Anise oil (0804). White or almost white powder.
Test solution. The substance to be examined. Liquid scintillation cocktaiL 1167300.
Content: minimum 98.0 per cent, calculated by the Commercially available solution for the determination of
normalisation procedure. radioactivity by liquid scintillation counting. It contains
Linalyl acetate. C 12 H 2oÜ2' (Mr 196.3). 1107200. [115-95-7]. one or more fluorescent agents and mostly one or more
(RS)-1,5- Dimethyl-l-vinylhex-4-enyl acetate.
emulsiíying agents in a suitable organie solvent or mixture of
organic solvents.
Colourless or slightly yellow liquid with a strong odour of
bergamot and laven der. Liquid scintiHation cocktail Rl. 1176800.
d~~: 0.895 to 0.912. To 1000 mL of dioxan R, add 0.3 g of methylphenyloxazolylben-
n~o: 1.448 to 1.451. zene R, 7 g of diphenyloxazole R and 100 g of naphthalene R.
bp: about 215 oc. Lithium. Li. (A r 6.94). 1048800. [7439-93-2].
Linalyl acetate used in gas ehromatography complies wíth the A soft metal whose freshly cut surface is silvery-grey. It rapidly
following additional test. tarnishes in contact with airo It reacts violently with water,
Assay. Gas chromatography (2.2.28) as prescribed in the yielding hydrogen and giving a solution of lithium hydroxide;
monograph Bitter-orange-flower oil (1175). soluble in methanol, yielding hydrogen and a solution of
Test solution. The substance to be examined. lithium methoxide; practically insoluble in light petroleum.
Content: minimum 95.0 per cent, calculated by the Storage: under Iight petroleum or liquid paraffin.
normalisation procedure.
Lithium carbonate. Li 2C0 3. (Mr 73.9). 1048900. [554-13-2].
Lindane. C6H 6 C1 6 • (Mr 290.8). 1128900. [58-89-9]. Dilithium carbonate.
y-Hexachlorocyclohexane. White or almost white, light powder, sparingly soluble
For the monograph Woolfat (0134), a suitable certified in water, very slightly soluble in ethanol (96 per cent). A
reference solution (lO ng/¡..tL in cyclohexane) maybe used. saturated solution at 20 oC contains about 13 giL of Li2CO}'

Linolek acid. ClsH3202' (Mr 280.5). 1143200. [60-33-3]. Lithium chloride. LíCl. (Mr 42.39). 1049000. [7447-41-8].
(9Z,12Z)-Octadeca-9,12-dienoie acid. Crystalline powder or granules or cubie crystals, deliquescent,
Colourless, oily liquido íreely soluble in water, soluble in acetone and in ethanol
(96 per cent). Aqueous solutions are neutral or slightly
d¡o: about 0.903.
alkaline.
n~o: about 1.470.
Storage: in an airtight container.
Linoleie acid used in the assay of total fatty acids in Saw
palmetto fruit (1848) complies with the following additional Lithium hydroxide. LiOH,H 20. (Mr 41.96). 1049100.
test. [1310-66-3]. Lithium hydroxide monohydrate.
Assay. Gas chromatography (2.2.28) as prescribed in the White or almost white, granular powder, strongly alkaline, it
monograph Saw palmetto fruit (1848). rapidly absorbs water and carbon dioxide, soluble in water,
Content: minimum 98 per cent, calculated by the sparingly soluble in ethanol (96 per cent).
normalisation procedure. Storage: in an airtight container.

General Notiees (1) apply to all monographs and other texts 481
4. L L Reagents EUROPEAN PHARMACOPOEIA 8.0

Lithium metaborate, anhydrous. LiB0 2. (M, 49.75). Absorbance (2.2.25). A solution in methanol R shows
1120000. [13453-69-5]. absorption maxima at 255 nm, 267 nm, 290 nm and 350 nm.
Lithium sulfate. Li 2S0 4,Hp. (Mr 128.0). 1049200. mp: about 247°C.
[10102-25-7]. Dilithium sulfate monohydrate.
Macrogol 23 lauryl ether. 1129000.
Colourless crystals, freely soluble in water, practically
See Macrogollauryl ether (1124), the number of moles of
insoluble in ethanol (96 per cent).
ethylene oxide reacted per mole of lauryl alcohol being 23
Lithium trifluoromethanesulfonate. CF 3 Li0 3 S. (Mr 156.0). (nominal value).
1173400. [33454-82-9].
Macrogo1200. 1099200. [25322-68-3]. Polyethyleneglycol
Litmus. 1049300. [1393-92-6]. 200.
Schultz No. 1386. Clear, colourless or almost colourless viscous liquid, very
Indigo-blue fragments prepared from various species of soluble in acetone and in anhydrous ethanol, practically
Rocella, Lecanora or other lichens, soluble in water, practically insoluble in fatty oils.
insoluble in ethanol (96 per cent). d~g: about 1.127.
Colour ehange: pH 5 (red) to pH 8 (blue). n~o: about 1.450.
Litmus paper, bIue. 1049301.
Macrogol200 Rl. 1099201.
BoillO parts of coarsely powdered litmus R for 1 h with
Introduce 500 mL of macrogol 200 R into a 1000 mL round
100 parts of ethanol (96 per cent) R. Decant the alcohol
bottom flask. Using a rotation evaporator remove any
and add to the residue a mixture of 45 parts of ethanol
volatile components applying for 6 h a temperature of 60 oC
(96 per cen!) R and 55 parts of water R. After 2 days decant
and a vacuum with a pressure of 1.5-2.5 kPa.
the clear liquido Impregnate strips of filter paper with the
solution and allow to dry. Macrogol300. 1067100. [25322-68-3]. Polyethyleneglycol
Test for sensitivity. Immerse a strip measuring 10 mm by 300.
60 mm in a mixture of 10 mL of 0.02 M hydrochloric acid See Macrogols (1444).
and 90 mL of water R. On shaking the paper turns red
within 45 s. Macrogol400. 1067200. [25322-68-3]. Polyethyleneglycol
400.
Litmus paper, red. 1049302.
To the blue litmus extract, add dilute hydrochloric acid R See Macrogols (1444).
dropwise until the blue colour becomes red. Impregnate MacrogollOOO. 1067300. [25322-68-3]. Polyethyleneglycol
strips of filter paper with the solution and allow to dry. 1000.
Test for sensitivity. Immerse a strip measuring 10 mm by See Macrogols (1444).
60 mm in a mixture of 10 mL of 0.02 M sodium hydroxide
and 90 mL of water R. On shaking the paper turns blue Macrogol1500. 1067400. [25322-68-3]. Polyethyleneglycol
within 45 s. 1500.
Loganin. C¡7H260¡O' (Mr 390.4). 1136700. [18524-94-2]. See Macrogols (1444).
Methyl (1 S,4aS,6S, 7R, 7aS)-l- (~- D-glucopyranosyloxy)-
6-hydroxy-7 -methyl-l ,4a,5,6, 7, 7a-hexahydro- Macrogol 20 000. 1067600. Polyethyleneglycol 20 000.
cyclopenta[ e] pyran -4-carboxylate. See Macrogols (1444).
mp: 220 oC to 221°C. Macrogo120 000 2-nitroterephthalate. 1067601.
Longifolene. C 1s H 24 . (Mr 204.4). 1150300. [475-20-7]. Polyethyleneglycol20 000 2-nitroterephthalate.
(1 S,3aR,4S,8aS)-4,8,8- Trimethyl-9-methylenedecahydro-1,4- Macrogol 20 000 R modified by treating with
methanoazulene. 2-nitroterephthalate acid.
Oily, colourless liquid, practically insoluble in water, miscible A hard, white or almost white, waxy solid, soluble in
with ethanol (96 per cent). acetone.
dl 8 : 0.9319.
Magnesium. Mg. (A r 24.30). 1049500. [7439-95-4].
n~o: 1.5050.
[al~o: + 42.7.
Silver-white ribbon, turnings or wire, or a grey powder.
bp: 254 oC to 256 oc. Magnesium acetate. C 4 H 6Mg04,4H 20. (Mr 214.5). 1049600.
Longifolene used in gas chromatography complies with the [16674-78-5]. Magnesium diacetate tetrahydrate.
following additional test. Colourless crystals, deliquescent, freely soluble in water and
Assay. Gas chromatography (2.2.28) as prescribed in the in ethanol (96 per cent).
monograph Turpentine oil, Pinus pinaster type (1627). Storage: in an airtight container.
Content: minimum 98.0 per cent, calculated by the
normalisation procedure. Magnesium chloride. 1049700. [7791-18-6].
See Magnesium ehloride hexahydrate (0402).
Low-vapour-pressure hydrocarbons (type L). 1049400.
Unctuous mass, soluble in benzene and in toluene. Magnesium nitrate. Mg(N0 3 )2,6Hp. (Mr 256.4). 1049800.
[13446-18-9]. Magnesiumnitrate hexahydrate.
Lumiflavine. C 13 H 12 NP2' (Mr 256.3). 1141000. [1088-56-8].
7,8,10-Trimethylbenzo [g]pteridine- 2,4(3H, 1OH)-dione. Colourless, clear crystals, deliquescent, very soluble in water,
freely soluble in ethanol (96 per cent).
Yellow powder or orange crystals, very slightly soluble in
water, freely soluble in methylene chloride. Storage: in an airtight container.

Luteolin-7-glucoside. C 21 H 2aÜll' (M, 448.4). 1163400. Magnesium nitrate solution. 1049801.


[5373-11-5]. 2-(3,4-Dihydroxyphenyl)-7 -(~-D­ Dissolve 17.3 g of magnesium nitrate R in 5 mL of water R
glucopyranosyloxy)- 5-hydroxy-4H-l-benzopyran -4-one. warming gently and add 80 mL of ethanol (96 per cent) R.
Yellow powder. Cool and dilute to 100.0 mL with the same solvent.

482 See the information seetion on general monographs (cover pages)


EUROPEAN PHARMACOPOEIA 8.0 4.1. L Reagents

Magnesium nitrate solution Rl. 1049802. Maleic anhydride. C4HP3' (Mr 98.1). 1050700. [108-31-6].
Dissolve 20 g of magnesium nitrate R (Mg(N0 3)2,6HP) Butenedioie anhydride. 2,5-Furandione.
in deionised distilled water R and dilute to 100 mL with White or almost white crystals, soluble in water forming
the same solvent. lmmediately before use, dilute 10 mL to maleic acid, very soluble in acetone and in ethyl acetate, freely
100 mL with deíonised distilled water R. A volume of 5 flL soluble in toluene, soluble in ethanol (96 per cent) with ester
will provide 0.06 mg of Mg (NOJ2' formation, very slightly soluble in light petroleum.
mp: about 52 oc.
Magnesium oxide. 1049900. [1309-48-4].
See Light magnesíum oxide (0040). Any residue insoluble in toluene do es not exeeed 5 per eent
(maleic acid).
Magnesium oxide Rl. 1049901.
Maleic anhydride solution. 1050701.
Complies with the requirements prescribed for magnesium
oxide R with the following modifications. Dissolve 5 g of maleie anhydride R in toluene R and dilute
to 100 mL with the same solvent. Use within one month.
Arseníe (2.4.2, Method A) : maximum 2 ppm. lf the solution becomes turbid, filter.
Dissolve 0.5 g in a mixture of 5 mL of water R and 5 mL of
hydrochloric acid R1. Maltitol. 1136800. [585-88-6].
Heavy metals (2.4.8): maximum 10 ppm. See Maltitol (1235).
Dissolve 1.0 g in a mixture of 3 mL of water R and 7 mL Maltotriose. C 1SH 3 P¡6' (M, 504.4). 1176300. [1109-28-0].
of hydrochloric acid R1. Add 0.05 mL of phenolphthalein a-o-Glucopyranosyl-(1-74)-a-D-glueopyranosyl-(1-74)- 0-
solution R and concentrated ammonia R until a pink colour glueose.
is obtained. Neutralise the excess of ammonia by the
addition of glacial acetic acid R. Add 0.5 mL in excess and White or almost white, erystalline powder, very soluble in
dilute to 20 mL with water R. Filter, if necessary. 12 mL of water.
the solution complies with test A. Prepare the reference mp: about 134 oc.
solution using a mixture of 5 mL of lead standard solution
(1 ppm Pb) R and 5 mL of water R. Mandelic add. CSHSO J . (M, 152.1). 1171300. [90-64-2].
2-Hydroxy-2-phenylaeetie acid.
Iron (2.4.9): maximum 50 ppm.
White crystalline flakes, soluble in water.
Dissolve 0.2 g in 6 mL of dilute hydrochloric acid R and
dilute to 10 mL with water R. mp: 118 to 121°C.

Magnesium oxide, heavy. 1050000. [1309-48-4]. Manganese sulfate. MnS0 4,Hp. (M, 169.0). 1050900.
[10034-96-5]. Manganese sulfate monohydrate.
See Heavy magnesium oxide (0041).
Pale-pink, crystalline powder or erystals, freely soluble in
Magnesium silicate for pestidde residue analysis. 1129100. water, practieally insoluble in ethanol (96 per cent).
[1343-88-0]. Loss on ignition: 10.0 per eent to 12.0 per eent, determined on
Magnesium silicate for chromatography (60-100 mesh). 1.000 g at 500 ± 50 oc.
Magnesium sulfate. 1050200. [10034-99-8]. Mannitol. 1051000. [69-65-8].
See Magnesium sulfate heptahydrate (0044). See Mannitol (0559).
Magnolol. C¡SH¡aÜ2' (Mr 266.3). 1182800. [528-43-8]. Mannose. C6H1206' (Mr 180.2). 1051100. [3458-28-4].
5,5'-Di(prop-2-enyl)biphenyl-2,2'-diol. 5,5'-Diallyl-2,2'- 0-(+ )-Mannose.
dihydroxybiphenyl. 5,5'-Di -2-propenyl- [1,1' -biphenyl]-2,2'- white or almost white, crystalline powder or small crystals,
diol. very soluble in water, slightly soluble in anhydrous ethanol.
Maize oH. 1050400. [al~o: + 13.7 + 14.7, determined on a 200 giL solution in
See Maize oil, refíned (1342). water R containing about 0.05 per eent of NH J.
mp: about 132 oC, with deeomposition.
Malachite green. C 23 H 2s ClN 2 • (Mr 364.9). 1050500.
[123333-61-9]. Marrubiin. C2oH2S04' (Mr 332.4). 1158300. [465-92-9].
Schultz No. 754. (2aS,SaS,6R, 7R,8aR,8bR)-6- [2- (Furan -3-yl)ethyl]-6-hydroxy-
2a,Sa, 7 -trimethyldeeahydro- 2H- naphtho [1 ,8-be] furan -2-one.
Colour lndex No. 42000.
[4- [[ 4- (Dimethylamino )phenyl] phenylmethylene] cyclohexa- Colourless, microerystalline powder.
2,S-dien -l-ylidene] dimethylammonium chloride. Marrubiin used in liquid chromatography complies with the
Green crystals with a metallie lustre, very soluble in water following additional test.
giving a bluish-green solution, soluble in ethanol (96 per eent) Assay. Liquid chromatography (2.2.29) as preseribed in the
and in methanol. monograph White horehound (1835).
Absorbanee (2.2.25). A 0.01 giL solution in ethanol (96 per Content: minimum 95.0 per cent, calculated by the
cent) R shows an absorption maximum at 617 nm. normalisation proeedure.
Malachite green solution. 1050501. Medozine dihydrochloride. 1051200. [1104-22-9].
A 5 giL solution in anhydrous acetic acid R. See Meclozine dihydrochloride (0622).
Malathion. C¡OH¡P6PS2' (M,. 330.3). 1129200. [121-75-5]. Melamine. CJH6N6' (M, 126.1). 1051300. [108-78-1].
bp: about 156 oc. 1,3,5-Triazine-2,4,6-triamine.
A suitable eertified referenee solution (lO ng/flL in iso-oetane) A white or almost white, amorphous powder, very slightly
may be used. soluble in water and in ethanol (96 per eent).

Maleic add. 1050600. [110-16-7]. Menadione. 1051400. [58-27-5].


See Maleic acid (0365). See Menadione (0507).

General Notices (1) apply to all monographs and other texts 483
4.1.1. Reagents EUROPEAN PHARMACOPOEIA 8.0

Menthofuran. ClQH140. (Mr 150.2). 1051500. Mercuric acetate. C4H6Hg04' (Mr 318.7). 1052000.
[17957 -94-7]. 3,9-Epoxy-p-mentha-3,8-diene. [1600-27-7]. Mercury diacetate.
3,6-Dimethyl-4,5,6, 7 -tetrahydro-benzofuran. White or almost white crystals, freely soluble in water, soluble
Slightly bluish liquid, very slightly soluble in water, soluble in in ethanol (96 per cent).
etharrol (96 per cerrt).
Mercuric acetate solution. 1052001.
dig: about 0.965.
Dissolve 3.19 g of mercuric acetate R in anhydrous acetic
n~o: about 1.480.
acid R and dilute to 100 mL with the same acid. If necessary,
[al~o: about + 93. neutralise the solution with 0.1 M perchloric acid using
bp: 196 oc. 0.05 mL of crystal violet solution R as indicator.
Menthofuran used in gas chromatography complies with the Mercurk bromide. HgBr z. (Mr 360.4). 1052100. [7789-47-1].
following additional test.
Mercury dibromide.
Assay. Gas chromatography (2.2.28) as prescribed in the
White or faintly yellow crystals or a crystalline powder, slightly
monograph Peppermint oil (0405).
soluble in water, soluble in ethanol (96 per cent).
Test solution. The substance to be examined.
Content: minimum 97.0 per cent, calculated by the Mercuric bromide papero 1052101.
normalisation procedure. In a rectangular dish place a 50 giL solution of mercuric
bromíde R in anhydrous ethanol R and immerse in it pieces
MenthoL 1051600. [2216-51-5]. of white filter paper weighing 80 g per square metre (speed
See Levomenthol (0619) and Racemic menthol (0623). of filtration = filtration time expressed in seconds for
Menthol used in gas chromatography complies with the 100 mL of water at 20 oC with a filter surface of 10 cm 2 and
following additíonal test. constant pressure of 6.7 kPa: 40 sto 60 s), each measuring
Assay. Gas chromatography (2.2.28) as prescribed in the 1.5 cm by 20 cm and folded in two. Allow the excess liquid
related substances test included in the monograph Racemic to drain and allow the paper to dry, protected from light,
menthol (0623). suspended over a non-metallic thread. Discard 1 cm from
each end of each strip and cut the remainder into 1.5 cm
Content: minimum 98.0 per cent, calculated by the
normalisation procedure. squares or discs of 1.5 cm diameter.
Storage: in a glass-stoppered container wrapped with black
Menthone. CIOH[p. (Mr 154.2). 1051700. [14073-97-3]. papero
(2S,5R)- 2-Isopropyl-5-methylcyclohexanone.
(- )-trans-p- Menthan-3-one. Mercurk chloride. 1052200. [7487-94-7].
Contains variable amounts of isomenthone. See Mercuric chloride (0120).
Colourless liquid, very slightly soluble in water, very soluble Mercuric chloride solution. 1052201.
in ethanol (96 per cent).
A 54 giL solution.
d~g: about 0.897.
n~o: about 1.450. Mercuric iodide. HgI2' (Mr 454.4). 1052300. [7774-29-0].
Mercury di-iodide.
Menthone used in gas chromatography complies with the
followíng additional test. Dense, scarlet, crystalline powder, slightly soluble in water,
sparingly soluble in acetone and in ethanol (96 per cent),
Assay. Gas chromatography (2.2.28) as prescribed in the
soluble in an excess of potassium iodide solution R.
monograph Peppermint oil (0405).
Storage: protected from light.
Test solution. The substance to be examined.
Content: minimum 90.0 per cent, calculated by the Mercuric nitrate. Hg(N0 3 )2,HzÜ. (Mr 342.6). 1052400.
normalisation procedure. [7783-34-8]. Mercury dinitrate monohydrate.
Colourless or slightly coloured crystals, hygroscopic, soluble
Menthyl acetate. C 11H 2P2' (Mr 198.3). 1051800. [2623-23-6].
in water in the presence of a small quantity of nitric acid.
2-Isopropyl-5-methylcyclohexyl acetate.
Colourless liquid, slightly soluble in water, miscible with
Storage: in an airtight container, protected from light.
ethanol (96 per cent). Mercuric oxide. HgOo (Mr 216.6). 1052500. [21908-53-2].
d~g: about 0.92. Yellow mercuric oxide. Mercury oxide.
n~o: about 1.447. A yellow to orange-yellow powder, practically insoluble in
bp: about 228 oc. water and in ethanol (96 per cent).
Menthyl acetate used in gas chromatography complies with the Storage: protected from light.
following additional test. Mercurk sulfate solution. 1052600. [7783-35-9].
Assay. Gas chromatography (2.2.28) as prescribed in the Dissolve 1 g of mercuric oxide R in a mixture of 20 mi of
monograph Peppermint oil (0405). water R and 4 mL of sulfuric acid R.
Test solution. The substance to be examined.
Content: minimum 97.0 per cent, calculated by the Mercurk thiocyanate. Hg(SCN)2' (Mr 316.7). 1052700.
normalisation procedure. [592-85-8]. Mercury di(thiocyanate).
White or almost white, crystalline powder, very slightly
2-Men:aptobenzimidazole. C 7 H 6N 1S. (Mr 150.2). 1170100. soluble in water, slightly soluble in ethanol (96 per cent),
[583-39-1]. lH-benzimidazole-2-thiol. soluble in solutions of sodium chloride.
mp: about 302 oc.
Mercuric thiocyanate solution. 1052701.
2-Mercaptoethanol. C2H 6 0S. (Mr 78.1). 1099300. [60-24-2]. Dissolve 0.3 g of mercuric thiocyanate R in anhydrous
Liquid, miscible with water. ethanol R and dilute to 100 mL with the same solvent.
d~g: about 1.116. Storage: use within 1 week.
bp: about 157 oc.
Mercury. Hg. (A, 200.6). 1052800. [7439-97-6].
Mercaptopurine. 1051900. [6112-76-1]. Silver-white liquid, breaking into spherical globules which do
See Mercaptopurine (0096). not leave a metallic trace when rubbed on papero

484 See the information section on general monographs (caver pages)


EUROPEAN PHARMACOPOEIA 8.0 4.1.1. Reagents

d~g: about 13.5. n~O : about 1.430.


bp: about 357 oc. Methanesulfonyl chloride. CH 3 CI0 2 S. (M, 114.6). 1181300.
Mercury, nitric add solution of. 1052801. [124-63-0).
Carefully dissolve 3 mL of mercury R in 27 mL of fuming Clear, colourless or slightly yellow liquido
nitric acid R. Dilute the solution with an equal volume of Content: minimum 99.0 per cent.
water R. Density: 1.48 g/cm 3 •
Storage: protected from light; use within 2 months. n~o : about 1.452.
Mesityloxide. C6H IO O. (M,. 98.1). 1120100. [141-79-7). bp: about 161°C.
4-Methylpent -3-en -2-one. Methanol. CHp. (M, 32.04). 1053200. [67-56-1).
Colourless, oily liquid, soluble in 30 parts of water, miscible Clear, colourless, flammable liquid, miscible with water and
with most organic solvents. with ethanol (96 per cent).
d~g : about 0.858. d~g: 0.791 to 0.793.
bp: 129 oC to 130 oc. bp: 64 oC to 65 oc.
Metanil yellow. ClsHI4N3Na03S, (11,11,375.4). 1052900. Methanol RL 1053201.
[587-98-4).
Complies with the requirements prescribed for methanol R
Schultz No. 169. and the following additional requirement.
Colour Index No. 13065. Minimum transmittance (2.2.25) using water R as
Sodium 3- [4-(phenylamino )phenylazo )benzenesulfonate. compensation liquid: 20 per cent at 210 nm, 50 per cent
A brownish-yellow powder, soluble in water and in ethanol at 220 nm, 75 per cent at 230 nm, 95 per cent at 250 nm,
(96 per cent). 98 per cent at 260 nm and at higher wavelengths.
Metanil yellow solution. 1052901. Methanol R2. 1053202.
A 1 giL solution in methanol R. Complies with the requirements prescribed for methanol R
Test for sensítivity. To 50 mL of anhydrous acetic acid R and the following additional requirements.
add 0.1 mL ofthe metanil yellow solution. Add 0.05 mL of Content: minimum 99.8 per cent.
0.1 M perchloric acid; the colour changes from pinkish-red Absorbance (2.2.25): maximum 0.17, determined at 225 nm
to violet. using water R as the eompensation liquido
Colour change: pH l.2 (red) to pH 2.3 (orange-yellow).
Methanol, hydrochloric. 1053203.
Metaphosphoric adJ. (HP0 3 )x' 1053000. [37267-86-0). Dilute 1.0 mL of hydrochloric acid Rl to 100.0 mL with
Glassy lumps or sticks containing a proportion of sodium methanol R.
metaphosphate, hygroscopic, very soluble in water.
Methanol, aldehyde-free. 1053300.
Nitra tes. Boill.O g with 10 mL of water R, cool, add 1 mL
of indigo carmine 50lution R, 10 mL of nitrogen-free sulfuric Dissolve 25 g of iodine R in 1 L of methanol R and pour the
acid R and heat to boiling. The blue colour is not entirely solution, with constant stirring, into 400 mL of 1 M sodium
discharged. hydroxide. Add 150 mL of water R and allow to stand for
16 h. Filter. Boil under a re flux condenser until the odour
Reducing substances: maximum 0.01 per cent, calculated as
of iodoform disappears. Distil the solution by fractional
H 3 P03"
distillation.
Dissolve 35.0 g in 50 mL of water R. Add 5 mL of a 200 giL
Aldehydes and ketones: maximum 0.001 per cent.
solution of sulfuric acid R, 50 mg of potassium bromide R and
5.0 mL of 0.02 M potassium bromate and heat on a water-bath Methanol, anhydrous. 1053400. [67-56-1).
for 30 mino AlIow to cool and add 0.5 g of potassium iodide R. Treat 1000 mL of methanol R with 5 g of magnesium R. lf
Titrate the liberated iodine with 0.1 M sodium thiosulfate, necessary initiate the reaction by adding 0.1 mL of mercuric
using 1 mL of starch solution R as indicator. Carry out a blank chloride solution R. When the evolution of gas has ceased,
test. distil the liquid and colleet the distillate in a dry container
1 mL of 0.02 M potassium bromate is equivalent to 4.10 mg protected from moisture.
ofH 3 P0 3 · Water (2.5.12): maximum 0.3 giL.
Storage: in an airtight container.
DL-Methionine. 1129400. [59-51-8).
Methacrylic add. C 4 H 60 2 . (M, 86.1). 1101800. [79-41-4). See DL-Methionine (0624).
2-Methylprop-2-enoic acid.
L- Methionine. 1053500. [63-68-3).
Colourless liquido
n~o : about 1.43l. See Methionine (1027).
bp: about 160 oc. (RS)-Methotrexate. C2oH22NsOs' 1120200. [60388-53-6).
mp: about 16 oc. (RS)-2- [4- [[ (2,4-diaminopteridin-6-yl)methyl)-
methylamino )benzoylamino )pentanedioic acid.
Methane. CH 4 • (Mr 16). 1166300. [74-82-8). Content: minimum 96.0 per cent.
Content: minimum 99.0 per cent V/V. mp: about 195 oc.
Methane Rl. CH 4 . (M, 16). 1176400. [74-82-8).
Methoxychlor. C I6 H 1S ClP2' (M, 345.7). 1129300. [72-43-5).
Content: minimum 99.995 per cent V/V. 1,1- (2,2,2-Trichloroethylidene) -bis( 4-methoxybenzene).
Methanesulfonic add. CHP3S, (M, 96.1). 1053100. Practically insoluble in water, freely soluble in most organic
[75-75-2). solvents.
Clear, colourless liquid, solidifying at about 20 oC, miscible bp: about 346 oc.
with water, slightly soluble in toluene, practically insoluble mp: 78 oC to 86 oc.
in hexane. A suitable certified reference solution (lO ng/llL in iso-octane)
d~g: about 1.48. maybe used.

General Notices (1) apply to all monographs and other texts 485
4.1.1. Reagents EUROPEAN PHARMACOPOEIA 8.0

trans-2-Methoxyeinnamaldehyde. C lO H lO 02' (M, 162.2). Content: minimum 99.5 per cent, determined by gas
1129500. [60125-24-8]. chromatography.
mp: 44 oC to 46 oc.
MethyI4-aminobenzoate. CSH 9N0 2. (M, 151.2). 1175600.
trans-2-Methoxycinnamaldehyde used in gas chromatography [619-45-4].
complíes wíth the foliowing additional test. mp: 110 oC to ll3 oc.
Assay. Gas chromatography (2.2.28) as prescribed in the
monograph Cassia oil (1496). 4-Methylaminophenol sulfate. C¡4H20NP6S, (Mr 344.4).
1053800. [55-55-0].
Content: minimum 96.0 per cent, calculated by the
normalisation procedure. Colourless crystals, very soluble in water, slightly soluble in
ethanol (96 per cent).
(lRS)-l-( 6-Methoxynaphthalen-2-yl)ethanoL mp: about 260 oc.
C 13H¡P2' (Mr 202.3). 1159600. [77301-42-9].
6-Methoxy-a -methyl-2-naphthalenemethanol. 3-(Methylamino)-1-phenylpropan-l-oL ClOH¡SNO.
White or almost white powder. (Mr 165.2). 1186400. [42142-52-9].
mp: about 113 oc. White or almost white powder.
mp: 59 oC to 64 oc.
1- (6- Methoxynaphthalen- 2-yl)ethanone. C13H120Z'
(M, 200.2). 1159700. [3900-45-6]. 6'-Methoxy-2'- Methyl anthranilate. CSH 9 N0 2. (M, 151.2). 1107300.
acetonaphthone. [134-20-3]. Methyl 2-aminobenzoate.
White or almost white powder. Colourless crystals or a colourless or yellowish Iiquid, soluble
mp: about 108 oc. in water, freely soluble in ethanol (96 per cent).
bp: 134 oC to 136 oc.
6-Methoxy-2-naphthoic aeid. C¡ZH lO 03' (Mr 202.2). mp: 24 oC to 25 oc.
1184200. [2471-70-7]. 6-Methoxynaphthalene-2-carboxylic
aeid. Methyl anthranílate used in gas chromatography complies with
the following additional test.
White or almost white, crystalline powder.
Assay. Gas chromatography (2.2.28) as prescribed in the
mp: 201 oC to 206 oc. monograph Bitter-orange-flower oil (1175).
Methoxyphenylacetic aeid. C9 H lO OJ' (Mr 166.2). 1053600. Test solution. The substance to be examined.
[7021-09-2]. (RS)-2-Methoxy-2-phenylacetic aeid. Content: minimum 95.0 per cent, calculated by the
White, crystalline powder or white or almost white crystals, normalisation procedure.
sparingly soluble in water, freely soluble in ethanol (96 per
Methyl arachidate. C21 H 4zÜ2' (Mr 326.6). 1053900.
cent).
[1120-28-1]. Methyl eicosanoate.
mp: about 70 oc.
Content: minimum 98.0 per cent, determined by gas
Methoxyphenylacetic reagent. 1053601. chromatography (2.4.22).
Dissolve 2.7 g of methoxyphenylacetic acíd R in 6 mL of White or yellow, crystalline mass, soluble in ethanol (96 per
tetramethylammonium hydroxide so/ution R and add 20 mL cent) and in light petroleum.
of anhydrous ethanol R. mp: about 46 oc.
Storage: in a polyethylene container. Methyl behenaíe. C23H4602' (Mr 354.6).1107500. [929-77-1].
3-Methoxy-L-tyrosine. ClOH13N04Hp. (Mr 229.2). 1164400. Methyl docosanoate.
[200630-46-2]. mp: 54 oC to 55 oc.
Off-white or yellow powder. Methyl benzenesulfonaíe. C7Hs03S, (M, 172.2). 1159800.
Methyl acetate. CJH602' (Mr 74.1). 1053700. [79-20-9]. [80-18-2].
Clear, colourless liquid, soluble in water, miscible with ethanol Clear, colourless liquido
(96 per cent). bp: about 148 oc.
d~g: about 0.933. Methyl benzoate. CSHsÜ2' (M, 136.2). 1164500. [93-58-3].
n5D: about 1.361. Benzoic acid, methyl ester.
bp: 56 oC to 58 oc. Colourless Iiquid.
d~o: 1.088.
MethyI4-acetylbenzoate. ClOHlO03' (Mr 178.2). 1154100.
[3609-53-8]. bp: about 200 oc.
mp: about 94 oc. Methylbenzothiazolone hydrazone hydrochloride.
C sH lO CIN 3S,Hp (Mr 233.7). 1055300. [38894-11-0].
Methy14-acetylbenzoate reagent. 1154101.
3-Methylbenzothiazol-2(3H)-one hydrazone hydrochloride
Dissolve 0.25 g of methyl4-acetylbenzoate R in a mixture of monohydrate.
5 mL of sulfuric acid R and 85 mL of cooled methanol R.
Almost white or yellowish, crystalline powder.
Methylal. C3HsÜ2' (Mr 76.1). 1173500. [109-87-5]. mp: about 270 oc.
Dimethoxymethane. Dioxapentane. Formaldehyde dimethyl Suitability far determination of aldehydes. To 2 mL of
acetal. Methylene dimethyl ether. aldehyde-free methanol R add 60 flL of a 1 giL solution of
Clear, colourless, volatile, flammable liquid, soluble in water propionaldehyde R in aldehyde-free methano/ R and 5 mL
and miscible with ethanol (96 per cent). of a 4 giL solution of methylbenzothiazolone hydrazone
d§g: about 0.860. hydrochloride. Mix. Allow to stand for 30 mino Prepare a
blank omitting the propionaldehyde solution. Add 25.0 mL of
n5D : about 1.354. a 2 giL solution of ferric chloride R to the test solution and to
bp: about 41°C. the blank, dilute to 100.0 mL with acetone R and mix. The
Methyla/ used in gas chromatography complies with the absorbance (2.2.25) of the test solution, measured at 660 11m
following additional test. using the blank as compensation Iiquid, is not less than 0.62.

486 See the information section on general monographs (cover pages)


EUROPEAN PHARMACOPOEIA 800 4.1.1. Reagents

(R)-( + )-a-Methylbenzyl isocyanate. CgHgNO. (Mr 147.2). Methyl decanoate. CllHzpz. (Mr 186.3). 1054000.
1171400. [33375-06-3]. (+)-(R)-a-Methylbenzyl isocyanate. [110-42-9].
(+)- [(lR)-l-Isocyanatoethyl]benzene. (+ )-(lR)-l-Phenylethyl Content: mínimum 99.0 per cent.
isocyanate. Clear, colourless or yellow liquid, soluble in Iight petroleum.
Content: minimum 99.0 per cent. d~g: 0.871 to 0.876.
Colourless liquido n5°: 1.425 to 1.426.
d~g: about 1.045. Foreign substances. Gas chromatography (2.2.28), injecting
ni?: about 1.513. equal volumes of each of the following:
bp: 55 oC to 56 oC at 2.5 mm Hg. A 0.02 giL solution of the substance to be examined in carbon
Enantiomeric purity: minimum 99.5. disulfide R (solution A), a 2 giL solution of the substance to
Storage: at a temperature of 2 oC to 8 oc. be examined in carbon dísulfide R (solution B), and carbon
disulfide R (solution C). Carry out the chromatographic
(S)-( - )-a-Methylbenzyl isocyanate. CgHgNO. (Mr 147.2). procedure under the conditions of the test for butylated
1170200. [14649-03-7]. (-)-(S)-a-Methylbenzyl isocyanate. hydroxytoluene prescribed in the monograph Wool fat (0134).
(-)- [(1S)-l- Isocyanatoethyl]benzene. (-)-(lS)-l-Phenylethyl The total area of any peaks, apart from the solvent peak
isocyanate. and the principal peak, in the chromatogram obtained with
Content: minimum 99.0 per cent. solution B is less than the are a of the principal peak in the
Colourless liquido chromatogram obtained with solution A.
d§g: about 1.045. Methyldopa, racemic. ClO H 13 N0 4,1 V,H 2 0. (Mr 238.2).
n5°: about 1.514. 1175100.
bp: 55 oC to 56 oC at 2.5 mm Hg. Mixture of equal volumes of (2S)- and (2R)-2-amino-3-(3,4-
Enantiomeric purity: minimum 99.5 per cent. dihydroxyphenyl)-2-methylpropanoic acids.
Storage: at a temperature of 2 oC to 8 oc. 3-0-Methyldopamine hydrochloride. C9H¡4CIN02'
NOTE: do not use the reagent if it is coloured. (Mr 203.7). 1055600. [1477-68-5]. 4-(2-Aminoethyl)-2-
methoxyphenol hydrochloride.
2-Methylhutane. CSH¡z. (Mr 72.2). 1099500. [78-78-4].
mp: 213 oC to 215 oc.
Isopentane.
Content: minimum 99.5 per cent of CSH¡z. 4-0-Methyldopamine hydrochloride. CgH¡4CIN02'
Very flammable colourless liquido (Mr 203.7). 1055700. [645-33-0]. 5-(2-Aminoethyl)-2-
d~g: about 0.621.
methoxyphenol hydrochloride.
mp: 207 oC to 208 oc.
n5° : about 1.354.
bp: about 29 oc. Methylenebisacrylamide. C7H¡ONzÜz. (Mr 154.2). 1056000.
Water (2.5.12): maximum 0.02 per cent. [110-26-9]. N,N"- Methylenebispropenamide.
Residue on evaporation: maximum 0.0003 per cent. Fine, white or almost white powder, slightly soluble in water,
Mínimum transmittance (2.2.25) using water Ras soluble in ethanol (96 per cent).
compensation liquid: 50 per cent at 210 nm, 85 per cent at mp: 300 oC, with decomposition.
220 nm, 98 per cent at 240 nm and at higher wavelengths. Methylene Mue. C¡6H¡sCIN3S,xHzÜ. (Mr 319.9 for the
2-Methylbut-2-ene. CSHlO' (Mr 70.1). 1055400. [513-35-9]. anhydrous substance). 1055800. [122965-43-9].
Very flammable liquid, practically insoluble in water, miscible Schultz No. 1038.
with ethanol (96 per cent). Colour Index No. 52015.
bp: 37.5 oC to 38.5 oc. 3,7 -Dimethylaminophenothiazin -5-ium chloride.
It occurs in different hydrated forms and may contain up to
Methyl caprate. 1054000. 22 per cent of water.
See Methyl decanoate R. Dark-green or bronze, crystalline powder, freely soluble in
Methyl caproate. C 7 H¡40Z' (Mr 130.2). 1120300. [106-70-7]. water, soluble in ethanol (96 per cent).
Methyl hexanoate. Methylene chloride. CH zC1 2. (Mr 84.9). 1055900. [75-09-2].
d~g: about 0.885. Dichloromethane.
n5°: about 1.405. Colourless liquid, sparingly soluble in water, miscible with
bp: 150 oC to 151°C. ethanol (96 per cent).
bp: 39 oC to 42 oc.
Methyl caprylate. CgH¡sÜz. (Mr 158.2). 1120400. [111-11-5].
Methyl octanoate. Methylene chloride used in fluorimetry complíes with the
following additional test.
d~g : about 0.876.
Fluorescence. Under irradiation at 365 nm, the fluorescence
n5°: about 1.417. (2.2.21) measured at 460 nm in a 1 cm cell is not more intense
bp: 193 oC to 194 oc. than that of a solution containing 0.002 ppm of quinine R in
Methykellulose 450. 1055500. [9004-67-5]. 0.5 M sulfuric acid measured in the same conditions.
See Methylcellulose (0345). Methylene chloride, addified. 1055901.
Nominal viscosity: 450 mPa·s. To 100 mL of methylene ehloride R add 10 mL of
hydrochloric acid R, shake, allow to stand and separate the
Methyl dnnamate. C lO H lO 02' (Mr 162.2). 1099400. two layers. Use the lower layer.
[103-26-4].
Colourless crystals practically insoluble in water, soluble in Methyl eicosenoate. C2¡H40 0 2 • (Mr 324.5). 1120500.
ethanol (96 per cent). [2390-09-2]. Methyl (11Z)-eicos-ll-enoate.
n50: about 1.56. Methyl erucate. C23H440Z' (Mr 352.6). 1146100. [1120-34-9].
bp: about 260 oc. Methyl (l3Z)-docos-13-enoate.
mp: 34 oC to 36 oc. d~g : about 0.871.

General Notices (1) apply to all monographs and other texts 487
4.1.1. Reagents EUROPEAN PHARMACOPOEIA 8.0

nj'¡D: about 1.456. d~g : about 0.80.


bp: about 115 oc.
3-0-Methylestrone. C 19 H 2P2' (Mr 284.4). 1137000.
[1624-62-0]. 3-Methoxy-1,3,5(lO)-estratrien-17 -one. Distillation range (2.2.11). DistillOO mL. The range of
temperature of distillation from 1 mL to 95 mL of distillate
White to yellowish-white powder. does not exceed 4.0 oc.
[al~D: about + 157. Residue on evaporation: maximum 0.01 per cent, determined
mp: about 173 oc. by evaporating on a water-bath and drying at 100-105 oc.

Methyl ethyl ketone. C"HsÜ. (Mr 72.1). 1054100. [78-93-3]. Methyl isobutyl ketone Rl. 1054301.
Ethyl methyl ketone. 2-Butanone. Shake 50 mL of freshly distilled methyl isobutyl ketone R
Clear, colourless, flammable liquid, very soluble in water, with 0.5 mL of hydrochloric acid Rl for 1 mino Allow the
miscible with ethanol (96 per cent). phases to separate and discard the lower phase. Prepare
immediately before use.
dsg: about 0.81.
bp: 79 oC to 80 oc. Methyl isobutyl ketone R3. 1054302.
Complies with the requirements for methyl isobutyl
Methyleugenol. CllH1402' (Mr 178.2).1182000. [93-15-2]. ketone R and with the following limits.
1,2-Dimethoxy-4-prop-2-enylbenzene. Cr: maximum 0.02 ppm.
Methyleugenol used in gas chromatography complies with Cu: maximum 0.02 ppm.
the following additional test. Pb: maximum 0.1 ppm.
Assay. Gas chromatography (2.2.28) as prescribed in the Ni: maximum 0.02 ppm.
monograph Niaouli oil, cineole type (2468).
Sn: maximum 0.1 ppm.
Content: minimum 97.0 per cent, calculated by the
normalisation procedure. Methyllaurate. C13H2602' (Mr 214.4).1054400. [111-82-0].
Methyl dodecanoate.
Methyl green. C26H33C12Ny (Mr 458.5). 1054200. [7114-03-6]. Content: minimum 98.0 per cent, determined by gas
Schultz No. 788. chromatography (2.4.22).
Colour Index No. 42585. Colourless or yellow liquid, soluble in ethanol (96 per cent)
4- [[ 4-(Dimethyl-amino )phenyl] [4-( dimethyliminio )cyelo- and in light petroleum.
hexa -2,5-dienylldene]-methylphenyl]trimethylammonium d~g: about 0.87.
dichloride. nj'¡°: about 1.431.
Green powder, soluble in water, soluble in sulfuric acid giving mp: about 5 oc.
a yellow solution turning green on dilution with water.
Methyllignocerate. C2sHsoOz' (M, 382.7). 1120600.
Methyl green-iodomercurate papero 1054201. [2442-49-1]. Methyl tetracosanoate.
Immerse thin strips of suitable filter paper in a 40 giL Flakes.
solution of methyl green R and allow to dry in airo Immerse mp: about 58 oc.
the strips for 1 h in a solution containing 140 giL of
potassium iodide R and 200 giL of mercuric iodide R. Wash Methyllinoleate. C 19 H 3"Oz' (M, 294.5). 1120700. [112-63-0].
with distilled water R until the washings are practically Methyl (9Z,12Z)-octadeca-9,12-dienoate.
colourless and allow to dry in airo d~g : about 0.888.
Storage: protected from light; use within 48 h. nj'¡° : about l.466.
bp: 207 oC to 208 oc.
Methy14-hydroxybenzoate. 1055000. [99-76-3].
See Methyl parahydroxybenzoate R. Methyllinolenate. C19H3202' (M, 292.5). 1120800.
[301-00-8]. Methyl (9Z,12Z,15Z)-octadeca-9, 12,15-trienoate.
l-Methylimidazole. C4H 6 N 2. (Mr 82.1). 1139700. [616-47-7]. Methyl a-linolenate.
1-Methyl-lH-imidazole. d~g: about 0.90l.
Colourless or slightly yellowish liquido nj'¡°: about l.47l.
nj'¡D: about 1.495. bp: about 207 oc.
bp: 195 oC to 197 oc. Methyl y-linolenate. C 19 H 3P2' (Mr 292.5). 1158400.
Storage: in an airtight container, protected from light. [16326-32-2]. Methyl (6Z,9Z, 12Z)-octadeca-6,9,12-trienoate.
Content: minimum 99.0 per cent, determined by gas
l-Methylimidazole Rl. 113970l. chromatography.
Complies with the requirements prescribed for
l-methylimidazole R with the following additional Methyl margarate. C 1s H 3P2' (Mr 284.5). 1120900.
requirement. [1731-92-6]. Methyl heptadecanoate.
White or almost white powder.
Content: minimum 95.0 per cent.
mp: 32 oC to 34 oc.
2-Methylimidazole. C4H6NZ' (Mr 82.1). 1143400. [693-98-1]. Methyl margarate used in the assay of total fatty acids in Saw
White or almost white, crystalline powder. palmetto fruit (1848) complies with the following additional
mp: about 145 oc. test.
Assay. Gas chromatography (2.2.28) as prescribed in the
Methyl iodide. CH 3I. (M, 141.9). 1166400. [74-88-4]. monograph Saw palmetto fruit (1848).
Iodomethane. Content: minimum 97 per cent, calculated by the
normalisation procedure.
Methyl ísobutyl ketone. C 6 H,p. (M, 100.2). 1054300.
[108-10-1]. 4-Methyl-2-pentanone. Methyl methacrylate. C SH S0 2. (Mr 100.1). 1054500.
CIear, colourless liquid, slightly soluble in water, miscible with [80-62-6]. MethyI2-methylprop-2-enoate.
most organic solvents. Colourless liquido

488 See the information section on general monographs (cover pages)


EUROPEAN PHARMACOPOEIA 8,0 4,1.1. Reagents

n~O: about 1.414, Test for sensitivity. A mixture of 0.1 mL of the methyl
bp: about 100 oC, orange solution and 100 mL of carbon dioxide-free water R
is yellow. Not more than 0.1 mL of 1 M hydrochloric acid is
mp: about - 48 oC,
required to change the colour to red.
It contains a suitable stabilising reagent Colour change: pH 3.0 (red) to pH 4.4 (yellow).
Methyl methanesulfonate. C2HP JS, (Mr 110,1), 1179500, Methyl palmitate. C 17H 3P2' (Me 270.5). 1054900. [112-39-0].
[66-27-3] , Methyl hexadecanoate.
Clear, colourless or slightly yellow liquid, Content: minimum 98.0 per cent, determined by gas
Content: mínimum 99,0 per cent chromatography (2.4.22).
Density: about 1.3 g/cm 3 (25 OC), White or yellow, crystalline mass, soluble in ethanol (96 per
n~o: about 1.414,
cent) and in light petroleum.
mp: about 30 oc.
bp: about 202 oC,
Methyl palmitoleate. C 17H 32 02' (Me 268.4). 1121000.
Methyl N-methylanthranilate. C9HllN02, (Mr 165.2), [1120-25-8]. Methyl (9Z)-hexadec-9-enoate,
1164600, [85-91-6], Methy12-(methylamino)benzoate,
ds8: about 0.876.
Pale yellow liquido
n~o : about 1.451.
d~o: about 1.128.
n~o: about 1.579.
Methyl parahydroxybenzoate. 1055000. [99-76-3].
bp: 255 oC to 258 oC, See Methyl parahydroxybenzoate (0409).

Methyl N-methylanthranilate used in gas chromatography Methyl pelargonate. C lO H 2oÜ2' (Mr 172.3), 1143500,
complies with the following additional test. [1731-84-6]. Methyl nonanoate.
Assay. Gas chromatography (2.2.28) as prescribed in the Clear, colourless liquido
monograph Mandarin oil (2355). d~o: about 0,873,
Test solution, The substance to be examined. n~o: about 1.422.
Content: minimum 97 per cent, calculated by the bp: 91°C to 92 oc.
normalisation procedure, Methyl pelargonate used in the assay of total fatty acids in Saw
palmetto fruit (1848) complies with the following additional
Methyl myristate. C jS H JaÜ2' (Mr 242.4). 1054600, [124-10-7]. test,
Methyl tetradecanoate,
Assay. Gas chromatography (2.2.28) as prescribed in the
Content: minimum 98.0 per cent, determined by gas monograph Saw palmetto fruit (1848).
chromatography (2.4,22).
Content: minimum 98 per cent, calculated by the
Colourless or slightly yellow liquid, soluble in ethanol (96 per normalisation procedure.
cent) and in light petroleum.
2-Methylpentane. C 6H 14 . (Mr 86.2). 1180400. [107-83-5].
ds8: about 0.87.
Isohexane,
n~o: about 1.437.
mp: about 20 oC,
ds8: about 0.653,
bp: about 60.0 oc.
Methyl nervonate. 1144800. [2733-88-2]. Colourless, flammable liquid, practically insoluble in water,
See Tetracos-15-enoic acid methyl ester R. miscible with anhydrous ethanoL

Methyloleate. C19H3602' (M,o 296.4). 1054700. [112-62-9]. 3-Methylpentan-2-one. C6 H 1P, (Mr 100.2). 1141100,
Methyl (9Z)-octadec-9-enoate. [565-61-7].
Content: minimum 98.0 per cent, determined by gas Colourless, flammable liquido
chromatography (2.4.22). d~g: about 0,815,
Colourless or slightly yellow liquid, soluble in ethanol (96 per n~o: about 1.400,
cent) and in light petroleum, bp: about 118 oC
d~g : about 0,88.
4-Methylpentan-2-ol. C6H140. (Me 102.2). 1114300.
n~o : about 1.452. [108-11-2] ,
CIear, colourless, volatile liquido
Methylorange. C14H14NJNa03S, (Me 327,3). 1054800,
[547-58-0], d~o : about 0.802.
Schultz No. 176, n~o: about 1.411.

Colour Index No. 13025. bp: about 132 oc.


Sodium 4'-( dimethylamíno )azobenzene-4-sulfonate. 4-Methylphenazone. C12H14N2o. (MI 202.3). 1182100.
Orange-yellow, crystalline powder, slightly soluble ín water, [56430-08-1]. 1,5-Dimethyl-2-( 4-methylphenyl)-1,2-dihydro-
practically insoluble in ethanol (96 per cent). 3H-pyrazol-3-one.
Methyl orange mued solution. 1054801, Methylphenyloxazolylbenzene. C26 H 20 NP2' (Mr 392.5).
Dissolve 20 mg of methyl orange R and 0.1 g of bromocresol 1056200. [3073-87-8]. 1,4-Bis[2-(4-methyl-5-phenyl)-
green R in 1 mL of 0,2 M sodium hydroxide and dilute to oxazolyl]benzene,
100 mL with water R. Fine, greenish-yellow powder with a blue fluorescence or
Colour change: pH 3.0 (orange) to pH 4.4 (olive-green), small crystals, soluble in ethanol (96 per cent), sparingly
soluble in xylene.
Methyl orange solution. 1054802, mp: about 233 oc.
Díssolve 0,1 g of methyl orange R in 80 mL of water R and Methylphenyloxazolylbenzene used for liquid scintillation is
dilute to 100 mL with ethanol (96 per cent) R. of a suitable analytical grade.

General Notices (1) apply to all mOl1ographs and other texts 489
4.1.1. Reagents EUROPEAN PHARMACOPOEIA 8.0

1-Methyl-4-phenyl-l,2,3,6-tetrahydropyridine. C 12 H IS N. Test for sensitivity. To 0.1 mL of the methyl red solution


(M, 173.3). 1137100. [28289-54-5]. MPTP. add 100 mL of carbon dioxide-free water R and 0.05 mL of
White or almost white, crystalline powder, slightly soluble in 0.02 M hydrochloric acid. The solution is red. Not more
water. than 0.1 mL of 0.02 M sodium hydroxide is required to
mp: about 41°C. change the colour to yellow.
Colour change: pH 4.4 (red) to pH 6.0 (yellow).
Methylpiperazine. C SH I2 N 2. (Mr 100.2). 1056300. [109-01-3].
1- Methylpiperazine. Methyl salicylate. 1146200. [119- 36-8].
Colourless liquid, miscible with water and with ethanol See Methyl salicylate (0230)
(96 per cent). Methyl stearate. C 19 H 3P2' (Mr 298.5). 1055200. [112-61-8].
d~g: about 0.90. Methyl octadecanoate.
n~o : about 1.466. Content: minimum 98.0 per cent, determined by gas
bp: about 138 oc. chromatography (2.4.22).
White or yellow, crystalline mas s, soluble in ethanol (96 per
4- (4-Methylpiperidin-l-yl)pyridine. C ll HI6NZ' (Mr 176.3).
1114400. [80965-30-6].
cent) and in light petroleum.
mp: about 38 oc.
Clear liquido
n~o: about 1.565. Methylthymol bIue. C37H40N2Na4013S, (Mr 845). 1158500.
[1945-77 -3]. Tetrasodium 2,2',2",2"'- [3H-2,l-benzoxathiol-
2-MethylpropanoL C4 HlQO. (Mr 74.1).1056400. [78-83-1]. 3-ylidenebis[ [6-hydroxy-2-methyl-5-(1-methylethyl)-3, 1-
Isobutyl alcohol. 2-Methylpropan-l-ol. phenylene] methylenenitrilo]] tetraacetate S,S-dioxide.
CIear colourless liquid, soluble in water, miscible with ethanol Produces a blue colour with calcium in alkaline solution.
(96 per cent).
d~g : about 0.80. Methylthymol biue mixture. 1158501.
nh5 : 1.397 to 1.399. A mixture of 1 part of methylthymol blue R and 100 parts
bp: about 107 oc. of potassium nítrate R.
Distillation range (2.2.11). Not less than 96 per cent distils N-Methyl-m-toluidine. CSH ll N. (Mr 121.2). 1175200.
between 107 oC and 109 oc. [696-44-6]. N,3- Dimethylaniline. N,3- Dimethylbenzenamine.
Methyl-m -tolylamine.
2-Methyl-2-propanoL C4 HlQO. (Mr 74.1). 1056500.
Content: minimum 97 per eent.
[75-65-0]. l,l-Dimethyl ethyl alcohol. tert-Butyl alcohol.
CIear, colourless liquid or crystalline mass, soluble in water, Methyl tricosanoate. C24 H 4P2' (Mr 368.6). 1111500.
miscible with ethanol (96 per cent). [2433-97 -8]. Tricosanoic acid methyl estero
Freezing point (2.2.18): about 25 oc. Content: minimum 99.0 per cent.
Distillation range (2.2.11). Not less than 95 per cent distils White or almost white crystals, practically insoluble in water,
between 81°C and 83 oc. soluble in hexane.
mp: 55 oC to 56 oc.
(l5R)-15-Methylprostaglandin F2u ' C 2I H 3 Ps' (Mr 368.5).
1159900. [35864-81-4]. (5Z)-7-[(1R,2R,3R,5S)-3,5- Methyl tridecanoate. C 14H 2P2' (Mr 228.4). 1121100.
Dihydroxy-2- [( lE) -( 3R) - 3-hydroxy -3-methyloct -1- [1731-88-0].
enyl] cyclopentyl]hept -5-enoic acid. Colourless or slightly yellow liquid, soluble in ethanol (96 per
Available as a 10 giL solution in methyl acetate R. cent) and in light petroleum.
Storage: at a temperature below - 15 oc. d~g: about 0.86.
N-Methylpyrrolidine. CSH ll N. (Mr 85.2). 1164700. n~o: about 1.441.
[ 120-94-5]. mp: about 6 oc.
Content: minimum 97.0 per cent. MethyI3,4,5-trimethoxybenzoate. CllHIPs' (Mr 226.23).
bp: about 80 oc. 1177200. [1916-07-0].
N-Methylpyrrolidone. CSHgNO. (Mr 99.1). 1164800. N- MethyItrimethylsilyl- trifluoroacetamide.
[872-50-4]. 1-Methylpyrrolidin-2-one. C 6 H 12 F3 NOSi. (Mr 199.3). 1129600. [24589-78-4].
d~g : about 1.028. 2,2,2-Trifluoro-N-methyl- N-( trimethylsilyl)acetamide.
bp: about 202 oc. n~o : about 1.380.
mp: about - 24 oc. bp: 130 oC to 132 oc.

Methyl red. C ls H IS NP2' (Mr 269.3). 1055100. [493-52-7]. Minocydine hyrlrochloride. 1146300.
Schultz No. 250. See Minocycline hydrochloride (1030).
Colour Index No. 13020. Molecular sieve. 1056600.
2-( 4-Dimethylamino-phenylazo )benzoic acid.
Molecular sieve composed of sodium aluminosilicate. It
Dark-red powder or violet crystals, practically insoluble in is available as beads with a pore size of 0.4 nm and with a
water, soluble in ethanol (96 per cent). diameter of 2 mm.
Methyl red mixed solution. 1055101. Molecular sieve for chromatography. 1129700.
Dissolve 0.1 g of methyl red R and 50 mg of methylene Molecular sieve composed of sodium aluminosilicate. The
blue R in 100 mL of ethanol (96 per cent) R. pore size is indicated after the name of the reagent in the tests
Colour change: pH 5.2 (red-violet) to pH 5.6 (green). where it is used. If necessary, the particle size is also indicated.
Methyl red solution. 1055102. Molybdovanadic reagent. 1056700.
Dissolve 50 mg in a mixture of 1.86 mL of 0.1 M sodium In a 150 mL beaker, mix 4 g of finely powdered ammonium
hydroxide and 50 mL of ethanol (96 per cent) R and dilute molybdate R and 0.1 g of finely powdered ammonium
to 100 mL with water R. vanadate R. Add 70 mL of water R and grind the particles

490 See the information section on general monographs (cover pages)


EUROPEAN PHARMACOPOEIA 8.0 4. L 1. Reagents

using a glass rod. A clear solution is obtained within a few ~-Myrcene. CIOHI6' (Mr 136.2). 1114500. [123-35-3].
minutes. Add 20 mL of nitric acid R and dilute to 100 mL 7 -Methyl-3-methylenocta-1,6-diene.
with water R. Oily liquid with a pleasant odour, practically insoluble in
water, miscible with ethanol (96 per cent), soluble in glacial
Monodocosahexaenoin. C2sH3S04' (M, 402.6). 1143600.
acetic acid. It dissolves in solutions of alkali hydroxides.
[124516-13-8]. Monoglyceride of docosahexaenoic
acid (C22:6). Glycerol monodocosahexaenoate. da o : about 0.794.
(all-Z)-Docosa-4,7,10,13,16,19-hexaenoic acid, monoester n~o : about 1.470.
with propane-1,2,3-triol. f3-Myrcene used in gas chromatography complíes with the
following additional test.
Mordant black H. C20H12N3Na07S, (M, 461.4). 1056800.
[1787-61-7]. Assay. Gas chromatography (2.2.28) as prescribed in the
monograph Peppermint oil (0405).
Schultz No. 241.
Test solution. The substance to be examined.
Colour Index No. 14645.
Sodium 2-hydroxy-l- [( l-hydroxynaphth-2-yl)azo ]-6- Content: minimum 90.0 per cent, calculated by the
nitronaph- thalene-4-sulfonate. Eriochrome black. normalisation procedure.
Brownish-black powder, soluble in water and in ethanol Myristic add. C¡4H2S02' (Mr 228.4). 1143700. [544-63-8].
(96 per cent). Tetradecanoic acid.
Storage: in an airtight container, protected from light. Colourless or white or almost white flakes.
Mordant black 11 triturate. 1056801. mp: about 58.5 oc.
Myristic acid used in the assay of total fatty acids in Saw
Mix 1 g of mordant black 11 R with 99 g of sodium
palmetto fruit (1848) complies with the following additional
chloride R.
test.
Test for sensitivity. Dissolve 50 mg in 100 mL of water R.
Assay. Gas chromatography (2.2.28) as prescribed in the
The solution is brownish-violet. On addition of 0.3 mL
monograph Saw palmetto fruit (1848).
of dilute ammonia Rl the solution turns blue. On the
subsequent addition of 0.1 mL of a 10 giL solution of Content: minimum 97 per cent, calculated by the
magnesium sulfate R, it turns violet. normalisation procedure.
Storage: in an airtight container, protected from light. Myristidne. C¡¡H 12 03' (Mr 192.2). 1099600. [607-91-0].
5-Allyl-1- methoxy-2,3-methylenedioxybenzene.
Mordant black 11 triturate Rl. 1056802. 4-Methoxy-6-(prop-2-enyl) -1 ,3-benzodioxole.
Mix 1.0 g of mordant black 11 R, 0.4 g of methyl orange R Oily colourless liquid, practically insoluble in water, slightly
and 100 g of sodium chloride R. soluble in anhydrous ethanol, miscible with toluene and with
Morphine hydrochloride. 1056900. xylene.
d~g: about 1.144.
See Morphine hydrochloride (0097).
n~o : about 1.540.
Morpholine. C 4H 9 NO. (M,. 87.1). 1057000. [110-91-8]. bp: 276 oC to 277 oc.
Tetrahydro-1,4-oxazine.
mp: about 173 oc.
Colourless, hygroscopic liquid, flammable, soluble in water
and in ethanol (96 per cent). Chromatography. Thin-Iayer chromatography (2.2.27)
as prescribed in the monograph Star anise (1153); the
d~g: about 1.01. chromatogram shows only one principal spot.
Distillation range (2.2.11). Not less than 95 per cent distils Myristicine used in gas chromatography complies with the
between 126 oC and 130 oc. following additional test.
Storage: in an airtight container. Assay. Gas chromatography (2.2.28) as prescribed in the
monograph Nutmeg oil (1552).
Morpholine for chrornatography. 1057001.
Content: minimum 95.0 per cent, calculated by the
Complies with the requirements prescribed for normalisation procedure.
morpholine R with the following additional requirement.
Storage: protected from light.
Content: minimum 99.5 per cent.
Myristyl alcohol. C¡4H300. (M, 214.4). 1121300. [112-72-1].
2- [N- Morpholino ]ethanesulfonic add. C6 H 13 N0 4S. Tetradecan-1-01.
(M, 195.2).1186500. [4432-31-9]. 2-(Morpholin-4-yl)sulfonic 20
acid. MES. d 20 : about 0.823.
mp: 38 oC to 40 oc.
White or almost white, crystalline powder, soluble in water.
mp: about 300 oc. Myrtillin. C2j H 2¡ClO¡2' (Mr 500.8). 1172300. [6906-38-3].
Delphinidin 3-0-g1ucoside chloride.
Murexide. CsH sN 6 0 6,Hp. (M, 302.2). 1137200.
5,5' -Nitrilobis(pyrimidine-2,4,6( 1H,3H,5H) -trione) Naphthalene. ClOH s' (M, 128.2).1057100. [91-20-3].
monoammonium salt. White or almost white crystals, practically insoluble in water,
Brownish-red crystalline powder, sparingly soluble in cold soluble in ethanol (96 per cent).
water, soluble in hot water, practically insoluble in ethanol mp: about 80 oc.
(96 per cent), soluble in solutions of potassium hydroxide or Naphthalene used for liquid scintillation is of a suitable
sodium hydroxide giving a bIue colour. analytical grade.
Myosrnine. Cg HlQN 2. (M, 146.2). 1121200. [532-12-7]. Naphtharson. C16HIlAsNlNap¡OS2' (M, 576.3). 1121400.
3- (4,5-Dihydro- 3H- pyrrol-2-yl)pyridine. [3688-92-4]. Thorin. Disodium 4- [(2-arsonophenyl)azo ]-3-
Colourless crystals. hydroxynaphthalene-2, 7-disulfonate.
mp: about 45 oc. Red powder, soluble in water.

General Notices (1) apply to all monographs and other texts 491
4.1.1. Reagents EUROPEAN PHARMACOPOEIA 8.0

Naphtharson solution. 1121401. Naphthylamine. ClOH9N. (Mr 143.2). 1057700. [134-32-7].


A 0.58 giL solution. 1-N aphthylamine.
Test for sensitivity. To 50 mL of ethanol (96 per cent) R, White or almost white, crystalline powder, turning pink on
add 20 mL of water R, 1 mL of 0.05 M sulfuric acid and exposure to light and air, slightly soluble in water, freely
1 mL of the naphtharson solution. Titrate with 0.025 M soluble in ethanol (96 per cent).
barium perchlorate; the colour changes from orange-yellow mp: about 51 oc.
to orange-pink. 5torage: protected from light.
Storage: protected from light; use within 1 week.
N aphthylethylenediamine dihydrochloride.
a-Naphthol. ClOHsO. (M, 144.2). 1057300. [90-15-3]. C12H16CI2N2' (Mr 259.2). 1057800. [1465-25-4].
1-Naphthol. N-( 1-Naphthyl)ethylene-diamine dihydrochloride.
White or almost white, crystalline powder or colourless or It may contain methanol of crystallisation.
white or almost white crystals, darkening on exposure to light, White or yellowish-white powder, soluble in water, slightly
slightly soluble in water, freely soluble in ethanol (96 per cent). soluble in ethanol (96 per cent).
mp: about 95 oc.
Storage: protected from light. Naphthylethylenediamine dihydrochloride solution.
1057801.
a-Naphthol solution. 1057301. Dissolve 0.1 g of naphthylethylenediamine dihydrochloride R
Dissolve 0.10 g of a-naphthol R in 3 mL of a 150 giL in water R and dilute to 100 mL with the same solvent.
solution of sodium hydroxide R and dilute to 100 mL with Prepare immediately before use.
water R. Prepare immediately before use.
Naringin. C17H 3P14' (M, 580.5). 1137300. [10236-47-2].
~-Naphthol. CIOHsO. (M, 144.2). 1057400. [135-19-3]. 7 -[[2-0-( 6- Deoxy-a-L-mannopyranosyl)-~-D­
2-Naphthol. glucopyranosyl] oxy ]-5-hydroxy-2- (4-hydroxyphenyl)-
White or slightly pink plates or crystals, very slightly soluble 2,3-dihydro-4H-chromen-4-one.
in water, very soluble in ethanol (96 per cent). White or almost white crystalline powder, slightly soluble in
mp: about 122 oc. water, soluble in methanol and in dimethylformamide.
Storage: protected from light. mp: about 171 oc.
Absorbance (2.2.25). Naringin dissolved in a 5 giL solution
~-Naphthol solution. 1057401. of dimethylformamide R in methanol R shows an absorption
Dissolve 5 g of freshly recrystallised f3-naphthol R in 40 mL maximum at 283 nm.
of dilute sodium hydroxide solution R and dilute to 100 mL
with water R. Prepare immediately before use. Neohesperidin. C2s H 3 P1S' (Mr 610.6). 1182200.
[13241-33-3]. Hesperetin-7 -neohesperidoside.
p-Naphthol solution Rl. 1057402. (25) -7 -[[ 2-0-( 6-Deoxy-a -L- mannopyranosyl) -~- D-
Dissolve 3.0 mg of f3-naphthol R in 50 mL of sulfuric acid R glucopyranosyl] oxy]-5-hydroxy-2- (3-hydroxy-4-
and dilute to 100.0 mL with the same acid. Use the recently methoxyphenyl)-2,3-dihydro-4H-l-benzopyran-4-one.
prepared solution.
trans-NeroHdol. C1s H 26 0. (M, 222.4). 1107900. [407l6-66-3].
Naphtholbenzein. C27H1S02' (M, 374.4). 1057600. 3,7,11-Trimethyldodeca-1,6, 10-trien-3-ol.
[145- 50-6]. a -Naphtholbenzein. 4- [( 4-Hydroxynaphthalen-l- Slightly yellow liquid, slight odour of lily and lily of the valley,
yl)(phenyl)methylidene] naphthalen-1 (4H)-one. practically insoluble in water and in glycerol, miscible with
Brownish-red powder or shiny brownish-black crystals, ethanol (96 per cent).
practically insoluble in water, soluble in ethanol (96 per cent) d§g: about 0.876.
and in glacial acetic acid. n~o: about 1.479.
Naphtholbenzein solution. 1057601. bp12: 145 oC to 146 oc.
A 2 giL solution in anhydrous acetic acid R. trans-Nerolidol used in gas chromatography complies with the
Test for sensitivity. To 50 mL of glacial acetic acid R add following additional test.
0.25 mL of the naphtholbenzein solution. The solution Assay. Gas chromatography (2.2.28) as prescribed in the
is brownish-yellow. Not more than 0.05 mL of 0.1 M monograph Bitter-orange-flower oil (1175).
perchloric acid is required to change the colour to green. Test solution. The substance to be examined.
Naphthol yeHow. ClOHsN1NaO s' (M, 256.2). 1136600. Content: minimum 90.0 per cent, calculated by the
2,4-Dinitro-1-naphthol, sodium salto normalisation pro ce dure.
Orange-yellow powder or crystals, freely soluble in water, Neryl acetate. C 12 H 2aÜ2' (Mr 196.3). 1108000. [141-12-8].
slightly soluble in ethanol (96 per cent). (Z)-3,7 - Dimethylocta-2,6-dieny! acetate.
Naphthol yellow S. CJOH 4N1Nap sS. (M, 358.2). 1143800. Colourless, oily liquido
[846-70-8]. d~g: about 0.907.
Colour Index No. 10316. n~o: about 1.460.
8-Hydroxy-5,7 -dinitro-2-naphthalenesulfonic acid disodium bp25: 134 oc.
salt. Disodium 5,7-dinitro-8-oxidonaphthalene-2-sulfonate.
Neryl acetate used in gas chromatography complies with the
Yellow or orange-yellow powder, freely soluble in water. following additional test.
l-Naphthylacetic add. C 12 H lO Oz. (M, 186.2). 1148400. Assay. Gas chromatography (2.2.28) as prescribed in the
[86-87-3]. (Naphthalen-l-yl)acetic acid. monograph Bitter-orange-flower oil (1175).
White or yellow crystalline powder, very slightly soluble in Test solution. The substance to be examined.
water, freely soluble in acetone. Content: minimum 93.0 per cent, calculated by the
mp: about 135 oc. normalisation procedure.

492 5ee the informatiol1 section on general mOl1ographs (ca ver pages)
EUROPEAN PHARMACOPOEIA 8.0 4.1.1. Reagents

Nickel-aluminium alloy. 1058100. Ninhydrin. C9HP3,Hp. (Mr 178.1). 1058300. [485-47-2].


Contains 48 per cent to 52 per cent of aluminium (Al; Ar 26.98) 1,2,3-Indanetrione monohydrate.
and 48 per cent to 52 per cent of nickel (Ni; Ar 58.70). White or very pale yellow, crystalline powder, soluble in water
and in ethanol (96 per cent).
Before use, reduce to a fine powder (180) (2.9.12).
Storage: protected from light.
It is practically insoluble in water and soluble in mineral acids.
Ninhydrin and stannous chloride reagent. 1058301.
Nickel-aluminium alloy (halogen-free). 1118100. Dissolve 0.2 g of ninhydrin R in 4 mL ofhot water R, add
Contains 48 per cent to 52 per cent of aluminium (Al; Ar 26.98) 5 mL of a 1.6 giL solution of stannous chloride R, allow to
and 48 per cent to 52 per cent of nickel (Ni; Ar 58.71). stand for 30 min, then filter and store at a temperature of
Fine, grey powder, practically insoluble in water, soluble in 2 oC to 8 oc. Immediately before use dilute 2.5 mL of the
mineral acids with formation of salts. solution with 5 mL of water R and 45 mL of 2-propanol R.
ehlorides: maximum 10 ppm. Ninhydrin and stannous chloride reagent Rl. 1058302.
Dissolve 0.400 g in 40 mL of a mixture of 67 volumes of sulfuric Dissolve 4 g of ninhydrin R in 100 mL of ethylene
acid R and 33 volumes of dilute nitric acid R. Evaporate the glyeol monomethyl ether R. Shake gently with 1 g of
solution nearly to dryness, dissolve the residue in water R and cation-exchange resin R (300 flm to 840 flm) and filter
dilute to 20.0 mL with the same solvent. To one half-aliquot (solution A). Dissolve 0.16 g of stannous chloride R in
of the solution, add 1.0 mL of 0.1 M silver nitrate. Filter 100 mL of buffer solution pH 5.5 R (solution B). Immediately
after 15 min and add 0.2 mL of sodium chloride solution before use, mix equal volumes of each solution.
(containing 10 flg of chlorides per millilitre) to the filtrate.
After 5 min the solution is more opalescent than a mixture of Ninhydrin solution. 1058303.
the second half-aliquot of the solution with 1.0 mL of 0.1 M A 2 giL solution of Ninhydrin R in a mixture of 5 volumes
si/ver nitrate. of dilute acetic acid R and 95 volumes of butanol R.

Nickel chloride. NiCl 2. (Mr 129.6). 1057900. [7718-54-9]. Ninhydrin solution Rl. 1058304.
Nickel chloride, anhydrous. Dissolve 1.0 g of ninhydrin R in 50 mL of ethanol (96 per
cent) R and add 10 mL of glacial acetic acid R.
Yellow, crystalline powder, very soluble in water, soluble in
ethanol (96 per cent). It sublimes in the absence of air and Ninhydrin solution R2. 1058305.
readily absorbs ammonia. The aqueous solution is acid. Dissolve 3 g of ninhydrin R in 100 mL of a 45.5 giL solution
of sodium metabisulfite R.
Nickel nitrate hexahydrate. Ni(N0 3)2,6Hp. (Mr 290.8).
1175300. [13478-00-7]. Ninhydrin solution R3. 1058306.
A 4 giL solution in a mixture of 5 volumes of anhydrous
Nickel sulfate. NiS0 4,7Hp. (Mr 280.9). 1058000. acetic acid R and 95 volumes of butanol R.
[10101-98-1]. Nickel sulfate heptahydrate.
Green, crystalline powder or crystals, freely soluble in water, Ninhydrin solution R4. 1058307.
slightly soluble in ethanol (96 per cent). A 3 giL solution of ninhydrin R in a mixture of 5 volumes
of glacial aeetie acid R and 95 volumes of 2-propanol R.
Nicotinamide-adenine dinudeotide. C21H27N7014P2'
(Mr 663). 1108100. [-84-9]. NAD+. Nitrazepam. 1143900. [146-22-5].
See Nitrazepam (0415).
White or almost white powder, very hygroscopic, freely
soluble in water. Nitric add. HN0 3 • (Mr 63.0). 1058400. [7697-37-2].
eontent: 63.0 per cent m/m to 70.0 per cent mimo
Nicotinamide-adenine dinudeotide solution. 1108101.
Clear, colourless or almost colourless liquid, miscible with
Dissolve 40 mg of nicotinamide-adenine dinucleotide R in water.
water R and dilute to 10 mL with the same solvent. Prepare 20
.
lmme d'late1y b elOre
e
use. d 20 : 1.384 to 1.416.
A 10 giL solution is strongly acid and gives the reaction of
Nicotinic acid. 1158600. [59-67-6]. nitrates (2.3.1).
See Nicotinic acid (0459). Appearance. Nitric acid is clear (2.2.1) and not more intensely
coloured than reference solution Y6 (2.2.2, Method II).
Nile bIue A. C2o H 21 Np sS. (Mr 415.5). 1058200. [3625-57-8]. ehlorides (2.4.4): maximum 0.5 ppm.
Schultz No. 1029. To 5 g add 10 mL of water R and 0.3 mL of silver nitrate
Colour Index No. 51180. solution R2 and allow to stand for 2 min protected from light.
5-Amino-9-(diethylamino)benzo[a]phenoxazinylium Any opalescence is not more ¡ntense than that of a standard
hydrogen sulfate. prepared in the same manner using 13 mL of water R, 0.5 mL
of nitric acid R, 0.5 mL of chlaride standard solution (5 ppm
Green, crystalline powder with a bronze lustre, sparingly el) R and 0.3 mL of si/ver nitrate solution R2.
soluble in ethanol (96 per cent), in glacial acetic acid and in Sulfates (2.4.13): maximum 2 ppm.
pyridine.
Evaporate 10 g to dryness with 0.2 g of sodium carbonate R.
Absorbance (2.2.25). A 0.005 giL solution in ethanol (50 per Dissolve the residue in 15 mL of distilled water R. Prepare the
cent V/V) R shows an absorption maximum at 640 nm. standard using a mixture of 2 mL of sulfate standard solution
(10 ppm SO,) R and 13 mL of distilled water R.
Nile bIue A solution. 1058201.
Arsenic (2.4.2, Method A): maximum 0.02 ppm.
A 10 giL solution in anhydrous acetic acid R.
Gently heat 50 g with 0.5 mL of sulfuric acid R until white
Test for sensitivity. To 50 mL of anhydrous acetic acid R fumes begin to evolve. To the residue add 1 mL of a 100 giL
add 0.25 mL of the Nile blue A solution. The solution is solution of hydroxylamine hydrochloride R and dilute to 2 mL
blue. On the addition of 0.1 mL of 0.1 M perchloric acid, with water R. Prepare the standard using 1.0 mL of arsenic
the colour changes to blue-green. standard solution (1 ppm As) R.
Colour change: pH 9.0 (blue) to pH 13.0 (red). Iron (2.4.9): maximum 1 ppm.

General Notiees (1) apply to all monographs and other texts 493
4.1.L Reagents EUROPEAN PHARMACOPOEIA 8.0

Dissolve the residue from the determination of sulfated ash in Source: lead hollow-cathode lampo
1 mL of dilute hydrochloric acid R and dilute to 50 mL with Wavelength: 283.3 nm or 217.0 nm.
water R. Dilute 5 mL of this solution to 10 mL with water R.
Atomisation device: air-acetylene flameo
Heavy metals (2.4.8): maximum 2 ppm.
Dilute 10 mL of the solution prepared for the limit test for Nitric add, lead-free RL 1058405.
iron to 20 mL with water R. 12 mL of the solution complies Nitric acid R containing not more than 1 flg/kg of lead.
with test A. Prepare the reference solution using lead standard
Nitric add, lead-free, dHute. 1058406.
solution (2 ppm Pb) R.
Dilute 5 g of lead-free nitric acid Rl to 100 mL with
Sulfated ash: maximum 0.001 per cent.
deionised distilled water R.
Carefully evaporate 100 g to dryness. Moisten the residue with
a few drops of sulfuric acid R and heat to dull red. Nitric add, nickel-free. 1058408.
Assay. To 1.50 g add about 50 mL of water R and titrate with Complies with the requirements prescribed for nitric acid R
1 M sodium hydroxide, using 0.1 mL of methyl red solution R with the following additional requirement.
as indicator. Nickel: maximum 0.005 ppm.
1 mL of 1 M sodium hydroxide is equivalent to 63.0 mg of
Nitric add, fuming. 1058500. [52583-42-3].
HNO y
Storage: protected from light.
Clear, slightly yellowish liquid, fuming 011 contact with airo
d~g : about l.5.
Nitric add, cadmium- and lead-free. 105840l.
Complies with the requirements prescribed for nitric acid R Nitrilotriacetic add. C6 H 9N0 6 • (M, 191.1). 1137400.
and with the following additional test. [139-13-9].
Test solution. To 100 g add 0.1 g of anhydrous sodium White or almost white crystalline powder, practically insoluble
carbonate R and evaporate to dryness. Dissolve the residue in water and in most organic solvents.
in water R heating slightly, and dilute to 50.0 mL with the mp: about 240 oC, with decomposition.
same solvent.
Nitroaniline. C6H6NPZ' (Mr 138.1). 1058600. [100-01-6].
Cadmium: maximum 0.1 ppm. 4- Nitroaniline.
Atomic absorption spectrometry (2.2.23, Method II). Bright yellow, crystalline powder, very slightly soluble in water,
Source: cadmium hollow-cathode lampo sparingly soluble in boiling water, soluble in ethanol (96 per
Wavelength: 228.8 nm. cent), forms water-soluble salts with strong mineral acids.
Atomisation device: air-acetylene or air-propane flameo mp: about 147 oc.
Lead: maximum 0.1 ppm. Nitrobenzaldehyde. C 7 H SNO J • (Mr 151.1). 1058700.
Atomic absorption spectrometry (2.2.23, Method JI). [552-89-6]. 2- Nitrobenzaldehyde.
Source: lead hollow-cathode lampo Yellow needles, slightly soluble in water, freely soluble in
Wavelength: 283.3 nm or 217.0 nm. ethanol (96 per cent), volatile in steam.
Atomisation device: air-acetylene flameo mp: about 42 oc.

Nitric add, dHule. 1058402. Nitrobenzaldehyde papero 1058701.


Contains about 125 giL of HNO J (M, 63.0). Dissolve 0.2 g of nitrobenzaldehyde R in 10 mL of a 200 giL
solution of sodium hydroxide R. Use the solution within 1 h.
Dilute 20 g of nitric acid R to 100 mL with water R.
Immerse the lower half of a slow filter paper strip 10 cm
Nitric add, dHule Rl. 1058407. long and 0.8-1 cm wide. Absorb the excess reagent between
Dilute 40 g of nitric acid R to 100 mL with water R. two sheets of filter paper. Use within a few minutes of
preparation.
Nitric acid, dilute R2. 1058409.
Nitrobenzaldehyde solution. 1058702.
Dilute 30 g of nitric acid R to 100 mL with water R.
Add 0.12 g of powdered nitrobenzaldehyde R to 10 mL
Nitric adJ, heavy metal-free. 1058404. of dilute sodium hydroxide solution R; allow to stand for
Complies with the requirements prescribed for nitric acid R 10 min shaking frequently and filter. Prepare immediately
with the following maximum contents of heavy metals. before use.
As: 0.005 ppm. Nitrobenzene. C6H sN0 2 • (Mr 123.1). 1058800. [98-95-3].
Cd: 0.005 ppm. Colourless or very slightly yellow liquid, practically insoluble
Cu: 0.001 ppm. in water, miscible with ethanol (96 per cent).
Fe: 0.02 ppm. bp: about 211°C.
Hg: 0.002 ppm. Dinitrobenzene. To 0.1 mL add 5 mL of acetone R, 5 mL of
Ni: 0.005 ppm. water R and 5 mL of strong sodium hydroxide solution R. Shake
and allow to stand. The upper layer is almost colourless.
Pb: 0.001 ppm.
Zn: 0.01 ppm. 4-Nitrobenzoic aeid. C 7 H sN0 4 . (Mr 167.1). 1144000.
[62-23-7].
Nitric adJ, lead-free. 1058403.
Yellow crystals.
Complies with the requirements prescribed for Nitric
mp: about 240 oc.
acid R with the following additional test.
Lead: maximum 0.1 ppm. Nitrobenzoyl chloride. C 7 H 4CINO)' (M, 185.6). 1058900.
Atomic absorption spectrometry (2.2.23, Method II). [122-04-3]. 4-Nitrobenzoyl chloride.
Test solution. To 100 g add 0.1 g of anhydrous sodium Yellow crystals or a crystalline mass, decomposing in moist
carbonate R and evaporate to dryness. Dissolve the residue air, completely soluble in sodium hydroxide solution giving a
in water R, heating slightly, and dilute to 50.0 mL with the yellowish-orange colour.
same solvent. mp: about 72 oc.

494 See the information section on general mOl1ographs (cover pages)


EUROPEAN PHARMACOPOEIA 8,0 4.1.1. Reagents

Nitrobenzyl chloride. C 7H 6CINO z' (M, 171.6), 1059000, 3-Nitrosalicylic add. C7 HsNO s' (Me 183,1), 1184300,
[100-14-1), 4-Nitrobenzyl chloride, [85-38-1), 2-Hydroxy-3-nitrobenzoic acid,
Pale-yellow crystals, lachrymatory, practically insoluble in Yellowish crystals, slightly soluble in water, freely soluble in
water, very soluble in ethanol (96 per cent), ethanol (96 per cent),
mp: 142 oC to 147 oC,
4-(4-Nitrobenzyl)pyridine. C 12 HlQNPZ' (M, 214,2),
1101900, [1083-48-3), N-Nitrosodiethanolamine. C 4 H lO NPJ' (Me 134.1), 1129800,
Yellow powder. [1116-54-7), 2,2' -(Nitrosoimino )diethanoL
mp: about 70 oC, Yellow liquid, miscible with anhydrous ethanoL
Nitrochromic reagent. 1059100, n~o: about 1.485,

Dissolve 0.7 g of potassium dichromate R in nitric acid R and bp: about 125 oC,
dilute to 100 mL with the same acid, N-Nitrosodiisopropanolamine. C 6H 14NP3' (Mr 162,2),
Nitroethane. C2 H sN0 2 , (M, 75,1), 1059200, [79-24-3), 1176500, [53609-64-6), 1,1' -(Nitrosoimino )bispropan-2-oL
Clear, oily, colourless liquid, bp: 122-124 oC,
bp: about 114 oC, Nitrosodipropylamine. C6 H 14Np, (Me 130,2), 1099900,
[621-64-7), Dipropylnitrosamine,
Nitrofurantoin. 1099700, [67-20-9),
Liquid, soluble in anhydrous ethanol and in strong acids,
See Nitrofurantoin (0101),
d~g: about 0,915,
Nitrogen. N z' (M, 28,01),1059300, [7727-37-9), bp: about 78 oC,
Nitrogen, washed and dried, Appropriate grade for chemiluminescence determination,
Nitrogen gas mixture. 1136900, Nitrosodipropylamine solution. 1099901,
Nitrogen R containing 1 per cent V/V of each of the Inject 78,62 g of anhydrous ethanol R through the septum
following gases: carbon dioxide R2, carbon monoxide R1 oí a vial containing nitrosodipropylamine R. Dilute 1/100
and oxygen Rl, in anhydrous ethanol R and place 0.5 mL aliquots in
Nitrogen,oxygen-free. 1059600, crimp-sealed vials,
Nitrogen R which has been freed from oxygen by passing it Storage: in the dark at 5 oC,
through alkaline pyrogallol solution R. Nitrotetrazolium biue. C4oH30ClzNlO06' (Mr 818),1060000,
Nitrogen Rl. N z' (M, 28,01), 1059400, [7727-37-9), [298-83-9), 3,3'-(3,3'-Dimethoxy-4,4' -diphenylene)di[2-
(4-nitrophenyl)-5-phenyl-2H-tetrazolium) dichloride,
Content: minimum 99,999 per cent V/V,
p-Nitro-tetrazolium blue,
Carbon monoxide: less than 5 ppm,
Crystals, soluble in methanol, giving a clear, yellow solution,
Oxygen: less than 5 ppm,
mp: about 189 oC, with decomposition,
Nitrogen dioxide. N0 2 , (M, 46,01), 1179600, [10102-44-0),
Nitrous oxide. NP, (Mr 44,01), 1108500,
Content: minimum 98,0 per cent V/V,
Content: minimum 99,99 per cent V/V.
Nitrogen for chromatography. N z' (Me 28,01), 1059500, Nitrogen monoxide: less than 1 ppm,
[7727-37-9), Carbon monoxide; less than 1 ppm,
Content: minimum 99,95 per cent V/V,
Nonivamide. C 17 H 27 N03" (Me 293,4), 1148500, [2444-46-4).
Nitrogen monoxide. NO, (Mr 30,01), 1108300, N- [( 4-Hydroxy-3-methoxyphenyl)methyl) nonanamide,
Content: minimum 98,0 per cent V/V, White or almost white, crystalline powder, practically
insoluble in cold water, freely soluble in anhydrous ethanoL
Nitromethane. CH 3NO z' (Me 6l.0), 1059700, [75-52-5),
Nonivamide used in the test for nonivamide in the monograph
Clear, colourless, oily liquid, slightly soluble in water, miscible
Capsicum (1859) complies with the following additional test.
with ethanol (96 per cent),
Assay. Liquid chromatography (2,2,29) as prescribed in the
dSg: 1.132 to 1.134, monograph Capsicum (1859),
n~o: 1.381 to 1.383,
Content: minimum 98,0 per cent, calculated by the
Distillation range (2.2,11), Not less than 95 per cent distils normalisation procedure,
between 100 oC and 103 oC,
Nonylamine. C9 H 21 N, (Mr 143,3), 1139800, [112-20-9),
Nitro- molybdovanadic reagent. 1060100, N onan -l-amine. 1-Aminononane,
Solution A. Dissolve 10 g of ammonium molybdate R in Corrosive, colourless, clear liquid,
water R, add 1 mL of ammonia R and dilute to 100 mL with
d~o : about 0.788,
water R.
n~o: about 1,433,
Solution B, Dissolve 2,5 g of ammonium vanadate R in hot
water R, add 14 mL of nitric acid R and dilute to 500 mL with Nordazepam. C 1sH ll CINp, (Mr 270.7), 1060200,
water R. [1088-11-5), 7-Chloro-2,3-dihydro-5-phenyl-lH-1,4-
To 96 mL of nitric acid R add 100 mL of solution A and benzodiazepin -2-one,
100 mL of solution B and dilute to 500 mL with water R. White or pale yellow, crystalline powder, practically insoluble
4-NitrophenoL C6H SN03" (M, 139,1), 1146400, [100-02-7), in water, slightly soluble in ethanol (96 per cent),
p- NitrophenoL mp: about 216 oc.
Content: minimum 95 per cent DL-Norleudne. C 6 HlJN0 2 , (Mr 131.2),1060300, [616-06-8),
Colourless or slightly yellow powder, sparingly soluble in (RS)-2-Aminohexanoic acid,
water and in methanoL Shiny crystals, sparingly soluble in water and in ethanol
mp: about 114 oC, (96 per cent), soluble in acids,

General Notices (1) apply to all monographs and other texts 495
4. L 1. Reagents EUROPEAN PHARMACOPOEIA 8.0

Noscapine hydl'Ochloride. 1060500. [912-60-7]. d~g: about 0.782.


See Noscapine hydrochloride (0515). bp: 175 oC to 179 oc.
Ochratoxin A solution. 1175700. Oleamide. C 1s H 3S NO. (Mr 281.5). 1060900.
50 flg/mL solution of (2S)-2-([[(3R)-5-chloro-8-hydroxy- (9Z)-Octadec-9-enoamide.
3 -methyl-1-oxo-3,4-dihydro-lH -2-benzopyran -7- Yellowish or white powder or granules, practically insoluble
yI] carbonyl] amino )-3-phenylpropanoic acid (ochratoxin A) in water, very soluble in methylene chloride, soluble in
in a mixture of 1 volume of acetic acid R and 99 volumes of anhydrous ethanol.
benzene R. mp: about 80 oc.
Octadecyl [3- [3,5- bis( 1, l-dimethylethyl)-4- Oleanoli.c aeid. C30 H 4S 03' (Mr 456.7). 1183000. [508-02-1].
hydroxyphenyl]-pl'Opionate]. C 3S H 6P3' (Mr 530.9). 3~-Hydroxyolean-12-en-28-oic acid. Astrantiagenin C.
1060600. [2082-79-3]. Octadecy13-(3,5-di-tert-butyl-4-
hydroxyphenyl)propionate. Olei.c aeid. C 1sH 3P2' (M, 282.5). 1144100. [112-80-1].
White or slightly yellowish, crystalline powder, practically (9Z)-Octadec-9-enoic acid.
insoluble in water, very soluble in acetone and in hexane, Clear, colourless liquid, practically insoluble in water.
slightly soluble in methanol. d~o: about 0.891.
mp: 49 oC to 55 oc. n~o: about 1.459.
mp: 13 oC to 14 oc.
Octanal. C SH 16 0. (Mr 128.2).1150400. [124-13-0]. Octyl
aldehyde. Oleíe acid used in the assay of total fatty acids in the monograph
Oily, colourless liquido Practically insoluble in water. Saw palmetto fruit (1848) camplies with the following
additíonal test.
Octanal used in gas chromatography complies with the
following additional test. Assay. Gas chromatography (2.2.28) as prescribed in the
monograph Saw palmetto fruit (1848).
Assay. Gas chromatography (2.2.28) as prescribed in the
monograph Sweet orange oil (1811). Content: minimum 98 per cent, calculated by the
normalisation procedure.
Content: minimum 99 per cent, calculated by the
normalisation procedure. Oleuropein. C 2S H 3Pl3' (Mr 540.5). 1152900. [32619-42-4].
2- (3,4-DihydroxyphenyI)ethyl[ (2S,3E,4S)-3-ethylidene- 2-(~­
Octane. C SH 1S ' (M, 114.2).1166500. [111-65-9]. D-glucopyranosyloxy)-5-(methoxycarbonyl)-3,4-dihydro-2H-
Octano!. C SH 1S O. (Mr 130.2). 1060700. [111-87-5]. pyran -4-yl] aceta te.
Octan-1-ol. Caprylic alcohol. Powder, soluble in methanol.
Colourless liquid, practically insoluble in water, miscible with Oleuropeín used in Olive lea! (1878) camplies with the following
ethanol (96 per cent). test.
d§g: about 0.828. Assay. Liquid chromatography (2.2.29) as prescribed in the
bp: about 195 oc. monograph Olive leaf (1878).
Content: minimum 80 per cent, calculated by the
3-0ctanone. CS H 16 0. (Mr 128.2). 1114600. [106-68-3]. normalisation procedure.
Octan-3-one. Ethylpentylketone.
Colourless liquid with a characteristic odour. Oleyl alcohol. C1sH360. (Mr 268.5). 1156000. [143-28-2].
d~g : about 0.822.
(9Z)-Octadec-9-en-1-01.
n~o: about 1.415.
bp: about 207 oc.
n~o: l.460.
bp: about 167 oc.
3-0ctanone used in gas chromatography complies with the Content: mínimum 85 per cent.
following additíonal test. Olive oH. 1061000. [8001-25-0].
Assay. Gas chromatography (2.2.28) as prescribed in the See Olive oil, virgin (0518).
monograph Lavender oil (1338).
Test solution. The substance to be examined. Oracet blue 2R. C2o H 14NP2' (Mr 314.3). 1061100.
[4395-65-7].
Content: minimum 98.0 per cent, calculated by the
normalisation procedure. Colour Index No. 61110.
1- Amino-4-(phenylamino )anthracene-9, 1O-dione.
OctoxinollO. C34 H 6Pll (average). (M, 647). 1060800. mp: about 194 oc.
[9002-93-1]. Q- [4-(1,1,3,3-Tetramethylbutyl)phenyl]-w-
hydroxypoly-(oxyethylene). Ordnol. C 7 H sO z,H 2 0. (Mr 142.2). 1108700. [6153-39-5].
Clear, pale-yellow, viscous liquid, miscible with water, with 5-Methylbenzene-1,3-diol monohydrate.
acetone and with ethanol (96 per cent), soluble in toluene. Crystalline powder, sensitive to light.
Storage: in an airtight container. bp: about 290 oc.
mp: 58 oC to 61°C.
Octreotide acetate. C49H66NlOOlOSZ,xCzH402' 1182900.
[79517-01-4]. (Acetate-free peptide: M r 1019. Organosilica polymer, amorphous, octadecylsilyl. 1144200.
[83150-76-9]). D-Phenylalanyl-L-cysteinyl-L-phenylalanyl- Synthetic, spherical hybrid partides, containing both
D-tryptophyl-L-lysyl-L-threonyl-N- [( 1R,2R)-2-hydroxy-1- inorganic (silica) and organic (organosiloxanes) components,
(hydroxymethyl)propyl]-L-cysteinamide cycIic (2-¿ 7)-disulfide chemically modified at the surface by trifunctionally bonded
acetate. It contains a variable quantity of acetic acid. octadecylsilyl groups.
White or almost white powder, freely soluble in water and
acetic acid. Organosilica polymer, amorphous, octadecylsilyl,
Content: minimum 96.0 per cent. end-capped. 1178600.
Synthetic, spherical hybrid particIes, containing both
Octylamine. CS H 19 N. (Mr 129.2). 1150500. [111-86-4]. inorganic (silica) and organic (organosiloxanes) components,
Octan -l-amine. chemically modified at the surface by trifunctionally bonded
Colourless liquido octadecylsilyl groups. To minimise any interactiol1 with basic

496 See the information section on general monographs (caver pages)


EUROPEAN PHARMACOPOEIA 8.0 4.1.1. Reagents

compounds, it is carefully end-capped to cover most of the Nitrogen and argon: less than 100 ppm.
remaining silanol groups. The particle size is indicated after Carbon dioxide: les s than 10 ppm.
the name of the reagent in the tests where it is used. Carbon monoxide: less than 5 ppm.
Organosilka polymer, amorphous, polar-embedded Oxygen Rl. 02' (Mr 32.00). 1137600.
octadecylsilyl, end-capped. 1150600.
Content: minimum 99 per cent V/V.
Synthetic, spherical hybrid particles containing both inorganic
(silica) and organic (organosiloxanes) components, chemically Oxytetracydine hydrochloride. 1146500.
modified at the surface by the bonding of polar embedded See Oxytetracycline hydrochloride (0198).
octadecylsilyl groups. To minimise any interaction with basic
compounds, it is carefully end-capped to cover most of the Palladillm. Pd. (A, 106.4). 1114700. [7440-05-3].
remaining silanol groups. The particle size is indicated after Grey white metal, soluble in hydrochloric acid.
the name of the reagent in the tests where it is used.
Palladium ehloride. PdCI 2. (MI 177.3). 1061500. [7647-10-1].
Organosilka polymer, amorphous, propyl-2-phenylsilyl, Red crystals.
end-capped. 1178100. mp: 678 oC to 680 oc.
Synthetic, spherical hybrid particles containing both inorganic
(silica) and organic (organosiloxanes) components, chemically Palladium chloride solution. 1061501.
modified at the surface by the bonding of propyl-2-phenylsilyl Dissolve 1 g of palladium chloride R in 10 mL oí warm
groups. To minimise any interaction with basic compounds, it hydroch/oric acid R. Dilute the solutiol1 to 250 mL with a
is carefully end-capped to cover most of the remaining silanol mixture of equal volumes of dilute hydroch/oric acid R and
groups. The particle size is indicated after the name of the water R. Dilute this solution immediately before use with
reagent in the tests where it is used. 2 volumes of water R.
Organosiliea polymer for mas s spectrometry, amorphous, Palmitic add. C 16 H 32 02' (Mr 256.4). 1061600. [57-10-3].
oetadeeylsilyl, end-capped. 1164900. Hexadecanoic acid.
Synthetic, spherical hybrid particles containing both inorganic White or almost white, crystalline scales, practically insoluble
(silica) and organic (organosiloxanes) components. To in water, freely soluble in hot ethanol (96 per cent).
minimise any interaction with basic compounds, it is carefully mp: about 63 oc.
end -capped to cover most of the remaining silanol groups. Chromatography. Thin-Iayer chromatography (2.2.27)
The particle size is indicated after the name of the reagent in as prescribed in the monograph Ch/oramphenicol
the tests where it is used. palmitate (0473); the chromatogram shows only one principal
Osmillm tetroxide. OS04' (MI 254.2). 1061200. [20816-12-0]. spot.
Light-yellow needles or a yellow, crystalline mass, hygroscopic, Palmitie acid used in the assay of total fatty acids in the
light sensitive, soluble in water and in ethanol (96 per cent). monograph Saw palmetto fruit (1848) comp/ies with the
following additional test.
Storage: in an airtight container.
Assay. Gas chromatography (2.2.28) as prescribed in the
Osmium tetroxide solution. 1061201. monograph Saw palmetto fruit (1848).
A 2.5 giL solution in 0.05 M sulfuric acid. Content: minimum 98 per cent, calculated by the
normalisation procedure.
Osthole. ClSH160y (MI 244.3). 1180500. [484-12-8].
7 -Methoxy-8-( 3-methylbut -2-enyl)-2H-1-benzopyran -2-one. Palmitoleic adJ. C16H3002' (MI 254.4). 1144400. [373-49-9].
7 -Methoxy -8-isopentenylcoumarin. (9Z)- Hexadec-9-enoic acid.
OxaHe add. C2 H 2 04,2Hp. (M, 126.1).1061400. [6153-56-6]. CIear, colourless liquido
Ethanedioic acid dihydrate. bp: about 162 oc.
White or almost white crystals, soluble in water, Íreely soluble Palmito/eie acid used in the assay of total fatty acids in Saw
in ethanol (96 per cent). palmetto fruit (1848) eomplíes with the following additional
test.
OxaHc acid and sulfuric add sollltion. 1061401.
Assay. Gas chromatography (2.2.28) as prescribed in the
A 50 giL solution of oxalic acid R in a cooled mixture of monograph Saw palmetto fruit (1848).
equal volumes of sulfuric acid R and water R.
Content: minimum 98 per cent, calculated by the
Oxazepam. 1144300. [604-75-1]. normalisation procedure.
See Oxazepam (0778). PalmityI alcohoL C16H3P, (MI 242.4). 1156100.
Ox brain, acetone-dried. 1061300. [36653-82-4]. Hexadecan-1-ol. Cetyl alcohol.
Cut into small pieces a fresh ox brain previously freed mp: about 48 oc.
from vascular and connective tissue. Place in acetone R Content: minimum 96 per cent.
for preliminary dehydration. Complete the dehydratioll by Pancreas powder. 1061700.
pounding in a mortar 30 g of this material with successive
quantities, each of 75 mL, of acetone R until a dry powder See Pancreas powder (0350).
is obtained aÍter filtration. Dryat 37 oC for 2 h or until the Papain. 1150700. [9001-73-4].
odour oí acetone is no longer present. A proteolytic enzyme obtained Írom the latex of the green
2,2' -Oxybis(N,N-dimethylethylami.ne). C S H 20 Np. fruit and leaves of Carica papaya L.
(Mr 160.3). 1141200. [3033-62-3]. Bis(2-dimethylaminoethyl) Papaverine hydrochloride. 1061800. [61-25-6].
ether.
See Papaverine hydrochloride (0102).
Colourless, corrosive liquido
d~g: about 0.85. Paper chromatography performance test sollltions.
n~o: about 1.430.
1150800.
Test so/ution (A). Sodium perteehnetate (99mTc) injection
Oxygen. 02' (MI 32.00). 1108800. (fission) (0124) or Sodium perteehnetate (99"'Tc) injection
Content: minimum 99.99 per cent V/V. (non-fission) (0283).

General Notices (1) app/y to all monographs and other texts 497
4.1.1. Reagents EUROPEAN PHARMACOPOEIA 8.0

Test solutíon (B). In a closed vial mix 100 flL of a 5 giL Parthenolide. C 1sH 2aÜ3" (Mr 248.3). 1129900. [20554-84-1].
solution of stannous chloride R in 0.05 M hydrochloric acid (4E)-(laR,7a5,10aS, 10bS)-la,5-Dimethyl-8-methylene-
and 100 MBq to 200 MBq of Sodium pertechnetate (99mTc) 2,3,6,7,7 a,8, lOa, 1Ob-octahydro-oxireno [9,10] cyclodeca[ 1,2-
injectíon (jission) (0124) or Sodium pertechnetate (99mTc) b] furan-9( 1aH)-one. (E) -( 55,65) -4,5-Epoxygermacra-
injection (non-fission) (0283) in a volume not exceeding 2 mL. 1(1 O), 11 (13 )-dieno-12( 6)-lactone.
White or almost white, crystalline powder, very slightly
Paper for chromatography. 1150900.
soluble in water, very soluble in methylene chloride, soluble
Pure cellulose grade thin paper with a smooth surface and in methanol.
a thickness of abont 0.2 mm.
[a]g: - 71.4, determined on a 2.2 giL solution in methylene
Chromatographic separation. To 2 strip s of paper for chloride R.
chromatography R apply separately 2-5 flL of test solution (a) mp: 115 oC to 116 oc.
and test solution (b) of paper chromatography performance
test solutions R. Develop over a pathlength of 3/4 of the paper Absorbance (2.2.25). A 0.01 giL solution in ethanol (96 per
height, using a mixture of equal volumes of methanol R cent) R shows an absorption maximum at 214 nm.
and water R. Allow to dry and determine the distribution Assay. Liquid chromatography (2.2.29) as prescribed in the
of radioactivity using a suitable detector. The paper is not monograph Feverfew (1516), at the concentration of the
satisfactory, unless the chromatogram obtained with test reference solution.
solution (a) shows a single radioactivity spot with an Rp value Content: minimum 90 per cent, calculated by the
in the range 0.8-1.0 and the chromatogram obtained with test normalisation procedure.
solution (b) shows a single radioactivity spot at the application
point (R p value in the range 0.0-0.1). PeniciHinase solution. 1062300.
Paracetamol. 1061900. [103-90-2]. Dissolve 10 g of casein hydrolysate, 2.72 g of potassium
dihydrogen phosphate R and 5.88 g of sodium citrate R in
See Paracetamol (0049). 200 mL of water R, adjust to pH 7.2 with a 200 giL solution
of sodium hydroxide R and dilute to 1000 mL with water R.
Paracetamol,4-aminophenol-free. 106190l.
Dissolve 0.41 g of magnesium sulfate R in 5 mL of water R
Recrystallise paracetamol R from water R and dry in vacuo and add 1 mL of a 1.6 giL solution of ferrous ammonium
at 70 oC; repeat the procedure until the product complíes sulfate R and sufficient water R to produce 10 mL. Sterilise
with the following test: dissolve 5 g of the dried substance both solutions by heating in an autoclave, cool, mix, distribute
in a mixture of equal volumes of methanol R and water R in shallow layers in conical flasks and inoculate with Bacillus
and dilute to 100 mL with the same mixture of solvents. cereus (NCTC 9946). Allow the flasks to stand at 18 oC to
Add 1 mL of a freshly prepared solution containing 10 giL 37 oC until growth is apparent and then maintain at 35 oC
of sodium nitroprusside R and 10 giL of anhydrous sodium to 37 oC for 16 h, shaking constantly to ensure maximum
carbonate R, mix and allow to stand for 30 min protected aeration. Centrifuge and sterilise the supernatant by filtration
from light. No blue or green colour is produced. through a membrane filter. 1.0 mL of penicillinase solution
contains not less than 0.4 microkatals (corresponding to the
Paraffin, liquido 1062000. [8042-47-5]. hydrolysis of not les s than 500 mg of benzylpenicillin to
See Líquid paraffin (0239). benzylpenicilloic acid per hour) at 30 oC and pH 7, provided
that the concentration of benzylpenicillin does not faH below
Paraffin, white 50ft. 1062100. the leve] necessary for enzyme saturation.
A semi-liquid mixture of hydrocarbons obtained from The Michaelis constant for benzylpenicillin of the penicillinase
petroleum and bleached, practically insoluble in water and in penicillinase solution is approximately 12 flg/mL.
in ethanol (96 per cent), soluble in light petroleum Rl, the
Sterility (2.6.1). It complíes with the test for sterility.
solution sometimes showing a slight opalescence.
Storage: at a temperature between O oC and 2 oC for 2 to
Paraldehyde. 1151000. [123-63-7]. 3 days. When freeze-dried and kept in sealed ampoules, it
See Paraldehyde (0351). may be stored for several months.

Pararosaniline hydrochloride. C19H1SClN3. (Mr 323.8). Pentaerythrityl tetrakis[3-(3,5-di(1,1-dimethylethyl)-


1062200. [569-61-9]. 4-hydroxyphenyl)propionate]. C73HIOS012. (Mr 1178).
1062400. [6683-19-8]. Pentaerythrityl tetrakis [3-
Schultz No. 779. (3,5-di - tert-butyl-4- hydroxyphenyl) propionate].
Colour Index No. 42500. 2,2' -Bis(hydroxymethyl)propane-l,3-diol tetrakis[3- [3,5-
4- [Bis( 4-aminophenyl)methylene] cyclohexa -2,5-dieniminium di( 1, l-dimethylethyl) -4-hydroxyphenyl]]propionate.
chloride. White or slight1y yellow, crystalline powder, practically
Bluish-red, crystalline powder, slightly soluble in water, insoluble in water, very soluble in acetone, soluble in
soluble in anhydrous ethanol. Solutions in water and methanol, slightly soluble in hexane.
anhydrous ethanol are deep-red; solutions in sulfuric acid and mp: 110 oC to 125 oc.
in hydrochloric acid are yellow.
a-form: 120 oC to 125 oC.
mp: about 270 oC, with decomposition.
~-form: 110 oC to 115 oc.
Dewlorised pararosaniHne solution. 1062201.
Pentaftuoropropanoic add. C3 HF s02. (Mr 164.0). 1151100.
To 0.1 g of pararosaniline hydrochloride R in a [422-64-0].
ground-glass-stoppered flask add 60 mL of water R and
Clear, colourless liquido
a solution of 1.0 g of anhydrous sodium sulfite R or 2.0 g
of sodium sulfite R or 0.75 g of sodium metabisulfite R in d~g: about 1.561.
10 mL of water R. Slowly and with stirring add 6 mL of n~o : about 1.284.
dilute hydrochloric acid R, stopper the flask and continue
bp: about 97 oc.
stirring until dissolution is complete. Dilute to 100 mL
with water R. Allow to stand for 12 h before use. Pentafluoropropionk anhydríde. C6FIO03. (Mr 310.0).
Storage: protected from light. 1177300. [356-42-3]. Pentafluoropropanoic anhydride.

498 See the information section on general monographs (caver pages)


EUROPEAN PHARMACOPOEIA 8,0 4,1.1. Reagents

Pentane. CSH 12 , (Mr 72,2), 1062500, [109-66-0], Permethrin. C2 ¡H 20 ClP3" (Mr 391.3).1130000. [52645-53-1].
Clear, colourless, flammable Iiquid, very slightly soluble in mp: 34 oC to 35 oc.
water, miscible with acetone and with anhydrous ethanoL A suitable certified reference solution (10 ng/ flL in
d~g : about 0,63, cydohexane) may be used.
n~o : about 1.359,
Peroxide test strips. 1147800.
bp: about 36 oc. Use commercial test strips with a suitable scale in the range
Pentane used in spectrophotometry complies with the following from O ppm to 25 ppm peroxide,
additional test,
Mínimum transmittance (2.2.25) using water R as Perylene. C 20 H¡2' (Mr 252.3), 1130100. [198-55-0].
compensation Iiquid: 20 per cent at 200 nm, 50 per cent at Dibenz [de,kl] anthracene.
210 nm, 85 per cent at 220 nm, 93 per cent at 230 nm, 98 per Orange powder.
cent at 240 nm, mp: about 279 oc.
1,2-Pentanediol. C SH 12 0 2 , (Mr 104,2), 1155800, [5343-92-0], Petroleum, light. 1063100. [8032-32-4]. Petroleum ether
(2RS) - Pentane-l ,2-dioL 50-70 oc.
d~o: about 0,971. Clear, colourless, flammable liquid without fluorescence,
n~o: about 1.439, practically insoluble in water, miscible with ethanol (96 per
bp: about 201°C. cent).
d~g: 0.661 to 0.664.
Pentanol. CSH¡p, (Mr 88,1), 1062600, [71-41-0], Distillation range (2.2.11): 50 oC to 70 oc.
Pentan -l-oL
Colourless liquid, sparingly soluble in water, miscible with Petroleum, light Rl. 1063101. Petroleum ether 40-60 oc.
ethanol (96 per cent), Complies with the requirements prescribed for light
n~o: about 1.410, petroleum R, with the following modifications.
bp: about 137 oc. d~g : 0.630 to 0.656.
Distillation range (2.2.11): 40 oC to 60 oc. It does not
3-Pentanone. C SHlQo. (Mr 86,13), 1173600, [96-22-0],
become doudy at O oc.
Pentan-3-one, Diethy! ketone,
Petroleum, light R2. 1063102. Petroleum ether 30-40 oc.
tert-Pentyl alcohoL C SH 12 0, (M, 88,1), 1062700, [75-85-4],
tert-Amyl alcohoL 2-Methyl-2-butanoL Complies with the requirements prescribed for light
petroleum R, with the following modifications.
Volatile, flammable liquid, freely soluble in water, miscible
with ethanol (96 per cent) and with glycerol. d~g : 0.620 to 0.630.
d~g: about 0,81. Distillation range (2.2.11): 30 oC to 40 oc. 1t does not
become doudy at O oc.
Distillation range (2.2,11), Not less than 95 per cent distils
between 100 oC and 104 oc. Petroleum, light R3. 1063103. Petroleum ether 100-120 oc.
Storage: protected from light Complies with the requirements prescribed for light
petroleum R, with the following modifications.
Pentetic add. C¡4H23NP¡O' (Mr 393,3), 1183100, [67-43-6],
[[ (Carboxymethyl)imino ]bis( ethylenenitrilo)] tetraacetic acid, d§g: about 0.720.
White or almost white powder, slightly soluble in water. Distillation range (2.2.11): 100 oC to 120 oc.
mp: 219 oC to 220 oC, with decomposition, Water (2.5.12): maximum 0.03 per cent.
Pepsin powder. 1062800, [9001-75-6], Petroleum, light R4. 1063104. Petroleum ether 80-100 oc.
See Pepsin powder (0682), Complies with the requirements prescribed for light
petroleum R, with the following modifications.
Peptide N-glycosidase F. 1186600, [83534-39-8],
d~g: about 0.70.
Peptide-N4-(N-acetyl-~-glucosaminyl)asparagine amidase
Distillation range (2.2.11): 80 oC to 100 oc.
(EC 3,5,1.52), PNGase F.
Perchloric acid. HCI0 4 • (Mr 100.5). 1062900. [7601-90-3].
pH indicator strip. 1178900.
PI as tic strip containing multiple segments of different
Content: 70.0 per cent m/m to 73.0 per cent mimo
dye-impregnated papers allowing visual determination of pH
Clear, colourless Iiquid, miscible with water. in the prescribed range by comparison with a master chart.
d~g: about 1.7,
a-Phellandrene. CIOHI6" (M,. l36.2). 1130400.
Assay. To 2.50 g add 50 mL of water R and titrate with 1 M
[4221-98-1]. (R)-5- IsopropyI-2-methyl-cydohexa-1,3-diene.
sodium hydroxide, using 0.1 mL of methyl red solution R as
(-)-p-Mentha-1,5-diene.
indicator.
n~o : about 1.471.
1 mL of 1 M sodium hydroxide is equivalent to 100.5 mg of
HCI0 4 • bp: 171°C to 174 oc.
a-Phellandrene used in gas chromatography complies with the
Perchloric add solution. 1062901. following additional test.
Dilute 8.5 mL of perchloric acid R to 100 mL with water R. Assay. Gas chromatography (2.2.28) as prescribed in the
Periodk acetk acid solution. 1063000. monograph Eucalyptus oil (0390).
Dissolve 0.446 g of sodium periodate R in 2.5 mL of a 25 per Test solution. The substance to be examined.
cent V/V solution of sulfuric acid R. Dilute to 100.0 mL with Content: 95.0 per cent, calculated by the normalisation
glacial acetic acid R. procedure.
Periodk acid. H s106' (M, 227.9). 1108900. [10450-60-9]. Phenanthrene. C¡4HJO" (M, 178.2). 1063200. [85-01-8].
Crystals, freely soluble in water and soluble in ethanol (96 per White or almost white crystals, practically insoluble in water,
cent). sparingly soluble in ethanol (96 per cent).
mp: about 122 oc. mp: about 100 oc.

General Notices (1) apply to all monographs and other texts 499
4.1.1. Reagents EUROPEAN PHARMACOPOEIA 8.0

Phenanthroline hydrochloride. C¡2H9CIN2,H20. (Mr 234.7). Phenoxyacetic acid. CSHgÜ3' (Mr 152.1). 1063800.
1063300. [3829-86-5]. 1,l0-Phenanthroline hydrochloride [122-59-8]. 2-Phenoxyethanoic acid.
monohydrate. Almost white crystals, sparingly soluble in water, freely soluble
White or almost white, crystalline powder, freely soluble in in ethanol (96 per cent), and in glacial acetic acid.
water, soluble in ethanol (96 per cent). mp: about 98 oc.
mp: about 215 oC, with decomposition. Chromatography. Thin-Iayer chromatography (2.2.27) as
prescribed in the monograph Phenoxymethylpenicillin (0148);
Phenazone. 1063400. [60-80-0]. the chromatogram shows only one principal spot.
See Phenazone (0421). 2-Phenoxyaniline. C¡2HllNO. (Mr 185.2). 1165500.
[2688-84-8]. 2-Phenoxybenzenamine. 2-Aminophenyl phenyl
Phenol. 1063500. [108-95-2].
ether.
See Phenol (0631).
Phenoxybenzamine hydrochloride. C¡SH 23 CI2NO.
Phenolphthalein. C10H¡P4' (Mr 318.3).1063700. [77-09-8]. (M r 340.3). 1063900. N-(2-Chloroethyl)-N-(l-methyl-2-
3,3-Bis( 4-hydroxyphenyl) -3H- isobenzofuran -l-one. phenoxyethyl)-benzylamine hydrochloride.
White or yellowish-white powder, practically insoluble in Content: 97.0 per cent to 103.0 per cent (dried substance).
water, soluble in ethanol (96 per cent). White or almost white, crystalline powder, sparingly soluble
in water, freely soluble in ethanol (96 per cent).
Phenolphthalein papero 1063704. mp: about 138 oc.
Immerse strips of filter paper for a few minutes in L055 on drying (2.2.32): maximum 0.5 per cent, determined
phenolphthalein solution R. Allow to dry. by drying over diphosphorus pentoxide R at a pressure not
exceeding 670 Pa for 24 h.
Phenolphthalein solution. 1063702.
Assay. Dissolve 0.500 g in 50.0 mL of ethanol-free chloroform R
Dissolve 0.1 g of phenolphthalein R in 80 mL of ethanol and extract with three quantities, each of 20 mL, of 0.01 M
(96 per cent) R and dilute to 100 mL with water R. hydrochloric acid. Discard the acid extracts, filter the
Test for sensitivity. To 0.1 mL of the phenolphthalein chloroform layer through cotton and dilute 5.0 mL of the
solution add 100 mL of carbon dioxide-free water R. The filtrate to 500.0 mL with ethanol-free chloroform R. Measure
solution is colourless. Not more than 0.2 mL of 0.02 M the absorbance of the resulting solution in a closed ceH at the
sodium hydroxide is required to change the colour to pink. maximum at 272 nm. Calculate the content of C¡SH 23 CI 2NO,
taking the speciflc absorbance to be 56.3.
Colour change: pH 8.2 (colourless) to pH 10.0 (red).
Storage: protected from light.
Phenolphthalein solution Rl. 1063703.
Phenoxyethanol. CSHlO02. (M, 138.2). 1064000. [122-99-6].
A 10 giL solution in ethanol (96 per cent) R. 2-Phenoxyethanol.
Clear, colourless, oily liquid, slightly soluble in water, freeIy
Phenol red. 1063600. [143-74-8].
soluble in ethanol (96 per cent).
Bright red or dark red, crystalline powder, very slightly soluble d~g : about 1.11.
in water, slightly soluble in ethanol (96 per cent).
n~o: about 1.537.
Phenol red solution. 1063601. Freezing point (2.2.18): minimum 12 oc.
Dissolve 0.1 g of phenol red R in a mixture of 2.82 mL of Phenylacetic add. C SHgÜ2' (Mr 136.2). 1160000. [103-82-2].
0.1 M sodium hydroxide and 20 mL of ethanol (96 per White or almost white powder, soluble in water.
cent) R and dilute to 100 mL with water R.
bp: about 265 oc.
Test for sensitivity. Add 0.1 mL of the phenol red solution
mp: about 75 oc.
to 100 mL of carbon dioxide-free water R. The solution is
yellow. Not more than 0.1 mL of 0.02 M sodium hydroxide Phenylalanine. 1064100. [63-91-2].
is required to change the colour to reddish-violet. See Phenylalanine (0782).
Colour change: pH 6.8 (yellow) to pH 8.4 (reddish-violet).
p-Phenylenediamine dihydrochloride. C 6 H lO CI 2N 2 •
Phenol red solution R2. 1063603. (M r 181.1). 1064200. [615-28-1]. 1,4-Diaminobenzene
dihydrochloride.
Solution A. Dissolve 33 mg of phenol red R in 1.5 mL of
dilute sodium hydroxide solution R and dilute to 100 mL Crystalline powder or white or slightly coloured crystals,
with water R. turning reddish on exposure to air, freely soluble in water,
slightly soluble in ethanol (96 per cent).
Solution B. Dissolve 25 mg of ammonium sulfate R in
235 mL of water R; add 105 mL of dilule sodium hydroxide a-Phenylglydne. C SH 9NO z' (Mr 151.2). 1064300.
solution R and 135 mL of dilute acetic acid R. [2835-06-5]. (RS)-2-Amino-2-phenylacetic acid.
Add 25 mL of solution A to solution B. If necessary, adjust D-Phenylglydne. C SH 9 NO z' (Mr 151.2).1144500. [875-74-1].
the pH of the mixture to 4.7. (2R)-2-Amino-2-phenylacetic acid.
Phenol red solution R3. 1063604. Content: minimum 99 per cent.
White or almost white, crystalline powder.
Solution A. Dissolve 33 mg of phenol red R in 1.5 mL of
dilute sodium hydroxide solution R and dilute to 50 mL Phenylhydrazine hydrochloride. C6 H 9 CIN 2 • (Mr 144.6).
with water R. 1064500. [59-88-1].
Solution B. Dissolve 50 mg of ammonium sulfate R in White or almost white, crystalline powder, becoming brown
235 mL of water R; add 105 mL of dilute sodium hydroxide on exposure to air, soluble in water and in ethanol (96 per
solution R and 135 mL of dilute acetic acid R. cent).
Add 25 mL of solution A to solution B; if necessary, adjust mp: about 245 oC, with decomposition.
the pH of the mixture to 4.7. Storage: protected from light.

500 See the information section on general monograph5 (caver pages)


EUROPEAN PHARl'v1ACOPOEIA 8.0 4.1.1. Reagents

Phenylhydrazine hydrochloride solution. 1064501. Storage: at 2 oC to 8 oc.


Dissolve 0.9 g of phenylhydrazine hydrochloride R in 50 mL Phosphomolybdotungsti.c reagent, dilute. 1065001.
of water R. Decolorise with activated chareoal R and filter.
To the filtrate add 30 mL of hydrochloric acid R and dilute To 1 volume of phosphomolybdotungstic reagent R add
to 250 mL with water R. 2 volumes of water R.

Phenylhydrazine-sulfmic acid solution. 1064502. Phosphoric aeid. 1065100. [7664-38-2).


Dissolve 65 mg of phenylhydrazine hydrochloride R, See Concentrated phosphoric acid (0004).
previously recrystallised from ethanol (85 per cent V/V) R, Phosphoric aci.d, dilute. 1065101.
in a mixture of 80 volumes of water R and 170 volumes of See Dilute phosphoric acid (0005).
sulfuric acid R and dilute to 100 mL with the same mixture
of solvents. Prepare immediately before use. Phosphoric aci.d, dilute Rl. 1065102.
Dilute 93 mL of dilute phosphoric acid R to 1000 mL with
PhenyI isothiocyanate. C7 H sNS. (Mr 135.2). 1121500.
water R.
[103-72-0).
Liquid, insoluble in water, soluble in ethanol (96 per cent). Phosphorous add. H 3P03' (M, 82.0). 1130600. [13598-36-2).
d~g: about 1.13. White or almost white, very hygroscopic and deliquescent
n~O : about 1.65. crystalline mass; slowly oxidised by oxygen (air) to H 3P0 4 •
bp: about 221 oc. Unstable, orthorhombic crystals, soluble in water, in ethanol
(96 per cent) and in a mixture of 3 volumes of ether and
mp: about - 21 oc.
1 volume of ethanol (96 per cent).
Use a grade suitable for protein sequencing.
d~l: 1.651.
I-Phenylpiperazine. C IO H 14 N 2 • (Mr 162.2). 1130500. mp: about 73 oc.
[92-54-6).
Phosphotungstic add solution. 1065200.
Slightly viscous, yellow liquid, not miscible with water.
Heat under a reflux condenser for 3 h, 10 g of sodium
d~o: about 1.07.
tungstate R with 8 mL of phosphoric aeid R and 75 mL of
n~o : about 1.588. water R. Allow to cool and dilute to 100 mL with water R.
Phloroglucide. C 12HIOOs' (Me 234.2). 1177400. [491-45-2). Phthalaldehyde. C SHP2' (Me 134.1). 1065300. [643-79-8).
2,3',4,5',6-Biphenylpentol. Benzene-1,2-dicarboxaldehyde.
White or almost white powder, hygroscopic, light sensitive. Yellow, crystalline powder.
Slowly discolours on exposure to light. mp: about 55 oc.
Phlorogludnol. C6 HP3,2Hp. (Mr 162.1). 1064600. Storage: protected from light and air.
[6099-90-7). Benzene-1,3,5-triol.
Phthalaldehyde reagent. 1065301.
White or yellowish crystals, slightly soluble in water, soluble
in ethanol (96 per cent). Dissolve 2047 g of borie acid R in 75 mL of water R, adjust to
pH lOA using a 450 giL solution of potassium hydroxide R
mp: about 223 oC (instantaneous method).
and dilute to 100 mL with water R. Dissolve 1.0 g of
Phloroglucinol solution. 1064601. phthalaldehyde R in 5 mL of methanol R, add 95 mL of the
To 1 mL of a 100 giL solution of phloroglucinol R in ethanol boric acid solution and 2 mL of thioglycollic acid R and
(96 per cent) R, add 9 mL of hydrochloric acid R. adjust to pH lOA with a 450 giL solution of potassium
hydroxide R.
Storage: protected from light.
Storage: protected from light; use within 3 days.
Phosalone. C12HlSCIN04PS2' (Mr 367.8). 1130200.
[2310-17-0]. Phthalazine. C SH 6N 2 • (Mr 130.1).1065400. [253-52-1).
mp: 45 oC to 48 oC Pale yellow crystals, freely soluble in water, soluble in
anhydrous ethanol, in ethyl acetate and in methanol.
A suitable certified reference solution (10 ngl flL in iso-octane)
mp: 89 oC to 92 oc.
may be used.
Phosphomolybdic acid. 12Mo0 3 ,H3 P0 4 ,xHp.1064900. Phthalein purple. C32H32N2012,xH20. (M, 637,
[51429-74-4). anhydrous substance). 1065500. [2411-89-4).
Metalphthalein. 2,2',2",2 "'- [o-Cresolphthalein -3',3"-
Orange-yellow, fine crystals, freely soluble in water, soluble in bis (methylenenitrilo ) )tetra-acetic acid. (l,3-Dihydro-3-
ethanol (96 per cent). oxo-isobenzofuran-1-ylidene)bis[ (6-hydroxy-5-methyl-3,1-
Phosphomolybdic aeid solution. 1064901. phenylene) bis (methyleneimino )diacetic acid).
Dissolve 4 g of phosphomolybdic acid R in water R and Yellowish-white or brownish powder, practically insoluble
dilute to 40 mL with the same solvent. Add cautiously and in water, soluble in ethanol (96 per cent). The product may
with cooling 60 mL of sulfuric acid R. Prepare immediately be found in commerce in the form of the sodium salt: a
before use. yellowish-white to pink powder, soluble in water, practically
insoluble in ethanol (96 per cent).
Phosphomolybdotungstic reagent. 1065000. Test for sensitivity. Dissolve 10 mg in 1 mL of concentrated
Dissolve 100 g of sodium tungstate R and 25 g of sodium ammonia R and dilute to 100 mL with water R. To 5 mL of
molybdate R in 700 mL of water R. Add 100 mL of hydrochloric the solution add 95 mL of water R, 4 mL of concentrated
acid R and 50 mL of phosphorie acid R. Heat the mixture ammonia R, 50 mL of ethanol (96 per cent) R and 0.1 mL
under a reflux condenser in a glass apparatus for 10 h. Add of 0.1 M barium eh/oride. The solution is blue-violet. Add
150 g of lithium sulfate R, 50 mL of water R and a few drops 0.15 mL of 0.1 M sodium edetate. The solution becomes
of bromine R. Boil to remove the excess of bromine (15 min), colourless.
allow to cool, dilute to 1000 mL with water R and filter. The
reagent should be yellow in colour. If it acquires a greenish Phthalic add. CSH 6 0 4. (Mr 166.1). 1065600. [88-99-3).
tint, it is unsatisfactory for use but may be regenerated by Benzene-1,2-dicarboxylic acid.
boiling with a few drops of bromine R. Care must be taken to White or almost white, crystalline powder, soluble in hot
remove the excess of bromine by boiling. water and in ethanol (96 per cent).

General Notices (1) apply to all monographs and other texts 501
4.1.1. Reagents EUROPEAN PHARMACOPOEIA 8.0

Phthalic anhydride. C S H 4 0 3 • (Mr 148.1). 1065700. [85-44-9]. Piperidine. CSH ll N. (M, 85.2). 1066000. [110-89-4].
Isobenzofuran-1,3-dione. Hexahydropyridine.
Content: minimum 99.0 per cent. Colourless to slightly yellow, alkaline liquid, miscible with
White or almost white flakes. water, with ethanol (96 per cent) and with light petroleum.
mp: 130 oC to 132 oc, bp: about 106 oc.
Assay. Dissolve 2.000 g in 100 mL of water R and boil under a Piperine. C¡7H19N03' (M, 285.3). 1183200. [94-62-2].
re flux condenser for 30 mino Cool and titrate with 1 M sodium (2E,4E)-1-(Piperidin-l-yl)-5-( 1,3-benzodioxol-5-yl)penta-
hydroxide, using phenolphthalein solution R as indicator. 2,4-dien-1-one. 1- Piperoyl-piperidine. 1- [(2E,4E)-5-(3,4-
1 mL of 1 M sodium hydroxide is equivalent to 74.05 mg of Methylenedioxyphenyl) -l-oxo-2,4-pentadienyl]piperidine.
CS H 4 °3'
Piperitone. ClQH¡60. (M, 152.2). 1151200. [89-81-6].
Phthalic anhydride solution. 1065701. 6-Isopropyl- 3-methyl-cyclohex -2-en -l-one.
Dissolve 42 g of phthalic anhydride R in 300 mL of
anhydrous pyridine R. Allow to stand for 16 h. Pirimiphos-ethyl. C13H24N303PS, (M, 333.4). 1130300.
[23505-41-1].
Storage: protected from light; use within 1 week.
mp: 15 oC to 18 oc.
Picein. C¡4H¡sÜ7' (Mr 298.3). 1130700. [530-14-3]. A suitable certified reference solution (lO ng/[lL in
1- [4- (~- D-Glucopyranosyloxy)phenyl] ethanone. cyclohexane) may be used.
p-(Acetylphenyl) -~- D-glucopyranoside.
mp: 194 oC to 195 oc, Plasma, platelet-poor. 1066100.
Withdraw 45 mL of human blood into a 50 mL plastic syringe
Piuic add. C6H3NP7' (M, 229.1). 1065800. [88-89-1].
containing 5 mL of a sterile 38 giL solution of sodium cUrate R.
2,4,6-Trinitrophenol.
Without delay, centrifuge at 1500 g at 4 oC for 30 mino Remove
Yellow prisms or plates, soluble in water and in ethanol (96 per the upper two-thirds of the supernatant plasma using a plastic
cent). syringe and without delay centrifuge at 3500 g at 4 oC for
Storage: moistened with water R. 30 mino Remove the upper two- thirds of the liquid and freeze
it rapidly in suitable amounts in plastic tubes at or below
Picric add solution. 1065801. - 40 oc. Use plastic or silicone-treated equipment.
A 10 giL solution.
Plasma substrate. 1066200.
Piuic add solution Rl. 1065802.
Separate the plasma from human or bovine blood collected
Prepare 100 mL of a saturated solution of picric acid R and into one-ninth its volume of a 38 giL solution of sodium
add 0.25 mL of strong sodium hydroxide solution R. citrate R, or into two-sevenths its volume of a solution
u-Pinene. ClQHl6' (Mr 136.2). 1130800. [7785-70-8]. containing 20 giL of disodium hydrogen citrate R and 25 giL
(lR,5R)-2,6,6- Trimethylbicyclo[3.1.1]hept -2-ene. of g/ucose R. With the former, prepare the substrate on the
day of collection of the blood. With the latter, prepare within
Liquid not miscible with water.
two days of collection of the blood.
d~g: about 0.859.
Storage: at - 20 oc.
nbo : about 1.466.
bp: 154 oC to 156 oc, Plasma substrate Rl. 1066201.
a-Pi nene used in gas chromatography complies with the Use water-repellent equipment (madefrom materials such
following additional test. as suitable plastics 01' suitably silicone-treated glass) for
Assay. Gas chromatography (2.2.28) as prescribed in the taking and handling blood.
monograph Bitter-orange-flower oil (1175). Collect a suitable volume of blood from each of at least five
Test solution. The substance to be examined. sheep; a 285 mL volume of blood collected into 15 mL
of anticoagulant solution is suitable but smaller volumes
Content: minimum 99.0 per cent, calculated by the may be collected, taking the blood, either from a live
normalisation procedure.
animal or at the time of slaughter, using a needle attached
~-Pinene. CJOH¡6' (Mr 136.2). 1109000. [127-91-3]. to a suitable cannula which is long enough to reach the
6,6-Dimethyl-2-methylenebicyclo [3.1.1]heptane. bottom of the collecting vessel. Discarding the first few
Colourless, oily liquid, odour reminiscent of turpentine, millilitres and collecting only free-flowing blood, collect
practically insoluble in water, miscible with ethanol (96 per the blood in a sufficient quantity of an anticoagulant
cent). solution containing 8.7 g of sodium citrate R and 4 mg of
aprotinin R per 100 mL of water R to give a final ratio of
f3-Pinene used in gas chromatography complies with the blood to anticoagulant solution of 19 to 1. During and
following additional test. immediately after colleetion, swirl the flask gently to ensure
Assay. Gas chromatography (2.2.28) as prescribed in the mixing but do not allow frothing to occur. When collection
monograph Bitter-orange-flower oil (1175). is complete, close the flask and cool to 10-15 oc. When
Test solution. The substance to be examined. cold, pool the contents of all the flasks with the exception
Content: minimum 95.0 per cent. of any that show obvious haemolysis or clots and keep the
pooled blood at 10-15 oc.
l,4-Piperazinediethanesulfonic aeid. CSH¡SN Z0 6S2 . As 500n as possible and within 4 h of collection, centrifuge
(M, 302.4). 1186700. [5625-37-6]. Piperazine-1,4- the pooled blood at 1000-2000 g at 10-15 oC for 30 mino
bis(2-ethanesulfonic acid). 2,2' -(Piperazine-l,4- Separate the supernatant and centrifuge it at 5000 g for
diyl)bis( ethanesulfonic acid). Piperazine-N,N' -bis(2- 30 mino (Faster centrifugation, for example 20 000 g for
ethanesulfonic acid). PIPES. 30 min, may be used if necessary to clarify the plasma,
Content: minimum 99 per cent. but filtration pro ce dures should not be used.) Separate
White, crystalline powder. the supernatant and, without delay, mix thoroughly and
distribute the plasma substrate into small stoppered
Piperazine hydrate. 1065900. [142-63-2]. containers in portions sufficient for a complete heparin
See Piperazine hydrate (0425). assay (for example 10 mL to 30 mL). Without delay, rapidly

502 See the information sectiol1 on general monographs (cover pages)


EUROPEAN PHARMACOPOEIA 8.0 4.1.1. Reagents

cool to a temperature below - 70 oC (for example by Poly[ (cyanopropyl) (phenyl)] [dimethyl] siloxane. 1114800.
immel'sing the containers into liquid nitro gen) and sto re at Stationary phase for gas chromatography.
a temperature below - 30 oc.
Contains 6 per cent of (cyanopropyl) (phenyl) groups and
The plasma is suitable fol' use as plasma substrate in the 94 per cent of dimethyl groups.
assay for heparin if, under the conditions of the assay,
it gives a clotting time appropriate to the method of Poly( cyanopropyl) (phenylmethyl)siloxane. 1066600.
detection used and if it provides reproducible, steep log
Stationary phase for gas chromatography.
dose-response curves.
Contains 90 per cent of cyanopropylgroups and 10 per cent of
When required for use, thaw a portion of the plasma phenylmethyl groups.
substrate in a water-bath at 37 oC, gently swirling until
thawing is complete; once thawed it should be kept at Poly( cyanopropyl) (7) (phenyl) (7) (methyl) (86) siloxane.
10-20 oC and used without del ay. The thawed plasma 1109200.
substrate may be lightly centrifuged if necessary; filtration
procedures should not be used. Stationary phase for gas chromatography.
Polysiloxane substituted with 7 per cent of cyanopropyl
Plasma substrate R2. 1066202. groups, 7 per cent of phenyl groups and 86 per cent of
dimethyl groups.
Prepare from human blood containing less than 1 per
cent of the normal amount of factor IX. Collect the blood Poly( cyanopropylphenyI) (14) (methyl) (86)siloxane.
into one-ninth its volume of a 38 giL solution of sadium 1173700.
citrate R.
Stationary phase for chromatography.
Starage: in small amounts in plastic tubes at a temperature
of - 30 oC or lower. Contains 14 per cent of cyanopropylphenyl groups and 86 per
cent of methyl groups.
Plasma substrate R3. 1066203.
Poly( cyanopropyl) sHoxane. 1066700.
Prepare from human blood containing less than 1 per Polysiloxane substituted with 100 per cent of cyanopropyl
cent of the normal amount of factor XI. Collect the blood groups.
into one-ninth its volume of a 38 giL solution of sadium
citrate R. Poly( dimethyl) (diphenyl) (divinyl) siloxane. 1100000.
Starage: in small amounts in plastic tubes at a temperature Stationary phase for gas chromatography.
of - 30 oC or lower.
Contains 94 per cent of methyl groups, 5 per cent of phenyl
Plasma substrate deficient in factor V. 1066300. groups and 1 per cent of vinyl groups. SE54.

Use preferably a plasma which is congenitally deficient, or Poly( dimethyl) (diphenyl)siloxane. 1066900.
prepare it as follows: separate the plasma from human bIood Stationary phase for gas chromatography.
collected into one tenth of its volume of a 13.4 giL solution
of sodium oxalate R. Incubate at 37 oC for 24 h to 36 h. The Contains 95 per cent of methyl groups and 5 per cent of
coagulation time determined by the method prescribed for phenyl groups. DB-5, SE52.
coagulatíon factor V solution R should be 70 s to 100 s. lf the
Poly( dimethyl) (diphenyl) siloxane, base-deactivated.
coagulation time is les s than 70 s, incubate again for 12 h to
1176600.
24 h.
Base-deactivated stationary phase for gas chromatography
Storage: in small quantities at a temperature of - 20 oC or
specially designed for amine anaIysis.
lower.
Contains 95 per cent of methyl groups and 5 per cent of
Plasminogen, human. 1109100. [9001-91-6]. phenyl groups.
A substance present in blood that may be activated to plasmin, Poly( dimethyl) (75) (diphenyl) (25)siloxane. 1171500.
an enzyme that lyses fibrin in blood dots.
Stationary phase for chromatography.
Plutonium -242 spiking solution. 1167400. Contains 75 per cent of methyl groups and 25 per cent of
phenyl groups.
Contains 50 Bq/L 242pU and a 134 giL solution of lanthanum
chloride heptahydrate R in a 284 giL solution of nitric acid R. Foly( dimethyl) (85) (diphenyl) (15)siloxane. 1154700.
Poloxamer 188. 1186800. Stationary phase for chromatography.
See Poloxamers (1464). Contains 85 per cent of methyl groups and 15 per cent of
phenyl groups. PS086.
Poly[ (cyanopropyl) methylphenylmethylsiloxane].
Poly( dimethyl) siloxane. 1066800.
1066500.
Silicone gum rubber (methyl). Organosilicon polymer with
See poly[(cyanapropyl)(methyl)] [(phenyl)( methyl)]siloxane R. the appearance of a semi-liquid, colourless gum.
Poly[ (cyanopropyl) (methyl)] [(phenyl) (methyI)] siloxane. The intrinsic viscosity, determined as follows is about
1066500. 115 mLK 1 Weigh 1.5 g, 1 g and 0.3 g of the substance to
be examined to the nearest 0.1 mg, ¡nto 100 mL volumetric
Contains 25 per cent of cyanopropyl groups, 25 per cent of flasks. Add 40-50 mL of toluene R, shake until the substance
phenyl groups and 50 per cent of methyl groups. (Average is completely dissolved and dilute to 100.0 mL with the
relative molecular mass 8000). same solvento Determine the viscosity (2.2.9) of each
A very viscous liquid (viscosity about 9000 mPa·s). solution. Determine the viscosity of toluene R under the same
conditions. Reduce the concentration of each solution by half
d~~ : about 1.10.
by diluting with toluene R. Determine the viscosity of these
n~5: about 1.502. solutions.

General Natices (1) apply ta all monographs and other texts 503
4.1.1. Reagents EUROPEAN PHARMACOPOEIA 8.0

Polymethacrylate geL 1181100.


e concentration in grams per 100 mL, A methacrylate-based size-exclusion stationary phase for
flow time of the solution to be examined, water-soluble samples.

flow time of toluene, Polymethacrylate gel, hydroxylated. 1151300.


Stationary phase for size-exclusion chromatography.
111 viscosity of the solution to be examined in
millipascal seconds, Gel based on hydroxylated methacrylic acid polymer.
112 viscosity of toluene in millipascal seconds, Polymethylphenylsiloxane. 1067900.
d1 relative density of the solution to be examined, Stationary phase for gas chromatography.
Contains 50 per cent of methyl groups and 50 per cent of
d2 relative density of toluene. phenyl groups. (Average relative molecular mas s 4000.)
To obtain the relative densities use the following data. Very viscous liquid (viscosity about 1300 mPa·s).
Concentration (g/lOO mL) Relative density (dl )
d§g: about l.09.
n~5 : about l.540.
0-0.5 1.000
Poly[methyl(95)phenyl(5)] siloxane. 1068000.
0.5 - 1.25 1.001
See Poly(dimethyl)(diphenyl)siloxane R.
1.25 - 2.20 1.002
Poly[methyl(94)phenyl(5)vinyl(l)] siloxane. 1068100.
2.20 - 2.75 1.003
See Poly(dimethyl)(diphenyl)(divinyl)siloxane R.
2.75 - 3.20 1.004
Poly[ methyl( trifluoropropylmethyl)siloxane l. 1171600.
3.20 - 3.75 1.005 Stationary phase for gas chromatography.
3.75 - 4.50 1.006 Contains 50 per cent of trifluoropropylmethyl groups and
50 per cent of methyl groups.
The speciflc viscosity is obtained from the following equation:
Polyoxyethylated castor oH. 1068200.
Tlsp = TlI - Tl2 = tId l _ 1
Light yellow liquido It becomes clear aboye 26 oC.
Tl2 t 2 d2
and the reduced viscosity from: Polysorbate 20. 1068300. [9005-64-5].
Tlsp See Polysorbate 20 (0426).
Tlred = -
e Polysorbate 80. 1068400. [9005-65-6].
The intrinsic viscosity (Il) is obtained by extrapolating the See Polysorbate 80 (0428).
preceding equation to e = O. This is done by plotting the curve
Polystyrene 900-1000. 1112200. [9003-53-6].
Ils/c or log Ils/c as a function of c. Extrapolation to e = O gives
'l. The intrinsic viscosity is expressed in millilitres per gram; Organic standard used for calibration in gas chromatography.
the value obtained must therefore be multiplied by 100. M 1V : about 950.
The infrared absorption spectrum (2.2.24) obtained by M)M n: 1.10.
applying the substance, if necessary dispersed in a few drops
Potassium acetate. 1175900. [127-08-2].
of carbon tetrachloride R, to a sodium chloride plate, does not
show absorption at 3053 cm~ 1, corresponding to vinyl groups. See Potassium acetate (1139).
Loss on drying (2.2.32): maximum 2.0 per cent, determined Potassium bicarbonate. 1069900. [298-14-6].
on 1.000 g by drying in vacuo at 350 oC for 15 min; maximum See Potassium hydrogen carbonate R.
0.8 per cent, determined on 2.000 g by drying at 200 oC for 2 h.
Potassium bicarbonate solution, saturated methanolic.
Polyether hydroxylated gel for chromatography. 1067000. 1069901.
Gel with a small particle size having a hydrophilic surface See potassium hydrogen carbonate solution, saturated
with hydroxyl groups. It has an exclusion limit for dextran of methanolic R.
relative molecular mass 2 x lOs to 2.5 X 10 6.
PotassÍum bl'Omate. KBrOJ' (Mr 167.0). 1068700.
Polyethyleneglycol adipate. (C SH 120 4 )11' (Me (172.2)11)' [7758-01-2].
1067700. White or almost white granular powder or crystals, soluble in
White or almost white, wax-like mass, practically insoluble in water, slightly soluble in ethanol (96 per cent).
water. Potassium bl'Omide. 1068800. [7758-02-3].
mp: about 43 oc. See Potassium bromide (0184).
Polyethyleneglycol, base-deactivated. 1170300. Potassium bromide used for infrared absorption
spectrophotometry (2.2.24) also complies with the following
Stationary phase for gas chromatography.
additional test.
Cross-linked, base-deactivated polyethyleneglycol specially A disc 2 mm thick prepared from the substance previously
designed for amine analysis. dried at 250 oC for 1 h, has a substantially flat baseline over
the range 4000 cm- 1 to 620 cm~ 1. It exhibits no maxima
Polyethyleneglycol, polar-deactivated. 1179000.
with absorbance greater than 0.02 aboye the baseline, except
Stationary phase for gas chromatography. maxima for water at 3440 cm- 1 and 1630 cm-l.
Polyethyleneglycol succinaíe. (C6HsÜ4)¡¡' (Mr (144.1)¡¡). Potassium carbonate. K2 COy (Mr 138.2). 1068900.
1067800. [584-08-7]. Dipotassium carbonate.
White or almost white, crystalline powder, practically White or almost white, granular powder, hygroscopic, very
insoluble in water. soluble in water, practically insoluble in anhydrous ethanoL
mp: about 102 oc. Storage: in an airtight container.

504 See the informatíon section on general monographs (cover pages)


EUROPEAN PHARMACOPOEIA 8.0 4.1.1. Reagents

Potassium chlorate. KCIOy (Mr 122.6). 1069000. Potassium dihydrogen phosphate. 1069600. [7778-77-0].
[3811-04-9]. See Potassium dihydrogen phosphate (0920).
A white or almost white powder, granules or crystals, soluble
in water. Poíassium dihydrogen phosphate, 0.2 M. 1069601.
A solution of potassium dihydrogen phosphate R containing
Poíassium chloride. 1069100. [7447-40-7]. the equivalent of27.22 g ofKH 2 P0 4 in 1000.0 mL.
See Potassium chloride (0185).
Potassium ferricyanide. K3 [Fe(CN)6]' (Mr 329.3). 1069700.
Potassium chloride used for infrared absorption
[13746-66-2]. Potassium hexacyanoferrate(III).
speetrophotometry (2.2.24) a/so complies with the following
additiona/ test. Red crystals, freely soluble in water.
A disc 2 mm thick, prepared from the substance previously Potassium ferricyanide solution. 1069701.
dried at 250 oC for 1 h, has a substantially flat baseline over
Wash 5 g of potassium ferricyanide R with a little water R,
the range 4000 cm- 1 to 620 cm- 1. It exhibits no maxima
dissolve and dilute to 100 rnL with water R. Prepare
with absorbance greater than 0.02 aboye the baseline, except
immediately before use.
maxima for water at 3440 cm- 1 and 1630 cm-l.
Potassium ferriperiodate solution. 1070801.
Potassium chloride, 0.1 M. 1069101.
Dissolve 1 g of potassium periodate R in 5 mL of a freshly
A solution of potassium ch/oride R containing the equivalent
prepared 120 giL solution of potassium hydroxide R. Add
of 7.46 g of KCI in 1000.0 mL.
20 mL of water R and 1.5 mL of ferric eh/oride so/ution R1.
Poíassium chromate. K2Cr0 4 • (Mr 194.2). 1069200. Dilute to 50 mL with a freshly prepared 120 giL solution of
[7789-00-6]. Dipotassium chromate. potassium hydroxide R.
Yellow crystals, freely soluble in water. Potassiurn ferrocyanide. K4[Fe(CN)6],3Hp. (M, 422.4).
Potassium chromate solution. 1069201. 1069800. [14459-95-1]. Potassium hexacyanoferrate(II).
Transparent yellow crystals, freely soluble in water, practically
A 50 giL solution.
insoluble in ethanol (96 per cent).
Potassium cífrate. 1069300. [6100-05-6].
Potassium ferrocyanide solution. 1069801.
See Potassium citrate (0400).
A 53 giL solution.
Potassium cyanide. KCN. (Mr 65.1). 1069400. [151-50-8].
Potassium fluoride. KF. (M r 58.1). 1137800. [7789-23-3].
White or almost white, crystalline powder or white or almost
white mass or granules, freely soluble in water, slightly soluble Colourless crystals or white or almost white crystalline
in ethanol (96 per cent). powder, deliquescent, soluble in water, practically insoluble in
ethanol (96 per cent).
Potassium cyanide solution. 106940 l.
Potassium hydrogen carbonate. KHCO J . (M, 100.1).
A 100 giL solution. 1069900. [298-14-6]. Potassium bicarbonate.
Poíassium cyanide solution, lead-free. 1069402. Transparent, colourless crystals, freely soluble in water,
Dissolve 10 g of potassium cyanide R in 90 mL of water R, practically insoluble in ethanol (96 per cent).
add 2 mL of strong hydrogen peroxide so/ution R diluted 1 Potassium hydrogen carbonate solution, saturated
to 5. Allow to stand for 24 h, dilute to 100 mL with water R methanolic. 1069901.
and filter.
Dissolve 0.1 g of potassium hydrogen carbonate R in 0.4 mL
The solution complíes with the following test: take 10 mL of
of water R, heating on water-bath. Add 25 mL of methanol R
the solution, add 10 mL of water R and 10 mL of hydrogen and swirl, keeping the solution on the water-bath until
sulfide solution R. No colour is evolved even after addition dissolution is complete. Use a freshly prepared solution.
of 5 mL of di/u te hydroch/oric acid R.
Poiassium hydrogen phthalate. C8HsK04' (Mr 204.2).
Potassium dichromate. KzCrpr (Mr 294.2). 1069500.
1070000. [877-24-7]. Potassium hydrogen benzene-l,2-
[7778-50-9]. Dipotassium dichromate.
dicarboxylate.
Potassium dichromate used for the calibration of
White or almost white crystals, soluble in water, slightly
spectrophotometers (2.2.25) contains not less than 99.9 per
soluble in ethanol (96 per cent).
cent of K2 Cr2 0 7 , calculated with reference to the substance
dried at 130 oc. Potassium hydrogen phthalate, 0.2 M. 1070001.
Orange-red crystals, soluble in water, practically insoluble in A solution of potassium hydrogen phtha/ate R containing
ethanol (96 per cent). the equivalent of 40.84 g of CsHsK04 in 1000.0 mL.
Assay. Dissolve 1.000 g in water R and dilute to 250.0 mL with
the same solvent. To 50.0 mL of this solution add a freshly Potassium hydrogen sulfate. KHS0 4. (Mr 136.2). 1070100.
prepared solution of 4 g of potassium iodide R, 2 g of sodium [7646-93-7].
hydrogen carbonate R and 6 mL of hydrochloric acid R in Colourless, transparent, hygroscopic crystals, freely soluble in
100 mL of water R in a 500 mL flask. Stopper the flask and water giving a strongly acid solution.
allow to stand protected from light for 5 mino Titrate with Storage: in an airtight container.
0.1 M sodium thiosulfate, using 1 mL of iodide-free starch
solution R as indicator. Potassium hydrogen tartrate. C4HsK06' (Mr 188.2).
1 mL of 0.1 M sodium thiosulfate is equivalent to 4.903 mg 1070200. [868-14-4]. Potassium hydrogen (2R,3R)-2,3-
of K 2Cr 20 r dihydroxybutane-1,4-dioate.
White or almost white, crystalline powder or colourless,
Potassium dkhromate solution. 106950l. slightly opaque crystals, slightly soluble in water, soluble in
A 106 giL solution. boiling water, practically insoluble in ethanol (96 per cent).
Potassium dichromate solution Rl. 1069502. Poíassium hydroxide. 1070300. [1310-58-3].
A 5 giL solution. See Potassium hydroxide (0840).

Genera/ Notices (1) app/y to all monographs and other texts 505
4.1.1. Reagents EUROPEAN PHARMACOPOEIA 8.0

Potassium hydroxide, akoholic, 2 M. 1070301. Storage: protected from light.


Dissolve 12 g of potassium hydroxide R in 10 mL of water R Potassium iodobismuthate solution R2. 1070602.
and diIute to 100 mL with ethanol (96 per cent) R.
Stock solution. Suspend 1.7 g of bismuth subnitrate R and 20 g
Potassium hydroxide in alcohol (10 per cent V/V), of tartaric acid R in 40 mL of water R. To the suspension add
0.5 M. 1070302. 40 mL of a 400 giL solution of potassium iodide R and stir fOI
Dissolve 28 g of potassium hydroxide R in 100 mL of 1 h. Filter. The solution may be kept for several days in brown
ethanol (96 per cent) R and diIute to 1000 mL with water R. bottles.
Spray solution. Mix immediately before use 5 mL of the stock
Potassium hydroxide solution, alcoholic. 1070303. solution with 15 mL of water R.
Dissolve 3 g of potassium hydroxide R in 5 mL of water R
and dilute to 100 mL with aldehyde-free alcohol R. Decant Potassium iodobismuthate solution R3. 1070604.
the clear solution. The solution should be almost colourless. Dissolve 0.17 g of bismuth subnitrate R in a mixture of 2 mL
of glacial acetic acid R and 18 mL of water R. Add 4 g of
Potassium hydroxide solution, akoholic Rl. 1070304. potassium iodide R, 1 g of iodine R and dilute to 100 mL with
Dissolve 6.6 g of potassium hydroxide R in 50 mL of water R dilute sulfuric acid R.
and diIute to 1000 mL with anhydrous ethanol R.
Poíassium iodobismuthate solution R4. 1070605.
Potassium iodate. KIOy (Mr 214.0). 1070400. [7758-05-6]. Dissolve 1.7 g of bismuth subnitrate R in 20 mL of glacial
White or aImost white, crystalline powder, soluble in water. acetic acid R. Add 80 mL of distilled water R, 100 mL of a
400 giL solution of potassium iodide R, 200 mL of glacial
Poíassium iodide. 1070500. [7681-11-0]. acetic acid R and dilute to 1000 mL with distilled water R. Mix
See Potassium iodide (0186). 2 volumes of this solution with 1 volume of a 200 giL solution
of barium chloride R.
Potassi.um iodide and starch solution. 1070501.
Dissolve 0.75 g of potassium iodide R in 100 mL of water R. Potassium iodobismuthate solution R5. 1070606.
Heat to boiling and add whilst stirring a solution of 0.5 g To 0.85 g of bismuth subnitrate R add 10 mL of glacial acetic
of soluble starch R in 35 mL of water R. Boil for 2 min and acid R and gently heat until completely dissolved. Add 40 mL
allow to cool. of water R and allow to cool. To 5 mL of this solution, add
Test for sensítivity. A mixture of 15 mL of the potassium 5 mL of a 400 giL solution of potassium iodide R, 20 mL of
iodide and starch solution, 0.05 mL of glacial acetic acid R glacial acetic acid R and 70 mL of water R.
and 0.3 mL of iodine solution R2 is blue.
Potassium nitrate. KN03' (MI 101.1).1070700. [7757-79-1].
Potassium iodide solution. 1070502. Colourless crystals, very soluble in water.
A 166 giL solution.
Potassium periodate. KI0 4• (Mr 230.0). 1070800.
Poíassium iodide solution, iodinated. 1070503. [7790-21-8].
Dissolve 2 g of iodine R and 4 g of potassium iodide R in White or almost white, crystalline powder or colourless
10 mL of water R. When solution is complete dilute to crystals, soluble in water.
100 mL with water R.
Potassium permanganate. 1070900. [7722-64-7].
Potassium iodide solution, iodinaíed Rl. 1070505. See Potassium permanganate (0121).
Dissolve 500 mg of iodine R and 1.5 g of potassium iodide R
in water R and d¡lute to 25 mL with the same solvento Potassium permanganate and pbosphoric add solution.
1070901.
Potassium iodide solution, saturated. 1070504. Dissolve 3 g of potassium permanganate R in a mixture of
A saturated solution of potassium iodide R in carbon 15 mL of phosphoric acid R and 70 mL of water R. Dilute to
dioxide-free water R. Make sure the solution remains 100 mL with water R.
saturated as indicated by the presence of undissolved
crystals. Potassium permanganate solution. 1070902.
Test by adding to 0.5 mL of the saturated potassium iodide A 30 giL solution.
solution 30 mL of a mixture of 2 volumes of chloroform R Potassium perrhenate. KRe0 4 . (Mr 289.3). 1071000.
and 3 volumes of glacial acetic acid R, as well as 0.1 mL [10466-65-6].
of starch solution R. Any blue colour formed should be
White or almost white, crystalline powder, soluble in water,
discharged by the addition of 0.05 mL of 0.1 M sodium
slightly soluble in ethanol (96 per cent), in methanol and in
thiosulfate.
propylene glycol.
Storage: protected from light.
Potassium persulfate. K2S20S' (Mr 270.3). 1071100.
Potassium iodobismuthate solution. 1070600. [7727 -21-1]. Dipotassium peroxodisulfate.
To 0.85 g of bismuth subnitrate R add 40 mL of water R, 10 mL Colourless crystals or white or almost white, crystalline
of glacial acetic acid R and 20 mL of a 400 giL solution of powder, sparingly soluble in water, practically insoluble in
potassium iodide R. ethanol (96 per cent). Aqueous solutions decompose at room
Potassium iodobismuthate solution, dilute. 1070603. temperature and more rapidly 011 warming.
Dissolve 100 g of tartaric acid R in 500 mL of water R and Poíassium plumbite solution. 1071200.
add 50 mL of potassium iodobismuthate solution Rl. Dissolve 1.7 g of lead acetate R, 3.4 g of potassium citrate R
Storage: protected from light. and 50 g of potassium hydroxide R in water R and dilute to
100 mL with the same solvent.
Potassium iodobismuthate solution Rl. 1070601.
Dissolve 100 g of tartaric acid R in 400 mL of water R and add Potassium pyroantimonate. KSb(OH)6' (Mr 262.9). 1071300.
8.5 g of bismuth subnitrate R. Shake for 1 h, add 200 mL of a [12208-13-8]. Potassium hexahydroxoantimoniate.
400 giL solution of potassium iodide R and shake well. Allow White or almost white, crystals or crystalline powder,
to stand for 24 h and filter. sparingly soluble in water.

506 See the informatíon section on general monographs (ca ver pages)
EUROPEAN PHARMACOPOEIA 8.0 4.1.1. Reagents

Potassium pyroantimonate solution. 1071301. 2-Propanol Rl. 1072101.


Dissolve 2 g of potassium pyroantimonate R in 95 mL ofhot Complies with the requirements prescribed for 2-propanol R
water R. Cool quickly and add a solution containing 2.5 g with the following additional requirements.
of potassium hydroxide R in 50mL of water R and 1 mL of n~o: about 1.378.
dilute sodium hydroxide solution R. Allow to stand for 24 h, Water (2.5.12): maximum 0.05 per cent, determined on
filter and dilute to 150 mL with water R. 10 g.
Potassium tartrate. C4H4KP6,1/2Hp. (M, 235.3). 1071400. Mínimum transmíttance (2.2.25) using water R as
[921-53-9]. Dipotassium (2R,3R)-2,3-dihydroxybutane-lA- compensation liquid: 25 per cent at 210 nm, 55 per cent
dio ate hemihydrate. at 220 nm, 75 per cent at 230 nm, 95 per cent at 250 nm,
98 per cent at 260 nm.
White or almost white, granular powder or crystals, very
soluble in water, very slightly soluble in ethanol (96 per cent). 2-Propanol R2. 1184900. [67-63-0].
Potassium tetraiodomercurate solution. 1071500. See Isopropyl alcohol (0970).
Dissolve 1.35 g of mercuríc chloride R in 50 mL of water R. Add Propetamphos. C lO H 20N0 4 PS. (Mr 281.3). 1130900.
5 g of potassium iodide R and dilute to 100 mL with water R. [31218-83-4].
A suitable certified reference solution (lO ng/flL in
Potassium tetraiodomercurate solution, alkaline. 1071600. cyclohexane) may be used.
Dissolve 11 g of potassium iodide R and 15 g of mercuric
iodide R in water R and dilute to 100 mL with the same solvent. Propidiurn iodide. C27H3412N4' (M, 668.4). 1154200.
Immediately before use, mix 1 volume of this solution with an [25535-16-4]. 3,8-Diamino-S- [3( diethylmethylammonio)-
equal volume of a 250 giL solution of sodium hydroxide R. propyl]-6-phenylphenanthridinium diiodide.
Dark red solido
Potassium tetroxalate. C4H3KOs,2H20. (Mr 254.2). 1071700.
[6100-20-5]. Propionaldehyde. C3Hp. (M, 58.1). 1072300. [123-38-6].
Propanal.
White or almost white, crystalline powder, sparingly soluble
in water, soluble in boiling water, slightly soluble in ethanol Liquid freely soluble in water, miscible with ethanol (96 per
(96 per cent). cent).
d~g: about 0.81.
Potassiurn thiocyanate. KSCN. (M, 97.2). 1071800. n~o : about 1.365.
[333-20-0].
bp: about 49 oc.
Colourless crystals, deliquescent, very soluble in water and in
mp: about - 81°C.
ethanol (96 per cent).
Storage: in an airtight container. Propionic add. C3H602' (Mr 74.1). 1072400. [79-09-4].
Oily liquid, soluble in ethanol (96 per cent), miscible with
Potassium thiocyanate solution. 1071801. water.
A 97 giL solution. d~g: about 0.993.
Povidone. 1068500. [9003-39-8]. n~o: about 1.387.

See Povidone (0685). bp: about 141°C.


mp: about - 21°C.
Procaine hydrochloride. 1109400.
Propionic anhydride. C6H1003' (Mr 130.1). 1072500.
See Procaine hydrochloride (0050). [123-62-6].
Proline. 1152200. [147-85-3]. CIear, colourless liquid, soluble in ethanol (96 per cent).
See Proline (0785). d~g: about 1.01.
bp: about 167 oc.
Propane-l,3-diol. C 3HP2' (Mr 76.1). 1185100. [504-63-2].
1,3-Dihydroxypropane. Propionic anhydride reagent. 1072501.
Colourless, viscous liquido Dissolve 1 g of toluenesulfonic acid R in 30 mL of glacial
acetic acid R, add 5 mL of propionic anhydride R and allow
bp: about 214 oc.
to stand for at least 15 min before use.
mp: about - 27 oc.
Storage: use within 24 h.
Propano!. C 3H sO. (Mr 60.1). 1072000. [71-23-8]. Propyl acetate. CSH1002' (M, 102.1). 1072600. [109-60-4].
Propan-l-ol.
d~g: about 0.888.
Clear colourless liquid, miscible with water and with ethanol bp: abont 102 oc.
(96 per cent).
mp: about - 95 oc.
d~g : about 0.802 to 0.806.
bp: about 97.2 oc. Propyl parahydroxybenzoate. 1072700. [94-13-3].
Distillation range (2.2.11). Not less than 95 per cent distils See Propyl parahydroxybenzoate (0431).
between 96 oC and 99 oc. D- Prolyl-L-phenylalanyl-L-arginine4- nitroanilide
dihydrochloride. C26H36C12NsOs' (M, 612). 1072800.
Propanol Rl. 1184400. [71-23-8].
See Propanol (2036). Propylene glycol. 1072900. [57-55-6].
See Propylene glycol (0430).
2-Propanol. C 3Hp. (Mr 60.1). 1072100. [67-63-0].
Propan-2-ol. Isopropyl alcohol. Propylene oxide. C 3Hp. (M, 58.1). 1121800. [75-56-9].
CIear, colourless, flammable liquid, miscible with water and Colourless liquid, miscible with ethanol (96 per cent).
with ethanol (96 per cent). Protamine sulfate. 1073000. [53597-25-4 (salmine)
d~g: about 0.785. 9007-31-2 (clupeine)].
bp: 81°C to 83 oc. See Protamíne sulfate (0569).

General Notices (1) apply to all monographs and other texts 507
4.1.1. Reagent§ EUROPEAN PHARMACOPOEIA 8.0

Protopine hydrochloride. C2oH2oCINOs' (Mr 389.8). Pyridylazonaphthol solution. 1073501.


1163500. [6164-47-2]. A 1 giL solution in anhydrous ethanol R.
5-Methyl-4,6, 7,14-tetrahydrobis [1 ,3]benzodioxolo [4,5-c:5',6'- Test for sensitivity. To 50 mL of water R add 10 mL of
g]azecin-13(5H)-one hydrochloride. acetate buffer solution pH 4.4 R, 0.10 mL of 0.02 M sodium
edetate and 0.25 mL of the pyridylazonaphthol solution.
Pteroic acid. C14H12N60y (Mr 312.3). 1144600.
[119-24-4]. 4-[ [(2-Amino-4-oxo-1,4-dihydropteridin-6- After addition of 0.15 mL of a 5 giL solution of copper
yl)methyl]amino]benzoic acid. sulfate R, the colour changes from light yellow to violet.
Crystals, soluble in solutions of alkali hydroxides. 4- (2-Pyridylazo )resorcinol monosodium salto
C ll H sN 3Na0 2, Hp. (Mr 255.2).1131500. [16593-81-0].
Puerarin. C21 H 2aÜ9' (Mr 416.4). 1180600. [3681-99-0].
7,4' -Dihydroxy-8-C-glucosyliso-haloprone. 8-~-D­ Orange crystalline powder.
Glucopyranosyl-7 -hydroxy-3-( 4-hydroxyphenyl)-4H-l- PyrocatechoL C6H602' (Mr 110.1). 1073600. [120-80-9].
benzopyran-4-one. Benzene-l,2-dioL
Pulegone. ClOH160. (M, 152.2). 1073100. [89-82-7]. Colourless or slightly yellow crystals, soluble in water, in
(R) - 2-Isopropylidene-5-methylcyclohexanone. acetone and in ethanol (96 per cent).
(+ )-p-Menth -4-en-3-one. mp: about 102 oc.
Oily, colourless liquid, practically insoluble in water, miscible Storage: protected from light.
with ethanol (96 per cent).
Pyrogallol. C6H603' (Mr 126.1). 1073700. [87-66-1].
dig: about 0.936. Benzene-l ,2,3-triol.
n~o: 1.485 to 1.489. White or almost white crystals, becoming brownish on
bp: 222 oC to 224 oc. exposure to air and light, very soluble in water and in ethanol
Pulegone used in gas chromatography complies with the (96 per cent), slightly soluble in carbon disulfide. On exposure
fol/owing additional test. to air, aqueous solutions, and more rapidly alkaline solutions,
Assay. Gas chromatography (2.2.28) as prescribed in the become brown owing to the absorption of oxygen.
monograph Peppermint oil (0405). mp: about 131°C.
Test solution. The substance to be examined. Storage: protected from light.
Content: minimum 98.0 per cent, calculated by the Pyrogallol solution, alkaline. 1073701.
normalisation procedure.
Dissolve 0.5 g of pyrogallol R in 2 mL of carbon dioxide-free
Putrescine. C4H¡2N2' (Mr 88.15). 1137900. [110-60-1]. water R. Dissolve 12 g of potassium hydroxide R in 8 mL
1,4-Butanediamine. Tetramethylenediamine. of carbon dioxide-free water R. Mix the two solutions
Colourless oily liquid, very soluble in water. Strong immediately before use.
piperidine-Iike odour. Pyrrolidine. C,¡H 9 N. (M, 71.1). 1165000. [123-75-1].
bp: about 159 oc. Content: minimum 99 per cent.
mp: about 23 oc. bp: 87 oC to 88 oc.
Pyrazine-2-carbonitrile. CsHl·J3. (Mr 105.1). 1183300. 2-Pyrrolidone. C4H 7 NO. (Mr 85.1). 1138000. [616-45-5].
[19847-12-2].2-Cyanopyrazine. Pyrrolidin-2-one.
CIear, pale yellow liquido
Liquid aboye 25 oC, miscible with water, with anhydrous
Content: minimum 99 per cent. ethanol and with ethyl acetate.
liliiii
d 245 : 1.116.
Pyridin-2-amine. C SH 6N 2. (Mr 94.1). 1073400. [504-29-0].
Water (2.5.12): maximum 0.2 per cent determined on 2.00 g.
2-Aminopyridine. .
Assay. Gas chromatography (2.2.28) : use the normalisation
Large crystals soluble in water and in ethanol (96 per cent).
procedure.
bp: about 210 oc.
Test solution. Dissolve 1.0 g in methanol R and dilute to
mp: about 58 oc. 10.0 mL with the same solvent.
Pyridine. CsHsN. (Mr 79.1). 1073200. [110-86-1]. Column:
Clear, colourless liquid, hygroscopic, miscible with water and - material: glass;
with ethanol (96 per cent). - size: 1 = 30 m; 0 = 0.53 mm;
bp: about 115 oc. - stationary phase: macrogol20 000 R (1.0 flm).
Storage: in an airtight container. Carrier gas: helium for chromatography R.
Pyridine, anhydrous. 1073300. Flow rate: adjusted so that the retention time of 2-pyrrolidone
is about 10 mino
Dry pyridine R over anhydrous sodium carbonate R. Filter
and distil. Split ratio: 1:20.
Water (2.5.12) : maximum 0.01 per cent mlm. Temperature:
Time Temperatnre
Pyridinium hydrobromide perbromide. CSH6Br3N. (min) (oC)
(Mr 319.8). 1166100. [39416-48-3]. Pyridinium
Colnmn 0-1 80
tribromide(l-).
Red crystals. 1 - 12 80 .¿ 190

12 - 32 190
Pyridylazonaphthol. C1sHllN30. (M, 249.3). 1073500.
[85-85-8]. 1-(2-Pyridylazo )-2-naphthoL Injection por! 200
Brick-red powder, practically insoluble in water, soluble in
ethanol (96 per cent), in methanol and in hot dilute alkali Detection: flame ionisation.
solutions. Injection: 1 flL of the test soJution.
mp: about 138 oc. Content: minimum 98.0 per cent.

508 See the information section 011 general monographs (ca ver pages)
EUROPEAN PHARMACOPOEIA 8.0 4.1. L Reagent§

Pyruvic acid. C J HP3' (Mr 88.1). 1109300. [127-17-3]. [a]~O: about + 260, determined on a 10 giL solution in
2-0xopropanoic acid. anhydrous ethanol R.
Yellawish liquid, miscible with water and with anhydrous mp: about 172 oc.
ethanol. Storage: protected from light.
d~g: about 1.267.
Quinidine sulfate. 1109500. [6591-63-5].
n~o: about 1.413.
See Quinidine sulfate (0017).
bp: about 165 oc.
Quinine. C2o H 24 NP2' (Mr 324.4). 1074100. [130-95-0].
Quercetin diliydrate. C1sHIO07,2Hp. (Mr 338.2). 1138100.
(R) - (6-Methoxyquinol-4-yl) [(2S,4S,5R) - 5-vinylquinuclidin -2-
2-(3,4-Dihydroxyphenyl)-3,5, 7-trihydroxy-4H-l-benzopyran-
yl]methanol.
4-ane.
White or almost white, microcrystalline powder, very slightly
Yellow crystals or yellawish powder, practically insoluble in
soluble in water, slightly soluble in boiling water, very soluble
water, soluble in acetone and in methanol.
in anhydrous ethanol.
Water (2.5.12): maximum 12.0 per cent, determined on
[a]~o: about - 167, determined on a 10 giL solution in
0.100 g.
anhydrous ethanol R.
Assay. Liquid chromatography (2.2.29) as prescribed in the
mp: about 175 oc.
monograph Ginkgo leaf (1828).
Content:minimum 90 per cent (anhydrous substance) Storage: protected from light.
calculated by the normalisation procedure. Quinine hydrochloride. 1074200. [6119-47-7].
Storage: protected from light. See Quinine hydrochloride (0018).
Quercitrin. C21H20011' (Mr 448.4). 1138200. Quinine sulfate. 1074300. [6119-70-6].
[522-12-3]. Quercetin 3-L-rhamnopyranoside.
3- [( 6-Deoxy-a-L-mannopyranosyl)oxy]-2-(3,4- See Quinine sulfate (0019).
dihydroxyphenyl) -5, 7-dihydroxy-4H-l-benzopyran -4-one. Rabbit erythrocyte suspension. 1074500.
Quercitroside.
Prepare a 1.6 per cent V/V suspension of rabbit erythrocytes
Yellow crystals, practically insoluble in cold water, soluble in as follows: defibrinate 15 mL of freshly drawn rabbit blood by
ethanol (96 per cent). shaking with glass beads, centrifuge at 2000 g for 10 min and
mp: 176 oC to 179 oc. wash the erythrocytes with three quantities, each of 30 mL,
Chromatography. Thin-layer chromatography (2.2.27) as of a 9 giL solution of sodium chloride R. Dilute 1.6 mL of
prescribed in the monograph Goldenrod (1892): apply 20 flL the suspension of erythrocytes to 100 mL with a mixture of
of the solution; after spraying, the chromatogram shows a 1 volume of phosphate buffer solution pH 7.2 R and 9 volumes
yellowish-brown fluorescent zone with an Rp of about 0.6. of a 9 giL solution of sodium chloride R.
Storage: at a temperature of 2 oC to 8 oc. Radopride tartrate. C,9H26CI2NP9' (M¡ 497.3). 1144700.
Quillai.a saponins, purified. 1184500. [98185-20-7]. Radopride L-tartrate.
A mixture of related saponins obtained from the barle of White 01' almost white solid, sensitive to light, soluble in water.
Quillaja saponaria Molina 5.1. [a]g: + 0.3, determined on a 3 giL solution.
Chromatography. Thin-Iayer chromatography (2.2.27) as mp: about 141 oc.
prescribed in the monograph Quillaia bark (1843): apply 5 flL
of the solution; after treating with a 10 per cent V/V solution Rapeseed oH. 1074600.
of sulfuric acid R in methanol R, heat at 120 oC for 5 min and See Rapeseed oil, refined (1369).
examine in daylight; the chromatogram shows 3 principal
zones in the upper part of the middle third. Redudng mixture. 1074700.
Grind the substances added in the following arder to obtain a
Quinaldinered. C 21 H 23 IN 2' (Mr 430.3). 1073800. [117-92-0]. homogeneous mixture: 20 mg of potassium bromide R, 0.5 g
2- [2- [4-(Dimethylamino )phenyl] ethenyl]-l-ethylquinolinium of hydrazine sulfate R and 5 g of sodium chloride R.
iodide.
Dark bluish-black powder, sparingly soluble in water, freely Reichstein's substance S. C21 H 3aÜ4' (M r 346.5). 1175400.
soluble in ethanol (96 per cent). [152-58-9].
Content: minimum 95.0 per cent.
Quinaldine red solution. 1073801. mp: about 208 oc.
Dissolve 0.1 g of quinaldine red R in methanol R and dilute
to 100 mL with the same solvento Resin for reversed-phase ion chromatography. 1131100.
Colour change: pH 1.4 (colourless) to pH 3.2 (red). A neutral, macroporous, high specific surface are a with a
non-polar character resin consisting of polymer lattice of
Quinhydrone. C ,2 H¡o04' (Mr 218.2). 1073900. [106-34-3]. polystyrene cross-linked with divinylbenzene.
Equimolecular compound of l,4-benzoquinone and
hydroquinone. Resin, weak cationic. 1096000.
Dark green, lustrous crystals ar a crystalline powder, slightly See weak cationic resin R.
soluble in water, sparingly soluble in hot water, soluble in
ethanol (96 per cent) and in concentrated ammonia. Resorcinol. 1074800. [108-46-3].
mp: about 170 oc. See Resorcinol (0290).

Quinidine. C 2o H 24NP2' (Mr 324.4). 1074000. [56-54-2]. Resorcinol reagent. 1074801.


(S) -( 6-MethoxyquinoJ -4-yI) [(2R,4S,5R) - 5-vinylquinuclidin -2- To 80 mL of hydrochloric acid R1 add 10 mL of a 20 giL
yl]methanol. solution of resorcinol R and 0.25 mL of a 25 giL solution
White or almost white crystals, very slightly soluble in water, of copper sulfate R and dilute to 100.0 mL with water R.
sparingly soluble in ethanol (96 per cent), slightly soluble in Prepare the solution at ¡east 4 h before use.
methanol. Storage: at 2 oC to 8 oC for 1 week.

General Notices (1) apply to all monographs and other texts 509
4.1.1. Reagents EUROPEAN PHARMACOPOEIA 8.0

ResveratroL C¡4H1203' (M, 228.2). 1186900. Sabinene. C lO H)6" (Mr 136.2). 1109700. [3387-41-5]. Thuj-
[501-36-0]. 3,4',5-Stilbenetriol. 5- [(E)-2-( 4-Hydroxy- 4( 10) -ene. 4-Methylene-1-isopropylbicyclo [3.1.0 ]hexane.
phenyl)ethenyl]benzene-l,3-diol. A colourless, oily liquido
Rhamnose. C6H120S,Hp. (M, 182.2).1074900. [6155-35-7]. Sabinene used in gas chromatography complies with the
L-(+)-Rhamnose.6-Deoxy-L-mannose. following additional test.
White or almost white, crystalline powder, freely soluble in Assay. Gas chromatography (2.2.28) as prescribed in the
water. monograph Bitter-orange-flower oil (1175).
[al~o: + 7.8 to + 8.3, determined on a 50 giL solution in Test solution. The substance to be examined.
water R containing about 0.05 per cent of NH y Content: minimum 95.0 per cent, ca!culated by the
normalisation procedure.
Rhaponticin. C21 H 2P9' (M, 420.4). 1075000. [155-58-8].
3-Hydroxy-5- [2-( 3-hydroxy -4-methoxyphenyl) ethenyl] phenyl Saccharin sodium. 1131400. [128-44-9].
~-D-glucopyranoside.
See Saccharin sodium (0787).
Yellowish-grey, crystalline powder, soluble in ethanol (96 per
cent) and in methanol. Safrole. C lO H lO 02' (Mr 162.2). 1131200. [94-59-7].
Chromatography. Thin-layer chromatography (2.2.27) 5- (Prop- 2-enyl) -1,3- benzodioxole. 4-Allyl-1,2-
as prescribed in the monograph Rhubarb (0291); the (methylenedioxy)benzene.
chromatogram shows only one principal spot. Colourless OI slightly yellow, oily liquid, with the odour of
sassafras, insoluble in water, very soluble in ethanol (96 per
Rhodamine 6 G. C2s H 3¡ClNpy (Mr 479.0). 1153300. cent), miscible with hexane.
[989-38-8].
d~g: 1.095 to 1.096.
Colour Index No. 45160.
n~o: 1.537 to 1.538.
9- [2- (Ethoxycarbonyl)phenyl]-3,6-bis( ethylamino) -2, 7-
dimethylxanthenylium chloride. bp: 232 oC to 234 oc.
Brownish-red powder. Freezing point: about 11 oc.
Safrole used in gas chromatography complíes with the following
Rhodamine B. C2s H 3¡CINP3' (M, 479.0). 1075100. [81-88-9].
additional test.
Schultz No. 864.
Assay. Gas chromatography (2.2.28) as prescribed in the
Colour Index No. 45170. monograph Cinnamon bark oil, Ceylon (1501).
[9- (2-Carboxyphen -yl) -6- (diethylamino )-3H- xanthen -3-
Content: minimum 96.0 per cent, ca!culated by the
ylidene] diethylammonium chloride.
normalisation procedure.
Green crystals or reddish-violet powder, very soluble in water
and in ethanol (96 per cent). Salidn. C 13H¡sOr (M, 286.3). 1131300. [138-52-3].
2-(Hydroxymethyl)phenyl-~-D-glucopyranoside. Salicoside.
Ribose. CSHlOOS' (Mr 150.1). 1109600. [50-69-1]. D-Ribose.
[al~o: - 62.5 ± 2.
Soluble in water, slightly soluble in ethanol (96 per cent).
mp: 88 oC to 92 oc. mp: 199 oC to 201°C.
Assay. Liquid chromatography (2.2.29) as prescribed in the
Rkinolek aeid. C¡SH3PY (Mr 298.5). 1100100. [141-22-0]. monograph Willow bark (1583) at the concentration ofthe
(9Z,12R)-12-Hydroxyoctadec-9-enoic acid. 12-Hydroxyoleic reference solution.
acid. Content: mínimum 99.0 per cent, ca!culated by the
Yellow or yellowish-brown viscous liquid, consisting of a normalisation procedure.
mixture of fatty acids obtained by the hydrolysis of castor
oil, practically insoluble in water, very soluble in anhydrous Salicylaldehyde. C7H602' (M, 122.1). 1075400. [90-02-8].
ethanol. 2-Hydroxybenzaldehyde.
d~g : about 0.942. Clear, colourless, oily liquido
n~o: about 1.472. d~g: about 1.167.
mp: about 285 oC, with decomposition. n~o: about 1.574.

Rosmarinic aeid. C¡SH¡Ps' (M, 360.3). 1138300. bp: about 196 oc.
[20283-92-5]. mp: about - 7 oc.
mp: 170 oC to 174 oc. Salicylaldehyde aúne. C 14H¡ZN2 0 2. (M, 240.3). 1075500.
Ruthenium red. [(NH3)sRuORu(NH3)40Ru(NH3)s]CI6,4Hp. [959-36-4]. 2,2' -Azinodimethyldiphenol.
(M, 858). 1075200. [11103-72-3]. Dissolve 0.30 g of hydrazine sulfate R in 5 mL of water R, add
Brownish-red powder, soluble in water. 1 mL of glacial acetic acid R and 2 mL of a freshly prepared
20 per cent V/V solution of salicylaldehyde R in 2-propanol R.
Ruthenium red solution. 107520l. Mix, allow to stand until a yellow precipate is formed. Shake
A 0.8 giL solution in lead acetate solution R. with two quantities, each of 15 mL, of methylene chloride R.
Combine the organic layers and dry over anhydrous sodium
Rutin. C 27 H 3Pl6'3Hp. (M, 665). 1075300. [153-18-4]. sulfate R. Decant or filter the solution and evaporate to
Rutoside. 3-(0-6-Deoxy-a-L-mannopyranosyl-( 1-76)-~- D- dryness. Recrystallise from a mixture of 40 volumes of
glucopyranosyloxy)-2-( 3,4-dihydroxyphenyl) -5, 7 -dihydroxy- methanol R and 60 volumes of toluene R with cooling. Dry the
4H-chromen-4-one. crystals in vacuo.
Yellow, crystalline powder, darkening in light, very slightly mp: about 213 oc.
soluble in water, soluble in about 400 parts of boiling water,
slightly soluble in ethanol (96 per cent), soluble in solutions of Chromatography. Thin-layer chromatography (2.2.27)
the alkali hydroxides and in ammonia. as prescribed in the test for hydrazine in the monograph
Povidone (0685); the chromatogram shows only one principal
mp: about 21 O oC, with decomposition.
spot.
Absorbance (2.2.25). A solution in ethanol (96 per cent) R
shows two absorption maxima at 259 nm and 362 nm. Salicylic aeid. 1075600. [69-72-7].
Storage: protected from light. See Salicylic acid (0366).

510 See the information section on general monographs (cover pages)


EUROPEAN PHARMACOPOEIA 8.0 4.1.1. Reagents

Salvianolic add B. C36 H 3P16' (Mr 719). 1184600. SDS-PAGE sample buffer for reducing conditions
[121521-90-2J. (2R)c2- [[ (2E)-3- [(2S,3S)-3- [[ (lR)-l- (concentrated). 1122100.
Carboxy-2- (3,4-dihydroxyphenyl)ethoxyJcarbonyl]-2-( 3,4- Dissolve 3.78 g of tris(hydroxymethyl)aminomethane R, 10.0 g
dihydroxyphenyl)-7 -hydroxy- 2,3-dihydrobenzofuran -4- of sodium dodeeyl sulfate R and 100 mg of bromophenol blue R
yl]prop-2-enoyIJoxy]-3-(3,4-dihydroxyphenyl)propanoic add. in water R. Add 50.0 mL of glyeerol R and dilute to 200 mL
Sand. 1075800. with water R. Add 25.0 mL of 2-mercaptoethanol R. Adjust
to pH 6.8 with hydrochloric acid R, and dilute to 250.0 mL
White or slightly greyish grains of silica with a particle size with water R.
between 150 11m and 300 ~lm.
Alternatively, dithiothreitol may be used as reducing
Sarafloxacin hydrochloride. C2oHlsCIF2NP3' (Mr 421.8). agent instead of 2-mercaptoethanol. In this case
1181400. [91296-87 -6J. 6-Fluoro-l-( 4-fluorophenyl)-4-oxo- prepare the sample buffer as follows: dissolve 3.78 g of
7-piperazin -1-yl-1 ,4-dihydroquinoline-3-carboxylic acid tris(hydroxymethyl)aminomethane R, 10.0 g of sodium dodecyl
hydrochloride. sulfate R and 100 mg of bromophenol blue R in water R. Add
50.0 mL of glycerol R and dilute to 200 mL with water R. Adjust
Schisandrin. C24 H 3zÜ7' (Mr 432.5). 1173800. to pH 6.8 with hydrochloric acid R, and dilute to 250.0 mL
[7432-28-2]. Schisandrol A. Wuweizichun A. with water R. Immediately before use, add dithiothreitol R to a
(6S,7S,12aR a )-5,6,7,8- Tetrahydro-l,2,3,10,11,12- final concentration of 100 mM.
hexamethoxy-6, 7-dimethyldibenzo [a,c] cyclooctan -6-01.
White or almost white, crystalline powder. Selenious add. H 2Se0 3. (M r 129.0). 1100200. [7783-00-8].
Schisandrin used in the assay in the monograph Schisandra Deliquescent crystals, freely soluble in water.
fruit (2428) complíes with the following additional test. Storage: in an airtight container.
Assay. Liquid chromatography (2.2.29) as prescribed in the
monograph Schisandra fruit (2428). Selenium. Se. (A r 79.0). 1075900. [7782-49-2J.
Content: minimum 95 per cent, calculated by the Brown-red or black powder or granules, practically insoluble
normalisation procedure. in water and in ethanol (96 per cent), soluble in nitric acid.
Storage: in an airtight container, at - 20 oC or below. mp: about 220 oc.

y-Schisandrin. C23 H 2sÜ6' (Mr 400.5). 1173900. Serine. 1076000. [56-45-1].


[61281-37-6J. Schisandrin B. Wuweizisu B. rac- See Serine (0788).
(6R,7 S,13aR a )-1,2,3, 13-Tetramethoxy-6,7 -dimethyl-5,6,7,8-
tetrahydrobenzo [3,4] cycloocta[ 1,2-J] [1,3 Jbenzodioxole. Sialic add. 1001100. [131-48-6].
White or almost white, crystalline powder. See N-acetylneuraminic aeid R.
Storage: in an airtight container, at - 20 oC or below.
Silibinin. C2S H 2zÜlO' (M, 482.4). 1151400. [22888-70-6J.
SdareoL C2oH3602' (Mr 308.5). 1139900. [515-03-7]. Silybin. (2R,3R)-3,5,7 -Trihydroxy-2- [(2R,3R)-3-( 4-hydroxy-
(lR,2R,4aS,8aS) -1- [( 3R) -3 -Hydroxy- 3-methylpent -4-enyl]- 3-methoxyphenyl)-2-(hydroxymethyl)-2,3-dihydro-1,4-
2,5,5,8a -tetramethyldecahydronaphthalen -2-01. benzodioxin -6-yl]- 2,3-dihydro-4H-1-benzopyran -4-one.
Odourless crystals. White or yellowish powder, practically insoluble in water,
[al~o: 6.7, determined with a solution in anhydrous ethanol. soluble in acetone and in methano1.
bP19mm: 218 oC to 220 oc. Silibinin used in the assay of Milk thistle fruit (1860) complíes
mp: 96 oC to 98 oc. with the following additional test.
Sclareol used in the chromatographic profile test in the Assay. Liquid chromatography (2.2.29) as prescribed in the
monograph Clary sage oil (1850) complies with the following monograph Milk thistle fruit (1860).
additional test. Test solution. Dissolve 5.0 mg of silibinin, dried in vacuo, in
Assay. Gas chromatography (2.2.28) as prescribed in the methanol R and dilute to 50.0 mL with the same solvent.
monograph Clary sage oil (1850). Silibinin A and silibinin B content: minimum 95.0 per cent,
Content: minimum 97 per cent, calculated by the ca!culated by the normalisation procedure.
normalisation procedure.
Silica geln-acceptor/n-donor [or chiral separations.
Scopoletin. ClOHsÜ4' (Mr 192.2). 1158700. [92-61-5]. 1160100.
7-Hydroxy-6-methoxy-2H-l-benzopyran-2-one. A very finely divided silica gel for chromatography consisting
7-Hydroxy-6-methoxycoumarin. of spherical particles to which 1-(3,5-dinitrobenzamido)-
Faintly beige, fine crystals. 1,2,3,4-tetrahydrophenantrene has been covalently bound,
mp: 202 oC to 208 oc. showing both n-electron acceptor and n-electron donor
characteristics. The particle size and the configuration are
SDS-PAGE running buffer. 1114900. indicated after the name of the reagent in the tests where it
Dissolve 151.4 g of tris(hydroxymethyl)aminomethane R, is used.
721.0 g of glycine R and 50.0 g of sodium laurilsulfate R
Silica gel for chiral separation, amylose derivative of.
in water R and dilute to 5000 mL with the same solvent.
1171700.
Immediately before use, dilute to 10 times its volume with
water R and mix. Measure the pH (2.2.3) of the diluted A very finely divided silica gel for chromatography (5 flm)
solution. The pH is between 8.1 and 8.8. coated with the following derivative:

t
SDS-PAGE sample buffer (concentrated). 1115000. CH20R
Dissolve 1.89 g of tris(hydroxymethyl)aminomethane R, 5.0 g
of sodium laurilsulfate R and 50 mg of bromophenol blue R in
o OR
water R. Add 25.0 mL of glyeerol R and dilute to 100 mL with
water R. Adjust the pH to 6.8 with hydrochloric aeid R, and R o OR OR
dilute to 125 mL with water R.

General Notices (1) apply to all monographs and other texts 511
4.1.1. Reagents EUROPEAN PHARMACOPOElA 8.0

Silica gel for chiral separation, cenuIose derivative of. Fine, white or almost white, homogeneous powder, practically
1110300. insoluble in water and in ethanol (96 per cent).
A very finely divided silica gel for chromatography (5 11m)
coated with the following derivative: Silica gel for chromatography, aminopropylmethylsilyl.
1102400.
Silica gel with a fine particle size (between 3 ¡.tm and 10 11m),
OR chemically modified by bonding aminopropylmethylsilyl
R = groups on the surface. The particle size is indicated aÍter the
name of the reagent in the tests where it is used.
Fine, white or almost white, homogeneous powder, practically
insoluble in water and in ethanol (96 per cent).
Silica gel AD fOl chiral separation. 1171700.
See Amylose derivative of si/iea gel for chiral separation R. Silica gel for chromatography, aminopropylsilyL 1077000.
Silica gel with a fine particle size (between 3 11m and 10 ~lm),
Silica gel AGP for chiral chromatography. 1148700.
chemically modified by bonding aminopropylsilyl groups on
See a1-Acid-glycoprotein silica gel for chiral separatíon R. the surface. The particle size is indicated after the name of the
Silica gel, anhydrous, 1076100. [112926-00-8]. reagent in the tests where it is used.
Partly dehydrated polymerised, amorphous silieic aeid, Fine, white or almost white, homogeneous powder, practically
absorbing at 20 oC abaut 30 per cent of its mass of water. insoluble in water and in ethanol (96 per cent).
Practically insoluble in water, partly soluble in solutions
of sodium hydroxide. It contains a suitable indicator for Silica gel for chromatography, aminopropylsilyl Rl.
detection of the humidity status, far which the colour change 1077001.
from the hydrated to anhydrous form is given on the label. Silica gel with a particle size of about 55 [lm, chemically
modified by bonding aminopropylsilyl groups on the surface.
Silica gel BC for chiral chromatography. 1161300.
A very finely divided silica gel for chromatography (5 [lm) Silica gel for chromatography, amylose derivative of.
coated with ~-cyclodextrin. Higher selectivity may be obtained 1109800.
when cyclodextrin has been derivatized with propylene oxide.
A very finely divided (10 [lm) silica gel, chemically modified
Silica gel fOl chiral chromatography, urea type. 1181000. at the surface by the bonding of an amylose derivative. The
A very finely divided silica gel (5 11m) coated with the particle size is indicated after the name of the reagent in the
following derivative: test where it is used.
O Fine, white or almost white, homogenous powder, practically

~¿H'- ~Y~YlrN02
insoluble in water and in ethanol (96 per cent).

f '\ H O Y Silica gel for chromatography, butylsilyL 1076200.


- N0 2
A very finely divided silica gel (3-10 ¡.tm), chemically modified
at the surface by the bonding of butylsilyl groups. The particle
Silica gel for chromatography. 1076900. size is indicated after the name of the reagent in the tests
A very finely divided (3-10 11m) silica gel. The particle size where it is used.
is indicated after the name of the reagent in the tests where Fine, white or almost white, homogeneous powder, practically
it is used. insoluble in water and in ethanol (96 per cent).
Fine, white or almost white, homogeneous powder, practically Spheroidal si/ica: 30 nm.
insoluble in water and in ethanol (96 per cent).
Pore volume: 0.6 cm 3!g.
Silica gel for chromatography, alkyl-bonded for use with
Specific surface area: 80 m 2! g.
highly aqueous mobile phases. 1160200.
A very finely divided silica gel with bonded alkyl groups SHica gel for chromatography, butylsilyl, end-capped.
suitable for use with highly aqueous mobile phases. 1170500.
Silica gel for chromatography, alkyl-bonded for use with A very fineIy divided silica (3-10 11m), chemically modified
highly aqueous mobile phases, end-capped. 1176900. at the surface by the bonding of butylsilyl groups. To
A very finely divided silica gel with bonded alkyl groups minimise any interaction with basic compounds, it is carefully
suitable for use with highly aqueous mobile phases. To end-capped to cover most of the remaining silanol groups.
minimise any interaction with basic compounds it is carefully The particle size is indicated after the name of the reagent in
end -capped to cover most of the remaining silanol groups. the tests where it is used.
The particle size is indicated after the name of the reagent in Fine, white or almost white, homogenous powder, practically
the tests where it is used. insoluble in water and in ethanol (96 per cent).
Silica gel for chromatography, amidohexadecylsilyl.
Silica gel for chromatography, crown-ether. 1178000.
1170400.
A very finely divided silica gel with a fine particle size, Stationary phase for liquid chromatography.
chemically modified at the surface by the bonding of Crown ether coated on silica gel.
amidohexadecylsilyl groups. The particle size is indicated
after the name of the reagent in the test where it is used. Silica gel for chromatography, cyanosHyL 1109900.
SHica gel for chromatography, aminohexadecylsilyl. A very finely divided silica gel chemically modified at the
1138400. surface by the bonding of cyanosilyl groups. The particle size
A very finely divided (3-10 [lm) silica gel with a fine particle is indicated after the name of the reagent in the tests where
size chemically modified at the surface by the bonding of it is used.
aminohexadecylsilyl groups. The particle size is indicated Fine, white or almost white, homogeneous powder, practically
after the name of the reagent in the test where it is used. insoluble in water and in ethanol (96 per cent).

512 See the information section on general monographs (cover pages)


EUROPEAN PHARMACOPOEIA 8.0 4.1.1. Reagents

Silica gel for chromatography, di-isobutyloctadecylsilyl. Silica gel for chromatography, human albumin coated.
1140000. 1138500.
A very finely divided silica gel chemically modified at the A very finely divided (3-10 flm) silica gel, chemically modified
surface by the bonding of di-isobutyloctadecylsilyl groups. at the surface by the bonding of human albumino The particle
The particle size is indicated afier the name of the reagent in size is indicated afier the name of the reagent in the tests
the tests where it is used. where it is used.
White or almost white, fine, homogeneous powder.
Silica gel for chromatography, diisopropylcyanopropylsilyl.
1168100. Silica gel for chromatography, hydrophilic. 1077200.
A very finely divided silica gel chemically modified at the A very finely divided (3-10 flm) silica gel whose surface has
surface by the bonding of diisopropylcyanopropylsilyl groups. been modified to provide hydrophilic characteristics. The
The particle size is indicated after the name of the reagent in particle size may be stated afier the name of the reagent in the
which the test is used. tests where it is used.
Silica gel for chromatography, nitrile. 1077300.
Silica gel for chromatography, dimethyloctadecylsilyl.
A very finely divided silica gel, chemically modified at the
1115100.
surface by the bonding of cyanopropylsilyl groups. The
A very finely divided silica gel (3-10 flm), chemically modified particle size is indicated afier the name of the reagent in the
at the surface by the bonding of dimethyloctadecylsilyl groups. test where it is used.
The particle size is indicated afier the name of the reagent in Fine white or almost white, homogenous powder, practicalIy
the tests where it is used. insoluble in water and in ethanol (96 per cent).
Fine, white or almost white, homogeneous powder, practically
insoluble in water and in ethanol (96 per cent). Irregular Silica gel for chromatography, nitrile R1. 1077400.
particle size. A very finely divided silica gel consisting of porous, spherical
particles with chemicalIy bonded nitrile groups. The particle
Specific surface area: 300 m 2/ g. size is indicated afier the name of the reagent in the test where
it is used.
Silica gel for chromatography, diol. 1110000.
Fine, white or almost white, homogeneous powder, practically
Spherical silica particles to which dihydroxypropyl groups are insoluble in water and in ethanol (96 per cent).
bonded. Pore size 10 nm.
Silica gel for chromatography, nitrile R2. 1119500.
Silica gel for chromatography, dodecylsilyl, end-capped. Ultrapure silica gel, chemicalIy modified at the surface by the
1179700. introduction of cyanopropylsilyl groups. Less than 20 ppm
A very finely divided silica gel, chemicalIy modified at of metals. The particle size is indicated after the name of the
the surface by the introduction of dodecylsilyl groups. To reagent in the tests where it is used.
minimise any interaction with basic compounds, it is carefulIy Fine white or almost white, homogenous powder, practicalIy
end-capped to cover most of the remaining silanol groups. insoluble in water and in ethanol (96 per cent).

Silica gel for chromatography, hexadecylamidylsilyl. Silica gel for chromatography, nitrile, end-capped. 1174500.
1162500. A very finely divided silica gel, chemicalIy modified at the
A very finely divided (5 flm) silica gel, chemicalIy surface by the bonding of cyanopropylsilyl groups. To
minimise any interaction with basic components it is carefully
modified at the surface by the introduction of
end -capped to cover most of the remaining silanol groups.
hexadecylcarboxamidopropyldimethylsilyl groups.
The particle size is indicated after the name of the reagent in
Silica gel for chromatography, hexadecylamidylsilyl, the test where it is used.
end-capped. 1172400. A fine, white or almost white, homogenous powder, practically
insoluble in water and in anhydrous ethanol.
A very finely divided (5 flm) silica gel, chemically
modified at the surface by the introduction of Silica gel for chromatography, 4-nitrophenylcarbamidesilyl.
hexadecylcarboxamidopropyldimethylsilyl groups. To 1185200.
minimise any interaction with basic compounds it is carefulIy A very finely divided silica gel, chemically modified at the
end-capped to caver most ofthe remaining silanol groups. surface by bonding of 4-nitrophenylcarbamide groups. The
particle size is indicated afier the name of the reagent in the
Silica gel for chromatography, hexylsilyl. 1077100. tests where it is used.
A very finely divided (3-10 flm) silica gel, chemically modified Fine, white or almost white, homogeneous powder, practically
at the surface by the bonding ofhexylsilyl groups. The particle insoluble in water and in ethanol (96 per cent).
size is indicated afier the name of the reagent in the tests
where it is used. Silica gel for chromatography, octadecanoylaminopropyl-
silyl. 1115200.
Fine, white or almost white, homogeneous powder, practicalIy
insoluble in water and in ethanol (96 per cent). A very finely divided (3-10 flm) silica gel, chemically modified
at the surface by the bonding of aminopropylsilyl groups
Silica gel for chromatography, hexylsilyl, end-capped. which are acylated with octadecanoyl groups. The particle
1174400. size is indicated afier the name of the reagent in the tests
where it is used.
A very finely divided (3-10 flID) silica gel, chemically modified
Fine, white or almost white, homogeneous powder, practically
at the surface by the bonding of hexylsilyl groups. To
insoluble in water and in ethanol (96 per cent).
minimise any interaction with basic compounds it is carefulIy
end-capped to cover most of the remaining silanol groups. Silica gel for chromatography, octadecylsilyl. 1077500.
The particle size is indicated after the name of the reagent in A very finely divided (3-10 flm) silica gel, chemicalIy modified
the tests where it is used. at the surface by the bonding of octadecylsilyl groups. The
A fine, white or almost white, homogeneous powder, particle size is indicated afier the name of the reagent in the
practically insoluble in water and in ethanol (96 per cent). tests where it is used.

General Natices (1) apply ta all manographs and ather texts 513
4.1.1. Reagents EUROPEAN PHARMACOPOEIA 8.0

Fine, white or almost white, homogeneous powder, practically carefully end-capped to cover most of the remaining silanol
insoluble in water and in ethanol (96 per cent). groups. The particle size is indicated after the name of the
reagent in the test where it is used.
Silica gel for chromatography, octadecylsilyl Rl. 1110100.
Fine, white or almost white, homogeneous powder, practically
A very finely divided ultrapure silica gel, chemically modified insoluble in water and in ethanol (96 per cent).
at the surface by the bonding of octadecylsilyl groups. The
particle size, the pore size and the carbon loading are indicated Silica gel for chromatography, octadecylsilyl, monolithic.
after the name of the reagent in the tests where it is used. Less 1154500.
than 20 ppm of metals. Monolithic rods of highly porous (greater than 80 per cent)
Silica gel for chromatography, octadecylsilyl R2. 1115300. metal-free silica with a bimodal pore structure, modified at
the surface by the bonding of octadecylsilyl groups.
A very finely divided (15 nm pore size) ultrapure silica
gel, chemically modified at the surface by the bonding of Silica gel for chromatogI'aphy, octadecylsilyl, with
octadecylsilyl groups (20 per cent carbon load), optimised embedded polar groups, end-capped. 1177900.
for the analysis of polycyclic aromatic hydrocarbons. The A very finely divided silica gel (3-10 11m). The particles are
particle size is indicated after the name of the reagent in the based on a mixture of silica chemically modified at the surface
tests where it is used. by the bonding of octadecylsilyl groups and silica chemically
Fine, white or almost white, homogeneous powder, practically modified with a reagent providing a surface with chains having
insoluble in water and in ethanol (96 per cent). embedded polar groups. Furthermore, the pacldng material is
end -capped. The particle size is indicated after the name of
SHica gel for chromatography, octadecylsilyl, the reagent in the tests where it is used.
base-deactivated. 1077600.
A very finely divided (3-10 11m) silica gel, pretreated before Silica gel for chromatogI'aphy, octadecylsilyl, with polar
the bonding of octadecylsilyl groups by careful washing incorpoI'ated groups, end-capped. 1165100.
and hydrolysing most of the superficial siloxane bridges to A very finely divided silica gel (3-10 11m). The particles are
minimise the interaction with basic components. The particle based 011 silica, chemically modified with a reagent providing
size is indicated after the name of the reagent in the tests a surface with chains having polar incorporated groups and
where it is used. terminating octadecyl groups. Furthermore, the packing
Fine, white or almost white, homogeneous powder, practically material is end -capped. The particle size is indicated after the
insoluble in water and in ethanol (96 per cent). name of the reagent in the tests where it is used.
Fine, white or almost white, homogeneous powder.
Silica gel for chromatography, octadecylsilyl, end-capped.
1115400. Silica gel for chI'omatography, octylsilyl. 1077700.
A very finely divided (3-10 11m) silica gel, chemically modified A very finely divided (3-10 11m) silica gel, chemical1y modified
at the surface by the bonding of octadecylsilyl groups. To at the surface by the bonding of octylsilyl groups. The particle
minimise any interaction with basic compounds it is carefully size is indicated after the name of the reagent in the tests
end -capped to cover most of the remaining silanol groups. where it is used.
The particle size is indicated after the name of the reagent in
the tests where it is used. Fine, white or almost white, homogeneous powder, practically
insoluble in water and in ethanol (96 per cent).
Fine, white or almost white, homogenous powder, practically
insoluble in water and in ethanol (96 per cent). Silica gel foI' chromatogI'aphy, octylsilyl Rl. 1077701.
Silica gel far chromatography, octadecylsilyl, A very finely divided (3-10 11m) silica gel, chemically modified
end-capped Rl. 1115401. at the surface by the bonding of octylsilyl and methyl groups
(double bonded phase). The particle size is indicated after the
A very finely divided (10 nm pore size) ultrapure silica name of the reagent in the tests where it is used.
gel, chemically modified at the surface by the bonding
of octadecylsilyl groups (19 per cent carbon load). To Fine, white or almost white, homogeneous powder, practically
minimise any interaction with basic compounds it is carefully insoluble in water and in ethanol (96 per cent).
end -capped to cover most oí the remaining silanol groups. The
Silica gel for chromatography, octylsilyl R2. 1077702.
particle size is indicated after the name of the reagent in the
tests where it is used. It contains less than 20 ppm of metals. Ultrapure very finely divided (lO nm pore size) silica gel,
chemically modified at the surface by the bonding of octylsilyl
Silica gel for chromatography, octadecylsilyl, end-capped, groups (19 per cent carbon load). Less than 20 ppm of metals.
base-deactivated. 1108600.
Silica gel for chromatography, octylsilyl R3. 1155200.
A very finely divided (3-10 11m) silica gel with a pore size
of 10 nm and a carbon loading of 16 per cent, pre-treated A very finely divided ultrapure silica gel, chemically modified
before the bonding of octadecylsilyl groups by washing and at the surface by the bonding of octylsilyl groups and sterically
hydrolysing most of the superficial siloxane bridges. To protected with branched hydrocarbons at the silanes. The
further minimise any interaction with basic compounds it is particle size is indicated after the name of the reagent in the
carefully end-capped to cover most of the remaining silanol tests where it is used.
groups. The particle size is indicated after the name of the
reagent in the test where it is used. Silica gel for chromatography, octylsilyl, base-deactivated.
1131600.
Fine, white or almost white, homogeneous powder, practically
insoluble in water and in ethanol (96 per cent). A very finely divided (3-10 11m) silica gel, pretreated before
the bonding of octylsilyl groups by careful washing and
Silica gel for chromatography, octadecylsHyl, end-capped, hydrolysing most of the superficial siloxane bridges to
base-deactivated Rl. 1162600. minimise the interaction with basic components. The particle
A very finely divided (3-10 11m) silica gel pre-treated size is indicated after the name of the reagent in the tests
before the bonding of octadecylsilyl groups by washing and where it is used.
hydrolysing most of the superficial siloxane bridges. To Fine, white or almost white, homogeneous powder, practically
further minimise any interaction with basic compounds it is insoluble in water and in ethanol (96 per cent).

514 See the information section on general monographs (ca ver pages)
EUROPEAN PHARMACOPOEIA 8.0 4.1.1. Reagents

Silica gel for chromatography, octylsilyl, end-capped. Silica gel for chromatography, phenylsilyI Rl. 1075700.
1119600. A very finely divided silica gel (5 ¡.tm), chemically modified at
A very finely divided (3-10 ¡.tm) silica gel, chemically modified the surface by the bonding of phenyl groups. The particle size
at the surface by the bonding of octylsilyl groups. To is indicated after the name of the reagent in the tests where
minimise any interaction with basic compounds, it is carefully it is used.
end-capped to cover most of the remaining silanol groups. Fine, white or almost white, homogeneous powder, practically
The particle size is indicated after the name of the reagent in insoluble in water, in ethanol (96 per cent) and in methylene
the tests where it is used. chloride.
Fine, white or almost white, homogeneous powder, practically Spheroidal silica: 8 nm.
insoluble in water and in ethanol (96 per cent). Specific sUlface area: 180 m 2 /g.
Carbon loading: 5.5 per cent.
Silica gel for chromatography, octylsilyl, end-capped,
base-deactivated. 1148800. Silica gel for chromatography, phenylsilyl, end-capped.
A very finely divided (3-10 ¡.tm) silica gel, pre-treated 1154900.
before the bonding of octylsilyl groups by washing and A very finely divided (5-10 ~lm) silica gel, chemically
hydrolysing most of the superficial siloxane bridges. To modified at the surface by the bounding of phenyl groups. To
further minimise any interaction with basic compounds it is minimise any interaction with basic compounds it is carefully
carefully end-capped to cover most of the remaining silanol end-capped to cover most of the remaining silanol groups.
groups. The particle size is indicated after the name of the The particle size is indicated after the name of the reagent in
reagent in the test where it is used. the tests where it is used.
Fine, white or almost white, homogeneous powder, practically Silica gel for chromatography, propoxybenzene,
insoluble in water and in ethanol (96 per cent). end-capped. 1174600.
A very finely divided (3-10 flm) silica gel, chemically modified
Silka gel for chromatography, octylsilyl, with polar at the surface by the bonding of propoxybenzene groups. The
incorporated groups, end-capped. 1152600. particle size is indicated after the name of the reagent in the
A very finely divided silica gel (3-10 ¡.tm). The particles are test where it is used.
based on silica, chemically modified with a reagent providing
a surface with chains having polar incorporated groups and Silica gel for chromatography, propylsilyL 1170700.
terminating octyl groups. Furthermore, the packing material A very finely divided silica gel (3-10 flm), chemically modified
is end-capped. The particle size is indicated after the name of at the surface by the bonding of propylsilyl groups. The
the reagent in the tests where it is used. particle size is indicated after the name of the reagent in the
test where it is used.
Fine, white or almost white, homogeneous powder.
Silica gel for chromatography, strong-anion-exchange.
Silica gel for chromatography, oxypropionitrilsilyL 1077800.
1184700. A very finely divided (3-10 ¡.tm) silica gel, chemically modified
A very finely divided silica gel chemically modified at the at the surface by the bonding of quaternary ammonium
surface by the bonding of oxypropionitrilsilyl groups. The groups. The particle size is indicated after the name of the
particle size is indicated after the name of the reagent in the reagent in the tests where it is used.
tests where it is used. Fine, white or almost white, homogeneous powder, practically
insoluble in water and in ethanol (96 per cent).
Silka gel for chromatography, palmitamidopropylsilyl,
end-capped. 1161900. pH limit of use: 2 to 8.
A very finely divided (3-10 flm) silica gel, chemically modified Silica gel for chromatograpny, strong cation-excnal1ge.
at the surface by the bonding of palmitamidopropyl groups 1161400.
and end-capped with acetamidopropyl groups. The particle A very finely divided (5-10 ¡.tm) silica gel, chemically modified
size is indicated after the name of the reagent in the tests at the surface by the bonding of sulfonic acid groups. The
where it is used. particle size is specified after the name of the reagent in the
Fine, white or almost white, homogeneous powder, practically tests where it is used.
insoluble in water and in ethanol (96 per cent). Silica gel for chromatograpny, trimethylsilyl. 1115500.
Silica gel for chromatography, phenylhexylsilyL 1153900. A very finely divided (3-10 ¡.tm) silica gel, chemically modified
at the surface by the bonding of trimethylsilyl groups. The
A very finely divided silica gel, chemically modified at the particle size is indicated after the name of the reagent in the
surface by the bonding of phenylhexyl groups. The particle tests where it is used.
size is indicated after the name of the reagent in the tests Fine, white or almost white, homogeneous powder, practically
where it is used. insoluble in water and in ethanol (96 per cent).
Silica gel for chromatography, phenylhexylsilyl, Silica gel for size-exclusion chromatography. 1077900.
end-capped. 1170600. A very finely divided silica gel (lO ¡.tm) with a very hydrophilic
A very finely divided silica gel (3 ¡.tm), chemically modified surface. The average diameter of the pores is about 30 nm.
at the surface by the bonding of phenylhexylsilyl groups. To It is compatible with aqueous solutions between pH 2 and 8
minimise any interaction with basic compounds, it is carefully and with organic solvents. It is suitable for the separation of
end -capped to cover most of the remaining silanol groups. proteins with relative molecular masses of 1 x 103 to 3 x 10 5 .
The particle size is indicated after the name of the reagent in
the tests where it is used. Silica gel G. 1076300. [112926-00-8].
Contains abont 13 per cent of calcium sulfate hemihydrate.
Silica gel for chromatography, phenylsilyl. 1110200. Fine, white or almost white, homogeneous powder with a
A very finely divided silica gel, chemically modified at the particle size of about 15 flm.
surface by the bonding of phenyl groups. The particle size Calcium sulfate contento Place 0.25 g in a ground-glass
is indicated after the name of the reagent in the tests where stoppered flask, add 3 mL of dilute hydrochlaric acid R
it is used. and 100 mL of water R and shake vigorously for 30 mino

General Natices (1) apply ta all monagraphs and ather texts 515
4.1.1. Reagents EUROPEAN PHARMACOPOEIA 8.0

Filter through a sintered-glass filter (2.1.2) and wash the solution to a separating funnel by means of 100 mL of water R,
residue. Carry out on the combined filtrate and washings the acidify (pH 2 to 3) with dilute hydrochloric acid R and shake
complexometric assay of calcium (2.5.11). with three quantities, each of 10 mL of chloroform R. Dry
1 mL of 0.1 M sodium edetate is equivalent to 14.51 mg of the combined chloroform extracts over anhydrous sodium
CaS0 4 ,1/2 H 20 . sulfate R, filter and evaporate to dryness on a water-bath.
Dissolve the residue in 50 mL of chloroform R. Examine by
pH (2.2.3). Shake 1 g for 5 min with 10 mL of carbon thin-Iayer chromatography (2.2.27), using silanised silica
dioxide-free water R. The pH of the suspension is about 7. gel HF 2s4 as the coating substance. Apply to the plate at each
Silica gel GF 254 • 1076400. [112926-00-8]. of three separate points 10 [.tL of the chloroformic solution.
Develop over a path of 14 cm with a mixture of 10 volumes of
Contains about 13 per cent of calcium sulfate hemihydrate glacial acetic acid R, 25 volumes of water R and 65 volumes of
and about 1.5 per cent of a fluorescent indicator having an dioxan R. Dry the plate at 120 oC for 30 mino Allow to cool,
optimal intensity at 254 nm. spray with a 35 giL solution of phosphomolybdic acid R in
Fine, white or almost white, homogeneous powder with a 2-propanol R and heat at 150 oC until the spots become visible.
particle size of about 15 [.tm. Treat the plate with ammonia vapour until the background
Calcium sulfate contento Determine by the method prescribed is white. The chromatograms show four clearIy separated,
for siliea gel G R. well-defined spots.
pH (2.2.3). Complies with the test prescribed for siliea gel G R. Silica gel OC for chiral separations. 1146800.
Fluoreseence. Thin-layer chromatography (2.2.27) using A very finely divided silica gel for chromatography (5 [.tm)
silica gel GF254 R as the coating substance. Apply separately coated with the following derivative:
to the plate at ten points increasing volumes from 1 [.tL to
10 [.tL of a 1 giL solution of benzoic acid R in a mixture of
10 volumes of anhydrous formic acid R and 90 volumes of
2-propanol R. Develop over a path of 10 cm with the same
mixture of solvents. After evaporating the solvents examine
the chromatogram in ultraviolet light at 254 nm. The benzoic
acid appears as dark spots on a fluorescent background in the
upper third of the chromatogram for quantities of 2 flg and
greater. Silka gel OD for chiral separati.ons. 1110300.
See Cellulose derivative of si/ica gel for chiral separation R.
Silica gel H. 1076500. [112926-00-8].
Fine, white or almost white, homogeneous powder with a Silica gel OI for chiral separations. 1179800.
particle size of about 15 [.tm. A very finely divided silica gel for chromatography
pH (2.2.3). Complies with the test prescribed for silica gel GR. consisting of spherical particles coated with ceHulose
tris( 4-methylbenzoate). The particle size is indicated after the
Silica gel H, silanised. 1076600. name of the reagent in the test where it is used.
Preparation of a thin layer. See silanised silica gel HF254 R.
Silicotungstic add. H 4 SiW 120 40 ,xHp. 1078000.
A fine, white or almost white homogeneous powder which, [11130-20-4].
after being shaken with water, floats on the surface because of
White or yellowish-white crystals, deliquescent, very soluble
its water-repellent properties. in water and in ethanol (96 per cent).
Chromatographic separation. Complies with the test
Storage: in an airtight container.
prescribed for silanised si/ica gel HF251 R.
Silicristin. C2S H 2PlO' (Mr 482.4). 1151500. [33889-69-9].
SiHca gel HF 2S4 ' 1076700. (2R,3R)-3,5,7 - Trihydroxy-2- [(2R,3S)-7 -hydroxy-2-( 4-
Contains about 1.5 per cent of a fluorescent indicator having hydroxy-3-methoxyphenyl)-3-hydroxymethyl-2,3-dihydro-l-
an optimal intensity at 254 nm. benzofuran-5-yl]chroman-4-one.
Fine, white or almost white, homogeneous powder with a White or yellowish powder, practically insoluble il1 water,
particle size of about 15 [.tm. soluble in acetone and in methanoL
pH. Complies with the test prescribed for silica gel G R.
Silidianin. C2SH 2PlO' (Mr 482.4). 1151600. [29782-68-1].
Fluorescence. Complies with the test prescribed for silica (3R,3aR,6R, 7aR,8R) -7 a -Hydroxy -8-( 4-hydroxy-3-
gel GF254 R. methoxyphenyl)-4- [(2R, 3R)-3,5, 7 -trihydroxy-4-oxochroman-
2-yl]-2,3,3a, 7a -tetrahydro-3,6-methano-l-benzofuran-
Silica gel HF 254 , sHanised. 1076800. 7(6aH)-one.
Contains about 1.5 per cent of a fluorescent indicator having White or yellowish powder, practically insoluble in water,
an optimal intensity at 254 nm. soluble in acetone and in methanol.
Fine, white or almost white, homogeneous powder which,
after shaking with water, floats on the surface because of its Silver diethyldithiocarbamate. C SH lOAgNS 2 • (Mr 256.1).
water- repellent properties. 1110400. [1470-61-7].
Preparation of a thin layer. Vigorously shake 30 g for 2 min Pale-yellow or greyish-yellow powder, practically insoluble
with 60 mL of a mixture of 1 volume of methanol R and in water, soluble in pyridine.
2 volumes of water R. Coat carefully cleaned plates with a It may be prepared as follows. Dissolve 1.7 g of silver nitrate R
layer 0.25 mm thick using a spreading device. Allow the in 100 mL of water R. Separately dissolve 2.3 g of sodium
coated plates to dry in air and then heat in an oven at 100 oC diethyldithiocarbamate R in 100 mL of water R. Cool both
to 105 oC for 30 mino solutions to 10 oC, then mix and while stirring collect the
Chromatographic separation. Introduce 0.1 g each of methyl yellow precipitate 011 a sintered-glass filter (2.1.2) and wash
laurate R, methyl myristate R, methyl palmitate R and methyl with 200 mL of cold water R. Dry the precipitate in vacuo for
stearate R into a 250 mL conical flask. Add 40 mL of alcoholic 2-3 h.
potassium hydroxide solution R and heat under a reflux Silver diethyldithiocarbamate may be used provided it has not
condenser on a water-bath for 1 h. Allow to cool, transfer the changed in colour or developed a strong odour.

516 See the information section on general monographs (caver pages)


EUROPEAN PHARMACOPOEIA 8.0 4.1.1. Reagents

Silver manganese papero 1078200. into a suitable 3 mL flask and evaporate to dryness under
Immerse strips of slow filter paper into a solution containing nitrogen R. To the residue add 100 [1L of a freshly prepared
8.5 giL of manganese sulfate R and 8.5 giL of silver nitrate R. mixture of 50 [1L of 1-methylimidazole R and 1.0 mL of
Maintain for a few minutes and allow to dry over diphosphorus heptafluoro-N-methyl-N-(trimethylsilyl)butanamide R. Close
pentoxide R protected from acid and alkaline vapours. the flask tightly and heat at 100 oC for 15 mino Allow to cool.
Injection: 1 [1L of the test solution.
Silver nitrale. 1078300. [7761-88-8].
See Silver nitrate (0009). Sodium. Na. (A r 22.99). 1078500. [7440-23-5].
A metal whose freshly cut surface is bright silver-grey. It
Silver nitrate reagent. 1078305. rapidly tarnishes in contact with air and is oxidised completely
To a mixture of 3 mL of concentrated ammonia R and to sodium hydroxide and converted to sodium carbonate. It
40 mL of 1 M sodium hydroxide, add 8 mL of a 200 giL reacts violently with water, yielding hydrogen and a solution
solution of silver nitrate R, dropwise, with stirring. Dilute of sodium hydroxide; soluble in anhydrous methanol, yielding
to 200 mL with water R. hydrogen and a solution of sodium methoxide; practically
insoluble in light petroleum.
Silver nitrate solution Rl. 1078301.
Storage: under light petroleum or liquid paraffin.
A 42.5 giL solution.
Storage: protected from light. Sodium acetate. 1078600. [6l31-90-4].
See Sodium acetate trihydrate (0411).
Silver nitrate solution R2. 1078302.
A 17 giL solution. Sodium acetate, anhydrous. C 2H 3Naü 2 • (Mr 82.0). 1078700.
[127 -09-3].
Storage: protected from light.
Colaurless crystals ar granules, very soluble in water, sparingly
Silver nitrate solution, ammoniacal. 1078303. soluble in ethanol (96 per cent).
Dissolve 2.5 g of silver nitrate R in 80 mL of water R and Loss on drying (2.2.32). Not more than 2.0 per cent,
add dilute ammonia R1 dropwise until the precipitate determined by drying in an oven at 105 oc.
has dissolved. Dilute to 100 mL with water R. Prepare
Sodium arsenite. NaAs0 2 • (Me 129.9).1165900. [7784-46-5].
immediately before use.
Sodium metaarsenite.
Silver nitrate solution in pyridine. 1078304.
Sodium arsenite solution. 1165901.
An 85 giL solution in pyridine R.
Dissolve 5.0 g of sodium arsenite R in 30 mL of 1 M sodium
Storage: protected from light. hydroxide. Cool to O oC and add, while stirring, 65 mL of
dilute hydrochloric acid R.
Silver oxide. AgzÜ. (Mr 231.7). 1078400. [20667-12-3].
Disilver oxide. Sodium ascorbate solution. lO78800. [l34-03-2].
Brownish-black powder, practically insoluble in water and in Dissolve 3.5 g of ascorbic acid R in 20 mL of 1 M sodium
ethanol (96 per cent), freely soluble in dilute nitric acid and hydroxide. Prepare immediately before use.
in ammonia.
Storage: protected from light. Sodium azide. NaN3' (Mr 65.0). 1078900. [26628-22-8].
White or almost white, crystalline powder or crystals, freely
Sinensetin. C2oH2007' (M, 372.4). 1110500. [2306-27-6]. soluble in water, slightly soluble in ethanol (96 per cent).
3',4' ,5,6, 7 -Pentamethoxyflavone.
Sodium bicarbonate. 1081300. [144-55-8].
White or almost white, crystalline powder, practically
insoluble in water, soluble in ethanol (96 per cent). See sodium hydrogen carbonate R.
mp: about 177 oc. Sodium bismuthate. NaBiO]' (Mr 280.0). 1079000.
Absorbance (2.2.25). A solution in methanol R shows [12232-99-4].
3 absorption maxima, at 243 nm, 268 nm and 330 nm. Content: minimum 85.0 per cent.
Assay. Liquid chromatography (2.2.29) as prescribed in the Yellow or yellowish-brown powder, slowly decomposing when
monograph Java tea (1229). moist or at a high temperature, practically insoluble in cold
Content: minimum 95 per cent, calculated by the water.
normalisation procedure. Assay. Suspend 0.200 g in 10 mL of a 200 giL solution of
potassium iodide R and add 20 mL of dilute sulfuric acid R.
Sinomenine. C19H23N04' (Mr 329.4). 1183400. [115-53-7].
Using 1 mL of starch solution R as indicator, titrate with 0.1 M
7,8-Didehydro-4-hydroxy-3,7 -dimethoxy-17 -methyl-
sodium thiosulfate until an orange colour is obtained.
9a, 13a, 14a -morphinan -6-one. Cucoline.
1 mL of 0.1 M sodium thiosulfate is equivalent to 14.00 mg
SitostanoL C 29 H sp. (Mr 416.7). 1140100. [19466-47-8]. ofNaBiO]'
Dihydro-~-sitosterol.
Sodium bromide. 1154300. [7647-15-6].
Content: minimum 95.0 per cent.
See Sodium bromide (0190).
~-Sitosterol. C 29 H saÜ. (Me 414.7). 1140200. [83-46-5].
Stigmast -5-en -3 ~-ol. 22,23-Dihydrostigmasterol. Sodium butanesulfonate. C 4 H 9Na0 3S. (Me 160.2). 1115600.
[2386-54-1].
White or almost white powder, practically insoluble in water,
sparingly soluble in tetrahydrofuran. White or almost white, crystalline powder, soluble in water.
Content: minimum 75.0 per cent mlm (dried substance). mp: greater than 300 oc.
Assay. Gas chromatography (2.2.28), as prescribed in the Sodium cakium edetate. 1174000. [62-33-9].
monograph Phytosterol (1911). See sodium calcium edetate (0231).
Test solution. Dissolve 0.100 g of the substance to be
examined in tetrahydrofuran R and dilute to 10.0 mL Sodium carbonate. 1079200. [6132-02-1].
with the same solvento Introduce 100 [1L of this solution See Sodium carbonate decahydrate (0191).

General Notices (1) apply to al! monographs and other texts 517
4.1.1. Reagents EUROPEAN PHARMACOPOEIA 8.0

Sodium carbonate, anhydrous. Na2C0 3 • (Mr 106.0). supernatant, measured at 260 nm using a mixture of 1 mL of
1079300. [497-19-8]. Disodium carbonate. imídazole buffer solutíon pH 6.5 R and 3 mL of perchloric acid
White or almost white powder, hygroscopic, freely soluble in (25 giL HCI0 4) as compensation liquid, is not greater than 0.3.
water. In each of two tubes, place 0.5 mL of solution A and 0.5 mL
When heated to about 300 oC it loses not more than 1 per of a solution of a reference preparation of streptodornase
cent of its mass. containing 10 IVlmL in ímidazole buffer solution pH 6.5 R.
To one tube add immediately 3 mL of perchloric acid (25 giL
Storage: in an airtight container.
HCI0 4 ). A precipitate is formed. Centrifuge and collect
Sodium carbonate solution. 1079301. supernatant A. Heat the ather tube at 37 oC for 15 min and add
A 106 giL solution of anhydrous sodium carbonate R. 3 mL of perchloric acid (25 giL HCIO J Centrifuge and collect
supernatant B. The absorbance of supernatant B, measured at
Sodium carbonate solution Rl. 1079302. 260 nm with reference to supernatant A is not less than 0.15.
A 20 giL solution of anhydrous sodium carbonate R in Sodium diethyldithiocarbamate. CSHlQNNaS 2 ,3H 2 0.
0.1 M sodium hydroxide. (M, 225.3). 1080000. [20624-25-3].
Sodium carbonate solution R2. 1079303. White or almost white or colourless crystals, freely soluble in
water, soluble in ethanal (96 per cent). The aqueous solution
A 40 giL solution of anhydrous sodium carbonate R in
is colourless.
0.2 M sodium hydroxide.
Sodium dihydrogen phosphate. 1080100. [13472-35-0].
Sodium carbonate monohydrate. 1131700. [5968-11-6].
See Sodíum díhydrogen phosphate díhydrate (0194).
See Sodium carbonate monohydrate (0192).
Sodium dihydrogen phosphate, anhydrous. NaH 2P0 4.
Sodium cetostearyl sulfate. 1079400. (Mr 120.0). 1080200. [7558-80-7].
See Sodíum cetostearyl sulfate (0847). White or almost white powder, hygroscopic.
Sodium chloride. 1079500. [7647-14-5]. Storage: in an airtight container.
See Sodíum chloríde (0193). Sodium dihydrogen phosphate monohydrate.
NaH 2 P04,H 20. (Me 138.0).1080300. [10049-21-5].
Sodium chloride solution. 1079502.
White or almost white, slightly deliquescent crystals or
A 20 per cent m/m solution. granules, freely soluble in water, practically insoluble in
Sodium chloride solution, saturated. 1079503. ethanol (96 per cent).
Mix 1 part of sodíum chloríde R with 2 parts of water R, Storage: in an airtight container.
shake from time to time and allow to stand. Before use, Sodium dioctyl sulfosucdnate. C2oH37Na07S, (Mr 444.6).
decant the solution from any undissolved substance and 1170800. [577-11-7]. Sodium 1,4-bis[(2-ethylhexyl)oxy]-
filter, if necessary. 1,4-dioxobutane-2-sulfonate. 1,4-Bis(2-ethylhexyl)
Sodium dtrate. 1079600. [6132-04-3]. sulfobutanedioate sodium salt.
See Sodium citrate (0412). White ar almost white, waxy solido
Sodium dithionite. Na 2SP4' (Mr 174.1). 1080400.
Sodium cobaltinitrite. Na 3[Co(N0 2)6]' (Mr 403.9).1079700.
[7775-14-6].
[13600-98-1]. Trisodium hexanitrocobaltate(IlI).
White or greyish-white, crystalline powder, oxidises in air,
Orange-yellow powder, freely soluble in water, slightly soluble
very soluble in water, slightly soluble in ethanol (96 per cent).
in ethanol (96 per cent).
Storage: in an airtight container.
Sodium cobaltinitrite solution. 1079701.
Sodium dodecyl sulfate. 1080500. [151-21-3].
A 100 giL solution. Prepare immediately before use.
See Sodíum laurílsulfate (0098).
Sodium decanesulfonate. ClQH 21 Na0 3 S. (Mr 244.3). 1079800. Content: minimum 99.0 per cent.
[13419-61-9].
Sodium edetate. 1080600. [6381-92-6].
Crystalline powder or flakes, white or almost white, freely
soluble in water, soluble in methanol. See Dísodíum edetate (0232).

Sodium decyl sulfate. ClQH Z1 Na0 4 S. (Me 260.3). 1138600. Sodium fluOl'esceinate. C20 HlQNa 20 S' (Mr 376.3). 1080700.
[142-87-0]. [518-47-8].
Schultz No. 880.
Content: minimum 95.0 per cent.
Colour Index No. 45350.
White or almost white powder, freely soluble in water.
Fluorescein sodium. Disodium 2-(3-oxo-6-oxido-3H-
Sodium deoxycholate. C24H39Na04' (Mr 414.6). 1131800. xanthen-9-yl)benzoate.
[302-95-4]. Sodium 3a,l2a-dihydroxy-5~-cholan-24-oate. Orange-red powder, freely soluble in water. Aqueous solutions
display al1 il1tense yellowish-green fluorescence.
Sodium deoxyribonudeate. (About 85 per cent has a relative
molecular mass of 2 x 10 7 or greater). 1079900. [73049-39-5]. Sodium fluoride. 1080800. [7681-49-4].
White or almost white, fibrous preparation obtained from calf See Sodíum fluoride (0514).
thymus.
Sodium formateo CHNaO z' (Mr 68.0).1122200. [141-53-7].
Test for suitabilíty. Dissolve 10 mg in imidazole buffer solutíon Sodium methanoate.
pH 6.5 R and dilute to 10.0 mL with the same buffer solution
White or almost white, crystalline powder or deliquescent
(solution A). Dilute 2.0 mL of solution A to 50.0 mL with
granules, soluble in water and in glycerol, slightly soluble in
ímídazole buffer solution pH 6.5 R. The absorbance (2.2.25) of
ethanol (96 per cent).
the solution, measured at 260 nm, is 0.4 to 0.8.
mp: about 253 oc.
To 0.5 mIo of solution A add 0.5 mIo of ímídazole buffer
solution pH 6.5 R and 3 mL of perchloric acid (25 giL HCI0 4). Sodium glucuronate. C6H9Na07,HzÜ. (Mr 234.1). 1080900.
A precipitate is formed. Centrifuge. The absorbance of the Sodium D-glucuronate monohydrate.

518 See the information section on general monographs (caver pages)


EUROPEAN PHARMACOPOEIA 8.0 4.1.1. Reagents

[ctl~O: about + 21.5, determined on a 20 giL solution. Sodium hydroxide solution, carbonate-free. 1081406.
Dissolve sodium hydroxide R in carbon dioxide-free water R
Sodium glycocholate. C26H42NNa06,2H20. to give a concentration of 500 giL and allow to stand.
(Mr 523.6). 1155500. [207300-80-9]. Sodium
Decant the clear supernatant, taking precautions to avoid
[( 3, 7,12-trihydroxy-5-cholan -24-oyl)amino] acetate dihydrate.
the introduction of carbon dioxide.
N- [( 3,5,7,12)-3,7,12-Trihydroxy-24-oxocholan -24-yl] glyeine
monosodium salt dihydrate. Sodium hydroxide solution, dHute. 1081402.
Content: minimum 97 per cent of C26H42NNa06,2H20. Dissolve 8.5 g of sodium hydroxide R in water R and dilute
to 100 mL with the same solvent.
Sodi.um heptanesulfonate. C 7H¡SNa0 3S. (M, 202.3). 1081000.
[22767-50-6]. Sodium hydroxide solution, methanolk. 1081403.
White or a1m05t white, crystalline mass, freely soluble in Dissolve 40 mg of sodium hydroxide R in 50 mL of water R.
water, soluble in methanol. Cool and add 50 mL of methanol R.
Sodium hydroxide solution, methanolic Rl. 1081405.
Sodium heptanesulfonate monohydrate. C 7H¡SNa0 3 S,H 2 0.
(M, 220.3). 1081100. Dissolve 200 mg of sodium hydroxide R in 50 mL of water R.
Cool and add 50 mL of methanol R.
Content: minimum 96 per cent (anhydrous substance).
White or almost white, crystalline powder, soluble in water, Sodium hydroxide solution, strong. 1081404.
very slightly soluble in anhydrous ethanol. Dissolve 42 g of sodium hydroxide R in water R and dilute
Water (2.5.12): maximum 8 per cent, determined on 0.300 g. to 100 mL with the same solvent.
Assay. Dissolve 0.150 g in 50 mL of anhydrous acetic acid R. Sodium 2-hydroxybutyrate. C 4H 7Na03' (Mr 126.1). 1158800.
Titrate with 0.1 M perchloric acid, determining the end-point [19054-57 -O]. Sodium (2RS)- 2-hydroxybutanoate.
potentiometrically (2.2.20).
Sodium hypobromite solution. 1081500.
1 mL of 0.1 M perehloric acid is equivalent to 20.22 mg of In a bath of iced water mix 20 mL of strong sodium hydroxide
C7H¡SNa0 3S. solution R and 500 mL of water R, add 5 mL of bromine
Sodium hexanesulfonate. C6H 13 Na0 3S. (M, 188.2). 1081200. solution R and stir gently until solution is complete. Prepare
[2832-45-3]. immediately before use.
White or almost white powder, freeIy soluble in water. _Sodium hypochlorite solution, strong. 1081600.
Content: 25 giL to 30 giL of active chlorine.
Sodium hexanesulfonate monohydrate. C6H 13 Na0 3S,H 20.
(Mr 206.2). 1161500. [207300-91-2].
Yellowish liquid with an alkaline reaction.
Assay. Introduce into a flask, successively, 50 mL of water R,
White or almost white powder, soluble in water.
1 g of potassium iodide R and 12.5 mL of dilute acetie acid R.
Sodium hexanesulfonate monohydrate for ion-paír Dilute 10.0 mL ofthe substance io be examined to 100.0 mL
chromatography. C6H13Na03S,H20. (Mr 206.2). 1182300. with water R. Introduce 10.0 mL of this solution into the flask
[207300-91-2]. and titrate with 0.1 M sodium thiosulfate, using 1 mL of starch
solution R as indicator.
Content: minimum 99.0 per cent.
1 mL of 0.1 M sodium thiosulfate is equivalent to 3.546 mg
Sodium hydrogen carbonate. 1081300. [144-55-8]. of active chlorine.
See Sodium hydrogen carbonate (0195). Storage: protected from light.

Sodium hydrogen carbonate solution. 1081301. Sodium hypophosphite. NaH 2 P0 2,Hp. (M, 106.0). 1081700.
[10039-56-2]. Sodium phosphinate monohydrate.
A 42 giL solution.
White or almost white, crystalline powder or colourless
Sodium hydrogen sulfate. NaHS0 4 • (M, 120.1). 1131900. crystals, hygroscopic, freely soluble in water, soluble in ethanol
[7681-38-1]. Sodium bisulfate. (96 per cent).
Freely soluble in water, very soluble in boiling water. It Storage: in an airtight container.
decomposes in ethanol (96 per cent) into sodium sulfate and Sodium iodide. 1081800. [7681-82-5].
free sulfuric aeid. See Sodium iodide (0196).
mp: about 315 oc.
Sodium laurilsulfaíe. 1081900. [151-21-3].
Sodium hydrogensulfite. NaH0 3S. (Mr 104.1). 1115700. See Sodium laurilsulfate (0098).
[7631-90-5].
Sodium lauryl sulfate. 1081900. [151-21-3].
White or almost white, crystalline powder, freely soluble in
water, sparingly soluble in ethanol (96 per cent). See Sodium laurilsulfate R.
On exposure to air, some sulfur dioxide is lost and the Sodium laurylsulfonate for chromatography. C¡2H2SNa03S,
substance is gradually oxidated to sulfate. (M, 272.4). 1132000. [2386-53-0].
White or almost white powder or crystals, freely soluble in
Sodium hydroxide. 1081400. [1310-73-2]. water.
See Sodium hydroxide (0677). Absorbance A~~rn (2.2.25), determined in water R: about 0.05
2 M Sodium hydroxide. 3009800. at 210 nm; about 0.03 at 220 nm; about 0.02 at 230 nm;
about 0.02 at 500 nm.
Dissolve 84 g of sodium hydroxide R in carbon dioxidelree
water R and dilute to 1000.0 mL with the same solvent. Sodium metabisulfite. 1082000. [7681-57-4].
See Sodium metabisulfite (0849).
Sodium hydroxide solution. 1081401.
Dissolve 20.0 g of sodium hydroxide R in water R and dilute Sodium methanesulfonate. CH 3S0 3 Na. (Mr 118.1). 1082100.
to 100.0 mL with the same solvent. Verify the concentration [2386-57 -4].
by titration with 1 M hydrochloric acid, using methyl orange White or almost white, crystalline powder, hygroscopic.
solution R as indicator, and adjust if necessary to 200 giL. Storage: in an airtight container.

General Notices (1) apply to all monographs and other texts 519
4.1.1. Reagents EUROPEAN PHARMACOPOEIA 8.0

Sodium molybdate. Na 2 Mo0 4 ,2H 2 0. (Mr 242.0). 1082200. Sodium perchlorate. NaCI0 4,H 20. (Mr 140.5). 1083100.
[10102-40-6]. Disodium molybdate dihydrate. [7791-07-3].
White or almost white, crystalline powder or colourless Content: minimum 99.0 per cent ofNaCI0 4 ,Hp.
crystals, freely soluble in water. White Ol" almost white, deliquescent crystals, very soluble in
Sodium naphthoquinonesulfonate. ClOHsNaOsS. (Mr 260.2). water.
1082300. [521-24-4]. Sodium l,2-naphthoquinone-4- Storage: in a well-closed container.
sulfonate.
Sodium periodate. NaI0 4 • (Mr 213.9). 1083200. [7790-28-5].
Yellow or orange-yellow, crystalline powder, freely soluble in Sodium metaperiodate.
water, practically insoluble in ethanol (96 per cent).
Content: minimum 99.0 per cent.
Sodium nitrate. NaN0 3 • (Mr 85.0). 1082400. [7631-99-4]. White or almost white, crystalline powder or crystals, soluble
White or almost white powder or granules or colourless, in water and in mineral acids.
transparent crystals, deliquescent in moist air, freely soluble in
water, slightly soluble in ethanol (96 per cent). Sodium periodate solution. 1083201.
Storage: in an airtight container. Dissolve 1.07 g of sodium periodate R in water R, add
5 mL of di/u te sulfuric acid R and dilute to 100.0 mL with
Sodium nitrite. NaN0 2. (Mr 69.0). 1082500. [7632-00-0]. water R. Use a freshly prepared solution.
Content: minimum 97.0 per cent.
Sodium phosphite pentahydrate. Na 2HP0 3 ,5H 20.
White or almost white, granular powder or a slightly yellow, (M, 216.0). 1132200. [13517-23-2].
crystalline powder, freely soluble in water.
White or almost white, crystalline powder, hygroscopic, freely
Assay. Dissolve 0.100 g in 50 mL of water R. Add 50.0 mL of soluble in water.
0.02 M potassium permanganate and 15 mL of dilute sulfuric
acid R. Add 3 g of potassium iodide R. Titrate with 0.1 M Storage: in an airtight container.
sodium thiosulfate, using 1.0 mL of starch solution R added Sodium picrate solution, alkaHne. 1083300.
towards the end of the titration as indicator.
Mix 20 mL of picric acid so/ution R and 10 mL of a 50 giL
1 mL of 0.02 M potassium permanganate is equivalent to solution of sodium hydroxide R and dilute to 100 mL with
3.450 mg of NaN0 2 • water R.
Sodium nitdte solution. 1082501. Storage: use within 2 days.
A 100 giL solution. Prepare immediately before use. Sodium potassium tartrate. C 4 H 4 KNaü 6 ,4H 2 0. (Mr 282.2).
Sodium nitroprusside. NaJFe( CNMNO) J,2Hp. 1083500. [6381-59-5].
(M, 298.0). 1082600. [13755-38-9]. Sodium Colourless, prismatic crystals, very soluble in water.
pentacyano-nitrosylferrate (nI) dihydrate.
Sodium pyrophosphate. Na4 pp7,10H,o. (Mr 446.1).
Reddish-brown powder or crystals, freely soluble in water,
1083600. [13472-36-1]. Tetrasodium diphosphate
slightly soluble in ethanol (96 per cent).
decahydrate.
Sodium octanesulfonate. CSH17Na03S, (Mr 216.3). 1082700. Colourless, slightly efflorescent crystals, freely soluble in water.
[5324-84-5].
Content: minimum 98.0 per cent. Sodium rhodizonate. C6NaP6' (Mr 214.0). 1122300.
[523-21-7]. [(3,4,5,6-Tetraoxocyclohex-l-en -1,2-
White or almost white, crystalline powder or flakes, freely ylene )dioxy] disodium.
soluble in water, soluble in methanol.
Violet crystals, soluble in water with an orange-yellow colour.
Absorbance (2.2.25): maximum 0.10, determined at 200 nm
and maximum 0.01, determined at 250 nm using a 54 giL Solutions are unstable and must be prepared on the day of use.
solution. Sodium salicylate. 1083700. [54-21-7].
Sodium octanesulfonate monohydrate. CSH17Na03S,H20. See Sodium salicy/ate (0413).
(Mr 234.3). 1176700. [207596-29-0].
Sodium sulfate, anhydrous. 1083800. [7757-82-6].
White or almost white powder.
Ignite at 600 oC to 700 oC anhydrous sodium sulfate complying
Sodium octyl sulfate. CSH 17 Na0 4 S. (Mr 232.3). 1082800. with the requirements prescribed in the monograph on
[142-31-4]. Anhydrous sodium sulfate (0099).
White or almost white, crystalline powder or flakes, freely Loss on drying (2.2.32) : maximum 0.5 per cent, determined by
soluble in water, soluble in methanol. drying in an oven at 130 oc.
Sodium oxalate. C2Nap4' (Mr 134.0). 1082900. [62-76-0]. Sodium sulfate, anhydrous Rl. 1083801.
White or almost white, crystalline powder, soluble in water, Complies with the requirements prescribed for anhydrous
practically insoluble in ethanol (96 per cent). sodium sulfate R with the following maximum contents.
Sodium pentanesulfonate. CSH ll Na0 3S. (Mr 174.2). 1083000. CI: 20 ppm.
[22767-49-3]. Pb: 10 ppm.
White or almost white, crystalline solid, soluble in water. As: 3 ppm.
Sodium pentanesulfonate monohydrate. CSH Il Na0 3S,H 20. Ca: 50 ppm.
(Mr 192.2). 1132100. [207605-40-1]. Fe: 10 ppm.
White or almost white crystalline solid, soluble in water. Mg: 10 ppm.

Sodium pentanesulfonate monohydrate Rl. Sodium sulfate decahydrate. Na2 SÜ 4,10Hp. (M r 322.2).
CSH ll Na0 3S,H 20. (M, 192.2). 1172500. [207605-40-1]. 1132300. [7727-73-3].
Content: minimum 99 per cent of C SH ll Na0 3S,H 2 0. See Sodium sulfate decahydrate (0100).

520 See the information section on general monographs (cover pages)


EUROPEAN PHARMACOPOEIA 8.0 4.1.1. Reagents

Sodium sulfide. Na 2S,9H 20. (M, 240.2). 1083900. Sodium thioglycoUate. C2H 3NaO zS. (M, 114.1). 1084500.
[1313-84-4]. Disodium sulfide nonahydrate. [367-51-1]. Sodium mercaptoacetate.
Colourless, rapidly yellowing crystals, deliquescent, very White or almost white, granular powder or crystals,
soluble in water. hygroscopic, freely soluble in water and in methanol, slightly
soluble in ethanol (96 per cent).
Storage: in an airtight container.
Storage: in an airtight container.
Sodium sulfide solution. 1083901.
Sodium thiosulfate. 1084600. [10102-17-7].
Dissolve 12 g of sodium sulfide R with heating in 45 mL
of a mixture of 10 volumes of water R and 29 volumes of See Sodium thiosulfate (0414).
glycerol (85 per cent) R, allow to cool and dilute to 100 mL Sodium thiosulfate, anhydrous. Na ZSp3" (M, 158.1).
with the same mixture of solvents. 1180700. [7772-98-7]. Disodium thiosulfate.
The solution should be colourless. Content: minimum 98.0 per cent.
Sodium sulfide solution Rl. 1083902. Sodium tungstate. Na2W0 4 ,2Hp. (Mr 329.9). 1084700.
Prepare by one of the following methods. [10213-10-2]. Disodium tungstate dihydrate.
_ Dissolve 5 g of sodium sulfide R in a mixture of 10 mL of White or almost white, crystalline powder or colourless
water R and 30 mL of glycerol R. crystals, freely soluble in water forming a clear solution,
practically insoluble in ethanol (96 per cent).
- Dissolve 5 g of sodium hydroxide R in a mixture of 30 mL
of water R and 90 mL of glyeerol R. Divide the solution Sorbitol. 1084800. [50-70-4].
into 2 equal portions. Saturate 1 portion with hydrogen See Sorbitol (0435).
sulfide R, with cooling. Mix the 2 portions.
Storage: in a well-filled container, protected from light; use Squalane. C30 H 62 . (Mr 422.8). 1084900. [111-01-3].
within 3 months. 2,6,10,15,19,23-Hexamethyltetracosane.
Colourless, oily liquid, freely soluble in fatty oils, slightly
Sodium sulfite. 1084000. [10102-15-5]. soluble in acetone, in ethanol (96 per cent), in glacial acetic
See Sodium sulfite heptahydrate (0776). acid and in methanol.
d~g: 0.811 to 0.813.
Sorlium sulfite, anhydrous. 1084100. [7757-83-7]. n~o: 1.451 to 1.453.
See Anhydrous sodium sulfite (0775).
Stannous chloride. SnCI 2,2Hp. (Mr 225.6). 1085000.
Sorlium tartrate. C 4H 4 NaP6,2Hp. (M,. 230.1). 1084200. [10025-69-1]. Tin dichloride dihydrate.
[6106-24-7]. Disodium (2R,3R) - 2,3-dihydroxybutanedioate Content: minimum 97.0 per cent of SnCl z,2H 20.
dihydrate. Colourless crystals, very soluble in water, freely soluble in
White or almost white crystals or granules, very soluble in ethanol (96 per cent), in glacial acetic acid and in dilute and
water, practicalIy insoluble in ethanol (96 per cent). concentrated hydrochloric acid.
Assay. Dissolve 0.500 g in 15 mL of hydrochlorie acid R in a
Sodium taurodeoxycholate. CZ6H44NNa06S,H20.
ground-glass-stoppered flask. Add 10 mL of water R and 5 mL
(Mr 539.7). 1155600. [110026-03-4]. Sodium
of chloroform R. Titrate rapidly with 0.05 M potassium iodate
2- [( 3, 12-dihydroxy-5-cholan -24-oyl)amino] ethanesulfonate
until the chloroform layer is colourless.
monohydrate. 2-[[(3,5,12)-3,12-Dihydroxy-24-oxocholan-24-
yl] amino] ethanesulfonic acid monosodium salt monohydrate. 1 mL of 0.05 M potassium iodate is equivalent to 22.56 mg of
SnCI 2,2Hp.
Content: minimum 94 per cent of CZ6H44NNa06S,H20.
Stannous chloride solution. 1085001.
Sorlium tetrahydroborate. NaBH 4. (Mr 37.8). 1146900.
Heat 20 g of tin R with 85 mL of hydrochloric acid R until
[16940-66-2]. Sodium borohydride.
no more hydrogen is released. Allow to cool.
Colourless, hygroscopic crystals, freely soluble in water, Storage: over an excess of tin R, protected from airo
soluble in anhydrous ethanol, decomposing at higher
temperature or in the presence of acids or certain metal salts Stannous chloride solution Rl. 1085002.
forming borax and hydrogen. Immediately before use, dilute 1 volume of stannous
Storage: in an airtight container. chloride solution R with 10 volumes of dilute hydroehlorie
aeid R.
Sodium tetrahydroborate redudng solution. 1146901.
Introduce about 100 mL of water R into a 500 mL Stannous chloride solution R2. 1085003.
volumetric flask containing a stirring bar. Add 5.0 g To 8 g of stannous chloride R add 100 mL of a 20 per
of sodium hydroxide R in pellets and 2.5 g of sodium cent V/V solution of hydroehlorie acid R. Shake until
tetrahydroborate R. Stir until complete dissolution, dilute dissolved, heating, if necessary, on a water-bath at
to 500.0 mL with water R and mix. Prepare immediately 50 oc. Pass a current of nitrogen R for 15 mino Prepare
before use. immediately before use.

Sodium tetraphenylborate. NaB(C 6 H s).¡' (Mr 342.2). Stanolone. C19H3002' (Mr 290.4). 1154400. [521-18-6].
1084400. [143-66-8]. 17~- Hydroxy-5a -androstan -3-one.

White or slightly yellowish, bulky powder, freely soluble in White or almost white powder.
water and in acetone. mp: about 180 oc.

Sodium tetraphenylborate solution. 1084401. Standard solution for the micro determination of water.
1147300.
Filter before use if necessary.
Commercially available standard solution for the coulometric
A 10 giL solution. titration of water, containing a certified content of water in a
Storage: use within 1 week. suitable solvent.

General Notices (1) apply to al! monographs and other texts 521
4.1.1. Reagents EUROPEAN PHARMACOPOEIA 8.0

Staphylococcus aureus strain V8 protease, type XVII-E. Stearic add. C¡SH 36 0 2 • (Mr 284.5). 1085200. [57-11-4].
1115800. [66676-43-5]. Octadecanoic acid.
Microbial extracellular proteolytic enzyme. A lyophilised White or almost white powder or flakes, greasy to the touch,
powder containing 500 units to 1000 units per milligram of practically insoluble in water, soluble in hot ethanol (96 per
solido cent).
mp: about 70 oc.
Starch, soluble. 1085100. [9005-84-9].
Stearic acid used in the assay of total fatty acids in Saw palmetto
White or almost white powder. fruit (1848) complies with the following additional test.
Prepare a 20 giL solution in hot water R. The solution is at Assay. Gas chromatography (2.2.28) as prescribed in the
most slightly opalescent and remains fluid on cooling. monograph Saw palmetto fruit (1848).
Content: minimum 98 per eent, ealculated by the
Starch iodate papero 1085101. normalisation proeedure.
Immerse strip s of filter paper in 100 mL of iodide-free
starch solution R containing 0.1 g of potassium iodate R. Stearyl alcohol. C¡SHJSO. (Mr 270.5). 1156400. [112-92-5].
Drain and allow to dry protected from light. Octadeean -1-01.
mp: about 60 oc.
Starch iodide papero 1085106. Content: mínimum 95 per eent.
Immerse strips of filter paper in 100 mL of starch solution R
eontaining 0.5 g of potassium iodide R. Drain and allow Stigmasterol. C29 H 4p· (Mr 412.7). 1141400. [83-48-7].
to dry proteeted from light. (22E)-Stigmasta-5,22-dien-3~-01. (22E)-24-Ethylcholesta-
5,22-dien-3~-01.
Test for sensitivity. Mix 0.05 mL of 0.1 M sodium nitrite
White or almost white powder, insoluble in water.
with 4 mL of hydrochloric acid R and dilute to 100 mL with
water R. Apply one drop of the solution to stareh iodide mp: about 170 oc.
paper; a blue spot appears. [a]g: about - 51, determined with a 20 giL solution in
chloroform R.
Starch solution. 1085103.
Triturate 1.0 g of soluble starch R with 5 mL of water R and Streptomydn sulfate. 1085300. [3810-74-0].
whilst stirring pour the mixture into 100 mL ofboiling See Streptomycin sulfate (0053).
water R containing 10 mg of mercuric iodide R.
Strongly addic ion-exchange resi.n. 1085400.
Carry out the test for sensitivity eaeh time the reagent is
used. See ion-exchange resin, strongly acidic R.

Test for sensitivity. To a mixture of 1 mL of the starch Strontium carbonate. SrC0 3 . (M, 147.6). 1122700.
solution and 20 mL of water R, add about 50 mg of [1633-05-2].
potassium iodide R and 0.05 mL of iodine solution Rl. The White or almost white, erystalline powder.
solution is blue.
Content: minimum 99.5 per cent.
Starch solution, iodide- free. 1085104.
Strontium chloride hexahydrate. SrCI 2 ,6H 2 0. (Mr 266.6).
Prepare the solution as preseribed for starch solution R 1167000. [10025-70-4].
omitting the mercuric iodide. Prepare immediately befare White or almost white crystals, very soluble in water.
use.
mp: about 115 oC (los5 ofwater) and 872 oc.
Starch solution Rl. 1085105.
Stnmtium selective extraction resino 1167100.
Mix 1 g of soluble starch R and a small amount of eold Commercially available resin prepared by loading a suspension
water R. Add this mixture, while stirring, to 200 mL of of 4,4' (5')-di -tert-butylcyclohexano-18-crown -6 (crown ether)
boiling water R. Add 0.25 g of salicylic acid R and boil for in octanol onto an inert ehromatographic support. The bed
3 mino Immediately remove from the heat and eool. density of this resin is approximately 0.35 g/mL.
Storage: long storage is required, the solution shall be
sto red at 4 oC to 10 oc. A fresh starch solution shall be Strontium-85 spiking solution. 1166800.
prepared when the end-point of the titration from blue to Dilute strontium-85 standard solution R to a radioaetivity
eolourless fails to be sharp. If stored under refrigeration, coneentration of approximately 10 kBq/mL with a 0.27 giL
the starch solution is stable for about 2 to 3 weeks. solution of strontium chloride hexahydrate R in a 1.03 giL
Test for sensitivity. A mixture of 2 mL of starch solution Rl, solution of hydrochloric acid R.
20 mL of water R, about 50 mg of potassium iodide R and Strontium-85 standard solution. 1166900.
0.05 mL of iodine solution R1 is blue.
A solution of strontium-85 in the form of Sr 2 + ions in a 51.5 giL
Starch solution R2. 1085107. solution of hydrochloric acid R.
Triturate 1.0 g of soluble starch R with 5 mL of water R and Styrene. CsH s' (Mr 104.2). 1151700. [100-42-5].
whilst stirring pour the mixture into 100 mL of boiling Ethenylbenzene.
water R. Use a freshly prepared solution. bp: about 145 oc.
Test for sensitivity. To a mixture of 1 mL of the starch Colourless, oily liquid, very slightly soluble in water.
solution and 20 mL of water R, add about 50 mg of
potassium iodide R and 0.05 mL of iodine solution R1. The Styrene-divinylbenzene copolymer. 1085500.
solution is blue. Porous, rigid, eross-linked polymer beads. Several grades are
available with different sizes of beads. The size range of the
Stavudine. 1187000. [3056-17-5]. beads is speeified after the name of the reagent in the tests
See Stavudine (2130). where it is used.

522 See the information section on general monographs (caver pages)


EUROPEAN PHARMACOPOEIA 8.0 4.1.1. Reagents

Succinic add. C 4 H 60 4. (Mr 118.1). 1085600. [110-15-6]. Sulfanilic add solution, diazotised. 1086202.
Butanedioic acid. Dissolve, with warming, 0.9 g of sulfanilic acid R in 9 mL
White or almost white, crystalline powder or colourless of hydrochloric acid R, and dilute to 100 mL with water R.
crystals, soluble in water and in ethanol (96 per cent). CoollO mL of this solution in iced water and add 10 mL
mp: 184 oC to 187 oc. of an ice-cold 45 giL solution of sodium nitrite R. Allow to
stand at O oC for 15 min (if stored at this temperature, the
Sucrose. 1085700. [57-50-1]. solution is stable for 3 days) and immediately before use
See Sucrose (0204). add 20 mL of a 100 giL solution of sodium carbonate R.
Sudan orange. C 16H 12 N l O. (Mr 248.3).1110700. [842-07-9]. Sulfomolybdi.c reagent R2. 1086400.
Colour Index No. 12055. Dissolve about 50 mg of ammonium molybdate R in 10 mL
1-(Phenylazo)naphthalen-2-ol. Sudan 1. of sulfuric acid R.
Orange-red powder, practically insoluble in water, soluble in Sulfomolybdic reagent R3. 1086500.
methylene chloride.
Dissolve with heating 2.5 g of ammonium molybdate R in
mp: about 131°C. 20 mL of water R. Dilute 28 mL of sulfuric acid R in 50 mL
Sudan red G. C17H14N20l' (Mr 278.3). 1085800. of water R, then cool. Mix the two solutions and dilute to
100 mL with water R.
Schultz No. 149.
Storage: in a polyethylene container.
Colour Index No. 12150.
Solvent Red 1. 1- [(2-Methoxyphenyl)azo ]naphtalen-2-01. Sulfosalicylic acid. C7H606S,2Hp. (Mr 254.2). 1086600.
Reddish-brown powder, practically insoluble in water. [5965-83-3]. 2-Hydroxy-5-sulfobenzoic acid.
Chromatography. Thin-Iayer chromatography (2.2.27) using White or almost white, crystalline powder or crystals, very
silica gel G R as the coating substance: apply 10 ¡.tL of a 0.1 giL soluble in water and in ethanol (96 per cent).
solution in methylene chloride R and develop over a path of mp: about 109 oc.
10 cm with the same solvent; the chromatogram shows only
one principal spot. Sulfuro 1110800. [7704-34-9].
See Sulfur for external use (0953).
Sulfanilamide. C 6H sNP l S. (Mr 172.2).1086100. [63-74-1].
4-Aminobenzenesulfonamide. Sulfur dioxide. SOl' (Mr 64.1). 1086700. [7446-09-5].
White or almost white powder, slightly soluble in water, freely SuIfurous anhydride.
soluble in boiling water, in acetone, in dilute acids and in A colourless gas. When compressed it is a colourless liquido
solutions of the alkali hydroxides, sparingly soluble in ethanol
(96 per cent) and in light petroleum. Sulfur dioxide Rl. SOl' (Mr 64.1). 1110900. [7446-09-5].
mp: about 165 oc. Content: minimum 99.9 per cent VIV.

Sulfathiazole. C9H9NPlSl' (Mr 255.3). 1086300. [72-14-0]. Sulfuric add. H 2 S0 4 • (Mr 98.1). 1086800. [7664-93-9].
4-Amino- N-( thiazol-2-yl)benzenesulfonamide. Content: 95.0 per cent mlm to 97.0 per cent mlm.
White or yellowish-white powder or crystals, very slightly Colourless, caustic liquid with an oily consistency, highly
soluble in water, soluble in acetone, slightly soluble in ethanolhygroscopic, miscible with water and with ethanol (96 per
(96 per cent). It dissolves in dilute mineral acids and in cent) producing intense heat.
solutions of alkali hydroxides and carbonates. d~g: 1.834 to 1.837.
mp: about 200 oc. A 10 giL solution is strongly acid and gives the reactions of
sulfates (2.3.1).
Sulfamic add. H 3N0 3S. (Mr 97.1).1085900. [5329-14-6].
Appearance. It is dear (2.2.1) and colourless (2.2.2, Method II).
White or almost white crystalline powder or crystals, freely
soluble in water, sparingly soluble in acetone, in ethanol Oxidisable substances. Pour 20 g cautiously, with cooling,
(96 per cent) and in methanol. into 40 mL of water R. Add 0.5 mL of 0.002 M potassium
permanganate. The violet colour persists for at least 5 mino
mp: about 205 oC, with decomposition.
Chlorides: maximum 0.5 ppm.
Sulfan bIue. C27H31NlNa06S2' (Mr 566.6). 1086000. Pour 10 g, carefully and while cooling, into 10 mL of water R
[129-17-9]. and after cooling dilute to 20 mL with the same solvent.
Schultz No. 769. Add 0.5 mL of silver nitrate solution R2. Allow to stand for
Colour Index No. 42045. 2 min protected from bright light. The solution is not more
Acid BIue 1. Patent BIue VE Disulfine blue. BIue VS. opalescent than a standard prepared at the same time using a
Sodium [[ [( 4-diethylamino )phenyl] (2,4-disulfonatophenyl)- mixture of 1 mL of chloride standard solution (5 ppm el) R,
methylene] cyclohexa -2,5-dien -1-ylidene] diethylammonium. 19 mL of water R and 0.5 mL of silver nitrate solution R2.
Violet powder, soluble in water. Dilute solutions are blue and Nitrates: maximum 0.5 ppm.
turn yellow on the addition of concentrated hydrochloric acid. Pour 50 g or 27.2 mL, carefully and while cooling, into 15 mL
of water R. Add 0.2 mL of a freshly prepared 50 giL solution
Sulfanilic add. C6H 7N0 3S. (Mr 173.2). 1086200. [121-57-3].
of brucine R in glacial acetic acid R. After 5 min any colour
4-Aminobenzenesulfonic acid. is less intense than that of a reference mixture prepared in
Colourless crystals, sparingly soluble in water, practically the same manner and containing 12.5 mL of water R, 50 g
insoluble in ethanol (96 per cent). of nitrogen-free sulfuric acid R, 2.5 mL of nitrate standard
solution (lO ppm NO) R and 0.2 mL of a 50 giL solution of
Sulfanilic add solution. 1086203.
brucine R in glacial acetic acid R.
Dissolve 0.33 g of sulfanilic acid R in 75 mL of water R
heating gently if necessary and dilute to 100 mL with glacial Ammonium: maximum 2 ppm.
acetic acid R. Pour 2.5 g, carefully and while cooling, into water R and dilute
to 20 mL with the same solvento Cool, and add dropwise
Sulfanilk acid solution RL 1086201. 10 mL of a 200 giL solution of sodium hydroxide R, followed by
Dissolve 0.5 g of sulfanilic acid R in a mixture of 75 mL of 1 mL of alkaline potassium tetraiodomercurate solution R. The
dilute acetic acid R and 75 mL of water R. colour of the solution is less intense than that of a mixture of

General Notices (1) apply to all monographs and other texts 523
4.1.1. Reagents EUROPEAN PHARMACOPOEIA 8.0

5 mL of ammonium standard solution (1 ppm NHJ R, 15 mL Hg: 0.005 ppm.


of water R, 10 mL of a 200 giL solution of sodium hydroxide R Ni: 0.002 ppm.
and 1 mL of alkaline potassium tetraiodomercurate solution R.
Pb: 0.001 ppm.
Arsenic (2.4.2, Method A): maximum 0.02 ppm.
Zn: 0.005 ppm.
To 50 g add 3 mL of nitric acid R and evaporate carefully
until the volume is reduced to about 10 mL. Coo!, add to the Sulfuric acid, nitro gen-free. 1086806.
residue 20 mL of water R and concentrate to 5 mL. Prepare Complies with the requirements prescribed for sulfuric
the standard using l.0 mL of arsenic standard solution (1 ppm acid R with the following additional test.
As) R. Ni tra tes. To 5 mL of water R add carefully 45 mL of the
Iron (2.4.9): maximum 1 ppm. sulfuric acid, allow to cool to 40 oC and add 8 mg of
Dissolve the residue on ignition with slight heating in 1 mL of diphenylbenzidine R. The solution is faint pink or very pale
dilute hydrochloric acid R and dilute to 50.0 mL with water R. blue.
Dilute 5 mL of this solution to 10 mL with water R.
Sulfuric add, nitrogen-free Rl. 1086808.
Heavy metals (2.4.8): maximum 2 ppm.
Complies with the requirements prescribed for nitrogen-jree
Dilute 10 mL of the solution obtained in the test for iron to sulfuric acid R.
20 mL with water R. 12 mL of the solution complíes with
Content: 95.0 per cent mlm to 95.5 per cent mlm.
test A. Prepare the reference solution using lead standard
solution (2 ppm Pb) R. Sunflower oil. 1086900.
Residue on ignition: maximum 0.001 per cent, determined See Sunflower oil, rejined (1371).
on 100 g by evaporating cautiously in a small crucible over a
naked flame and igniting the residue to redness. Swertiamarin. C16H2201O' (M, 374.3). 1163600.
Assay. Weigh accurately a ground-glass-stoppered flask [17388-39-5]. Swertiamaroside. (4R,5R,6S)-5- Ethenyl-6-(~- D-
containing 30 mL of water R, introduce 0.8 mL of the sulfuric glucopyranosyloxy)-4a-hydroxy-4,4a,5,6-tetrahydro-lH,3H-
acid, cool and weigh again. Titrate with 1 M sodium hydroxide, pyrano [3,4-c ]pyran -l-one.
using 0.1 mL of methyl red solution R as indicator. Tagatose. C6H1206' (M, 180.16). 1111000. [87-81-0].
1 mL of 1 M sodium hydroxide is equivalent to 49.04 mg of D-lyxo- Hexulose.
H 2 S0 4 • White or almost white powder.
Storage: in a ground-glass-stoppered container made of glass [al~o: - 2.3 determined on a 21.9 giL solution.
or other inert material.
mp: 134 oC to 135 oc.
Sulfuric add, akoholic, 2.5 M. 1086801.
Tale. 1087000. [14807-96-6].
Carefully and with constant cooling, stir 14 mL of sulfuric
See Tale (0438).
acid R into 60 mL of anhydrous ethanol R. AlIow to cool
and dilute to 100 mL with anhydrous ethanol R. Prepare Tannk add. 1087100. [1401-55-4].
immediately before use. Yellowish or light-brown, glistening scales or amorphous
Sulfuric add, akoholic, 0.25 M. 1086802. powder, very soluble in water, freely soluble in ethanol (96 per
cent), soluble in acetone.
Dilute 10 mL of 2.5 M alcoholic sulfuric acid R to 100 mL
with anhydrous ethanol R. Prepare immediately before use. Storage: protected from light.

Sulfuric add, alcoholic solution of. 1086803. Tartaric acid. 1087200. [87-69-4].
Carefully and with constant cooling, stir 20 mL of sulfurie See Tartaric acid (0460).
acid R into 60 mL of ethanol (96 per cent) R. Allow to cool TaxifoHn. C 1S H¡P7' (Mr 304.3). 1151800. [480-18-2].
and dilute to 100 mL with ethanol (96 per cent) R. Prepare (2R,3R) - 2-(3,4-Dihydroxyphenyl) -3,5,7 -trihydroxy- 2,3-
immediately before use. dihydro-4H-1-benzopyran-4-one.
Sulfuric acid, dilute. 1086804. White or almost white powder, slightly soluble in anhydrous
Contains 98 giL of H 2S0 4• ethanol.
Add 5.5 mL of sulfurie aeid R to 60 mL of water R, allow to Absorbance (2.2.25). A solution in anhydrous ethanol R shows
cool and dilute to 100 mL with the same solvent. an absorption maximum at 290 nm.
Assay. 1nto a ground-glass-stoppered flask containing Tecnazene. C 6 HC14 N0 2 • (Mr 260.9). 1132400. [117-18-0].
30 mL of water R, introduce 10.0 mL of the dilute sulfuric bp: about 304 oc.
acid. Titrate with 1 M sodium hydroxide, using 0.1 mL of
mp: 99 oC to 100 oc.
methyl red solution R as indicator.
A suitable certified reference solution (10 ng/¡.tL in
1 mL of 1 M sodium hydroxide is equivalent to 49.04 mg
cyclohexane) may be used.
ofH 2 S0 4 •
a-Terpinene. ClOHl6' (Mr 136.2). 1140300. [99-86-5].
Sulfuric add-formaldehyde reagent. 1086805.
1- Isopropyl-4-methylcyclohexa -1 ,3-diene.
Mix 2 mL ofjormaldehyde so/ution R with 100 mL of
Clear, almost colourless liquido
sulfuric acid R.
d~o: about 0.837.
Sulfuric add, heavy metal-free. 1086807. nto: about 1.478.
Complies with the requirements prescribed for sulfurie bp: about 174 oc.
aeid R with the following maximum contents of heavy
(X- Terpinene used in gas chromatography camplies with the
metals.
jollowing additional test.
As: 0.005 ppm.
Assay. Gas chromatography (2.2.28) as prescribed in the
Cd: 0.002 ppm. monograph Tea tree oil (1837).
Cu: 0.001 ppm. Content: minimum 90 per cent, calculated by the
Fe: 0.05 ppm. normalisation procedure.

524 See the information section on monographs (ca ver pages)


EUROPEAN PHARMACOPOEIA 8.0 4.1.1. Reagents

y- Terpinene. ClQH16' (M, 136.2). 1115900. [99-85-4]. 1,3,4,6-Tetra-O-acetyl- p- D-mannopyranose. C¡4H20010'


1-Isopropyl-4-methylcyclohexa -1 ,4-diene. (M, 348.3).1174100. [18968-05-3].
Oily liquido Colourless or white powder or crystals.
y- Terpinene used in gas chromatography complies with the mp: 160 oC to 161°C.
following additional test. [al~o: - 68, determined on a 7 giL solution in methylene
Assay. Gas chromatography (2.2.28) as prescribed in the ehloride R.
monograph Peppermint oil (0405).
Test solution. The substance to be examined. Tetrabutylammonium bromide. C16H36BrN. (M, 322.4).
1087500. [1643-19-2].
Content: minimum 93.0 per cent, calculated by the
normalisation procedure. White or almost white crystals.
mp: 102 oC to 104 oc.
Terpinen-4-ol. ClQH1SO. (M, 154.2). 1116000.
[562-74-3]. 4-Methyl-1-( 1-methylethyl)cyclohex-3-en-1-ol. Tetrabutylammonium dihydrogen phosphate. C¡6HJSN04P.
p- Menth -l-en -4-01. (M, 339.5). 1087600. [5574-97-0].
Oily, colourless liquido White or almost white powder, hygroscopic.
Terpinen-4-01 used in gas chromatography complies with the pH (2.2.3): about 7.5 for a 170 giL solution.
following additional test. Absorbance (2.2.25): about 0.10 determined at 210 nm using a
Assay. Gas chromatography (2.2.28) as prescribed in the 170 giL solution.
monograph Lavender oil (1338). Storage: in an airtight container.
Test solution. The substance to be examined.
Tetrabutylammoni.um hydrogen sulfate. C 16 H J7 NO¡S.
Content: minimum 90.0 per cent, calculated by the (M, 339.5). 1087700. [32503-27-8].
normalisation procedure.
Crystalline powder or colourless crystals, freely soluble in
a- Terpineol. ClOH1SO. (M,. 154.2). 1087300. [98-55-5]. water and in methanol.
(RS) -2-( 4-Methylcyclohex -3 -enyl) -2-propanol. mp: 169 oC to 173 oc.
Colourless crystals, practically insoluble in water, soluble in Absorbanee (2.2.25) : maximum 0.05, determined between
ethanol (96 per cent). 240 nm and 300 nm using a 50 giL solution.
d~g: about 0.935.
Tetrabutylammonium hydrogen sulfate Rl. 1087701.
n~o: about 1.483.
Complies with the requirements prescribed for
[al~o: about 92.5.
tetrabutylammonium hydrogen sulfate R with the following
mp: about 35 oc. additional requirement.
It may contain 1 to 3 per cent of ~-terpineol. Absorbance (2.2.25) : maximum 0.02, determined between
a- Terpineol used in gas chromatography complies with the 215 nm and 300 nm using a 50 giL solution.
following test.
Tetrabutylammonium hydroxide. C J6 H 37NO,30Hp.
Assay. Gas chromatography (2.2.28) as prescribed in the (M, 800). 1087800. [2052-49-5].
monograph Anise oil (0804).
Content: minimum 98.0 per cent of C16HJ7NO,30H20.
Test solutíon. A 100 giL solution in hexane R.
White or almost white crystals, soluble in water.
Content: minimum 97.0 per cent, calculated by the
normalisation procedure. Assay. Dissolve 1.000 g in 100 mL of water R. Titrate
immediately with 0.1 M hydroehloric acid determining the
Terpinolene. ClOHI6' (M, 136.2). 1140400. [586-62-9]. end-point potentiometrically (2.2.20). Carry out a blank
p- Mentha-1,4(8)-diene. 4-Isopropylidene-l- titration.
methylcyclohexene. 1 mL of 0.1 M hydrochlorie aeid is equivalent to 80.0 mg
Clear, almost colourless liquid. C16H37NO,30HzO.
d~o: about 0.863.
Tetrabutylammonium hydroxide solutlon (104 giL).
n~o : about 1.488. 1087801.
bp: about 184 oc. A solution containing 104 giL of C¡6H37NO (M, 259.5),
Terpinolene used in gas ehromatography complies with the prepared by dilution of a suitable reagent grade.
following additional test.
Tetrabutylarnrnonium hydroxide solution (400 gIL).
Assay. Gas chromatography (2.2.28) as prescribed in the
1087802.
monograph Tea tree oil (1837).
A solution containing 400 giL of C¡6H37NO (M, 259.5) of
Content: minimum 90 per cent, calculated by the
a suitable grade.
normalisation procedure.
Tetrabutylarnmoniurn iodide. C16HJ6IN. (M, 369.4).
Testosterone. 1116100. [58-22-0].
1087900. [311-28-4].
See Testosterone (1373).
Content: minimum 98.0 per cent.
Tesiosterone propionate. 1087400. [57-85-2]. White or slightly coloured, crystalline powder or crystals,
See Testosterone propionate (0297). soluble in ethanol (96 per cent).
Sulfated ash (2.4.14): maximum 0.02 per cent.
1,2,3,4- Tetra-O-acetyl-~-D-glucopyranose. C14H2001O'
Assay. Dissolve l.200 g in 30 mL of water R. Add 50.0 mL of
(M, 348.3). 1172600. [13100-46-4].
0.1 M silver nitrate and 5 mL of dilute nítric acid R. Titrate the
White or almost white powder, soluble in water with gentle excess of silver nitrate with 0.1 M ammonium thioeyanate,
heating. using 2 mL oÍ ferric ammonium sulfate solution R2 as indicator.
[a 1~o : + 11, determined on a 6 giL solution in ehloroform R. 1 mL of 0.1 M sílver nitrate is equivalent to 36.94 mg of
mp: 126 oC to 128 oc. C¡6HJ6 IN .

General Notices (1) apply to all monographs and other texts 525
4.1.1. Reagents EUROPEAN PHARMACOPOEIA 8.0

Tetrachlofoethane. C 2H zCI 4• (M, 167.9). 1088000. [79-34-5]. Tetrahexylammonium hydrogen sulfate. C24Hs3N04S.
1,1,2,2-Tetrachloroethane. (Mr 451.8). 1116300. [32503-34-7]. N,N,N- Trihexylhexan-
CIear, colourless liquid, slightly soluble in water, miscible with l-aminium hydrogen sulfate.
ethanol (96 per cent). White or almost white crystals.
d~g: about 1.59. mp: 100 oC to 102 oc.
n~o: about 1.495.
Tetrahydrofuran. C4HgÜ. (M, 72.1). 1088500. [109-99-9].
Distillation range (2.2.11). Not less than 95 per cent distils Tetramethylene oxide.
between 145 oC and 147 oc.
Clear, colourless, flammable liquid, miscible with water, with
Tetrachlorvinphos. ClOH9ClP4P, (M, 366.0). 1132500. ethanol (96 per cent).
[22248-79-9]. d~g: about 0.89.
mp: about 95 oc. Do not distil if the tetrahydrofuran does not comply with the
A suitable certified reference solution (lO ng/flL in iso-octane) test for peroxides.
may be used. Peroxides. Place 8 mL of potassium iodide and starch solution R
Tetracos-15-enoic acid methyl estero C2sH4S02' (M, 380.7). in a 12 mL ground-glass-stoppered cylinder about 1.5 cm in
1144800. [2733-88-2]. 15-Tetracosaenoic acid methyl estero diameter. Fill completely with the substance to be examined,
Methyl tetracos-15-enoate. Nervonic acid methyl estero shake vigorously and allow to stand protected from light for
30 mino No colour is produced.
Content: minimum 99.0 per cent, determined by gas
chromatography. Tetrahydrofuran used in spectrophotometry complies with the
following additional test.
Liquid.
Minimum transmittance (2.2.25) using water R as
Tetracydine hydrochloride. 1147000. compensation liquid: 20 per cent at 255 nm, 80 per cent at
See Tetracycline hydrochloride (0210). 270 nm, 98 per cent at 310 nm.

Tetradecane. C¡,¡H 30 • (M, 198.4). 1088200. [629-59-4]. Tetrahydrofuran for chromatography R. 1147100.
n-Tetradecane. Complies with the requirements prescribed for
Content: minimum 99.5 per cent mlm. tetrahydrofuran R with the following additional requirements:
A colourless liquido d2~ = 0.8892.
d~g: about 0.76. bp: about 66 oc.
n~o: about 1.429. Content: minimum 99.8 per cent of C4H sO.
bp: about 252 oc.
a- Tetralone. ClOHlOO. (M, 146.2). 1171800. [529-34-0].
mp: about - 5 oc. 1-0xotetraline. 3,4-Dihydronaphthalen -1 (2H) -one.
Tetradecylammonium bromide. C 4o H s4 BrN. (M, 659). bp: about 115 oc.
1088300. [14937-42-9]. Tetrakis(decyl)ammonium bromide. mp: about 5 oc.
White or slightly coloured, crystalline powder or crystals.
mp: 88 oC to 89 oc. Tetramethylammonium bromide. C4H¡2BrN. (M, 154.l).
1156600. [64-20-0]. N,N,N- Trimethylmethanaminium
Tetraethylammonium hydrogen sulfate. CSH 2 ¡N04S. bromide.
(M, 227.3). 1116200. [16873-13-5]. White OI slightly yellow crystals, freely soluble in water.
Hygroscopic powder. mp: about 285 oC, with decomposition.
mp: about 245 oc.
Tetramethylammonium chloride. C4H 12 CIN. (Mr 109.6).
Tetraethylammonium hydroxide solution. C SH 2 ¡NO. 1100400. [75-57-0].
(M, 147.3). 1100300. [77-98-5]. Colourless crystals, soluble in water and in ethanol (96 per
A 200 giL solution. cent).
Colourless liquid, strongly alkaline. mp: about 300 oC, with decomposition.
d~g: about 1.01.
Tetramethylammonium hydrogen sulfate. C 4H 13 N0 4S.
n~o: about 1.372.
(M, 171.2). 1116400. [80526-82-5].
HPLCgrade.
Hygroscopic powder.
Tetraethylene pentamine. CSH 23 N s' (M, 189.3). 1102000. mp: about 295 oc.
[112-57 -2]. 3,6,9-Triazaundecan-l,ll-diamine.
Colourless liquid, soluble in acetone. Tetramethylammonium hydroxíde. C4H13NO,5H20.
(M, 181.2). 1122800. [10424-65-4]. Tetramethylammonium
n~o : about1.506.
hydroxide pentahydrate.
Storage: protected from humidity and heat. Suitable grade for HPLC.
Tetraheptylammonium bromide. C2s H óo BrN. (M, 490.7).
Tetramethylammonium hydroxide solution. 1088600.
1088400. [4368-51-8].
[75-59-2].
White or slightly coloured, crystalline powder or crystals.
Content: minimum 10.0 per cent mlm of C4H 13 NO. (Mr 91.2).
mp: 89 oC to 91°C.
Clear, colourless or very pale yellow liquid, miscible with
Tetrahexylammonium bromide. C 24 H s2 BrN. (M, 434.6). water and with ethanol (96 per cent).
1152500. [4328-13 -6]. N,N,N- Trihexylhexan -l-aminium Assay. To 1.000 g add 50 mL of water R and titrate with 0.05 M
bromide. sulfuric acid, using 0.1 mL of methyl red solution R as indicator.
White or almost white, crystalline powder, hygroscopic. 1 mL of 0.05 M sulfuric acid is equivalent to 9.12 mg of
mp: about 100 oc. C 4 HlJNO.

526 See the information section on general rnonographs (cover pages)


EUROPEAN PHARMACOPOEIA 8.0 4.1.1. Reagents

Tetramethylammonium hydroxide solution, dilute. Tetrazolium bIue. C4oH32C12Ns02' (M, 728). 1089000.
108860l. [1871-22-3]. 3,3' -(3,3'-Dimethoxy[1,l'-biphenyl]-4,4'-
Dilute 10 mL of tetramethylammonium hydroxide diyl)bis [2,5-diphenyl-2H-tetrazolium] dichloride.
solution R to 100 mL with aldehyde-free alcohol R. Prepare Yellow crystals, slightly soluble in water, freely soluble in
immediately before use. ethanol (96 per cent) and in methanol, practically insoluble
in acetone.
Tetramethylbenzidine. CI6H10N2' (Mr 240.3). 1132600. mp: about 245 oC, with decomposition.
[54827 -17 -7]. 3,3',5,5'-Tetramethylbiphenyl-4,4' -diamine.
Powder, practically insoluble in water, very soluble in Tetrazolium bromide. ClsH16BrNsS. (M, 414.3).
methanol. 1152700. [298-93-1]. 3-(4,5-Dimethylthiazol-2-yl)-2,5-
mp: about 169 oc. diphenyltetrazolium bromide. MTT.
TetrazoHum salto C20HJ7NS06S2' (M, 487.5). 1174200.
1,1,3,3- Tetramethylbutylamine. CSH I9N. (Mr 129.3).
[138169-43-4]. 5-(3-Carboxymethoxyphenyl)-3-( 4,5-
1141500. [107-45-9]. 2-Amino-2,4,4-trimethylpentane.
dimethylthiazol-2-yl) -2 -(4-sulfophenyl) -2H-tetrazolium,
Clear, colourless liquido inner salto MTS.
d§g: about 0.805.
Thallous sulfate. T12S0 4. (M, 504.8). 1089100. [7446-18-6].
nbo: about 1.424. Dithallium sulfate.
bp: about 140 oc. White or almost white, rhomboid prisms, slightly soluble in
water, practically insoluble in ethanol (96 per cent).
Tetramethyldiaminodiphenylmethane. C J 7H22N 2'
(M, 254.4). 1088700. [101-61-1]. 4,4'-Methylenebis-(N,N- Thebaine. C19H21N03' (M, 311.4). 1089200. [115-37-7].
dimethylaniline) . (5R,9R,13S)-4,5-Epoxy-3,6-dimethoxy-9a-methylmorphina-
White or bluish-white crystals or leaflets, practically insoluble 6,8-diene.
in water, slightly soluble in ethanol (96 per cent), soluble in White or pale yellow, crystalline powder, very slightly soluble
mineral acids. in water, soluble in hot anhydrous ethanol and in toluene.
mp: about 90 oc. mp: about 193 oc.
Tetramethyldiaminodiphenylmethane reagent. 1088701. Chromatography (2.2.27). Thin-layer chromatography (2.2.27)
as prescribed in identification test B in the monograph Raw
Solution A. Dissolve 2.5 g of tetramethyldiaminodiphenyl- opium (0777): apply to the plate as a band (20 mm x 3 mm)
methane R in 10 mL of glacial acetic acid R and add 50 mL 20 flL of a 0.5 giL solution; the chromatogram shows an
of water R. orange-red or red principal band with an Rp of about 0.5.
Solution B. Dissolve 5 g of potassium iodide R in 100 mL
of water R. Theobromine. 1138800. [83-67-0].
Solution C. Dissolve 0.30 g of ninhydrin R in 10 mL of See Theobromine (0298).
glacial acetic acid R and add 90 mL of water R. TheophyUine. 1089300. [58-55-9].
Mix solution A, solution B and 1.5 mL of solution C. See Theophylline (0299).
Tetramethylethylenediamine. C6H 16 N 2. (Mr 116.2). 1088800. Thiamazole. C4 H 6N 2S. (M, 114.2). 1089400. [60-56-0].
[110-18-9]. N,N,N;N'- Tetramethylethylenediamine. Methimazole. 1-Methyl-1H-imidazole-2-thiol.
Colourless liquid, miscible with water and with ethanol White 01' almost white, crystalline powder, freely soluble
(96 per cent). in water, soluble in ethanol (96 per cent) and in methylene
d~g: about 0.78. chloride.
nbo: about 1.418. mp: about 145 oc.
bp: about 121 oc. 2-(2-Thienyl)acetk adJ. C6H602S. (M, 142.1). 1089500.
[1918-77-0].
Tetramethylsilane. C4H 11 Si. (Mr 88.2). 1088900. [75-76-3].
TMS. Brown powder.
Clear, colourless liquid, very slightly soluble in water, soluble mp: about 65 oc.
in acetone and in ethanol (96 per cent). Thioacetamide. C 2H sNS. (M, 75.1). 1089600. [62-55-5].
d§g: about 0.64. Crystalline powder or colourless crystals, freely soluble in
nbo: about 1.358. water and in ethanol (96 per cent).
bp: about 26 oc. mp: about 113 oc.
Tetramethylsílane used in nuclear magnetic resonance Thioacetamide reagent. 1089601.
spectrometry complíes with the following additíonal test.
To 0.2 mL of thioacetamide solution R add 1 rnL of a
In the nuclear magnetic resonance spectrum of mixture of 5 mL of water R, 15 mL of 1 M sodium hydroxide
an approximately 10 per cent V/V solution of the and 20 mL of glycerol (85 per cent) R. Heat in a water-bath
tetramethylsilane in deuterated chloroform R, the intensity of for 20 S. Prepare immediately before use.
any foreign signa!, excluding those due to spinning side bands
and to chloroform, is not greater than the intensity of the C-13 Thioacetamide solution. 1089602.
satellite signals located at a distan ce of 59.1 Hz on each side of A 40 giL solution.
the principal signal of tetramethylsilane.
Thiobarbituric adJ. C4H4NP2S, (M, 144.2). 1111200.
Tetrandrine. C3s H 42 NP6' (M, 623). 1178500. [518-34-3]. [504-17 -6]. 4,6-Dihydroxy-2-sulfanylpyrimidine.
Tetrapropylammonium chloride. C11 H 2S CIN. (Mr 221.8). Thiodiethylene glycoL C4H IO 0 2S. (M, 122.2). 1122900.
1151900. [5810-42-4]. [111-48-8]. Di(2-hydroxyethyl) sulfide.
White or almost white, crystalline powder, sparingly soluble Colourless or yellow, viscous liquido
in water. Content: minimum 99.0 per cent.
mp: about 241 oc. d~g: about 1.18.

General Notices (1) apply lo all monographs and other texts 527
4.1.1. Reagents EUROPEAN PHARMACOPOEIA 8.0

ThioglycoHk acid. C2 H 4 0 2S. (Mr 92.1). 1089700. [68-11-1]. Thymine. CSH 6N 20 2 . (Mr 126.1). 1090400. [65-71-4].
2-Mercaptoacetic acid. 5-Methylpyrimidine-2,4( lH,3H)-dione.
Colourless liquid, miscible with water, soluble in ethanol Short needles or plates, slightly soluble in cold water, soluble
(96 per cent). in hot water. It dissolves in dilute solution of alkali hydroxides.

Thiomalic add. C 4 H 6 0 4 S. (Mr 150.2). 1161600. [70-49-5]. Thymol. 1090500. [89-83-8]. See Thymol (0791).
(2RS)-2-Sulfanylbutanedioic acid. Thymol used in gas chromatography complies with the following
mp: 150 oC to 152 oc. additional test.
Assay. Gas chromatography (2.2.28) as prescribed in the
Thimnersal. C9H9HgNa01S, (Mr 404.8). 1089800. monograph Peppermint oil (0405).
[54-64-8]. Sodium mercurothiolate. Sodium Test solution. Dissolve 0.1 g in about 10 mL of acetone R.
2- [( ethylmercurio )thio ]benzoate.
Content: minimum 95.0 per cent, calculated by the
Light, yellowish-white, crystalline powder, very soluble in normalisation procedure.
water, freely soluble in ethanol (96 per cent).
Thymol bIue. C27H3uÜsS, (Mr 466.6). 1090600. [76-61-9].
Thiourea. CH 4N 2 S. (Mr 76.1). 1089900. [62-56-6]. Thymolsulfonphthalein. 4,4' -(3H-2,1- Benzoxathiol-3-
White or almost white, crystalline powder or crystals, soluble ylidene) bis(2-isopropyl- 5-methylphenol) S, S-dioxide.
in water and in ethanol (96 per cent). Brownish-green or greenish-blue, crystalline powder, slightly
mp: about 178 oc. soluble in water, soluble in ethanol (96 per cent) and in dilute
solutions of alkali hydroxides.
Threonine. 1090000. [72-19-5]. Thymol bIue solution. 1090601.
See Threonine (1049). Dissolve 0.1 g of thymol blue R in a mixture of 2.15 mL
of 0.1 M sodium hydroxide and 20 mL of ethanol (96 per
Thrombin, bovine. 1090200. [9002-04-4].
cent) R and dilute to 100 mL with water R.
A preparation of the enzyme, obtained from bovine plasma, Test for sensitivity. To 0.1 mL of the thymol blue solution
that converts fibrinogen into fibrin. add 100 mL of carbon dioxide-free water R and 0.2 mL of
A yellowish-white powder. 0.02 M sodium hydroxide. The solution is bIne. Not more
Storage: at a temperature below O oC. than 0.15 mL of 0.02 M hydrochloric acid is required to
change the colour to yellow.
Thrombin, human. 1090100. [9002-04-4]. Colour change: pH 1.2 (red) to pH 2.8 (yellow); pH 8.0
Dried human thrombin. A preparation of the enzyme which (olive-green) to pH 9.6 (bIue).
converts human fibrinogen into fibrin. It is obtained from
Thymolphthalein. C 2S H 30 0,j' (Mr 430.5), 1090700,
liquid human plasma and may be prepared by precipitation
[125-20-2]. 3,3-Bis( 4-hydroxy-5-isopropyl-2-methylphenyl)-
with suitable salts and organic solvents under controlled
3H-isobenzo-furan -l-one.
conditions of pH, ionic strength and temperature.
White or yellowish-white powder, practically insoluble in
Yellowish-white powder, freely soluble in a 9 giL solution of water, soluble in ethanol (96 per cent) and in dilute solutions
sodium chloride forming a doudy, paje yellow solution. of alkali hydroxides.
Storage: in a sealed, sterile container under nitro gen, protected
from light, at a temperature below 25 oc. Thymolphthalein solution. 1090701.
A 1 giL solution in ethanol (96 per cent) R.
Thrombin solution, human. 1090101. Test for sensitivity. To 0.2 mL of the thymolphthalein
Reconstitute human thrombin R as directed by solution add 100 mL of carbon dioxide-free water R. The
the manufacturer and dilute to 5 IU/mL with solution is colourless, Not more than 0.05 mL of 0.1 M
tris(hydroxymethyl)aminomethane sodium chloride buffer sodium hydroxide is required to change the colour to blue.
solution pH 7.4 R. Colour change: pH 9.3 (colourless) to pH 10.5 (blue).
Thrombin solution, human Rl. 1090102. Tin. Sn, (A r 118.7). 1090800. [7440-31-5].
Reconstitute human thrombin R as directed by the Silvery-white granules, soluble in hydrochloric acid with
manufacturer and dilute to 2.5 IU/mL with phosphate releas e of hydrogen.
buffer solution pH 6.5 R. Arsenic (2.4.2, Method A) : maximum 10 ppm, determined
on 0.1 g.
Thromboplastin. 1090300.
A preparation containing the membrane glycoprotein tissue Titan yellow. ClsH19NsNap6S4' (Mr 696). 1090900.
factor and phospholipid, either purified from animal brain [ 1829-00-1].
(usually rabbit) or human placenta or manufactured using Schultz No. 280.
recombinant DNA technology with added phospholipid. The Colour Index No. 19540.
preparation is formulated for routine use in the prothrombin Thiazol yellow. Disodium 2,2'-[(1-triazene-1,3-diyl)di-4,1-
time test and may contain calcium. phenylene ]bis- [6-methylbenzothiazole-7 -sulfonate].
A yellowish-brown powder, freely soluble in water and in
Thujone. ClOH160. (Mr 152.2), 1116500. [76231-76-0].
ethanol (96 per cent),
4-Methyl-l-( 1-methylethyl)bicyclo [3.1.0]hexan-3-one.
Colourless or almost colourless liquid, practically insoluble Titan yellow papel'. 1090901.
in water, soluble in ethanol (96 per cent) and in many other Immerse strips of filter paper in titan yellow solution R and
organic solvents, leave for a few minutes. Allow to dry at room temperature.
Thymidine. C lO H l4Nps' (Mr 242.2). 1158900. 1-(2-Deoxy-~­ Titan yeHow solution. 1090902.
D-erythro-pentofuranosyl)-5-methylpyrimidine-2,4(lH,3H)- A 0.5 giL solution.
dione. Test for sensitivity. To 0,1 mL of the titan yellow solution
Needles, soluble in water, in hot ethanol (96 per cent) and in add 10 mL of water R, 0.2 mL of magnesium standard
glacial acetic acid. solution (10 ppm Mg) R and 1.0 mL of 1 M sodium

528 See the information section on general monographs (ca ver pages)
EUROPEAN PHARMACOPOEIA 8.0 4.1. L Reagents

hydroxide. A distinct pink colour is visible by comparison spot ofbromocresol green with an Rp value les s than 0.15, the
with a reference solution prepared in a similar manner spot of methyI orange with an Rp value in the range of 0.1 to
omitting the magnesium. 0.25, the spot of methyl red with an Rp value in the range of
0.35 to 0.55 and the spot of Sudan red G with an Rp value in
Titanium. Ti. (A r 47.88). 1091000. [7440-32-6]. the range of 0.75 to 0.98.
Content: minimum 99 per cent.
Metal powder, fine wire (diameter not more than 0.5 mm), TLC silica gel F254 plateo 1116800.
sponge. Complies with the requirements prescribed for TLC silica gel
plate R with the following modification.
mp: about 1668 oc.
It contains a fluorescent indicator having a maximum
Density: about 4.507 g/cm J •
absorbance at 254 nm.
Titanium dioxide. 1117900. [13463-67-7]. Fluorescence suppression. Apply separately to the plate at five
See Titanium dioxide (0150). points increasing volumes (l I1L to 10 I1L for normal TLC
plates and 0.2 I1L to 2 I1L for fine partide size plates) of a
Titaniurn trichloride. TiCl)" (Mr 154.3). 1091200. 1 giL solution of benzoic acid R in a mixture of 15 volumes
[7705-07 -9]. Titanium(III) chloride. of anhydrous ethanol R and 85 volumes of cyclohexane R.
Reddish-violet crystals, deliquescent, soluble in water and in Develop over a pathlength half of the plate height with the
ethanol (96 per cent). same mixture of solvents. After evaporating the solvents
mp: about 440 oc. examine the chromatogram in ultraviolet light at 254 nm. Por
Storage: in an airtight container. normal TLC plates the benzoic acid appears as dark spots on
a fluorescent background approximately in the middle of the
Titanium trichloride solution. 1091201. chromatogram for quantities of 2 flg and greater. Por fine
d~g: about 1.19. particle size plates the benzoic acid appears as dark spots on
A 150 giL solution in hydrochloric acid (100 giL HC!). a fluorescent background approximately in the middle of the
chromatogram for quantities of 0.2 I1g and greater.
Titanium trichloride-sulfuric add reagent. 1091202.
TLC silica gel F 254, silanised plate. 1117200.
Carefully mix 20 mL of titanium trichloride solution R with
It complies with the requirements prescribed for TLC silanised
13 mL of sulfuric acid R. Add sufficient strong hydrogen
silica gel plate R with the following modification.
peroxide solution R to give a yellow colour. Heat uniíl white
fumes are evolved. Allow to cool. Dilute with water R It contains a fluorescent indicator having a maximum
and repeat the evaporation and addition of water R until absorbance at 254 nm.
a colourless solution is obtained. Dilute to 100 mL with TLC silica gel G plateo 1116900.
water R.
Complies with the requirements prescribed for TLC silica gel
TLC aluminium oxide G plateo 1165200. pIate R with the following modification.
Support of metal, glass or plastic, coated with a layer of It contains calcium sulfate hemihydrate as binder.
aluminium oxide (particle size 5-40 11m) containing about
TLC silica gel GF 254 plateo 1117000.
10 per cent of calcium sulfate hemihydrate as a binder.
Complies with the requirements prescribed for TLC si/ica gel
TLC octadecylsilyl silica gel plateo 1148600. pIate R with the following modifications.
Support of glass, metal or plastic coated with a layer of It contains calcium sulfate hemihydrate as binder and a
octadecylsilyl silica gel. The plate may contain an organic fluorescent indicator having a maximum absorbance at
binder. 254 nm.
TLC octadecylsHyl sHica gel F254 pIate R. 1146600. Fluorescence suppression. Complies with the test prescribed
for TLC silica gel F254 plate R.
Support of glass, metal or plastic coated with a layer of
octadecylsilyl silica gel. TLC silica gel pIate for aminopolyether test. 1172700.
It contains a fluorescent indicator having a maximum Immerse a TLC silica gel plate R in iodoplatinate reagent Rl for
absorbance in ultraviolet Iight at 254 nm. 5-10 s. Dry at room temperature for 12 h, protected from light.
Storage: protected from light, in an open container; use within
TLC performance test solution. 1116600.
30 days after preparation.
Prepare a mixture of 1.0 mL of each of the following solutions
and dilute to 10.0 mL with acetone R: a 0.5 giL solution of TLC silica gel plate for chiral separations, octadecylsilyl.
Sudan red G R in toluene R, a 0.5 giL solution of methyl 1137700.
orange R in ethanol R prepared immediately before use, a Support of glass, metal or plastic, coated with a layer of
0.5 giL solution of bromocresol green R in acetone R and a octadecylsilyl silica gel, impregnated with Cu2+ ions and
0.25 giL solution of methyl red R in acetone R. enantiomerically pure hydroxyproline. The plate may contain
an organic binder.
TLC silica gel plateo 1116700.
Support of glass, metal or plastic, coated with a layer of silica TLC silica gel, silanised plateo 1117100.
gel of a suitable thickness and particle size (usually 2 11m to Support of glass, metal or plastic, coated with a ¡ayer of
10 11m for fine particle size [High Performance Thin-Layer silanised silica gel of a suitable thickness and particle size
Chromatography, HPTLC] plates and 5 11m to 40 11m for (usually 2 11m to 10 11m for fine particle size [High Performance
normal TLC plates). If necessary, the particle size is indicated Thin-Layer Chromatography, HPTLC] plates and 5 11m to
after the name of the reagent in the tests where it is used. 40 flm for normal TLC plates). If necessary, the particle size
The plate may contain an organic binder. is indicated after the name of the reagent in the tests where
Chromatographic separation. Apply to the plate an appropriate it is used.
volume (10 I1L for a normal TLC plate and 1 I1L to 2 I1L The plate may contain an organic binder.
for a fine particle size plate) of TLC performance test Chromatographic separation. Introduce 0.1 g each of methyl
solution R. Develop over a pathlength two-thirds of the plate laurate R, methyl myristate R, methyl palmitate R and methyl
height, using a mixture of 20 volumes of methanol R and stearate R into a 250 mL conical flask. Add 40 mL of alcoho/ic
80 volumes of toluene R. The plate is not satisfactory, unless potassium hydroxide solution R and heat under a reflux
the chromatogram shows four clearly separated spots, the condenser on a water-bath for 1 h. Allow to coo1, transfer

General Notices (1) apply to all monographs and other texts 529
4.1.1. Reagents EUROPEAN PHARMACOPOEIA 8.0

the solution to a separating funnel by means of 100 mL of p-Toluenesulfonamide. 1091500. [70-55-3].


water R, acidify (pH 2 to 3) with dilute hydrochloric acid R See toluenesulfonamide R.
and shake with three quantitites each of 10 mL of methylene
chloride R. Dry the combined methylene chloride extracts over Toluenesulfonic add. C7Hs03S,Hp. (M, 190.2). 1091600.
anhydrous sodium sulfate R, filter and evaporate to dryness [6192-52-5]. 4-Methylbenzenesulfonic acid.
on a water-bath. Dissolve the residue in 50 mL of methylene Content: mínimum 87.0 per cent of C7Hs03S,
ehloride R. Examine by thin-Iayer chromatography (2.2.27), White or almost white, crystalline powder or crystals, freely
using TLC silanised si/ica gel plate R. Apply an appropriate soluble in water, soluble in ethanol (96 per cent).
quantity (about 10 flL for normal TLC plates and about 1 flL
to 2 flL for fine particle size plates) of the methylene chloride Toluenesulfonylurea. CSHIONz03S. (Mr 214.2).
solution at each of three separate points. Develop over a 1177000. [1694-06-0]. 4- Methylbenzenesulfonylurea.
pathlength two-thirds of the plate height with a mixture of p- Toluenesulfonylurea. (4-Methylphenyl)sulfonylurea.
10 volumes of glacial acetic acid R, 25 volumes of water R and White or almost white, crystalline powder.
65 volumes of dioxan R. Dry the plate at 120 oC for 30 mino mp: 196 to 198 oc.
Allow to cool, spray with a 35 giL solution of phosphomolybdic
acid R in 2-propanol R and heat at 150 oC until the spots 0- Toluidine. C7H9N. (M r 107.2). 1091700. [95-53-4].

become visible. Treat the plate with ammonia vapour until the 2-Methylaniline.
background is white. The chromatograms show four clearly Pale-yellow liquid becoming reddish-brown on exposure to air
separated, well-defined spots. and light, slightly soluble in water, soluble in ethanol (96 per
cent) and in dilute acids.
«-Tocopherol. 1152300. [10191-41-0].
d~g : about 1.01.
See all-rac-a- Tocopherol (0692).
n~o: about 1.569.
«-Tocopheryl acetate. 1152400. [7695-91-2]. bp: about 200 oc.
See all-rac-a- Tocopheryl acetate (0439). Storage: in an airtight container, protected from light.
a-Tolidine. CI4HJ6NZ' (M r 212.3). 1123000. [119-93-7]. 0- Toluidinehydrochloride. C 7H IO CIN. (M, 143.6).
3,3' -Dimethylbenzidine. 1117300. [636-21-5]. 2-Methylaniline hydrochloride.
Content: mínimum 97.0 per cent. 2-Methylbenzenamine hydrochloride.
Light brownish, crystalline power. Content: minimum 98.0 per cent.
mp: about 130 oc. mp: 215 oC to 217 oc.

a-Tolidine solution. 1123001. p-Toluidine. C7H9N. (Mr 107.2). 1091800. [106-49-0].


Dissolve 0.16 g of o-tolidine R in 30.0 mL of glacial acetic 4-Methylaniline.
acid R, add 1.0 g of potassium iodide R and dilute to Lustrous plates or flakes, slightly soluble in water, freely
500.0 mL with water R. soluble in acetone and in ethanol (96 per cent).
mp: about 44 oc.
Toluene. C7H8' (Mr 92.1). 1091300. [108-88-3].
Methylbenzene. Toluidine bIue. ClsH16ClN3S, (M, 305.8).1091900. [92-31-9].
Clear, colourless, flammable liquid, very slightly soluble in Schultz No. 1041.
water, miscible with ethanol (96 per cent). Colour Index No. 52040.
d§g: 0.865 to 0.870. Toluidine Blue O. 3-Amino-7-dimethylamino-2-
bp: about 11 O oc. methylphenothiazin -5 -ium chloride.
Dark-green powder, soluble in water, slightly soluble in
Toluene, sulfur-free. 1091301. ethanol (96 per cent).
Complies with the requirements prescribed fOl" toluene R
with the following additional requirements. Tosylarginine methyl ester hydrochloride.
C14H23CIN404S, (Mr 378.9). 1092000. [1784-03-8].
Sulfur compounds. To 10 mL add 1 mL of anhydrous N-Tosyl-L-arginine methyl ester hydrochloride. Methyl
ethanol R and 3 mL of potassium plum bite solution R and (S)-5-guanidino-2- (4-methylbenzenesulfonamido )valerate
boil under a reflux condenser for 15 mino Allow to stand hydrochloride.
for 5 mino No darkening is produced in the aqueous layer.
[al~o: - 12 to - 16, determined on a 40 giL solution.
Thiophen-related substances. Shake 2 mL with 5 mL of
isatin reagent R for 5 min and allow to stand for 15 mino mp: about 145 oc. .
No blue colour is produced in the lower layer. Tosylarginine methyl ester hydrochloride solution.
Toluenesulfonamide. C 7H 9N0 2S. (Mr 171.2). 1092001.
1091500. [70-55-3]. 4-Methylbenzenesulfonamide. To 98.5 mg of tosylarginine methyl ester hydrochloride R add
p- Toluenesulfonamide. 5 mL of tris(hydroxymethyl)aminomethane buffer solution
White or almost white, crystalline powder, slightly soluble pH 8.1 R and shake to díssolve. Add 2.5 mL of methyl red
in water, soluble in ethanol (96 per cent) and in solutions of mixed solution R and dilute to 25.0 mL with water R.
alkali hydroxides. Tosyl-lysyl-chloromethane hydrochloride.
mp: about 136 oc. C14HzzC12NP3S, (Mr 369.3). 1092100. [4238-41-9].
Chromatography. Thin-Iayer chromatography (2.2.27) as N-Tosyl-L-Iysyl-chloromethane hydrochloride. (35)-7-Amino-
prescribed in the monograph Tolbutamide (0304); the 1-chloro-3-( 4-methylbenzenesulfonamido )heptan -2-one
chromatogram shows only one principal spot. hydrochloride.
[al~o: - 7 to - 9, determined 011 a 20 giL solution.
0-Toluenesulfonamide. C 7H 9N0 2 S. (M r 171.2). 1091400.
[88-19-7]. 2-Methylbenzenesulfonamide. mp: about 155 oC, with decomposition.
White or almost white, crystalline powder, slightly soluble Ai~m: 310 to 340, determined at 230 nm in water R.
in water, soluble in ethanol (96 per cent) and in solutions of Tosylphenylalanylchloromethane. C I7 H 1S CINO)S.
alkali hydroxides. (M, 351.9). 1092200. [402-71-1]. N-Tosyl-L-
mp: about 156 oc. phenylalanylchloromethane.

530 See the information section on general monographs (cover pages)


EUROPEAN PHARMACOPOEIA 8.0 4.1.1. Reagents

[al~O: - 85 to - 89, determined on a 10 giL solution in ethanol 1, 1, l-Trichlol'Oethane. C 2H 3Cl3" (Mr 133.4). 1092600.
(96 per cent) R. [71-55-6]. Methylchloroform.
mp: about 105 oc. Non-flammable liquid, practically insoluble in water, soluble
Ai~m: 290 to 320, determined at 228.5 nm in ethanol (96 per in acetone and in methanol.
cent) R. d§g : about 1.34.
Toxaphene. 1132800. [8001-35-2]. n~o : about 1.438.

A mixture of polychloro derivatives. bp: about 74°C.


mp: 65 oC to 90 oc. Trichloroethylene. C2HCI 3. (Mr 131.4). 1102100. [79-01-6].
A suitable eertified referenee solution (lO ng/[lL in iso-oetane) Colourless liquid, praetically insoluble in water, miscible with
maybe used. ethanol (96 per cent).
Tragacanth. 1092300. [9000-65-1]. d~g : about 1.46.
See Tragacanth (0532). n~o: about 1.477.

Tanshinone HA' C J9 HJpY (M, 294.3). 1184800. [568-72-9]. Trichlorotrifluoroethane. C 2CI3 F3" (Mr 187.4). 1092700.
1,6,6-Trimethyl-6, 7,8,9-tetrahydrophenanthro [1 ,2-b] furan- [76-13-1]. 1,1,2-Trichloro-1 ,2,2-trifluoroethane.
10,1l-dione. Colourless, volatile liquid, praetically insoluble in water,
miscible with acetone.
Triacetin. C9H1406' (Mr 218.2). 1092400. [102-76-1].
Propane-1,2,3-triyl triaeetate. Glycerol triacetate. d~g: about 1.58.
Almost clear, colourless to yellowish liquid, soluble in water, Distillation range (2.2.11). Not less than 98 per cent distils
miscible with ethanol (96 per eent). between 47 oC and 48 oc.
d~g: about 1.16. Tricine. C6HJ3NOS' (Mr 179.2). 1138900. [5704-04-1].
n~o: about 1.43. N- [2- Hydroxy-1, 1-bis(hydroxymethyl)ethyl] glycine.
bp: about 260 oc. Use eleetrophoresis-grade reagent.
Triamdnolone. C21 H 27 F0 6. (M, 394.4). 1111300. [124-94-7]. mp: about 183 oc.
9-Fluoro-l1 ~,16a, 17,21-tetrahydroxypregna-1,4-diene-3,20-
Tricosane. C23 H 4S ' (M, 324.6). 1092800. [638-67-5].
dione.
White or almost white crystals, praetically insoluble in water,
A erystalline powder.
soluble in hexane.
mp: 262 oC to 263 oc.
mp: about 48 oc.
Triamdnolone acetonide. 1133100. [76-25-5].
Tridocosahexaenoin. C69H9S06' (M, 1023.5). 1144900.
See Triamcinolone acetonide (0533). [124596-98-1]. Triglyceride of docosahexaenoic acid
Tribromophenol. C6H3Br30. (Mr 330.8). 1165300. [118-79-6]. (C22:6). Glycerol tridocosahexaenoate. Propane-1,2,3-triyl
2,4,6-Tribromophenol. tri -( all-Z) -do cosa -4, 7,10,13,16,19-hexaenoate.
The reagent from Nu-Chek Prep, 1ne. has been found suitable.
Tributyl dtrate. C 1s H 3P7' (Mr 360.4). 1152800. [77-94-1].
Tributyl 2-hydroxypropane-1 ,2,3-tricarboxylate. Triethanolamine. 1092900. [102-71-6].
d~o : about l.043. See Trolamine (1577).
n~o : about 1.445.
Triethylamine. C 6H 1S N. (M, 101.2).1093000. [121-44-8].
Tributyl phosphate. C 12H 27 0 4 P. (Mr 266.3). 1179900. N, N- Diethylethanamine.
[126-73-8]. Tributoxyphosphine oxide. Tributoxyphosphane Colourless liquid, slightly soluble in water at a temperature
oxide. below 18.7 oc, miscible with ethanol (96 per cent).
Colourless liquid, slightly soluble in water, soluble in the usual d~g: about 0.727.
organic solvents.
n~o: about 1.401.
d3g: about 0.976. bp: about 90 oc.
n~5 : about 1.422.
bp: about 289 oC, with decompositioll. Triethylamine Rl. C6H 1S N. (Mr 101.2). 1093001.
[121-44-8]. N,N- Diethylethanamine.
Tributylphosphine. C 12 H 27 P. (M, 202.3). 1187100. [998-40-3].
Complies with the requirements prescribed for
Clear, colourless liquido triethylamine R with the following additional requirements.
bp: about 240 oc. Content: minimum 99.5 per eent, determined by gas
mp: about - 60 oc. ehromatography.
Trichlorethylene. 1102100. Water: maximum 0.1 per eent.
See Trichloroethylene R. Use freshly distilled or from a freshly opened container.

Trichloroacetic acid. C2HClP2' (M, 163.4). 1092500. Triethylamine R2. C6 H 1S N. (Mr 101.2). 1093002.
[76-03-9]. [121-44-8]. N,N-Diethylethanamine.
Colourless erystals or a crystalline mass, very deliquescent, Complies with the requirements prescribed for
very soluble in water and in ethanol (96 per cent). triethylamine R and with the following additional
Storage: in an airtight container. requirements.
Content: minimum 99.5 per cent, determined by gas
Trichloroacetic acid solution. 1092501. chromatography.
Dissolve 40.0 g of trichloroacetic acid R in water R and
Water: maximum 0.2 per cent.
dilute to 1000.0 mL with the same solvento Verify the
concentration by titration with 0.1 M sodium hydroxide It is suitable for gradient elution in liquid chromatography.
and adjust if necessary to 40 ± 1 giL. Use freshly distilled or from a freshly opened container.

General Notices (1) apply to all monographs and other texts 531
4. L 1. Reagents EUROPEAN PHARMACOPOEIA 8.0

Triethylenediamine. C 6H J2 N2. (Mr 112.2). 1093100. Absorbance (2.2.25). Not more than 0.07 from 220 nm to
1,4-Diazabicyclo [2.2.2] octane. 360 nm, determined using water R as the compensation
Crystals, very hygroscopic, sublimes readily at room liquido
temperature, freely soluble in water, in acetone and in Trimethylpentane for chromatography. 1093402.
anhydrous ethanol.
Complies with the requirements prescribed for
bp: about 174 oc.
trimethylpentane R with the following additional
mp: about 158 oc. requirement.
Storage: in an airtight container. Residue on evaporation: maximum 2 mg/L.
Triethyl phosphonoformate. C)HlSOSP. (Mr 210.2). 1132900. N,O-bis(TrimethylsilyI)acetamide. CSH 21 NOSi2. (Mr 203.4).
[1474-78-8]. Ethyl( diethoxyphosphoryl)formate. 1093600. [10416-59-8].
Colourless liquido Colourless liquido
bpJ2 mm: about 135 oc. d~g : about 0.83.

Triflumuron. C1s H lO ClF 3NP3' (Mr 358.7). 1180800. N- Trimethylsilylimidazole. C6 H 12 N 2Si. (M, 140.3). 1100500.
[64628-44-0]. 1-(2-Chlorobenzoyl)-3-( 4-triflumoromethoxy- [18156-74-6]. 1-Trimethylsilylimidazole.
phenyl)urea. Colourless, hygroscopic liquido
White or almost white crystalline powder, practically insoluble d~g : about 0.96.
in water, sparingly soluble in acetone and in methylene
nbo: about 1.48.
chloride.
Storage: in an airtight container.
Tl"ifluol"oacetic add. C 2 HF 30 2 • (Mr 114.0). 1093200.
[76-05-1]. N,O-bis(Trimethylsilyl)trifluoroacetamide. CSHlSF3NOSi2'
(Mr 257.4). 1133200. [25561-30-2]. BSTFA.
Content: minimum 99 per cent.
Colourless liquido
Liquid, miscible with acetone and with ethanol (96 per cent).
d~g : about 1.53.
d§g: about 0.97.
nbo : about 1.38.
bp: about 72 oc.
bP12mm: about 40 oC
Use a grade suitable for protein sequencing.
Storage: in an airtight container. Trimethylsulfonium hydroxide. C3H lO OS. (Mr 94.2).
1145000. [17287-03-5].
Tl"ifluoroacetic anhydride. C4F603' (Mr 210.0). 1093300. d~o: abollt 0.81.
[407-25-0].
Colourless liquido Trimethyltin chloride. C3H 9ClSn. (Mr 199.3). 1170900.
[1066-45-1]. Chlorotrimethylstannane.
d~g : about 1.5.
2,4,6-Trinitrobenzene sulfonic add. C6H3NP9S,3H20.
3- Trifluoromethylaniline. C)H6F3N. (Mr 161.1). 1171900.
(Mr 347.2). 1117500. [2508-19-2].
[98-16-8]. 3-(Trifluoromethyl)aniline. a,u,u-Trifluoro-m-
toluidine. 3-(Trifluoromethyl)benzenamide. White oy almost white, crystalline powder, soluble in water.
Colourless liquido mp: 190 oC to 195 oc.
Density: 1.30 g/cm 3 (20 OC). Triolein. C S)H lO P6' (M, 885.4). 1168200. [122-32-7].
Propane-l,2,3-triyl tris[ (9Z)-octadec-9-enoate]. sn-Glyceryl
4-TrifluoromethylphenoL C)HSFp. (Mr 162.1). 1161700.
trioleate. GlyceroJ trioleate. Oleyl triglyceride.
[402-45-9].
Content: minimum 99.0 per cent.
White or light yellow, crystalline solid or powder.
mp: about 46 oc. TriphenylmethanoL C19HIP. (Mr 260.3). 1093700.
[76-84-6]. Triphenylcarbinol.
TrigoneUine hydrochloride. C)H SCIN0 2. (M, 173.6). Colourless crystals, practically insoluble in water, freely
1117400. [6138-41-6]. 3-Carboxy-1-methylpyridinium
soluble in ethanol (96 per cent).
chloride. Nicotinic acid N-methylbetaine hydrochloride.
Crystalline powder, very soluble in water, soluble in ethanoJ Triphenyltetrazolium chloride. C19HlSClN4. (Mr 334.8).
(96 per cent). 1093800. [298-96-4]. 2,3,5-Triphenyl-2H-tetrazolium
mp: about 258 oc. chloride.
Content: minimum 98.0 per cent of C 19 H 1S CIN,¡.
Trimethylpentane. CSH 18 . (Mr 114.2). 1093400. [540-84-1]. Paje or dull-yellow powder, soluble in water, in acetone and in
Iso-octane. 2,2,4-Trimethylpentane. ethanol (96 per cent).
Colourless, flammable liquid, practically insoluble in water, mp: about 240 oC, with decomposition.
soluble in anhydrous ethanol.
Assay. Dissolve l.000 g in a mixture of 5 mL of dilute nitric
d~g: 0.691 to 0.696. acid R and 45 mL of water R. Add 50.0 mL of 0.1 M si/ver
nbo: 1.391 to 1.393. nitrate and heat to boiling. Allow to cool, add 3 mL of
Distillation range (2.2.11). Not less than 95 per cent distils dibutyl phthalate R, shake vigorously and titrate with 0.1 M
between 98 oC and 100 oc. ammonium thiocyanate, using 2 mL ofjerric ammonium
Trimethylpentane used in spectrophotometry complies with the sulfate solution R2 as indicator.
jollowing additional test. 1 mL of 0.1 M si/ver nitrate is equivalent to 33.48 mg of
Minimum transmittance (2.2.25) using water R as C19HlSCIN4'
compensation liquid: 98 per cent from 250 nm to 420 nm. Storage: protected from light.

Trimethylpentane Rl. 1093401. Triphenyltetrazolium chloride solution. 1093801.


Complies with the requirements prescribed for A 5 giL solution in aldehyde-jree alcohol R.
trimethylpentane R with the following modification. Storage: protected from light.

532 See the injormation section on general monographs (cover pages)


EUROPEAN PHARMACOPOEIA 8.0 4.1.1. Reagents

Triscyanoethoxypropane. C 12H 17Npy (M,. 251.3). 1093900. Tyrosine. C9H ll NOy (Mr 181.2). 1094800. [60-18-4].
1,2,3-Tris(2-cyanoethoxy)propane. 2-Amino-3-( 4-hydroxyphenyl)propionic acid.
Viscous, brown-yellow liquid, soluble in methanol. Used as a White or almost white, crystalline powder, or colourless
stationary phase in gas chromatography. or white or almost white crystals, slightly soluble in water,
d~g : about 1.11. practically insoluble in acetone and in anhydrous ethanol,
Viscosity (2.2.9): about 172 mPa·s. soluble in dilute hydrochloric acid and in solutions of alkali
hydroxides.
1,3,5-Tris [3,5-di( 1,1-dimethylethyI)-4-hydroxybenzyl]-
1,3,5-triazine- 2,4,6( lH,3H,5H) - trione. C 4s H 6P 6 N 3" Umbelliferone. C9H60y (M, 162.1). 1137500. [93-35-6].
(M, 784.1). 1094000. [27676-62-6]. 7-Hydroxycoumarin.7-Hydroxy-2H-1-benzopyran-2-one.
White or almost white, crystalline powder. Needles from water.
mp: 218 oC to 222 oc. mp: 225 oC to 228 oc.

Tris[2,4-di(1,1-dimethylethyl)phenyl] phosphite. Uracil. C4 H 4NP2' (M, 112.1).1161800. [66-22-8].


C 42 H 6P 3P. (Mr 647). 1094100. [31570-04-4]. Content: minimum 95.0 per cent.
White or almost white powder.
Urea. 1095000. [57-l3-6].
mp: 182 oC to 186 oc.
See Urea (0743).
Tris(hydroxymethyl)aminomethane. 1094200. [77-86-1].
See Trometamol (1053). Uridine. C 9H 12 NP6' (M, 244.2). 1095100. [58-96-8].
1- ~- D-Ribofuranosyluracil.
Tris(hydroxymethy!)aminomethane solution. 1094201. White or almost white, crystalline powder, soluble in water.
A solution containing the equivalent of24.22 g of C 4H 11 N0 3 mp: about 165 oc.
in 1000.0 mL.
Ursolic acid. C 30 H 4S Oy (M,. 456.7). 1141600. [77-52-l].
Tris(hydroxymethyl)aminomethane solution Rl. 3~- Hydroxyurs-12-en -28-oic acid.
1094202.
White or almost white powder, practically insoluble in water,
Dissolve 60.6 mg of tris(hydroxymethyl)aminomethane R
sparingly soluble in methanol, slightly soluble in ethanol
and 0.234 g of sodium chloride R in water R and dilute to
(96 per cent).
100 mL with the same solvent.
Storage: at 2 oC to 8 oC; use within 3 days. [ct]g: about 67.50, determined on a 10 giL solution in a
56.1 giL solution of potassium hydroxide R in ethanol (96 per
Tripotassium phosphate trihydrate. K 3P0 4 ,3Hp. (Mr 266.3). cent) R.
1155300. [22763-03-7]. mp: 285 oC to 288 oc.
White or almost white crystalline powder, freely soluble in
water. Valencene. C 1s H 24 . (Mr 204.4). 1152100. [4630-07-3].
4~H,5a- Eremophila-l(lO),ll-diene. (lR,7R,8a5)-
Trisodium phosphate dodecahydrate. Na 3P0 4 ,12Hp. 1,8a -Dimethyl-7 -( 1-methylethenyl) -1 ,2,3,5,6, 7,8,8a-
(M, 380.1).1094300. [10101-89-0]. octahydronaphthalene.
Colourless or white or almost white crystals, freely soluble in Oily, colourless or pale yellow liquid, with a characteristic
water. odour, practically insoluble in water, soluble in ethanol (96 per
cent).
Tropic add. C 9H lO Oy (M, 166.17). 1172000. [529-64-6].
(2RS)-3- Hydroxy-2-phenylpropanoic acid. d~o: about 0.918.
n¡;o: about 1.508.
Troxerutin. C33 H 4 P19' (M,. 743). 1160300. [7085-55-4].
Trihydroxyethylrutin. 3',4',7-Tris [O-(2-hydroxyethyl) ]rutin. bp: about 123 oc.
2- [3,4-Bis(2-hydroxyethoxy)phenyl]-3- [[ 6-0-( 6-deoxy -Q - L- Valencene used in gas chromatography complies with the
mannopyranosyl)- ~- D-glucopyranosyl] oxy ]-5-hydroxy-7 -(2- following additional test.
hydroxyethoxy)-4H-l-benzopyran-4-one. Assay. Gas chromatography (2.2.28) as prescribed in the
mp: 168 oC to 176 oc. monograph 5weet orange oil (1811).
Trypsin. 1094500. [9002-07-7]. Content: minimum 80 per cent, calculated by the
normalisation procedure.
A proteolytic enzyme obtained by activation of trypsinogen
extracted from the pancreas ofbeef (Bos taurus L.). Valerenic add. C 1s H 22 0 2. (M, 234.3). 1165700. [3569-10-6].
White or almost white, crystalline or amorphous powder, (2E)- 3- [( 45, 7R, 7aR)- 3, 7-Dimethyl-2,4,5,6, 7, 7a -hexahydro-lH-
sparingly soluble in water. inden -4-yl]- 2-methylprop-2-enoic acid.
mp: l34 oC to 138 oc.
Trypsin for peptide mapping. 1094600. [9002-07-7].
Trypsin of high purity treated to eliminate chymotryptic Valerk add. CSHlO02. (M, 102.1). 1095200. [109-52-4].
activity. Pentanoic acid.
Tryptophan. C ll H 12 NP2' (Mr 204.2). 1094700. [73-22-3]. Colourless liquid, soluble in water, freely soluble in ethanol
(96 per cent).
White or yellowish-white, crystalline powder or colourless
crystals, slightly soluble in water, very slightly soluble in d~g : about 0.94.
ethanol (96 per cent). n¡;o: about 1.409.
[ct]~o: about - 30, determined on a 10 giL solution. bp: about 186 oc.
Tyramine. CSHllNO. (M, l37.2). 1117600. [51-67-2]. Valine. 1185300. [72-18-4].
4- (2-Aminoethyl)phenol. See Valine (0796).
Crystals, sparingly soluble in water, soluble in boiling
anhydrous ethanol. VaniJHn. 1095300. [121-33-5].
mp: 164 oC to 165 oc. See Vanillin (0747).

General Notices (1) apply to all monographs and other texts 533
4.1.1. Reagents EUROPEAN PHARMACOPOEIA 8.0

Vanillin reagent. 109530l. Assay. Gas chromatography (2.2.28): use the normalisation
Carefully add, dropwise, 2 mL of sulfuric acid R to 100 mL procedure.
of a 10 giL solution of vanillin R in ethanol (96 per cent) R. Column:
5torage: use within 48 h. - material: fused -silica;
- size: 1= 30 m, el = 0.5 mm;
VaniHin solution, phosphoric. 1095302.
- stationary phase: macrogol20 000 R.
Dissolve 1.0 g of vanillin R in 25 mL of ethanol (96 per
cent) R. Add 25 mL of water R and 35 mL of phosphoric Carrier gas: helíum for ehromatography R.
acid R. Temperature:

Veratrole. CSHIO02. (Mr 138.2). 1165400. [91-16-7]. Time Temperature


1,2-Dimethoxybenzene. (mill) (OC)
Column O- 1 80
d~o: 1.085.
n~o: 1.534. 1 - 12 80 ." 190

bp: about 206 oc. 12 - 27 190


mp: about 22 oc. Injection port 190

Vel'benone. CIOHI40. (Mr 150.2). 1140500. [1196-01-6]. Deteetíon: flame- ionisation.


(1 S,55)-4,6,6- Trimethylbicyclo [3 .1.1]hept- 3-en -2-one.
Injection: 0.3 ¡.tL of the substance to be examined.
Oil with a characteristic odour, practically insoluble in water,
miscible with organic solvents. Adjust the flow rate of the carrier gas so that the retention
time ofthe peak corresponding to 1-vinylpyrrolidin-2-one
d~g : about 0.978. is about 17 mino
ng: about 1.49.
Vitexin. C21H200JO' (Mr 448.4). 1133300. [3681-93-4].
[a]g: about + 249.6.
Apigenin 8-g1ucoside.
bp: 227 oC to 228 oc.
Yellow powder.
mp: about 6.5 oc.
Storage: in an airtight container, protected from light.
Verbenone used in gas chromatography complies with the
following additional test. Water. 1095500. [7732-18-5].
Assay. Gas chromatography (2.2.28) as prescribed in the See Purified water (0008).
monograph Rosemary oil (1846).
Water RL 1095509.
Content: minimum 99 per cent, calculated by the
normalisation procedure. Prepared from distilled water R by multiple distillation.
Remove carbon dioxide by boiling for at least 15 min before
Vinyl acetate. C 4 H 6 0 2• (Mr 86,10). 1111800. [108-05-4]. use in a boiling flask of fused silica or borosilicate glass and
Ethenyl acetate. cooL Any other suitable method may be llsed. The boiling
d~g : about 0.930. flask has been already used for the test or has been filled
with water R and kept in an autoclave at 121°C for at least
bp: about 72 oc. 1 h prior to first use. When tested immediately before use,
Vinyl chloride. C 2H 3 Cl. (M, 62.5). 1095400. [75-01-4]. water Rl is neutral to methyl red solution R, i.e. it shall
produce an orange-red (l1ot a violet-red or yellow) colour
Colourless gas, slightly soluble in organic solvents. corresponding to pH 5.5 ± 0.1 when 0.05 mL of methyl red
Vinyl polymel' for chromatography, octadecyL 1155400. solution R is added to 50 mL of the water to be examined.
Spherical particles (5 I.uu) of a vinyl alcohol copolymer Conductivíty: maximum 1 ¡.tS·cm- 1, determined at 25 oC by
chemically modified by bonding of octadecyl groups on the an in-line conductivity meter (see Purified water (0008)).
hydroxyl groups. Water, ammonium-free. 1095501.
Vinyl polymer for chromatography, octadecylsilyl. To 100 mL of water R add 0.1 mL of sulfuric acid R. Distil
1121600. using the apparatus described for the determination of
Spherical particles (5 ¡.tm) of a vinyl alcohol copolymer bonded Distillation range (2.2.11). Reject the first 10 mL and collect
to an octadecylsilane. Carbon content of 17 per cent. the following 50 mL.
Water, carbon dioxide-free. 1095502.
2-Vinylpyridine. C7H7N. (Mr 105.1).1102200. [100-69-6].
Water R which has been boiled for a few minutes and
Yellow liquid, miscible in water.
protected from the atmosphere during cooling and storage
d~g: about 0.97. or deionised water R with a resistivity of not less than
n~o: about 1.549. 0.18 MO·m.

4-Vinylpyridine. C7H7N. (Mr 105.l). 1187200. [100-43-6]. Water for chromatography. 1095503.
4- Ethenylpyridine. Deionised water R with a resistivity of not less than
Clear, deep yellowish-brown liquido 0.18 MO·m.
bp: 58-61°C. Water, distiHed. 1095504.
l-Vinylpyrrolidin-2-one. C6H9NO. (Mr 111.1). 1111900. Water R prepared by distillation.
[88-12-0]. 1- Ethenylpyrrolidin-2-one.
Water, distilled, deionised. 1095508.
Content: minimum 99.0 per cent.
Deionised water R prepared by distillation with a resistivity
Clear colourless liquido of not less than 0.18 MO-m.
Water (2.5.12): maximum 0.1 per cent, determined on
2.5 g. Use as the solvent, a mixture of 50 mL of anhydrous Water for injections. 1095505.
methanol R and 10 mL of butyrolactone R. See Water for injections (0169).

534 See the information section on general monographs (caver pages)


EUROPEAN PHARMACOPOElA 8.0 4.LL Reagents

Water, nitrate-free. 1095506. o-Xylene. CsHIO" (Mr 106.2). 1100600. [95-47-6].


To 100 mL of water R add a few milligrams of potassium 1,2-Dimethylbenzene.
permanganate R and of barium hydroxide R. Distil using the Clear, colourless, flammable liquid, practically insoluble in
apparatus described for the determination of Distillation water, miscible with ethanol (96 per cent).
range (2.2.11). Reject the first 10 mL and colleet the d~g: about 0.88l.
following 50 mL. nbo: about 1.505.
Water, partide-free. 1095507. bp: about 144 oc.
Filter water R through a membrane with a pore size oí mp: about - 25 oc.
0.22 fIm. Xylenol orange. C31H2SN2Na40!3S, (Mr 761).
1096300. [3618-43-7]. Tetrasodium 3,3'-(3H-2,1-
Weak cationic resino 1096000.
benzoxathiol-3-ylidene )bis [( 6-hydroxy-S-methyl-3,1-
Polymethacrylic resin, slightly acid, with carboxyl groups phenylene )methyleneiminobisacetate] S,S-dioxide.
present in a protonated formo
Reddish-brown crystalline powder, soluble in water.
Particle size: 75 fIm to 160 fIm.
Xylenol orange triturate. 1096301.
pH limits of use: 5 to 14.
Triturate 1 part of xylenol orange R with 99 parts of
Maximum temperature of use: 120 oc.
potassium nitrate R.
Wedelolactone. C16HlQOr (Mr 314.3). 1187300. Test for sensitivity. To 50 mL of water R add 1 mL
[524-12-9]. 1,8,9-Trihydroxy-3-methoxy-6H-benzofuro [3,2- of dilute acetic acid R, 50 mg of the xylenol orange
e] [1]benzopyran-6-one. triturate and 0.05 mL of lead nitrate solution R. Add
hexamethylenetetramine R until the colour changes from
Xanthydrol. C13HlQ02' (Mr 198.2). 1096100. [90-46-0]. yellow to violet-red. After addition of 0.1 mL of 0.1 M
9-Xanthenol. sodium edetate the colour changes to yellow.
Content: minimum 90.0 per cent.
Xylose. 1096400. [58-86-6].
White or pale-yellow powder, very slightly soluble in water, See Xy/ose (1278).
soluble in ethanol (96 per cent) and in glacial aeetic acid.
It is also available as a methanolic so!ution containing 90 giL Zinc. Zn. (A r 65.4). 1096500. [7440-66-6].
to 110 giL of xanthydrol. Content: minimum 99.5 per cent.
mp: about 123 oc. Silver-white cylinders, granules, pellets or filings with a blue
Assay. In a 250 mL flask dissolve 0.300 g in 3 mL of methanol R sheen.
or use 3.0 mL of solution. Add 50 mL of glacial acetic acid R Arsenic (2.4.2, Method A): maximum 0.2 ppm.
and, dropwise with shaking, 25 mL of a 20 giL solution oí Dissolve 5.0 g in a mixture of the 15 mL of hydroehlorie acid R
urea R. Allow to stand Íor 12 h, collect the precipitate on a and 25 mL of water R prescribed.
sintered-glass filter (16) (2.1.2), wash with 20 mL of ethanol
(96 per cent) R, dry in an oven at 100 oC to 105 oC and weigh. Zinc, activated. 109650l.
Place the zinc cylinders or pellets to be activated in a conieal
1 g of precipitate is equivalent to 0.9429 g oí xanthydrol.
flask and add a sufficient quantity of a 50 ppm solution of
Storage: protected from light. If a methanolic solution is chloroplatinic acid R to cover the metal. Allow the metal to
used, store in small sealed ampoules and filter before use if remain in contact with the solution for 10 min, wash, drain
necessary. and dry immediately.
Xanthydrol Rl. 1096101. Arsenic (2.4.2, Method A). To 5 g of the activated zinc add
15 mL of hydrochloric acid R, 25 mL of water R, 0.1 mL of
Complies with the requirements prescribed for xanthydrol R stannous chloride solution R and 5 mL of potassium iodide
with the following requirement. solution R. No stain is produced on the mercuric bromide
Content: minimum 98.0 per cent of C!3HlQOz. paper R.
Xanthydrol solution. 1096102. Activity. Repeat the test for arsenic using the same reagents
and adding a solution containing 1 fIg of arsenic. An
To 0.1 mL of a 100 giL solution of xanthydrol R in appreciable stain appears on the mercuric bromide paper R.
methanol R add 100 mL of anhydrous acetie acid R and
1 mL of hydrochloric acid R. Allow to stand for 24 h before Zinc acetate. (C 2Hp2)ZZn,2H 20. (Mr 219.5). 1102300.
using. [5970-45-6]. Zinc acetate dihydrate.
Bright white or almost white crystals, slightly efflorescent,
Xylene. CsHIO" (Mr 106.2). 1096200. [1330-20-7]. freely soluble in water, soluble in ethanol (96 per cent). It loses
Mixture of isomers. Clear, colourless, flammable liquid, its crystallisation water at 100 oc.
practically insoluble in water, miscible with ethanol (96 per d~g: about 1.735.
cent).
mp: about 237 oc.
d~g: about 0.867.
n~o: about 1.497.
Zinc acetate soll.ltion. 110230l.
Mix 600 mL of water R with 150 mL of glacial aeetic acid R,
bp: about 138 oc.
54.9 g of zinc acetate R and stir to dissolve. Continue
m-Xylene. CsHIO" (Me 106.2). 1117700. [108-38-3]. stirring while adding 150 mL of coneentrated ammonia R.
1,3-Dimethylbenzene. Cool to room temperature and adjust with ammonia R to
pH 6.4. Dilute the mixture to 1 L with water R.
Clear, colourless, flammable liquid, practically insoluble in
water, miscible with ethanol (96 per cent). Zinc chloride. 1096600. [7646-85-7].
d~g : about 0.884. See Zinc chloride (0110).
nbo: about 1.497. Zinc chloride-formk add soluti.on. 1096601.
bp: about 139 oc. Dissolve 20 g of zinc chloride R in 80 g of an 850 giL
mp: about - 47 oc. solution of anhydrous formic acid R.

General Notices (1) apply to all monographs and other texts 535
4.1.2. Standard solutions for limit tests EUROPEAN PHARMACOPOEIA 8.0

Zinc chloride solution, iodinated. 1096602. Aluminium standard solution (200 ppm Al). 5000200.
Dissolve 20 g of zinc chloride R and 6.5 g of potassium Dissolve in water R a quantity of aluminium potassium
iodide R in 10.5 mL of water R. Add 0.5 g of iodine R and sulfate R equivalent to 0.352 g of AIK(S04)2,12H 20. Add 10 111L
shake for 15 mino Filter if necessary. of dilute sulfuric acid R and dilute to 100.0 mL with water R.
Storage: protected from light.
Aluminium standard solution (100 ppm Al). 5000203.
Zinc iodide and starch solution. 1096502. Immediately before use, dilute with water R to 10 times its
To a solution of 2 g of zinc chloride R in 10 mL of water R volume a solutiol1 containing 8.947 g of aluminium chloride R
add 0.4 g of soluble starch R and heat until the starch has in 1000.0 mL of water R.
dissolved. After cooling to room temperature add 1.0 mL of
a colourless solution containing 0.10 g zinc Ras filings and Aluminium standard solution (10 ppm Al). 5000201.
0.2 g of iodine R in water R. Dilute the solution to 100 mL Immediately before use, dilute with water R to 100 times
with water R and filter. its volume in a solution containing aluminium nitrate R
Storage: protected from light. equivalent to 1.39 g of AI(N0 3 )3,9HP in 100.0 mL.
Test for sensitivity. Dilute 0.05 mL of sodium nitrite solution R Aluminium standard solution (2 ppm Al). 5000202.
to 50 mL with water R. To 5 mL of this solution add 0.1 mL of
dilute sulfuric acid R and 0.05 mL of the zinc iodide and starch Immediately before use, dilute with water R to 100 times its
solution and mix. The solution becomes bIue. volume a solution containing aluminium potassium sulfate R
equivalent to 0.352 g of AIK(S04)2,12HP and 10 mL of dilute
Zinc oxide. 1096700. [1314-13-2]. sulfuric acid R in 100.0 mL.
See Zinc oxide (0252). Ammonium standard solution (lOO ppm NH 4 ). 5000300.
Zinc powder. Zn. (A, 65.4). 1096800. [7440-66-6]. Immediately before use, dilute to 25 mL with water R 10 mL
Content: minimum 90.0 per cent. of a solution containing ammonium chloride R equivalent to
Very fine, grey powder, soluble in dilute hydrochloric acid R. 0.741 g ofNH 4Cl in 1000 mL.

Zinc sulfate. 1097000. [7446-20-0]. Ammonium standard solution (3 ppm NH 4). 5006100.
See Zinc sulfate (0111). Immediately before use, dilute with water R to 100 times its
volume a solution containing ammonium chloride R equivalent
Zirconyl chloride. A basic salt corresponding approximately to 0.889 g ofNH 4 Cl in 1000.0 mL.
to the formula ZrClp, 8Hp. 1097100. [15461-27-5].
Content: minimum 96.0 per cent of ZrCI 20,8H 20. Ammonium standard solution (2.5 ppm NH 4 ). 5000301.
White or almost white, crystalline powder or crystals, freely Immediately before use, dilute withwater R to 100 times its
soluble in water and in ethanol (96 per cent). volume a solution containing ammonium chloride R equivalent
to 0.741 g ofNH 4 CI in 1000.0 mL.
Assay. Dissolve 0.600 g in a mixture of 5 mL of nitric acid R
and 50 mL of water R. Add 50.0 mL of 0.1 M silver nitrate Ammonium standard solution (l ppm NH 4 ). 5000302.
and 3 mL of dibutyl phthalate R and shake. Using 2 mL of
Immediately before use, dilute ammonium standard solution
ferric ammonium sulfate solution R2 as indicator, titrate with
(2.5 ppm NHJ R to 2.5 times its volume with water R.
0.1 M ammonium thiocyanate until a reddish-yellow colour is
obtained. Antimony standard solution (lOO ppm Sb). 5000401.
1 mL of 0.1 M silver nitrate is equivalent to 16.11 mg of Dissolve antimony potassium tartrate R equivalent to 0.274 g
ZrClp,8Hp. of C 4 H 4K0 7 Sb, 'i2H 2 0 in 500 mL of 1 M hydrochloric acid and
Zirconyl nitrate. A basic salt corresponding approximately to dilute the clear solution to 1000 mL with water R.
the formula ZrO(N0 3 )2,2Hp. 1097200. [14985-18-3]. Antimony standard solution (l ppm Sb). 5000400.
A white or almost white powder or crystals, hygroscopic, Dissolve antimony potassium tartrate R equivalent to 0.274 g
soluble in water. The aqueous solution is a clear or at most
of C4H4K07Sb,l/2HP in 20 mL of hydrochloric acid R1 and
slightly opalescent liquido dilute the clear solution to 100.0 mL with water R. To 10.0 mL
Storage: in an airtight container. of this solution add 200 mL of hydrochloric acid R1 and d¡lute
to 1000.0 mL with water R. To 100.0 mL of this solution add
Zirconyl nitrate solution. 1097201.
300 mL of hydrochloric acid R1 and dilute to 1000.0 mL with
A 1 giL solution in a mixture of 40 mL of water R and water R. Prepare the dilute solutions immediately before use.
60 mL of hydrochloric acid R.
Arsenic standard solution (lO ppm As). 5000500.
Immediately before use, dilute with water R to 100 times its
04/2010:40102 volume a solution prepared by dissolving arsenious trioxide R
equivalent to 0.330 g of ASP3 in 5 mL of dilute sodium
4.1.2. STANDARD SOLUTIONS FOR hydroxide solution R and diluting to 250.0 mL with water R.
LIMIT TESTS Arsenic standard solution (l ppm As). 5000501.
Acetaldehyde standard solution (100 ppm C 2 H 4 0). 5000100. Immediately before use, dilute arsenic standard solution
Dissolve 1.0 g of acetaldehyde R in 2-propanol R and dilute to (lO ppm As) R to 10 times its volume with water R.
100.0 mL with the same solvento Dilute 5.0 mL of the solution
Arsenic standard solution (0.1 ppm As). 5000502.
to 500.0 mL with 2-propanol R. Prepare immediately before
use. Immediately before use, dilute arsenic standard solution
(1 ppm As) R to 10 times its volume with water R.
Acetaldehyde standard solution (lOO ppm C 2H 4 0) RL
5000101. Barium standard solution (0.1 per cent Ba). 5000601.
Dissolve 1.0 g of acetaldehyde R in water R and dilute to Dissolve barium chloride R equivalent to 0.178 g ofBaC1 2,2HP
100.0 mL with the same solvent. Dilute 5.0 mL of the solution in distilled water R and dilute to 100.0 mL with the same
to 500.0 mL with water R. Prepare immediately before use. solvent.

536 See the information section on general monographs (cover pages)


EUROPEAN PHARMACOPOEIA 8.0 4.1.2. Standard solutions for limit tests

Barium standard solution (50 ppm Ba). 5000600. Chromium Hposoluble standard solution (l000 ppm Cr).
Immediately befare use, dilute with distilled water R to 5004600.
20 times its volume a solution in distilled water R containing A chromium (metal) organic compound in an oil.
barium chloride R equivalent to 0.178 g of BaCl2,2H 20 in
100.0 mL. Chromium standard solution (0.1 per cen! Cr). 5001002.
Dissolve potassium dichromate R equivalent to 2.83 g of
Barium standard solution (2 ppm Ba). 5005600. K2 Cr 2 0 7 in water R and dilute to 1000.0 mL with the same
Immediately before use, dilute barium standard solution solvent.
(50 ppm Ea) R to 25 times its volume with distilled water R.
Chromium standard solution (100 ppm Cr). 5001000.
Bismuth standard solution (100 ppm Bi). 5005300.
Dissolve potassium dichromate R equivalent to 0.283 g of
Dissolve bismuth R equivalent to 0.500 g of Bi in 50 mL of K2 Cr 2 0 7 in water R and dilute to 1000.0 mL with the same
nitric acid R and dilute to 500.0 mL with water R. Dilute solvent.
the solution to 10 times its volume with dilute nitric acid R
immediately befare use. Chromium standard solution (0.1 ppm Cr). 500100l.
Immediately before use, dilute chromium standard solution
Cadmium standard solution (0.1 per cent Cd). 5000700.
(100 ppm Cr) R to 1000 times its volume with water R.
Dissolve cadmium R equivalent to 0.100 g of Cd in the
smallest necessary amount of a mixture of equal volumes of Cobalt standard soiution (100 ppm Col. 5004300.
hydrochloric acid R and water R and dilute to 100.0 mL with a Dissolve cobalt nitrate R equivalent to 0.494 g of
1 per cent V/V solution of hydrochloric acid R. CO(N0 3 )2,6HP in 500 mL of 1 M nitric acid and dilute the
clear solution to 1000 mL with water R.
Cadmium standard solution (lO ppm Cd) . 500070l.
Immediately before use, dilute cadmium standard solution Copper liposoluble standard solutlon (1000 ppm Culo
(0.1 per cent Cd) R to 100 times its volume with a 1 per 5004700.
cent V/V solution of hydrochloric acid R. A copper (metal) organic compound in an oil.
Caldum standard solution (400 ppm Ca). 5000800. Copper standard solution (0.1 per cent Cu). 5001100.
Immediately before use, dilute with distilled water R to
Dissolve copper sulfate R equivalent to 0.393 g of CuSO'1'5H 2 0
10 times its volume a solution in distilled water R containing
in water R and dilute to 100.0 mL with the same solvent.
calcium carbonate R equivalent to 1.000 g of CaC0 3 and 23 mL
of 1 M hydrochloric acid in 100.0 mL. Copper standard solution (lO ppm Cu). 5001101.
Calci.um standard solution (lOO ppm Ca). 5000801. Immediately before use, dilute copper standard solution
(0.1 per cent Cu) R to 100 times its volume with water R.
Immediately before use, dilute with distilled water R to
10 times its volume a solution in distilled water R containing Copper standard solution (0.1 ppm Culo 5001102.
calcium carbonate R equivalent to 0.624 g of CaC0 3 and 3 mL
of acetic acid R in 250.0 mL. Immediately before use, dilute copper standard solution
(10 ppm Cu) R to 100 times its volume with water R.
Caldum standard solution (lOO ppm Ca) RL 5000804.
Ferrocyanide standard solution (lOO ppm Fe(CN)6)'
Immediately before use, dilute with water R to 10 times its 5001200.
volume a solution containing anhydrous calcium chloride R
equivalent to 2.769 g of CaCl 2 in 1000.0 mL of dilute Immediately before use, dilute with water R to 10 times
hydrochloric acid R. its volume a solution containing potassium ferrocyanide R
equivalent to 0.20 g ofK 4Fe(CN)6,3HP in 100.0 mL.
Cakium standard solution (100 ppm Ca), alcoholic.
5000802. Ferricyanide standard solution (50 ppm Fe(CN)6)' 5001300.
Immediately before use, dilute with ethanol (96 per cent) R to Immediately before use, dilute with water R to 100 times
10 times its volume a solution in distilled water R containing its volume a solution containing potassium ferricyanide R
calcium carbonate R equivalent to 2.50 g of CaC0 3 and 12 mL equivalent to 0.78 g of K3Fe( CN)6 in 100.0 mL.
of acetic acid R in 1000.0 mL.
Fluoride standard solution (10 ppm F). 5001400.
Caldum standard solution (10 ppm Ca). 5000803. Dissolve in water R sodium fluoride R previously dried at
Immediately before use, dilute with distilled water R to 300 oC for 12 h, equivalent to 0.442 g of NaF, and dilute to
100 times its volume a solution in distilled water R containing 1000.0 mL with the same solvent (1 mL = 0.2 mg F). Store in
calcium carbonate R equivalent to 0.624 g of CaC0 3 and 3 mL a polyethylene container. Immediately before use, dilute the
of acetic acid R in 250.0 mL. solution to 20 times its volume with water R.

Chloride standard solution (50 ppm CI). 5004100. Fluoride standard solution (l ppm F). 5001401.
Immediately before use, dilute with water R to 10 times its Immediately before use, dilute fluoride standard solution
volume a solution containing sodium chloride R equivalent to (10 ppm F) R to 10 times its volume with water R.
0.824 g ofNaCl in 1000.0 mL.
Formaldehyde standard solution (5 ppm CH 2 0). 5001500.
Chloride standard solution (8 ppm CI). 5000900. Immediately before use, dilute with water R to 200 times its
Immediately before use, dilute with water R to 100 times its volume a solution containing 1.0 g of CH 20 per litre prepared
volume a solution containing sodium chloride R equivalent to from formaldehyde solution R.
1.32 g of NaCl in 1000.0 mL.
Germanium standard solution (100 ppm Ge). 5004400.
Chloride standard solution (5 ppm CI). 500090l. Dissolve ammonium hexafluorogermanate(IV) R equivalent
Immediately before use, dilute with water R to 100 times its to 0.307 g of (NH4)2GeF6 in a 0.01 per cent V/V solution of
volume a solution containing sodium chloride R equivalent to hydrofluoric acid R. Dilute the clear solution to 1000 mL with
0.824 g of NaCl in 1000.0 mL. water R.

General Notices (1) apply to all monographs and other texts 537
4.1.2. Standard solutions ior limit tests EUROPEAN PHARMACOPOEIA 8.0

Glyoxal standard solution (20 ppm C 2 H z0 2 ). 5003700. Lead standard solution (lO ppm Pb). 5001702.
In a 100 mL graduated flask weigh a quantity of glyoxal Immediately before use, dilute lead standard solution (100 ppm
solution R corresponding to 0.200 g of C2 HP2 and make up Pb) R to 10 times its volume with water R.
to volume with anhydrous ethanol R. Immediately before use
dilute the solution to 100 times its volume with the same Lead standard solution (lO ppm Pb) Rl. 5001706.
solvent. Immediately before use, dilute with water R to 10 times its
volume a solution containing 0.160 g of lead nitrate R in
Glyoxal standard solution (2 ppm C 2HP2)' 5003701. 100 mL of water R, to which is added 1 mL of lead-free nitrie
Immediately before use, dilute glyoxal standard solution acid R and dilute to 1000.0 mL.
(20 ppm C2 H 2 0) R to 10 times its volume with anhydrous
ethanol R. Lead standard solution (lO ppm Pb) R2. 5005401.
Dilute lead standard solution (0.1 per eent Pb) R1 to 100 times
Hydrogen peroxide standard solution (la ppm HzOz). its volume with dilute lead-free nitrie aeid R. Use within
5005200. 1 week.
Dilute 10.0 mL of dilute hydrogen peroxide solution R to
300.0 mL with water R. Dilute 10.0 mL of this solution to Lead standard solution (2 ppm Pb). 5001703.
1000.0 mL with water R. Prepare immediately before use. Immediately before use, dilute lead standard solution (lO ppm
Pb) R to 5 times its volume with water R.
Iodide standard solution (lO ppm 1). 5003800.
Immediately before use, dilute with water R to 100 times its Lead standard solution (l ppm Pb). 5001704.
volume a solution containing potassium iodide R equivalent Immediately before use, dilute lead standard solution (10 ppm
to 0.131 g ofKI in 100.0 mL. Pb) R to 10 times its volume with water R.

Iron standard solution (0.1 per cent Fe). 5001605. Lead standard solution (0.5 ppm Pb). 5005402.
Dissolve 0.100 g of Fe in the smallest amount necessary of a Dilute lead standard solution (10 ppm Pb) R2 to 20 times its
mixture of equal volumes of hydrochloric acid R and water R volume with dilute lead-free nitrie aeid R. Use within 1 day.
and dilute to 100.0 mL with water R.
Lead standard solution (0.25 ppm Pb). 5006000.
lron standard solution (250 ppm Fe). 5001606. Immediately before use, dilute lead standard solution (1 ppm
Immediately before use, dilute with water R to 40 times its Pb) R to 4 times its volume with water R.
volume a solution containing 4.840 g of ferrie ehloride R in a
Lead standard solution (0.1 ppm Pb). 5001705.
150 giL solution of hydroehloric aeid R diluted to 100.0 mL.
Immediately before use, dilute lead standard solution (1 ppm
Iron standard solution (20 ppm Fe). 5001600. Pb) R to 10 times its volume with water R.
Immediately before use, dilute with water R to 10 times its
Magnesium standard solution (0.1 per cent Mg). 5001803.
volume a solution containingferrie ammonium sulfate R
equivalent to 0.863 g of FeNH 4 (S04)2,12HP and 25 mL of Dissolve magnesium sulfate R equivalent to 1.010 g of
dilute sulfuric acid R in 500.0 mL. MgS0 4,7HP in distilled water R and dilute to 100.0 mL with
the same solvent.
Iron standard solution (10 ppm Fe). 5001601.
Magnesium standard solution (l000 ppm Mg). 5006200.
Immediately before use, dilute with water R to 100 times its
volume a solution containingferrous ammonium sulfate R Dissolve 5.275 g of magnesium nitrate R in 16 mL of dilute
equivalent to 7.022 g of Fe(NH 4 lz(S04)2,6HP and 25 mL of nitrie acid R and dilute to 500.0 mL with water R.
dilute sulfuric acid R in 1000.0 mL. Standardisation: carry out the determination of magnesium
by complexometry (2.5.11).
Iron standard soIution (8 ppm Fe). 5001602.
Immediately before use, dilute with water R to 10 times its Magnesium standard solution (lOO ppm Mg). 5001800.
volume a solution containing 80 mg of iron R and 50 mL of Immediately before use, dilute with water R to 10 times its
hydroehlorie acid R (220 giL Hel) in 1000.0 mL. volume a solution containing magnesium sulfate R equivalent
to 1.010 g of MgS0 4 ,7HzÜ in 100.0 mL.
!ron standard solution (2 ppm Fe). 5001603.
Immediately before use, dilute iron standard solution (20 ppm Magnesium standard solution (lO ppm Mg). 5001801.
Fe) R to 10 times its volume with water R. Immediately before use, dHute magnesium standard solution
(100 ppm Mg) R to 10 times its volume with water R.
Iron standard solution (1 ppm Fe). 5001604.
Immediately before use, dilute iron standard solution (20 ppm Magnesium standard solution (lO ppm Mg) Rl. 5001802.
Fe) R to 20 times its volume with water R. Immediately before use, dilute with water R to 100 times its
volume a solution containing 8.365 g of magnesium ehloride R
Lead li.posoluble standard solution (1000 ppm Pb). in 1000.0 mL of dilute hydroehloric acid R.
5004800.
A lead (metal) organic compound in an oil. Manganese standard solution (1000 ppm Mn). 5005800.
Dissolve manganese sulfate R equivalent to 3.08 g of
Lead standard solution (0.1 per cent Pb). 5001700. MnS0 4,Hp in 500 mL of 1 M nitric acid and dilute the
Dissolve lead nitrate R equivalent to 0.400 g of Pb(N0 3)2 in solution to 1000 mL with water R.
water R and dilute to 250.0 mL with the same solvent.
Manganese standard solution (lOO ppm Mn). 5004500.
Lead standard solution (0.1 per cent Pb) Rl. 5005400. Dissolve manganese sulfate R equivalent to 0.308 g of
Dissolve in dilute lead-free nitrie acid R a quantity of lead MnS0 4,Hp in 500 mL of 1 M nitrie acid and dilute the clear
nitrate R equivalent to 0.400 g of Pb (N0 3)2 and dilute to solution to 1000 mL with water R.
250.0 mL with the same solvent.
Men::ury standard solution (1000 ppm Hg). 5001900.
Lead standard solution (lOO ppm Pb). 5001701. Dissolve mereurie ehloride R equivalent to 1.354 g of HgC12
Immediately before use, dilute lead standard solution (0.1 per in 50 mL of dilute nitric acid R and dilute to 1000.0 mL with
cent Pb) R to 10 times its volume with water R. water R.

538 See the information section on general monographs (cover pages)


EUROPEAN PHARMACOPOEIA 8.0 4.1.2. Standard solntions for Hmit tests

Mercury standard solution (10 ppm Hg). 5001901. Potassium standard solution (600 ppm K). 5005100.
Immediately before use, dilute with water to 100 times its Immediately before use, dilute with water R to 20 times its
volume a solution containing mercuric chloride R equivalent volume a solution eontaining dipotassium sulfate R equivalent
to 0.338 g of HgCl 2 in 250.0 mL. to 2.676 g ofK 2S0 4 in 100.0 mL.

Nickelliposoluble standard solution (1000 ppm Ni). Potassium standard solution (lOO ppm K). 5002400.
5004900. Immediately before use, dilute with water R to 20 times its
A niekel (metal) organie eompound in an oil. volume a solution eontaining dipotassium sulfate R equivalent
to 0.446 g ofK 2 S0 4 in 100.0 mL.
Nkkel standard solution (lO ppm Ni). 5002000.
Potassium standard solution (20 ppm K). 5002401.
Immediately before use, dilute with water R to 100 times its
volume a solution eontaining nickel sulfate R equivalent to Immediately befare use, dilute potassium standard solution
4.78 g ofNiS0 4,7HP in 1000.0 mL. (lOO ppm K) R to 5 times its volume with water R.
Selenium standard solution (100 ppm Se). 5002500.
Nickel standard solution (5 ppm Ni). 5005900.
Dissolve 0.100 g of selenium R in 2 mL of nitric acid R.
Immediately before use dilute nickel standard solution (10 ppm
Evaporate to dryness. Take up the residue in 2 mL of water R
Ni) R to twiee its volume with water for chromatography R.
and evaporate to dryness; carry out three times. Dissolvethe
Nickel standard solution (0.2 ppm Ni). 5002002. residue in 50 mL of dilute hydrochloric acid R and dilute to
1000.0 mL with the same aeid.
Immediately before use, dilute nickel standard solution
(lO ppm Ni) R to 50 times its volume with water R. Selenium standard solution (l ppm Se). 5002501.
Nickel standard solution (0.1 ppm Ni). 5002001. Immediately before use, dilute with water R to 40 times its
volume a solution eontaining selenious acid R equivalent to
Immediately before use, dilute nickel standard solution 6.54 mg of H 2Se0 3 in 100.0 mL.
(10 ppm Ni) R to 100 times its volume with water R.
Silver standard solution (5 ppm Ag). 5002600.
Nitrate standard solution (100 ppm N0 3 ). 5002100.
Immediately before use, dilute with water R to 100 times its
Immediately befare use, dilute with water R to 10 times its volume a solution eontaining si/ver nitrate R equivalent to
volume a solution eontaining potassium nitrate R equivalent 0.790 g of AgN0 3 in 1000.0 mL.
to 0.815 g of KN0 3 in 500.0 mL.
Sodium standard solution (l000 ppm Na). 5005700.
Nitrate standard solution (10 ppm N0 3 ). 5002101. Dissolve a quantity of anhydrous sodium carbonate R
Immediately before use, dHute nitrate standard solution equivalent to 2.305 g of Na2 C0 3 in a mixture of 25 mL of
(100 ppm NO) R to 10 times its volume with water R. water R and 25 mL of nitric acid R and dilute to 1000.0 mL
with water R.
Nitrate standard solution (2 ppm N0 3 ). 5002102.
Immediately before use, dilute nitrate standard solution Sodium standard solution (200 ppm Na). 5002700.
(lO ppm NO) R to 5 times its volume with water R. Immediately before use, dilute with water R to 10 times its
volume a solution containing sodium chloride R equivalent to
PaUadium standard solution (500 ppm Pd). 5003600. 0.509 g ofNaCl in 100.0 mL.
Dissolve 50.0 mg of palladium R in 9 mL of hydrochloric
Sodium standard solution (50 ppm Na). 5002701.
acid R and dilute to 100.0 mL with water R.
Dilute the sodium standard solution (200 ppm Na) R to four
PaUadi.um standard solution (20 ppm Pd). 5003602. times its volume with water R.
Dissolve 0.333 g of palladium chloride R in 2 mL of warm Strontium standard solution (1.0 per cent Sr). 5003900.
hydrochloric acid R. Dilute the solution to 1000.0 mL with a
mixture of equal volumes of dilute hydrochloric acid R and Cover with water R, strontium carbonate R equivalent to
water R. Immediately before use dilute to 10 times its volume 1.6849 g of SrC03' Cautiously add hydrochloric acid R until
with water R. al! the solid has dissolved and there is no sign of further
efferveseenee. Dilute to 100.0 mL with water R.
Palladium standard solution (0.5 ppm Pd). 5003601.
Sulfate standard solution (100 ppm S04)' 5002802.
Dilute 1 mL of palladium standard solution (500 ppm Pd) R
Immediately before use, dilute with distilled water R to
to 1000 mL with a mixture of 0.3 volumes of nitric acid R and 10 times its volume a solution in distilled water R eontaining
99.7 volumes of water R.
dipotassium sulfate R equivalent to 0.181 g of K2S0 4 in
Phosphate standard solution (200 ppm P0 4 ). 5004200. 100.0 mL.
Dissolve potassium dihydrogen phosphate R equivalent to Sulfate standard solution (10 ppm SO 4)' 5002800.
0.286 g of KH 2 PO.¡ in water R and dilute to 1000.0 mL with Immediately before use, dilute with distilled water R to
the same solvent. 100 times its volume a solution in distilled water R eontaining
Phosphate standard solution (5 ppm PO.¡). 5002200. dipotassium sulfate R equivalent to 0.181 g ofK 2S0 4 in
100.0 mL.
Immediately before use, dilute with water R to 100 times
its volume a solution containing potassium dihydrogen Sulfate standard solution (lO ppm SO,) Rl. 5002801.
phosphate R equivalent to 0.716 g ofKH 2 P0 4 in 1000.0 mL. Immediately befare use, dilute with ethanol (30 per cent V/V) R
to 100 times its volume a solution eontaining dipotassium
Platinum standard solution (30 ppm Pt). 5002300. sulfate R equivalent to 0.181 g ofK2 S0 4 in 100.0 mL of ethanol
Immediately before use, dilute with 1 M hydrochloric acid (30 per cent V/V) R.
to 10 times its volume a solution containing 80 mg of
chloroplatinic acid R in 100.0 mL of 1 M hydrochloric acid. Sulfite standard solution (80 ppm S02)' 5005500.
Dissolve 3.150 g of anhydrous sodium sulfite R in freshly
Potassium standard solution (0.2 per cení K). 5002402. prepared distilled water R and dilute to 100.0 mL with the
Dissolve dipotassium sulfate R equivalent to 0.446 g of K2SO 4 in same solvent. Dilute 0.5 mL to 100.0 mL with freshly prepared
distilled water R and dilute to 100.0 mL with the same solvent. distilled water R.

General Notíces (1) apply to all monographs and other texts 539
4.1.3. Buffer soluHons EUROPEAN PHARMACOPOEIA 8.0

Sulfite standard solution (1.5 ppm S02)' 5002900. Phosphate buffer solution pH 2.0. 4007900.
Dissolve sodium metabisulfite R equivalent to 0.152 g of Dissolve 8.95 g of disodium hydrogen phosphate R and 3.40 g
Na2 S2 0 S in water R and dilute to 100.0 mL with the same of potassium dihydrogen phosphate R in water R and dilute to
solvento Dilute 5.0 mL of this solution to 100.0 mL with 1000.0 mL with the same solvent. lf necessary adjust the pH
water R. To 3.0 mL of the resulting solution, add 4.0 mL of with phosphoric acid R.
0.1 M sodium hydroxide and dilute to 100.0 mL with water R.
Sulfate buffer solution pH 2.0. 4008900.
ThaUium standard solution (lO ppm TI). 5003000. Dissolve 132.1 g of ammonium sulfate R in water R and dilute
Dissolve thallous sulfate R equivalent to 0.1235 g of Tl 2 S0 4 in to 500.0 mL with the same solvent (Solution A). Carefully and
a 9 giL solution of sodium chloride R and dilute to 1000.0 mL with constant cooling stir 14 mL of sulfuric acid R into about
with the same solution. Dilute 10.0 mL of the solution to 400 mL of water R; allow to cool and dilute to 500.0 mL with
100.0 mL with the 9 giL solution of sodium ehloride R. water R (Solution B). Mix equal volumes of solutions A and B.
Adjust the pH if necessary.
Tin liposoluble standard solution (1000 ppm Sn). 5005000.
A tin (metal) organic compound in an oil. Buffer solution pH 2.2. 4010500.
Mix 6.7 mL of phosphorie acid R with 55.0 mL of a 40 giL
Tiu standard solution (5 ppm Su). 5003100. solution of sodium hydroxide R and dilute to 1000.0 mL with
Dissolve tin R equivalent to 0.500 g of Sn in a mixture of 5 mL water R.
of water R and 25 mL of hydroehlorie acid R and dilute to
1000.0 mL with water R. Dilute the solution to 100 times its Buffer solution pH 2.5. 4000300.
volume with a 2.5 per cent V/V solution of hydrochloric aeid R Dissolve 100 g of potassium dihydrogen phosphate R in 800 mL
immediately before use. of water R; adjust to pH 2.5 with hydrochlorie acid R and
dilute to 1000.0 mL with water R.
Tin standard solution (0.1 ppm Su). 50031Ol.
Immediately before use, dilute tin standard so/ution (5 ppm Buffer solution pH 2.5 Rl. 4000400.
Sn) R to 50 times its volume with water R. To 4.9 g of dilute phosphoric acid R add 250 mL of water R.
Adjust the pH with dilute sodium hydroxide solution R and
Titauium standard solution (100 ppm Ti). 5003200. dilute to 500.0 mL with water R.
Dissolve 100.0 mg of titanium R in 100 mL of hydrochloric
acid R diluted to 150 mL with water R, heating if necessary. 0.2 M Phosphate buffer solution pH 2.5. 4014100.
Allow to cool and dilute to 1000 mL with water R. Dissolve 27.2 g of potassium dihydrogen phosphate R in about
900 mL of water R, adjust to pH 2.5 with phosphorie acid R
Vanadium standard solution (l gIL V). 5003300. and dilute to 1.0 L with water R.
Dissolve in water R ammonium vanadate R equivalent to
0.230 g ofNH 4V0 3 and dilute to 100.0 mL with the same Phosphate buffer solutíon pH 2.8. 4010600.
solvent. Dissolve 7.8 g of sodium dihydrogen phosphate R in 900 mL of
water R, adjust to pH 2.8 with phosphoric acid R and dilute to
Zinc standard solution (5 mg/mL Zn). 5003400. 1000 mL with the same solvent.
Dissolve 3.15 g of zinc oxide R in 15 mL of hydroch/orie add R
and dilute to 500.0 mL with water R. Buffer solution pH 3.0. 4008000.
Dissolve 21.0 g of citrie acid R in 200 mL of 1 M sodium
Zinc standard solution (100 ppm Zn). 5003401. hydroxide and dilute to 1000 mL with water R. Dilute 40.3 mL
Immediately before use, dilute with water R to 10 times its of this solution to 100.0 mL with 0.1 M hydrochloric acid.
volume a solution containing zinc sulfate R equivalent to
0.440 g of ZnS0 4 ,7HP and 1 mL of acetic acid R in 100.0 mL. 0.25 M Ciírate buffer solution pH 3.0. 4012600.
Dissolve 5.3 g of eitrie acid R in 80 mL of water R. Adjust the
Zinc standard solution (10 ppm Zn). 5003402. pH with 1 M sodium hydroxide and dilute to 100.0 mL with
Immediately before use, dilute zine standard solution (100 ppm water R.
Zn) R to 10 times its volume with water R.
0.1 M Phosphate buffer solution pH 3.0. 4011500.
Zinc standard solution (5 ppm Zn). 5003403.
Dissolve 12.0 g of anhydrous sodium dihydrogen phosphate R
Immediately before use, dilute zine standard solution (100 ppm in water R, adjust the pH with dilute phosphoric aeid R1 and
Zn) R to 20 times its volume with water R. dilute to 1000 mL with water R.
Zirconium standard solution (l gIL Zr). 5003500. Phosphate buffer solution pH 3.0. 4000500.
Dissolve zirconyl nitrate R equivalent to 0.293 g of Mix 0.7 mL of phosphoric acid R with 100 mL of water R.
ZrO(N0 3)2,2HP in a mL'Cture of 2 volumes of hydrochloric Dilute to 900 mL with the same solvent. Adjust to pH 3.0 with
acid R and 8 volumes of water R and dilute to 100.0 mL with strong sodium hydroxide solution R and dilute to 1000 mL
the same mixture of solvents. with water R.
Phosphate buffer solution 3.0 Rl. 4010000.
01/2014:40103 Dissolve 3.40 g of potassium dihydrogen phosphate R in
900 mL of water R. Adjust to pH 3.0 with phosphorie acid R
4.1.3. BUFFER SOLUTIONS and dilute to 1000.0 mL with water R.
Buffered acetone solution. 4000100. Phosphate buffer solution pH 3.2. 4008100.
Dissolve 8.15 g of sodium acetate R and 42 g of sodium To 900 mL of a 4 giL solution of sodium dihydrogen
eh/oride R in water R, add 68 mL of 0.1 M hydrochlorie acid phosphate R, add 100 mL of a 2.5 giL solution of phosphoric
and 150 mL of acetone R and dilute to 500 mL with water R. acid R. Adjust the pH if necessary.
Buffer solution pH 2.0. 4000200. Phosphate buffer solution pH 3.2 Rl. 4008500.
Dissolve 6.57 g of potassium eh/oride R in water R and add Adjust a 35.8 giL solution of disodium hydrogen phosphate R
119.0 mL of 0.1 M hydrochloric add. Dilute to 1000.0 mL to pH 3.2 with dihtte phosphoric aeid R. Dilute 100.0 mL of
with water R. the solution to 2000.0 mL with water R.

540 See the informatíon section on monographs (cover pages)


EUROPEAN PHARMACOPOEIA 8,0 4.1.3. Buffer solutious

Buffer solutiou pH 3.5. 4000600. dithizone R in chloroform R. Shake with carbon tetrachloride R
Dissolve 25.0 g of ammonium acetate R in 25 mL of water R until the extraet is eolourless. Filter the aqueous layer to
and add 38.0 mL of hydrochloric acid Rl. Adjust the remove traces of carbon tetraehloride.
pH if neeessary with dilute hydrochloric acid R or dilute Acetate buffer solution pH 4.7 Rl. 4013600.
ammonia R1. Dilute to 100.0 mL with water R.
Dissolve 136.1 g of sodium acetate R in 500 mL of water R. Mix
Phosphate buffer solution pH 3.5. 4000700. 250 mL of this solution with 250 mL of dilute acetic acid R.
Dissolve 68.0 g of potassium dihydrogen phosphate R in Acetate buffer solution pH 5.0. 4009100.
water R and dilute to 1000.0 mL with the same solvent. Adjust
the pH with phosphoric acid R. To 120 mL of a 6 giL solution of glacial acetic acid R add
100 mL of 0.1 M potassium hydroxide and about 250 mL of
Buffer solution pH 3.6. 4000800. water R. Mix. Adjust the pH to 5.0 with a 6 giL solution of
To 250.0 mL of 0.2 M potassium hydrogen phthalate R add acetic acid R or with 0.1 M potassium hydroxide and dilute to
11.94 mL of 0.2 M hydrochloric acid. Dilute to 1000.0 mL 1000.0 mL with water R.
with water R. Citrate buffer solution pH 5.0. 4010700.
Buffer solution pH 3.7. 4000900. Prepare a solution containing 20.1 giL of citrie acid R and
To 15.0 mL of acetic acid R add 60 mL of ethanol (96 per 8.0 giL of sodium hydroxide R. Adjust the pH with dilute
cent) R and 20 mL of water R. Adjust to pH 3.7 by the addition hydrochloric acid R.
of ammonia R. Dilute to 100.0 mL with water R.
0.2 M Deuterated sodium phosphate buffer solution
Buffered copper sulfate solution pH 4.0. 4001000. pH 5.0. 4013900.
Dissolve 0.25 g of capper sulfate R and 4.5 g of ammonium Dissolve 2.76 g of sodium dihydrogen phosphate monohydrate R
acetate R in dilute acetic acid R and dilute to 100.0 mL with in 90 mL of deuterium oxide R, adjust the pH with a deuterated
the same solvent. solution of phosphoric acid R or a deuterated 1 M solution of
sodium hydroxide R, dilute to 100 mL with deuterium oxide R
0.1 M Sodium acetate buffer solution pH 4.0. 4013800. and mix.
Dissolve 822 mg of sodium acetate R in 100 mL of water R
Phosphate buffer solution pH 5.0. 4011300.
(solution A). Dilute 1.44 mL of glacial acetic acid R in 250 mL
of water R (solution B). Titrate 100 mL of solution B using Dissolve 2.72 g of potassium dihydrogen phosphate R in 800 mL
about 20 mL of solution A. of water R. Adjust the pH with 1 M potassium hydroxide and
dilute to 1000 mL with water R.
Acetate buffer solution pH 4.4. 4001100.
Buffer solution pH 5.2. 4001700.
Dissolve 136 g of sodium acetate R and 77 g of ammonium
acetate R in water R and dilute to 1000.0 mL with the same Dissolve 1.02 g of potassium hydrogen phthalate R in 30.0 mL
solvent; add 250.0 mL of glacial acetic acid R and mix. of 0.1 M sodium hydroxide. Dilute to 100.0 mL with water R.

Phthalate buffer solution pH 4.4. 4001200. 0.067 M Phosphate buffer solution pH 5.4. 4012000.
Dissolve 2.042 g of potassium hydrogen phthalate R in 50 mL Mix appropriate volumes of a 23.99 giL solution of disodium
of water R, add 7.5 mL of 0.2 M sodium hydroxide and dilute hydrogen phosphate R with a 9.12 giL solution of sodium
to 200.0 mL with water R. dihydrogen phosphate monohydrate R to obtain pH 5.4.

Acetate buffer solution pH 4.5. 4012500. Acetate-edetate buffer solution pH 5.5. 4001900.
Dissolve 77.1 g of ammonium acetate R in water R. Add 70 mL Dissolve 250 g of ammonium acetate R and 15 g sodium
of glacial acetic acid R and dilute to 1000.0 mL with water R. edetate R in 400 mL of water R and add 125 mL of glacial
acetic acid R.
0.5 M Ammonium acetate buffer solution pH 4.5. 4014200.
Mix 14.3 mL of glacial acetic acid R and 470 mL of water R Buffer solution pH 5.5. 4001800.
and adjust to pH 4.5 with cancentrated ammonia R. Dilute to Dissolve 54.4 g of sodium acetate R in 50 mL of water R,
500.0 mL with water R. heating to 35 oC if necessary. After cooling, slowlyadd 10 mL
of anhydrous acetie acid R. Shake and dilute to 100.0 mL with
0.05 M Phosphate buffer solution pH 4.5. 4009000. water R.
Dissolve 6.80 g of potassium dihydrogen phosphate R in
1000.0 mL of water R. The pH of the solution is 4.5.
Phosphate buffer solution pH 5.5. 4002000.
Dissolve 13.61 g of potassium dihydrogen phosphate R in
Sodium acetate buffer solution pH 4.5. 4010100. water R and dilute to 1000.0 mL with the same solvent
Dissolve 63 g of anhydrous sodium acetate R in water R, (solution A). Dissolve 35.81 g of disodium hydrogen
add 90 mL acetic acid R and adjust to pH 4.5, and dilute to phosphate R in water R and dilute to 1000.0 mL with the same
1000 mL with water R. solvent (solution B). Mix 96.4 mL of solution A and 3.6 mL
of solution B.
Aceta!e buffer solution pH 4.6. 4001400.
Dissolve 5.4 g of sodium acetate R in 50 mL of water R, add Phosphate-citrate buffer solution pH 5.5. 4008700.
2.4 g of glacial acetic acid R and dilute to 100.0 mL with Mix 56.85 mL of a 28.4 giL solution of anhydrous disodium
water R. Adjust the pH if necessary. hydrogen phosphate R and 43.15 mL of a 21 giL solution of
citric acid R.
Succinaíe buffer solution pH 4.6. 4001500.
Disssolve 11.8 g of succinic acid R in a mixture of 600 mL of Phosphate buffer solution pH 5.6. 4011200.
water R and 82 mL of 1 M sodium hydroxide and dilute to Dissolve 0.908 g of potassium dihydrogen phosphate R
1000.0 mL with water R. in water R and dilute to 100.0 mL with the same solvent
(solution A). Dissolve 1.161 g of dipotassium hydrogen
Acetaie buffer solution pH 4.7. 4001600. phosphate R in water R and dilute to 100.0 mL with the same
Dissolve 136.1 g of sodium aceta te R in 500 mL of water R. Mix solvent (solution B). Mix 94.4 mL of solution A and 5.6 mL
250 mL of this solution with 250 mL of dilute aeetic acid R. of solution B. If necessary, adjust to pH 5.6 using solution A
Shake twice with a freshly prepared, filtered, 0.1 giL solution of or solution B.

General Notices (1) apply to al! monographs and other texts 541
4,1.3. Buffer soIutions EUROPEAN PHARMACOPOEIA 8.0

Phosphate buffer solution pH 5.8. 4002100. 0.1 M Phosphate buffer solution pH 6.7. 4014300.
Dissolve 1.19 g of disodium hydrogen phosphate dihydrate R Dissolve 15.6 g of sodium dihydrogen phosphate R in water R
and 8.25 g of potassium dihydrogen phosphate R in water R and dilute to 1.0 L with the same solvent. Dissolve 17.8 g of
and dilute to 1000.0 mL with the same solvento disodium hydrogen phosphate dihydrate R in water R and
dilute to 1.0 L with the same solvento Mix the solutions, check
Acetate buffer solution pH 6.0. 4002200. the pH and ifnecessary adjust to pH 6.7.
Dissolve 100 g of ammonium acetate R in 300 mL of water R,
add 4.1 mL of glacial acetic acid R, adjust the pH if necessary Phosphate buffered saHne pH 6.8. 4003200.
using ammonia R or acetic acid R and dilute to 500.0 mL with Dissolve 1.0 g of potassium dihydrogen phosphate R, 2.0 g
water R. of dipotassium hydrogen phosphate R and 8.5 g of sodium
ehloride R in 900 mL of water R, adjust the pH if necessary
Diethylammonium phosphate buffer solution pH 6.0. and dilute to 1000.0 mL with the same solvent.
4002300.
Dilute 68 mL of phosphoric acid R to 500 mL with water R. Phosphate buffer solution pH 6.8. 4003300.
To 25 mL of this solution add 450 mL of water R and 6 mL Mix 77.3 mL of a 71.5 giL solution of disodium hydrogen
of diethylamine R, adjust to pH 6 ± 0.05, if necessary, using phosphate R with 22.7 mL of a 21 giL solution of citrie aeid R.
diethylamine R or phosphorie aeid R and dilute to 500.0 mL
with water R. Phosphate buffer solution pH 6.8 Rl. 4003400.
To 51.0 mL of a 27.2 giL solution of potassium dihydrogen
Phosphate buffer solution pH 6.0. 4002400.
phosphate R add 49.0 mL of a 71.6 giL solution of disodium
Mix 63.2 mL of a 71.5 giL solution of disodium hydrogen hydrogen phosphate R. Adjust the pH if necessary.
phosphate R and 36.8 mL of a 21 giL solution of citrie acid R. Storage: at 2 oC to 8 oc.
Phosphate buffer solution pH 6.0 Rl. 4002500.
1 M tris-hydrochloride buffer solution pH 6.8. 4009300.
Dissolve 6.8 g of sodium dihydrogen phosphate R in water R
Dissolve 60.6 g of tris(hydroxymethyl)aminomethane R in
and dilute to 1000.0 mL with water R. Adjust the pH with
400 mL of water R. Adjust the pH with hydrochloric acid R
strong sodium hydroxide solution R.
and dilute to 500.0 mL with water R.
Phosphate buffer solution pH 6.0 R2. 4002600.
Buffer solution pH 7.0. 4003500.
To 250.0 mL of 0.2 M potassium dihydrogen phosphate R add
To 1000 mL of a solution containing 18 giL of disodium
28.5 mL of 0.2 M sodium hydroxide and dilute to 1000.0 mL
hydrogen phosphate R and 23 giL of sodium chloride R add
with water R.
sufficient (about 280 mL) of a solution containing 7.8 giL
Phosphate buffer solution pH 6.4. 4002800. of sodium dihydrogen phosphate R and 23 giL of sodium
ehloride R to adjust the pH. Dissolve in the solution sufficient
Dissolve 2.5 g of disodium hydrogen phosphate R, 2.5 g of
sodium azide R to give a 0.2 giL solution.
sodium dihydrogen phosphate R and 8.2 g of sodium ehloride R
in 950 mL of water R. Adjust the pH of the solution to 6.4 with Maleate buffer solution pH 7.0. 4003600.
1 M sodium hydroxide or 1 M hydrochloric acid, if necessary.
Dilute to 1000.0 mL with water R. Dissolve 10.0 g of sodium ehloride R, 6.06 g of
tris(hydroxymethyl)aminomethane R and 4.90 g of maleie
0.5 M Phthalate buffer solution pH 6.4. 4009200. anhydride R in 900 111L of water R. Adjust the pH using a
170 giL solution of sodium hydroxide R. Dilute to 1000.0 mL
Dissolve 100 g of potassium hydrogen phthalate R in water R
with water R.
and dilute to 1000.0 mL with the same solvent. Adjust the pH
if necessary, using strong sodium hydroxide solution R. Storage: at 2 oC to 8 oC; use within 3 days.

Buffer solution pH 6.5. 4002900. 0.025 M Phosphate buffer solutlon pH 7.0. 4009400.
Dissolve 60.5 g of disodium hydrogen phosphate R and 46 g of Mix 1 volume of 0.063 M phosphate buffer solution pH 7.0 R
potassium dihydrogen phosphate R in water R. Add 100 mL of with 1.5 volumes of water R.
0.02 M sodium edetate and 20 mg of mercuric chloride R and
dilute to 1000.0 mL with water R. 0.03 M Phosphate buffer solution pH 7.0. 4010300.
Dissolve 5.2 g of dipotassium hydrogen phosphate R in
Imidazole buffer solution pH 6.5. 4003000. 900 mL of water jor chromatography R. Adjust the solution to
Dissolve 6.81 g of imidazole R, 1.23 g of magnesium sulfate R pH 7.0 ± 0.1 usingphosphoric acid R and dilute to 1000 mL
and 0.73 g of caleium sulfate R in 752 mL of 0.1 M hydrochlorie with water jor chromatography R.
acid. Adjust the pH if necessary and dilute to 1000.0 mL with
water R. 0.05 M Phosphate buffer solution pH 7.0. 4012400.
Mix 34 mL of water R and 100 mL of 0.067 M phosphate buffer
0.1 M phosphate buffer solution pH 6.5. 4010800. solution pH 7.0 R.
Dissolve 13.80 g of sodium dihydrogen phosphate
monohydrate R in 900 mL of distilled water R. Adjust the pH 0.063 M Phosphate buffer solution pH 7.0. 4009500.
using a 400 giL solution of sodium hydroxide R. Dilute to Dissolve 5.18 g of anhydrous disodium hydrogen phosphate R
1000 mL with distilled water R. and 3.65 g of sodium dihydrogen phosphate monohydrate R in
950 mL of water R and adjust the pH with phosphoric acid R;
Phosphate buffer solution pH 6.5. 4012800. dilute to 1000.0 mL with water R.
Dissolve 2.75 g of sodium dihydrogen phosphate R and 4.5 g of
sodium chloride R in 500 mL of water R. Adjust the pH with 0.067 M Phosphate buffer solution pH 7.0. 4003800.
phosphate buffer solution pH 8.5 R. Dissolve 0.908 g of potassium dihydrogen phosphate R
in water R and dilute to 100.0 mL with the same solvent
Buffer solution pH 6.6. 4003100. (solution A). Dissolve 2.38 g of disodium hydrogen phosphate R
To 250.0 mL of 0.2 M potassium dihydrogen phosphate R add in water R and dilute to 100.0 mL with the same solvent
89.0 mL of 0.2 M sodium hydroxide. Dilute to 1000.0 mL with (solution B). Mix 38.9 mL of solutiol1 A and 61.1 mL of
water R. solution B. Adjust the pH if necessary.

542 See the injormation section on general monographs (cover pages)


EUROPEAN PHARMACOPOEIA 8.0 4.1.3. Buffer solutions

0.1 M Phosphate buffer solution pH 7.0. 4008200. Imidazole buffer solution pH 7.3. 4004500.
Dissolve 1.361 g of potassium dihydrogen phosphate R in Dissolve 3.4 g of imidazole R and 5.8 g of sodium chloride R in
water R and dilute to 100.0 mL with the same solvent. water R, add 18.6 mL of 1 M hydrochloric acid and dilute to
Adjust the pH using a 35 giL solution of disodium hydrogen 1000.0 mL with water R. Adjust the pH if necessary.
phosphate R.
Barbital buffer solution pH 7.4. 4004700.
Phosphate buffer solution pH 7.0. 4003700. Mix 50 mL of a solution in water R containing 19.44 giL of
Mix 82.4 mL of a 71.5 giL solution of disodium hydrogen sodium acetate R and 29.46 giL of barbital sodium R with
phosphate R with 17.6 mL of a 21 giL solution of citrie acid R. 50.5 mL of 0.1 M hydrochloric acid, add 20 mL of an 85 giL of
sodium chloride R and dilute to 250 mL with water R.
Phosphate buffer solution pH 7.0 Rl. 4003900.
Buffer solution pH 7.4. 4004600.
Mix 250.0 mL of 0.2 M potassium dihydrogen phosphate R and Dissolve 0.6 g of potassium dihydrogen phosphate R, 6.4 g
148.2 mL of a 8 giL solution of sodium hydroxide R, adjust the of disodium hydrogen phosphate R and 5.85 g of sodium
pH if necessary. Dilute to 1000.0 mL with water R. chloride R in water R, and dilute to 1000.0 mL with the same
solvent. Adjust the pH if necessary.
Phosphate buffer solution pH 7.0 R2. 4004000.
Mix 50.0 mL of a 136 giL solution of potassium dihydrogen Phosphate buffered saline pH 7.4. 4005000.
phosphate R with 29.5 mL of 1 M sodium hydroxide and dilute Dissolve 2.38 g of disodium hydrogen phosphate R, 0.19 g
to 100.0 mL with water R. Adjust the pH to 7.0 ± O.l. of potassium dihydrogen phosphate R and 8.0 g of sodium
chloride R in water. Dilute to 1000.0 mL with the same solvento
Phosphate buffer solution pH 7.0 R3. 4008600. Adjust the pH if necessary.
Dissolve 5 g of potassium dihydrogen phosphate R and 11 g
of dipotassium hydrogen phosphate R in 900 mL of water R. Phosphate buffer solution pH 7.4. 4004800.
Adjust to pH 7.0 with dilute phosphoric acid R or dilute sodium Add 250.0 mL of 0.2 M potassium dihydrogen phosphate R to
hydroxide solution R. Dilute to 1000 mL with water R and mix. 393.4 mL of 0.1 M sodium hydroxide.

Phosphate buffer solution pH 7.0 R4. 4010200. Tris(hydroxymethyl)aminomethane buffer solution pH 7.4.
4012100.
Dissolve 28.4 g of anhydrous disodium hydrogen phosphate R
Dissolve 30.3 g of tris(hydroxymethyl)aminomethane R in
and 18.2 g of potassium dihydrogen phosphate R in water R
approximately 200 mL of water R. Add 183 mL of 1 M
and dilute to 500 mL with the same solvento
hydrochloric acid. Dilute to 500.0 mL with water R. Note:
Phosphate buffer solution pH 7.0 R5. 4011400. the pH is 7.7-7.8 at room temperature and 7.4 at 37 oc. This
solution is stable jor several months at 4 oc.
Dissolve 28.4 g of anhydrous disodium hydrogen phosphate R
in 800 mL of water R. Adjust the pH using a 30 per cent mlm Tris(hydroxymethyl)aminomethane sodium chloride
solution of phosphoric acid R and dilute to 1000 mL with buffer solution pH 7,4. 4004900.
water R. Dissolve 6.08 g of tris(hydroxymethyl)aminomethane R, 8.77 g
of sodium chloride R in 500 mL of distilled water R. Add 10.0 g
Tetrabutylammonium buffer solution pH 7.0. 4010900. of bovine albumin R. Adjust the pH using hydrochloric acid R.
Dissolve 6.16 g of ammonium acetate R in a mixture of 15 mL Dilute to 1000.0 mL with distilled water R.
of tetrabutylammonium hydroxide solution (400 giL) R and
185 mL of water R. Adjust the pH with nitric acid R. Tris(hydroxymethyl)aminomethane sodium chloride
buffer solution pH 7,4 RL 4012200.
Buffered salí solution pH 7.2. 4004300. Dissolve 0.1 g of bovine albumin R in a mixture containing
Dissolve in water R 8.0 g of sodium chloride R, 0.2 g of 2 mL of tris(hydroxymethyl)aminomethane buffer solution
potassium chloride R, 0.1 g of anhydrous calcium chloride R, pH 7.4 R and 50 mL of a 5.84 mg/mL solution of sodium
0.1 g of magnesium chloride R, 3.18 g of disodium hydrogen chloride R. Dilute to 100.0 mL with water R.
phosphate R and 0.2 g of potassium dihydrogen phosphate R
Tris-sodium acetate buffer solution pH 7.4. 4012900.
and dilute to 1000.0 mL with water R.
Dissolve 6.3 g of tris(hydroxymethyl)aminomethane R and
Buffer solution pH 7.2. 4004100. 4.9 g of anhydrous sodium acetate R in 900 mL of water R.
To 250.0 mL of 0.2 M potassium dihydrogen phosphate R add Adjust to pH 7.4 with sulfuric acid R and dilute to 1000 mL
175.0 mL of 0.2 M sodium hydroxide. Dilute to 1000.0 mL with water R.
with water R. Adjust the pH if necessary. Tris-sodium acetate-sodium chloride buffer solution
pH 7.4. 4013000.
Phosphate-albumin buffered saline pH 7.2. 4004400.
Dissolve 30.0 g of tris(hydroxymethyl)aminomethane R, 14.5 g
Dissolve 10.75 g of disodium hydrogen phosphate R, 7.6 g of of anhydrous sodium acetate R and 14.6 g of sodium chloride R
sodium chloride R and 10 g of bovine albumin R in water R and in 900 mL of water R. Add 0.50 g oí bovine albumin R. Adjust
dilute to 1000.0 mL with the same solvent. Immediately before to pH 7.4 with sulfuric acid R and dilute to 1000 mL with
use adjust the pH using dilute sodium hydroxide solution R water R.
or dilute phosphorie acid R.
Borate buffer solution pH 7.5. 4005200.
Phosphate-albumin buffered saline pH 7.2 Rl. 4009600. Dissolve 2.5 g of sodium chloride R, 2.85 g of disodium
Dissolve 10.75 g of disodium hydrogen phosphate R, 7.6 g of tetraborate R and 10.5 g of boric aeid R in water R and dilute to
sodium ehloride R and 1 g of bovine albumin R in water R and 1000.0 mL with the same solvento Adjust the pH if necessary.
dilute to 1000.0 mL with the same solvent. Immediately before Storage: at 2 oC to 8 oc.
use adjust the pH using dilute sodium hydroxide solution R
or dilute phosphorie acid R. Buffer (HEPES) solution pH 7.5. 4009700.
Dissolve 2.38 g of 2-[4-(2-hydroxyethyl)piperazin-1-
Phosphate buffer solution pH 7.2. 4004200. yl]ethanesulfonic acid R in about 90 mL of water R. Adjust the
Mix 87.0 mL of a 71.5 giL solution of disodium hydrogen pH to 7.5 with sodium hydroxide so/ution R. Dilute to 100 mL
phosphate R with 13.0 mL of a 21 giL solution of eitrie acid R. with water R.

General No/ices (1) apply ío all monographs and other texts 543
4.1.3. Buffer solutions EUROPEAN PHARMACOPOEIA 8.0

0.05 M Phosphate buffer solution pH 7.5. 4014400. 1 M Tris-hydrochloride buffer solution pH 8.0. 4012700.
Dissolve 0.89 g of disodium hydrogen phosphate dihydrate R Dissolve 121.1 g of tris(hydroxymethyl)aminomethane R and
in about 80 mL of water R. Adjust to pH 7.5 with an 8.5 per 1.47 g of calcium ehloride R in 900 mL of water R. Adjust the
cent V/V solution of phosphorie acid R and dilute to 100.0 mL pH with hydroehloric acid R and dilute to 1000.0 mL with
with water R. water R.
0.2 M Phosphate buffer solution pH 7.5. 4005400. Tris-hydrochloride buffer solution pH 8.0. 4012300.
Dissolve 27.22 g of potassium dihydrogen phosphate R in Dissolve 1.21 g of tris(hydroxymethyl)aminomethane R and
930 mL of water R, adjust to pH 7.5 with a 300 giL solution of 29.4 mg of calcium chloride R in water R. Adjust the pH with
potassium hydroxide R and dilute to 1000.0 mL with water R. 1 M hydroehloric acid and dilute to 100.0 mL with water R.
0.33 M Phosphate buffer solution pH 7.5. 4005300. Tris-sodium acetate buffer solution pH 8.0. 4013100.
Dissolve 119.31 g of disodium hydrogen phosphate R in water R Dissolve 6.3 g of tris(hydroxymethyl)aminomethane R and
and dilute to 1000.0 mL with the same solvent (solution A). 4.9 g of anhydrous sodium acetate R in 900 mL of water R.
Dissolve 45.36 g of potassium dihydrogen phosphate R in Adjust to pH 8.0 with sulfurie acid R and dilute to 1000 mL
water R and dilute to 1000.0 mL with the same solvent with water R.
(solution B). Mix 85 mL of solution A and 15 mL of solution B.
Adjust the pH if necessary. Tris-sodium acetate-sodium chloride buffer solution
pH 8.0. 4013200.
0.05 M Tris-hydrochloride buffer solution pH 7.5. 4005600.
Dissolve 30.0 g oftris(hydroxymethyl)aminomethane R, 14.5 g
Dissolve 6.057 g of tris(hydroxymethyl)aminomethane R in
of anhydrous sodium aeetate R and 14.6 g of sodium ehloride R
water R and adjust the pH with hydroehloric acid R. Dilute to
in 900 mL of water R. Add 0.50 g of bovine albumin R. Adjust
1000.0 mL with water R.
to pH 8.0 with sulfurie acid R and dilute to 1000 mL with
1 M Tris-hydrochloride buffer solution pH 7.5. 4014500. water R.
Dissolve 12.11 g of tris(hydroxymethyl)aminomethane R in Tris(hydroxymethyl)aminomethane buffer solution pH 8.1.
90 mL of water R, adjust to pH 7.5 with hydroehloric aeid R 4006200.
and dilute to 100.0 mL with water R.
Dissolve 0.294 g of ealcium chloride R in 40 mL of
Tris(hydroxymethyl)aminomethane buffer solution pH 7.5. tris(hydroxymethyl)aminomethane solution R and adjust the
4005500. pH with 1 M hydrochloric acid. Dilute to 100.0 mL with
Dissolve 7.27 g of tris(hydroxymethyl)aminomethane R and water R.
5.27 g of sodium chloride R in water R, and adjust the pH if
Tris-glydne buffer solution pH 8.3. 4006300.
necessary. Dilute to 1000.0 mL with water R.
Dissolve 6.0 g of tris(hydroxymethyl)aminomethane R and
Sodium dirate buffer solution pH 7.8 (0.034 M sodium 28.8 g of glyeine R in water R and dilute to 1000.0 mL with the
dtrate, 0.101 M sodium chloride). 4009800. same solvento Dilute 1 volume to 10 volumes with water R
Dissolve 10.0 g of sodium citrate R and 5.90 g of sodium irnmediately before use.
ehloride R in 900 mL of water R. Adjust the pH by addition of
hydroehloric acid R and dilute to 1000 mL with water R. Tris-hydrochloride buffer solution pH 8.3. 4011800.
Dissolve 9.0 g of tris(hydroxymethyl)aminomethane R in 2.9 L
0.0015 M Borale buffer solution pH 8.0. 4006000. of water R. Adjust the pH with 1 M hydroehlorie acid. Adjust
Dissolve 0.572 g of disodium tetraborate R and 2.94 g of the volume to 3 L with water R.
ealeium eh/oride R in 800 mL of water R. Adjust the pH with
1 M hydroch/oric acid. Dilute to 1000.0 mL with water R. 0.05 M Tris-hydrochloride buffer solution pH 9.0. 4013500.
Dissolve 0.605 g of tris(hydroxymethyl)aminomethane R in
Buffer solution pH 8.0. 4005900.
water R. Adjust the pH with 1 M hydrochlorie acid and dilute
To 50.0 mL of 0.2 M potassium dihydrogen phosphate R add to 100.0 mL with water R.
46.8 mL of 0.2 M sodium hydroxide. Dilute to 200.0 mL with
water R. Barbital buffer solution pH 8.4. 4006400.
Buffer solution pH 8.0 Rl. 4010400. Dissolve 8.25 g of barbital sodium R in water R and dilute to
1000.0 mL with the same solvento
Dissolve 20 g of dipotassium hydrogen phosphate R in 900 mL
of water R. Adjust the pH with phosphorie acid R. Dilute to Tris-EDTA BSA buffer solution pH 8.4. 4006500.
1000 mL with water R. Dissolve 6.1 g of tris(hydroxymethyl)aminomethane R, 2.8 g
0.02 M Phosphate buffer solution pH 8.0. 4006100. of sodium edetate R, 10.2 g of sodium ehloride R and 10 g
of bovine albumin R in water R, adjust to pH 8.4 using 1 M
To 50.0 mL of 0.2 M potassium dihydrogen phosphate R add
hydroehloric acid and diIute to 1000.0 mL with water R.
46.8 mL of 0.2 M sodium hydroxide. Dilute to 500.0 mL with
water R. Tris(hydroxymethyl)aminomethane- EDTA buffer solution
0.02 M Sodium phosphate buffer solution pH 8.0. 4013700. pH 8.4. 4006600.
Dissolve 0.31 g of sodium dihydrogen phosphate R in 70 mL of Dissolve 5.12 g of sodium ehloride R, 3.03 g of
water R and adjust to pH 8.0 with 1 M sodium hydroxide, then tris(hydroxymethyl)aminomethane R and 1.40 g of sodium
dilute to 100 mL with water R. edetate R in 250 mL of distilled water R. Adjust the pH to 8.4
using hydroehlorie aeid R. Dilute to 500.0 mL with distilled
0.1 M Phosphate buffer solution pH 8.0. 4008400. water R.
Dissolve 0.523 g of potassium dihydrogen phosphate R and
Guanidine- tris (hydroxymethyl) aminomethane-EDTA
16.73 g of dipotassium hydrogen phosphate R in water R and
buffer solution pH 8.5. 4014600.
dilute to 1000.0 mL with the same solvent.
Dissolve 1.0 g of sodium edetate R, 12.1 g of
1 M Phosphate buffer solution pH 8.0. 4007800. tris(hydroxymethyl)aminomethane R and 57.0 g of
Dissolve 136.1 g of potassium dihydrogen phosphate R in guanidine hydrochloride R in 35 mL of water R. Adjust to
water R, adjust the pH with 1 M sodium hydroxide. Dilute to pH 8.5 with hydroehloric acid R and dilute to 100 mL with
1000.0 mL with water R. water R.

544 See the information seetion on general monographs (eover pages)


EUROPEAN PHARMACOPOEIA 8.0 4.2.1. Primary standards for volumetric solutions

Phosphate buffer solution pH 8.5. 4013300. Buffer solution pH 10.9. 4007600.


Dissolve 3.5 g of dipotassium hydrogen phosphate R and 4.5 g Dissolve 6.75 g of ammonium chloride R in ammonia R and
of sodium chloride R in 500 mL of water R. Adjust the pH dilute to 100.0 mL with the same solvent.
with a mixture of equal volumes of dilute phosphoric acid R
and water R. Total-ionic-strength-adjustment buffer. 4007700.
Dissolve 58.5 g of sodium chloride R, 57.0 mL of glacial
Tri.s acetate buffer solution pH 8.5. 4006700. acetic acid R, 61.5 g of sodium acetate R and 5.0 g of
Dissolve 0.294 g of calcium chloride R and 12.11 g of cyclohexylenedinitrilotetra-acetic acid R in water R and dilute
tris(hydroxymethyl)aminomethane R in water R. Adjust the to 500.0 mL with the same solvent. Adjust to pH 5.0 to 5.5
pH with acetic acid R. Dilute to 1000.0 mL with water R. with a 335 giL solution of sodium hydroxide R and dilute to
1000.0 mL with distilled water R.
Barbital buffer solution pH 8.6 Rl. 4006900.
Dissolve in water R 1.38 g of barbital R, 8.76 g of barbital Total-ionic-strength-adjustment buffer RL 4008800.
sodium R and 0.38 g of calcium lactate R and dilute to Dissolve 210 g of citric acid R in 400 mL of distilled water R.
1000.0 mL with the same solvent. Adjust to pH 7.0 with concentrated ammonia R. Dilute to
1000.0 mL with distilled water R (solution A). Dissolve
1.5 NI tris-hydrochloride buffer solution pH 8.8. 4009900.
132 g of ammonium phosphate R in distilled water R and
Dissolve 90.8 g of tris(hydroxymethyl)aminomethane R in dilute to 1000.0 mL with the same solvent (soIution B). To a
400 mL of water R. Adjust the pH with hydrochloric acid R suspension of 292 g of (ethylenedinitrilo )tetra-acetic acid R
and dilute to 500.0 mL with water R. in about 500 mL of distilled water R, add about 200 mL of
Buffer (phosphate) solution pH 9.0. 4008300. concentrated ammonia R to dissolve. Adjust the pH to 6 to
7 with concentrated ammonia R. Dilute to 1000.0 mL with
Dissolve 1.74 g of potassium dihydrogen phosphate R in 80 mL distilled water R (solution C). Mix equal volumes of solution A,
of water R, adjust the pH with 1 M potassium hydroxide and B, and C and adjust to pH 7.5 with concentrated ammonia R.
dilute to 100.0 mL with water R.
Buffer solution pH n. 4014000.
Buffer solution pH 9.0. 4007000.
Dissolve 6.21 g of borie aeid R, 4.00 g of sodium hydroxide R
Dissolve 6.18 g of borie acid R in 0.1 M potassium ehloride R
and 3.70 g of potassium chloride R in 500 mL of water R and
and dilute to 1000.0 mL with the same solvent. Mix 1000.0 mL
dilute to 1000 mL with the same solvento
of this solution and 420.0 mL of 0.1 M sodium hydroxide.
Buffer solution pH 9.0 Rl. 4007100.
Dissolve 6.20 g of boric acid R in 500 mL of water R and adjust 4.2. VOLUMETRIC ANALYSIS
the pH with 1 M sodium hydroxide (about 41.5 mL). Dilute to
1000.0 mL with water R.
Ammonium chloride buffer solution pH 9.5. 4007200. 04/2010:40201
Dissolve 33.5 g of ammonium ehloride R in 150 mL of water R,
add 42.0 mL of concentrated ammonia R and dilute to 4.2.1. PRIMARY STANDARDS FOR
250.0 mL with water R.
Storage: in a polyethylene container.
VOLUMETRIC SOLUTIONS
Primary standards for volumetrie solutions are indicated
Ammonium chloride buffer solution pH 10.0. 4007300.
by the suffix RV. Primary standards of suitable quality may
Dissolve 5.4 g of ammonium ehloride R in 20 mL of water R, be obtained from eommercial sourees or prepared by the
add 35.0 mL of ammonia R and dilute to 100.0 mL with following methods.
water R.
Arsenious trioxide. ASP3' (Me 197.8). 2000100. [1327-53-3].
Diethanolamine buffer solution pH 10.0. 4007500.
Sublime arsenious trioxide R in a suitable apparatus.
Dissolve 96.4 g of diethanolamine R in water R and diIute
to 400 mL with the same solvent. Add 0.5 mL of an 186 giL Storage: over anhydrous si/ica gel R.
solution of magnesium chloride R and adjust the pH with 1 M Benzoic add. C7H602' (Me 122.1).2000200. [65-85-0].
hydrochloric acid. Dilute to 500.0 mL with water R.
Sublime benzoie acid R in a suitable apparatus.
0.1 M Ammonium carbonate buffer solution pH 10.3.
4011900. Potassium bromate. KBr03' (Mr 167.0). 2000300.
[7758-01-2].
Dissolve 7.91 g of ammonium carbonate R in 800 mL
of water R. Adjust the pH with dilute sodium hydroxide Crystallise potassium bromate R from boiling water R. Collect
solution R. DiIute to 1000.0 mL with water R. the erystals and dry to eonstant mass at 180 oc.

Ammonium chloride buffer solution pH lOA. 4011000. Potassium hydrogen phthalate. CsHsK04' (Mr 204.2).
2000400. [877-24-7].
Dissolve 70 g of ammonium chloride R in 200 mL of water R,
add 330 mL of concentrated ammonia R and diIute to Reerystallise potassium hydrogen phthalate R from boiling
1000.0 mL with water R. lf neeessary, adjust to pH 10.4 with water R, eolleet the erystals at a temperature aboye 35 oC and
ammonia R. dry to eonstant mass at 110 oc.

Borate buffer solution pH lOA. 4011100. Sodium carbonate. Na2C0 3 . (Mr 106.0). 2000500.
Dissolve 24.64 g of boric acid R in 900 mL of distilled water R. [497-19-8].
Adjust the pH using a 400 giL solution of sodium hydroxide R. Filter at room temperature a saturated solution of sodium
Dilute to 1000 mL with distilled water R. carbonate R. Introduce slowly into the filtrate a stream of
carbon dioxide R with eonstant eooling and stirring. After
Ammonium chloride buffer solution pH 10.7. 4013400. about 2 h, eolleet the precipitate on a sin te red -glass filter
Dissolve 67.5 g of ammonium chloride R in water R, add (2.1.2). Wash the filter with ieed water R eontaining carbon
570 mL of concentrated ammonia R and dilute to 1000.0 mL dioxide. After drying at 100 oC to 105 oC, heat to eonstant
with water R. mass at 270-300 oC, stirring from time to time.

General Notices (1) apply to all monographs and other texts 545
4.2.2. Volumetric solutions EUROPEAN PHARMACOPOEIA 8.0

Sodium chloride. NaCl. (M, 58.44).2000600. [7647-14-5]. Standardisation. To 25.0 mL of the ammonium and cerium
To 1 volume of the saturated sodium ehloride solution R add sulfate solution add 2.0 g of potassium iodide R and 150 mL
2 volumes of hydrochloric acid R. Collect the crystals formed of water R. Titrate immediately with 0.1 M sodium thiosulfate,
and wash with hydrochloric acid R1. Remove the hydrochloric using 1 mL of starch solution R as indicator.
acid by heating on a water-bath and dry the crystals to
constant mas s at 300 oc. 0.01 M Ammonium ana cerium sulfate. 3000400.
To 100.0 mL of 0.1 M ammonium and cerium sulfate add, with
Sulfanilic add. C 6 H 7 N0 3 S. (M, 173.2). 2000700. [121-57-3]. cooling, 30 mL of sulÍuric acid R and dilute to 1000.0 mL
Recrystallise sulfanilic acid R from boiling water R. Filter and with water R.
dry to constant mass at 100-105 oc.
0.1 M Ammonium thiocyanate. 3000500.
Zinc. Zn. (Mr 65.4). 2000800. [7440-66-6].
Dissolve 7.612 g of ammonium thiocyanate R in water R and
Content: minimum 99.9 per cent. dilute to 1000.0 mL with the same solvent.
Standardisation. To 20.0 mL of 0.1 M sílver nitrate add 25 mL
04/2010:40202 of water R. 2 mL of dilute nitric acid R and 2 mL oí ferric
ammonium sulfate solution R2. Titrate with the ammonium
4.2.2. VOLUMETRIC SOLUTIONS thiocyanate solution until a reddish-yellow colour is obtained.
Volumetric solutions are prepared according to the usual 0.1 M Bal"ium chloride. 3000600.
chemical analytical methods. The accuracy of the apparatus
Dissolve 24.4 g of barium chloride R in water R and dilute to
used is verified to ensure that it is appropriate for the intended 1000.0 mL with the same solvento
use.
The concentration of volumetric solutions is indicated in terms Standardisation. To 10,0 mL of the barium chloride solution
of molarity. Molarity expresses, as the number of moles, the add 60 mL of water R, 3 mL of concentrated ammonia R and
amount of substance dissolved in 1 L of solution. A solution 0.5-1 mg of phthalein purple R, Titrate with 0.1 M sodium
which contains x moles of substance per litre is said to be x M. edetate. When the solution begins to decolorise, add 50 mL
of ethanol (96 per cent) R and continue the titratiol1 until the
Volumetric solutions do not differ from the prescribed blue-violet colour disappears.
strength by more than 10 per cent. The molarity of the
volumetric solutions is determined by an appropriate number 0.05 M Barium perchlorate. 3000700.
of titrations. The repeatability does not exceed 0.2 per cent
Dissolve 15.8 g of barium hydroxide R in a mixture of 7.5 mL
(relative standard deviation).
of perchloric acid R and 75 mL of water R, adjust the solution
Volumetric solutions are standardised by the methods to pH 3 by adding perchloric acid R and filter if necessary. Add
described below. When a volumetric solution is to be used 150 mL of ethanol (96 per cent) R and dilute to 250 mL with
in an assay in which the end-point is determined by an water R. Dilute to 1000.0 mL with buffer solution pH 3.7 R.
electrochemical process (for example, amperometry or
potentiometry) the solution is standardised by the same Standardisation. To 5.0 mL of 0.05 M sulfuric acíd add 5 mL
method. The composition of the medium in which a of water R, 50 rnL of buffer solution pH 3.7 R and 0.5 mL of
volumetric solution is standardised should be the same as that alizarin S so/ution R. Titrate with the barium perchlorate
in which it is to be used. solutiol1 ul1til an orange-red colour appears. Standardise
immediately before use.
Solutions more dilute than those described are obtained
by dilution with carbon dioxide-free water R of the 0.025 M Barium perchlorate. 3009600.
least -concentrated solution that describes a standardisation.
The correction Íactors of these solutions are the same as those Dilute 500.0 mL of 0.05 M barium perchlorate to 1000.0 mL
from which the dilutions were prepared. with buffer solution pH 3.7 R.

0.1 M Acetic add, 3008900. 0.004 M Benzethonium chloride. 3000900.


Dilute 6.0 g of glacial acetic acid R to 1000.0 mL with water R. Dissolve in water R 1.792 g of benzethonium chloride R,
Standardisation. To 25.0 mL of acetic acid add 0.5 mL of previously dried to constant mass at 100-105 oC, and dilute to
phenolphthalein solution R and titrate with 0.1 M sodium 1000.0 mL with the same solvent.
hydroxide. Standardisation. Calculate the molarity of the solution from
the content of C27H42CIN02 in the dried benzethonium
0.1 M Arnmonium and cerium nitrate. 3000100. chloride determined as follows. Dissolve 0.350 g of the dried
Shake for 2 min a solution containing 56 mL of sulfuric acid R substance in 30 mL of anhydrous acetic acid R and add 6 mL
and 54.82 g of ammonium and cerium nitrate R, add five of mercuric acetate solution R. Titrate with 0.1 M perchloric
successive quantities, each of 100 mL, of water R, shaking after acíd, using 0.05 mL of crystal violet solution R as indicator.
each addition. Dilute the clear solution to 1000.0 mL with Carry out a blank titration.
water R. Standardise the solution after 10 days.
1 mL of 0.1 M perchloric acid is equivalent to 44.81 mg of
Standardisation. To 25.0 mL of the ammonium and cerium Cn H 42 CIN0 2 •
nitrate solution add 2.0 g of potassium iodide R and 150 mL
of water R. Titrate immediately with 0.1 M sodium thiosulfate, 0.01 M Bismuth nitrate. 3010000.
using 1 mL of starch solution R as indicatoL Dissolve 4.86 g of bismuth nitrate pentahydrate R in 60 mL of
Storage: protected from light. di/u te nitric acid R and dilute to 1000.0 mL with water R.
0.01 M Arnmonium and cerium nitrate. 3000200. Standardisation. To 25.0 mL of the bismuth nitrate solution,
To 100.0 mL of 0.1 M ammonium and cerium nitrate add, add 50 mL of water R and titrate with 0.01 M sodium edetate
with cooling, 30 mL of sulfuric acid R and dilute to 1000.0 mL using 0.05 mL of a 1 giL solution of xylenol orange R as
with water R. indicator.

0.1 M Ammonium and cerium sulfate. 3000300. 0.0167 M Bromide-bromaíe. 3001000.


Dissolve 65.0 g of ammonium and cerium sulfate R in a Dissolve 2.7835 g of potassium bromate RV and 13 g of
mixture of 500 mL of water R and 30 mL of sulfuric acid R. potassium bromide R in water R and dilute to 1000.0 mL with
Allow to cool and dilute to 1000.0 mL with water R. the same solvent.

546 See the informatiol1 section on general monographs (ca ver pages)
EUROPEAN PHARMACOPOEIA 8.0 4.2.2. Volumetric solutioll§

0.1 M Cerium sulfate. 3001100. Standardisation. To 20.0 mL of the iodine solution add 1 mL
Dissolve 40.4 g of cerium sulfate R in a mixture of 500 mL of of dilute acetic acid R and 30 mL of water R. Titrate with 0.1 M
water R and 50 mL of sulfuric acid R. Allow to cool and dilute sodium thiosulfate, using starch solution R as indicator.
to 1000.0 mL with water R. Storage: protected from light.
Standardisation. To 20.0 mL of the cerium sulfate solution,
0.01 M Iodine. 3002900.
add 1.6 g of potassium iodide R, 100 mL of water R and 40 mL
of dilute sulfuric acid R. Titrate immediately with 0.1 M sodium Add 0.3 g of potassium iodide R to 20.0 mL of 0.05 M iodine
thiosulfate using 0.8 mL of stareh solution R as indicator. and dilute to 100.0 mL with water R.

0.02 M Copper sulfate. 3001200. 0.1 M Lanthanum nitrate. 3010100.


Dissolve 5.0 g of eopper sulfate R in water R and dilute to DissoIve 43.30 g of lanthanum nitrate R in water R and diIute
1000.0 mL with the same solvent. to 1000.0 mL with the same solvent.
Standardisation. To 20.0 mL of the copper sulfate solution Standardisation. To 20 mL of the lanthanum nitrate solution,
add 2 g of sodium aceta te R and 0.1 mL of pyridylazonaphthol add 15 mL of water R and 25 mL of 0.1 M sodium edetate.
solution R. Titrate with 0.02 M sodium edetate until the colour Add about 50 mg of xylenol orange triturate R and about 2 g
changes from violet-blue to bright green. Titrate slowly of hexamethylenetetramine R. Titrate with 0.1 M zine sulfate
towards the end of the titration. until the colour changes from yellow to violet-pink.
1 mL of 0.1 M sodium edetate is equivalent to 43.30 mg of
0.1 M Ferric ammonium sulfate. 3001300. La(N0 3 )3,6Hp.
Dissolve 50.0 g ofjerric ammonium sulfate R in a mixture of
6 mL of sulfuric acid R and 300 mL of water R and dilute to 0.1 M Lead nitrate. 3003100.
1000.0 mL with water R. Dissolve 33 g of lead nitrate R in water R and dilute to
Standardisation. To 25.0 mL of the ferric ammonium sulfate 1000.0 mL with the same solvent.
solution, add 3 mL of hydroehloric aeid R and 2 g of potassium Standardisation. Take 20.0 mL of the lead nitrate solution
iodide R. Allow to stand for 10 mino Titrate with 0.1 M sodium and carry out the determination of lead by complexometry
thiosulfate, using 1 mL of stareh so/ution R as indicator. (2.5.11).
1 mL of 0.1 M sodium thiosulfate is equivalent to 48.22 mg
0.05 M Lead nitrate. 3009700.
of FeNH 4 (S04)2,12Hp.
Dilute 50.0 mL of 0.1 M Lead nitrate to 100.0 mL with water R.
0.1 M Ferrous sulfate. 3001400.
Dissolve 27.80 g of jerrous sulfate R in 500 mL of dilute sulfuric 0.1 M Lithium methoxide. 3003300.
acid R and dilute to 1000.0 mL with water R. Dissolve 0.694 g of lithium R in 150 mL of anhydrous
methanol R and dilute to 1000.0 mL with toluene R.
Standardisation. To 25.0 mL of the ferrous sulfate solution add
3 mL of phosphoric acid R and titrate immediately with 0.02 M Standardisation. To 10 mL of dimethylformamide R add
potassium permanganate. Standardise immediately before use. 0.05 mL of a 3 giL solution of thymol blue R in methanol R
and titrate with the lithium methoxide solution until a
1 M Hydrochloric add. 3001800. pure blue colour is obtained. Immediately add 0.200 g of
Dilute 103.0 g of hydrochloric acid R to 1000.0 mL with benzoic acid RV. Stir to effect solution and titrate with the
water R. lithium methoxide solution until the pure blue colour is again
Standardisation. Dissolve 1.000 g of sodium carbonate RV in obtained. Protect the solution from atmospheric carbon
50 mL of water R, add 0.1 mL of methyl orange solution R dioxide throughout the titration. From the volume of titrant
and titrate with the hydrochloric acid until the solution just used in the second titration ascertain the exact strength of the
becomes yellowish-red. Boil for 2 mino The solution reverts to lithium methoxide solution. Standardise immediately before
yellow. Cool and continue the titration until a yellowish-red use.
colour is obtained. 1 mL of 0.1 M lithium methoxide is equivalent to 12.21 mg
1 mL of 1 M hydroehloric acid is equivalent to 53.00 mg of ofC 7 H 6 0 2 ·
Na 2 C0 3 • 0.1 M Magnesium chloride. 3003400.
0.1 M Hydrochloric add. 3002100. Dissolve 20.33 g of magnesium chloride R in water R and dilute
Dilute 100.0 mL of 1 M hydrochloric acid to 1000.0 mL with to 1000.0 mL with the same solvent.
water R. Standardisation. Carry out the determination of magnesium
Standardisation. Carry out the titration described for 1 M by complexometry (2.5.11).
hydrochloric aeid using 0.100 g of sodium carbonate RV
1 M Nitric add. 3003600.
dissolved in 20 mL of water R.
Dilute 96.6 g of nitrie acid R to 1000.0 mL with water R.
1 mL of 0.1 M hydroehloric acid is equivalent to 5.30 mg of
Na 2 C0 3 • Standardisation. Dissolve 1.000 g of sodium carbonate RV in
50 mL of water R, add 0.1 mL of methyl orange solution R
0.1 M Hydrochloric add, alcoholic. 3008800. and titrate with the nitric acid until the solution just becomes
Dilute 9.0 mL of hydrochloric acid R to 1000.0 mL with reddish-yellow; boil for 2 mino The solution reverts to yellow.
aldehyde-jree alcohol R. Cool and continue the titration until a reddish-yellow colour
is obtained.
0.5 M Iodine. 3009400.
1 mL of 1 M nitrie acid is equivalent to 53.00 mg of Na2C0 3.
Dissolve 127 g of iodine R and 200 g of potassium iodide R in
water R and dilute to 1000.0 mL with the same solvent. 0.1 M Perchloric add. 3003900.
Standardisation. To 2.0 mL of the iodine solution add 1 mL of Place 8.5 mL of perchlorie acid R in a volumetric flask
dilute acetic acid R and 50 rnL of water R. Titrate with 0.1 M containing about 900 mL of glacial acetic acid R and mix. Add
sodium thiosulfate, using starch solution R as indicator. 30 mL of acetic anhydride R, dilute to 1000.0 mL with glacial
Storage: protected from light. acetic acid R, mix and allow to stand for 24 h. Determine the
water content (2.5.12) without addition of methanol and,
0.05 M Iodine. 3002700. if necessary, adjust the water content to 0.1-0.2 per cent by
Dissolve 12.7 g of iodine R and 20 g of potassium iodide R in adding either acetie anhydride R or water R. Allow to stand
water R and dilute to 1000.0 mL with the same solvent. for 24 h.

General Notices (1) apply to all monographs and other texts 547
4.2.2. Volumetric solutions EUROPEAN PHARMACOPOEIA 8.0

Standardisation. Dissolve 0.350 g of potassium hydrogen 0.5 M Potassium hydroxide in alcohol (60 per cent V/V).
phthalate RV in 50 mL of anhydrous acetie aeid R, warming 3004900.
gently if necessary. Allow to cool protected from the air, and Dissolve 3 g of potassium hydroxide R in aldehyde-free
titrate with the perchloric acid solution, using 0.05 mL of alcohol R (60 per cent V/V) and dilute to 100.0 mL with the
crystal violet solution R as indicator. Note the temperature of same solvent.
the perchloric acid solution at the time of the titration. If the
Standardisation. Titrate 20.0 mL of the alcoholic potassium
temperature at which an assay is carried out is different from
that at which the 0.1 M perchlorie acid has been standardised, hydroxide solution (60 per cent V/V) with 0.5 M hydrochloric
aeid, using 0.5 mL of phenolphthalein solution R as indicator.
the volume used in the assay beco mes :
0.5 M Potassium hydl'Oxide, alcoholic. 3005000.
Dissolve 3 g of potassium hydroxide R in 5 mL of water R and
temperature during standardisation, dilute to 100.0 mL with aldehyde-free alcohol R.
Standardisation. Titrate 20.0 mL of the alcoholic potassium
temperature during the assay,
hydroxide solution with 0.5 M hydrochlorie acid, using 0.5 mL
corrected volume, of phenolphthalein solution R as indicator.
v observed volume. 0.1 M Potassium hydroxirle, akoholic. 3005100.
1 mL of 0.1 M perchlorie acid is equivalent to 20.42 mg Dilute 20.0 mL of 0.5 M alcoholic potassium hydroxide to
ofC sH sK0 4 · 100.0 mL with aldehyde-free alcohol R.

0.05 M Perchloric add. 3004000. 0.01 M Potassium hydroxide, alcohoHc. 3009000.


Dilute 50.0 mL of 0.1 M perchlorie acid to 100.0 mL with Dilute 2.0 mL of 0.5 M alcoholic potassium hydroxide to
anhydrous acetie acid R. 100.0 mL with aldehyde-free alcohol R.

0.02 M Perchloric add. 3009900. 0.05 M Potassium iodate. 3005200.


Dilute 20.0 mL of 0.1 M perchlorie acid to 100.0 mL with Dissolve 10.70 g of potassium iodate R in water R and dilute to
anhydrous aeetie acid R. 1000.0 mL with the same solvent.
Standardisation. Dilute 25.0 mL of the potassium iodate
0.033 M Poiassium bromate. 3004200.
solution to 100.0 mL with water R. To 20.0 mL of this solution
Dissolve 5.5670 g of potassium bromate RV in water R and add 2 g of potassium iodide R and 10 mL of dilute sulfuric
dilute to 1000.0 mL with the same solvent. acid R. Titrate with 0.1 M sodium thiosulfate, using 1 mL of
starch solution R, added towards the end of the titration, as
0.02 M Potassium bromate. 3004300.
indicator.
Dissolve 3.340 g of potassium bromate RV in water R and
dilute to 1000.0 mL with the same solvent. 0.001 M Potassium iodide. 3009200.
0.0167 M Poiassium bromate. 3004400. Dilute 10.0 mL of potassium iodide solution R to 100.0 mL
with water R. Dilute 5.0 mL of this solution to 500.0 mL with
Prepare by diluting 0.033 M Potassium bromate. water R.
0.0083 M Poíassium bromate. 3004500. 0.02 M Potassium permanganate. 3005300.
Prepare by diluting 0.033 M Potassium bromate. Dissolve 3.2 g of potassium permanganate R in water R and
0.0167 M Potassium dichromate. 3004600. dilute to 1000.0 mL with the same solvento Heat the solution
for 1 h on a water-bath, allow to cool and filter through a
Dissolve 4.90 g of potassium dichromate R in water R and sintered-glass filter (2.1.2).
dilute to 1000.0 mL with the same solvent.
Standardisation. To 20.0 mL of the potassium permanganate
Standardisation. To 20.0 mL of the potassium dichromate solution, add 2 g of potassium iodide R and 10 mL of di/ute
solution add 1 g of potassium iodide R and 7 mL of di/u te sulfuric acid R. Titrate with 0.1 M sodium thiosulfate, using
hydrochloric acid R. Add 250 mL of water R and titrate with 1 mL of starch solution R, added towards the end of the
0.1 M sodium thiosulfate, using 3 mL of starch solution R as titratiol1, as indicator. Stalldardise immediately before use.
indicator, until the colour changes from blue to light green.
Storage: protected from light.
0.1 M Potassium hyrll'Ogen phthalate. 3004700.
0.1 M Silver nitrate. 3005600.
In a conical flask containing about 800 mL of anhydrous acetic
acid R, dissolve 20.42 g of potassium hydrogen phthalate RV. Dissolve 17.0 g of si/ver nitrate R in water R and dilute to
Heat on a water-bath until completely dissolved, protected 1000.0 mL with the same solvento
from humidity. Cool to 20 oC and dilute to 1000.0 mL with Standardisation. Dissolve 0.100 g of sodium chloride RVin
anhydrous aeetic acid R. 30 mL of water R. Titrate with the sil ver nitrate solution,
determining the end-point potentiometrically (2.2.20).
1 M Potassium hyrlroxide. 3009100.
1 mL of 0.1 M si/ver nitrate is equivalent to 5.844 mg ofNaCl.
Dissolve 60 g of potassium hydroxide R in carbon dioxide-free
Storage: protected from light.
water R and dilute to 1000.0 mL with the same solvent.
Standardisation. Titrate 20.0 mL of the potassium hydroxide 0.001 M Silver nitrate. 3009300.
solution with 1 M hydrochloric acid, using 0.5 mL of Dilute 5.0 mL of silver nitrate 0.1 M to 500.0 mL with water R.
phenolphthalein solution R as indicator.
0.1 M Sodium arsenite. 3005800.
0.1 M Potassium hyrlroxide. 3004800.
Dissolve arsenious trioxide RV equivalent to 4.946 g of ASP3
Dissolve 6 g of potassium hydroxide R in carbon dioxide-free in a mixture of 20 mL of strong sodium hydroxide solution R
water R and dilute to 1000.0 mL with the same solvent. and 20 mL of water R, dilute to 400 mL with water R and add
Standardisation. Titrate 20.0 mL of the potassium hydroxide dilute hydrochloric acid R until the solution is neutral to litmus
solution with 0.1 M hydrochloric acid, using 0.5 mL of paper R. Dissolve 2 g of sodium hydrogen carbonate R in the
phenolphthalein solution R as indicator. solution and dilute to 500.0 mL with water R.

548 See the information section on general monographs (cover pages)


EUROPEAN PHARMACOPOEIA 8.0 4.2.2. Volumetdc solutions

0.1 M Sodium edetate. 3005900. Standardisation. Dissolve 0.100 g of benzoic acid RVin 2 mL
Dissolve 37.5 g of sodium edetate R in 500 mL of water R, add of water R and 10 mL of ethanol (96 per cent) R. Titrate
100 mL of 1 M sodium hydroxide and dilute to 1000.0 mL with the ethanolic sodium hydroxide solution, using 0.2 mL
with water R. of thymolphthalein solution R as indicator. Standardise
immediately before use.
Standardisation. Dissolve 0.120 g of zinc RV in 4 mL of
1 mL of 0.1 M ethanolic sodium hydroxide is equivalent to
hydrochloric acid R1 and add 0.1 mL of bromine water R.
12.21 mg of C7HP2'
Drive off the excess of bromine by boiling, add dilute sodium
hydroxide solution R until the solution is weakly acid or neutral 0.1 M Sodium methoxide. 3007100.
and carry out the assay of zinc by complexometry (2.5.11).
Cool175 mL of anhydrous methanol R in iced water R and add,
1 mL of 0.1 M sodium edetate is equivalent to 6.54 mg of Zn. in small portions, about 2.5 g of freshly cut sodium R. When
Storage: in a polyethylene container. the metal has dissolved, dilute to 1000.0 mL with toluene R.
Standardisation. To 10 mL of dimethylformamide R add
0.02 M Sodium edetate. 3006000. 0.05 mL of a 3 giL solution of thymol blue R in methanol R,
Dissolve 7.444 g of sodium edetate R in water R and dilute to and titrate with the sodium methoxide solution until a
1000.0 mL with the same solvent. pure blue colour is obtained. Immediately add 0.200 g of
benzoic acid RV. Stir to effect solution and titrate with the
Standardisation. Dissolve 0.100 g of zinc RV in 4 mL of sodium methoxide solution until the pure blue colour is again
hydrochloric acid R1 and add 0.1 mL of bromine water R. Drive obtained. Protect the solution from atmospheric carbon
off the excess of bromine by boiling. Transfer the solution to a dioxide throughout the titratiol1. From the volume of titrant
volumetric flask and dilute to 100.0 mL with water R. Transfer used in the second titration ascertain the exact strength of the
25.0 mL of the solution to a 500 mL conical flask and dilute sodium methoxide solution. Standardise immediately before
to 200 mL with water R. Add about 50 mg of xylenol orange use.
triturate R and hexamethylenetetramine R until the solution
1 mL of 0.1 M sodium methoxide is equivalent to 12.21 mg
becomes violet-pink. Add 2 g of hexamethylenetetramine R
ofC 7H 6 0 2 •
in excess. Titrate with the sodium edetate solution until the
violet-pink colour changes to yellow. 0.1 M Sodium nitrite. 3007200.
1 mL of 0.02 M sodium edetate is equivalent to 1.308 mg of Zn. Dissolve 7.5 g of sodium nitrite R in water R and dilute to
1000.0 mL with the same solvent.
1 M Sodium hydroxide. 3006300.
Standardisation. Dissolve 0.300 g of sulfanilic acid RV in 50 mL
Dissolve 42 g of sodium hydroxide R in carbon dioxide-free of dilute hydrochloric acid R and carry out the determination
water R and dilute to 1000.0 mL with the same solvento of primary aromatic amino-nitrogen (2.5.8), using the
Standardisation. Titrate 20.0 mL of the sodium hydroxide sodium nitrite solution and determining the end-point
solution with 1 M hydrochloric acid using the indicator e1ectrometrically. Standardise immediate1y before use.
prescribed in the assay in which 1 M sodium hydroxide is used. 1 mL of 0.1 M sodium nitrite is equivalent to 17.32 mg of
lf sodium hydroxide free from carbonate is prescribed, prepare C6 H 7 N0 3S.
it as follows. Dissolve sodium hydroxide R in water R to give a 0.1 M Sodium periodate. 3009500.
concentration of 400-600 giL and allow to stand. Decant the
clear supernatant, taking precautions to avoid the introduction Dissolve 21.4 g of sodium periodate R in about 500 mL of
of carbon dioxide, and dilute with carbon dioxide-free water R water R and dilute to 1000.0 mL with the same solvent.
to the required molarity. The solution complies with the Standardisation. In a stoppered flask, introduce 20.0 mL of the
following test. Titrate 20.0 mL of hydrochloric acid of the sodium periodate solutiol1 and add 5 mL of perchloric acid R.
same molarity with the solution of sodium hydroxide, using Close the flask and shake. Adjust the solution to pH 6.4 using
0.5 mL of phenolphthalein solution R as indicator. At the a saturated solution of sodium hydrogen carbonate R. Add
end-point add just sufficient of the acid to discharge the pink 10 mL of potassium iodide solution R, close, shake and allow to
colour and concentrate the solution to 20 mL by boiling. stand for 2 mino Titrate with 0.025 M sodium arsenite until the
During boiling add just sufficient acid to discharge the pink yellow colour almost disappears. Add 2 mL of starch solution R
colour, which should 110t reappear after prolonged boiling. and titrate slowly until the colour is completely discharged.
The volume of acid used does not exceed 0.1 mL.
0.1 M Sodium thiosulfate. 3007300.
0.1 M Sodium hydroxide. 3006600. Dissolve 25 g of sodium thiosulfate R and 0.2 g of sodium
Dilute 100.0 mL of 1 M sodium hydroxide to 1000.0 mL with carbonate R in carbon dioxide-free water R and dilute to
carbon dioxide-free water R. 1000.0 mL with the same solvent.
Standardisation. To 10.0 mL of 0.033 M potassium bromate,
Standardisation. Titrate 20.0 mL of the sodium hydroxide
add 40 mL of water R, 10 mL of potassium iodide solution R
solution with 0.1 M hydrochloric acid, USi11g the e11d-poi11t
and 5 mL of hydrochloric acid Rl. Titrate with the sodium
detection prescribed for the assay in which the 0.1 M sodium
thiosulfate solution, using 1 mL of starch solution R, added
hydroxide is used.
towards the end of the titration, as indicator.
Standardisation (for use in the assay of halide salts of organic
bases). Dissolve 0.100 g of benzoic acid RV in a mixture of 0.5 M Sulfuric add. 3007800.
5 mL of 0.01 M hydrochloric acid and 50 mL of ethanol (96 per Dissolve 28 mL of sulfuric acid R in water R and dilute to
cent) R. Carry out the titration (2.2.20), using the sodium 1000.0 mL with the same solvent.
hydroxide solution. Note the volume added between the 2
Standardisation. Dissolve 1.000 g of sodium carbonate RV in
points of inflexion.
50 mL of water R, add 0.1 mL of methyl orange solution R,
1 mL of 0.1 M sodium hydroxide is equivalent to 12.21 mg and titrate with the sulfuric acid until the solution begins to
of C7H60Z' turn reddish-yellow. Boil for about 2 mino The colour of the
solutions reverts to yellow. Cool and titrate again until the
0.1 M Sodium hydroxide, ethanolic. 3007000. reddish -yellow colour reappears.
To 250 mL of anhydrous ethanol R add 3.3 g of strong sodium 1 mL of 0.5 M sulfuric acid is equivalent to 53.00 mg of
hydroxide solution R. Na 2 C0 3 •

General Notices (1) apply to all monographs and other texts 549
4.2.2. Volumetric solutions EUROPEAN PHARMACOPOEIA 8.0

0.05 M Sulfmic add. 3008000. blue colour is again obtained. Protect the solution from
Dilute 100.0 mL of 0.5 1VI sulfuric acid to 1000.0 mL with atmospheric carbon dioxide throughout the titration. From
water R. the volume of titrant used in the second titration ascertain the
exact strength of the tetrabutylammonium hydroxide solution.
Standardisation. Carry out the titration described for 0.5 M
Standardise immediately before use.
sulfuric acid, using 0.100 g of sodium carbonate RV, dissolved
in 20 mL of water R. 1 mL of 0.1 M tetrabutylammonium hydroxide is equivalent
to 12.21 mg of C7H602'
1 mL of 0.05 M sulfuric acid is equivalent to 5.30 mg of
Na 2C0 3 • 0.1 M Tetrabutylammonium hydroxide in 2-propanol.
3008400.
0.1 M Tetl'abutylammonium hydroxide. 3008300.
Prepare as described for 0.1 M tetrabutylammonium hydroxide
Dissolve 40 g of tetrabutylammonium iodide R in 90 mL of using 2-propanol R instead of toluene R and standardise as
anhydrous methanol R, add 20 g of finely powdered si/ver described.
oxide R and shake vigorously for 1 h. Centrifuge a few
millilitres of the mixture and test the supernatant for iodides. 0.05 M Zinc chloride. 3008500.
If a positive reaction is obtained, add an additional2 g of si/ver Dissolve 6.82 g of zinc chloride R, weighed with appropriate
oxide R and shake for a further 30 mino Repeat this procedure precautions, in water R. If necessary, add dropwise dilute
until the liquid is free from iodides, filter the mixture through hydrochloric acid R until 'che opalescence disappears. Dilute to
a fine sintered-glass filter (2.1.2) and rinse the reaction vessel 1000.0 mL with water R.
and filter with three quantities, each of 50 mL, of toluene R. Standardisation. To 20.0 mL of the zinc chloride solution add
Add the washings to the filtrate and dilute to 1000.0 mL with 5 mL of dilute acetic acid R and carry out the determination of
toluene R. Pass dry carbon dioxide-free nitro gen through the zinc by complexometry (2.5.11).
solution for 5 mino
0.1 M Zinc sulfate. 3008600.
Standardisation. To 10 mL of dimethylformamide R add
0.05 mL of a 3 giL solution of thymol blue R in methanol R Dissolve 29 g of zinc sulfate R in water R and dilute to
and titrate with the tetrabutylammonium hydroxide solution 1000.0 mL with the same solvent.
unti! apure blue colour is obtained. Immediately add 0.200 g Standardisation. To 20.0 mL of the zinc sulfate solution add
of benzoic acid RV. Stir to effect solution, and titrate with 5 mL of dilute acetic acid R and carry out the determination of
the tetrabutylammonium hydroxide solution until the pure zinc by complexometry (2.5.11).

550 See the information section on general monographs (ca ver pages)
EUROPEAN PHARMACOPOEIA 8.0

5 General texts
@

General Natices (1) apply ta all managraphs and ather texts 551
EUROPEAN PHARMACOPOEIA 8.0

552 See the information section on general monographs (caver pages)


EUROPEAN PHARMACOPOEIA 8.0

5.1. General texts on microbiology


5.1. General texts on microbiology. ....................................... 555 5.1.6. Alternative methods for control of microbiological
5.1.1. Methods of preparation of sterile products ................ 555 quality....................................................................................... 560
5.1.2. Biological indicators of sterilisation ............................ 556 5.1.7. Viral safety. ...................................................................... 571
5.1.3. Efficacy of antimicrobial preservation ........................ 557 5.1.8. Microbiological quality ofherbal medicinal products for
5.1.4. Microbiological quality of non-sterile pharmaceutical oral use and extracts used in their preparation .................. 571
preparations and substances for pharmaceutical use ......... 559 5.1.9. Guidelines for using the test for sterility..................... 572
5.1.5. Application of the Fo concept to steam sterilisation of 5.1.10. Guidelines for using the test for bacterial
aqueous preparations ............................................................. 560 endotoxins ............................................................................... 572

General Notices (1) apply to all monographs and other texts 553
EUROPEAN PHARMACOPOEIA 8.0

554 See the information section on general monographs (cover pages)


EUROPEAN PHARMACOPOEIA 8,0 5,1.1. Methods of preparation of sterile products

5.1. GENERAL TEXTS ON guaranteed nor can it be demonstrated, The inactivation


of micro-organisms by physical or chemical means follows
MICROBIOLOGY an exponentiallaw; thus there is always a finite statistical
probability that a micro-organism may survive the sterilising
0112008:50101 process, For a given process, the probability of survival
is determined by the number, types and resistance of the
5.1.1. METHODS OF PREPARATION OF the organisms existpresent micro-organisms and by the environment in which
during treatmenL The SAL of a sterilising
STERILE PRODUCTS process is the degree of assurance with which the process in
Sterility is the absence of viable micro-organisms, The sterility question renders a population of items sterile, The SAL for a
of a product cannot be guaranteed by testing; it has to be given process is expressed as the probability of a non-sterile
item in that population, An SAL of 10- 6, for example, denotes
assured by the application of a suitably validated production
process, lt is essential that the effect of the chosen sterilisation a probability of not more than one viable micro-organism
procedure on the product (induding its final container or in 1 x 10 6 sterilised items of the final producL The SAL of

package) is investigated to ensure effectiveness and the a process for a given product is established by appropriate
validation studies,
integrity of the product and that the procedure is validated
before being applied in practice, It is recommended that METHODS AND CONDITIONS OF STERILISATION
the choice of the container is such as to allow the optimum
Sterilisation may be carried out by one of the methods
sterilisation to be applied, Failure to follow meticulously a
described be!ow, Modifications to, or combinations of, these
validated process involves the risk of a non-sterile product
methods may be used provided that the chosen procedure
or of a deteriorated product. Revalidation is carried out
is validated both with respect to its effectiveness and the
whenever major changes in the sterilisation procedure,
integrity of the product including its container or package,
induding changes in the load, take place, It is expected that
the principIes of good manufacturing practice (as described For all methods of sterilisation the critical conditions of
in, for example, the European Community Guide to GMP) the operation are monitored in order to confirm that the
will have been observed in the design of the process including, previously determined required conditions are achieved
in particular, the use of: throughout the batch during the whole sterilisation process
This applies in all cases including those where the reference
- qualified personnel with appropriate training,
conditions are used,
- adequate premises,
TERMINAL STERILISA TION
- suitable production equipment, designed for easy cleaning
and sterilisation, For terminal sterilisation it is essential to take into account the
non-uniformity of the physical and, where relevant, chemical
- adequate precautions to minimise the bioburden prior to conditions within the sterilising chamber, The location within
sterilisation, the sterilising chamber that is least accessible to the sterilising
- validated procedures for all critical production steps, agent is determined for each loading configuration of each
- environmental monitoring and in-process testing type and size of container or package (for example, the coolest
procedures, location in an autoclave), The minimum lethality delivered by
The precautions necessary to minimise the pre-sterilisation the sterilising cycle and the reproducibility of the cyele are also
bioburden indude the use of components with an acceptable determined in order to ensure that allloads will consistently
low degree of microbial contamination, Microbiological receive the specified treatment
monitoring and setting of suitable action Iimits may be Having established a terminal sterilisation process, knowledge
advisable for ingredients which are liable to be contaminated of its performance in routine use is gained wherever possible,
because of their origin, nature or method of preparation, by monitoring and suitably recording the physica! and, where
The methods described here apply mainly to the inactivation re!evant, chemical conditions achieved within the load in the
or removal of bacteria, yeasts and moulds, For biological chamber throughout each sterilising cycleo
products of animal or human origin or in cases where such Steam sterilisation (Heating in an autoclave), Sterilisation
material has been used in the production process, it is necessary by saturated steam under pressure is preferred, wherever
during validation to demonstrate that the process is capable of applicable, especially for aqueous preparations, For this
the removal or inactivation of relevant viral contamination, method of terminal sterilisation the reference conditions for
Guidance on this aspect is provided in, for example, the aqueous preparations are heating at a minimum of 121°C for
appropriate European Community Notes for Guidance, 15 min, Other combinations of time and temperature may be
Wherever possible, a process in which the product is sterilised used provided that it has been satisfactorily demonstrated that
in its final container (terminal sterilisation) is chosen, When the process chosen delivers an adequate and reproducible level
a fully validated terminal sterilisation method by steam, dry of lethality when operating routinely within the established
heat or ionising radiation is used, parametric re!ease, that is tolerances, The procedures and precautions employed are
the reIease of a batch of sterilised items based on process data such, as to give an SAL of 10- 6 or betteL Guidance concerning
rather than on the basis of submitting a sample of the items to validation by means of the Fo concept is provided below (5,15),
sterility testing, may be carried out, subject to the approval of Knowledge of the physical conditions (temperature and
the competent authority, pressure) within the autoclave chamber during the sterilisation
lf terminal sterilisation is not possible, filtration through pro ce dure is obtained, The temperature is usually measured
a bacteria -retentative filter or aseptic processing is used; by means of temperature-sensing elements inserted into
wherever possible, appropriate additional treatment of the representative container s together with additional e!ements at
product (for example, heating of the product) in its final the previously established coolest part of the loaded chamber,
container is applied, In al! cases, the container and elosure are The conditions throughout each cycle are suitably recorded,
required to maintain the sterility of the product throughout for example, as a temperature- time chart, or by any other
its shelf-lik suitable means,
Sterility Assurance Leve! (SAL) Where a biological assessment is carried out, this is obtained
Where appropriate reference is made within the methods using a suitable biological indicator (5,12),
described below, to a "sterility assurance leve!" or "SAL', The Dry heat sterilisation, For this method of terminal
achievement of sterility within any one ítem in a population sterilisation the reference conditions are a minimum of 160 oC
of items submitted to a sterilisation process cannot be for at least 2 h, Other combinations of time and temperature

General No/ices (1) apply to all monographs and o/her texts 555
5.1.2. Biologkal indicators of sterilisation EUROPEAN PHARMACOPOEIA 8.0

may be used provided that it has been satisfactorily satisfactory by means of a microbial challenge test using a
demonstrated that the process chosen delivers an adequate and suitable test micro-organismo A suspension of Pseudo monas
reproducible leve! of lethality when operated routinely within diminuta (ATCC 19146, NCIMB 11091 or CIP 103020) may
the established tolerances. The procedures and precautions be suitable. It is recommended that a challenge of at least
employed are such as to give an SAL of 10- 6 or better. 10 7 CPU per cm 2 of active filter surface is used and that the
Dry heat sterilisation is carried out in an oven equipped suspension is prepared in tryptone soya broth which, after
with forced air circulation or other equipment specially passage throug the filter, is collected aseptically and incubated
designed for the purpose. The steriliser is loaded in such aerobically at 32 oc. Such products need special precautions.
a way that a uniform temperature is achieved throughout The production process and environment are designed to
the load. Knowledge of the temperature within the steriliser minimise microbial contamination and are regularly subjected
during the sterilisation procedure is usually obtained to appropriate monitoring procedures. The equipment,
by means of temperature-sensing e!ements inserted into containers and closures and, wherever possible, the ingredients
representative containers together with additional e!ements at are subjected to an appropriate sterilisation process. It is
the previously established coolest part of the loaded steriliser. recommended that the filtration process is carried out as
The temperature throughout each cyde is suitably recorded. close as possible to the filling point. The operations following
Where a biological assessment is carried out, this is obtained filtration are carried out under aseptic conditions.
using a suitable biological indicator (5.1.2). Solutions are passed through a bacteria-retentive membrane
Dry heat at temperatures greater than 220 oC is frequently with a nominal pore size of 0.22 flm or less or any other
used for sterilisation and depyrogenation of glassware. In this type of filter known to have equivalent properties of bacteria
case demonstration of a 3 10g¡O reduction in heat resistant retention. Appropriate measures are taken to avoid 10ss of
endotoxin can be used as a replacement for biological solute by adsorption on to the filter and to avoid the releas e
indicators (5.1.2). of contaminants from the fiJter. Attention is given to the
bioburden prior to filtration, fiIter capacity, batch size and
Ionising radiation sterilisation. Sterilisation by this method duration of filtration. The filter is not used for a longer period
is achieved by exposure of the product to ionising radiation than has been approved by validation of the combination of
in the form of gamma radiation from a suitable radioisotopic the filter and the product in question.
source (such as cobalt 60) or of a beam of e1ectrons energised
The integrity of an assembled sterilising filter is verified before
by a suitable electro n accelerator.
use and confirmed after use by carrying out tests appropriate
In sorne countries there are regulations that lay down rules to the type of filter used and the stage of testing, for example
for the use of ionising radiation for sterilisation purposes, for bubble-point, pressure hold or diffusion rate tests.
example, in the appropriate European Community Notes for
Due to the potential additional risks of the filtration method
Guidance.
as compared with other sterilisation processes, a prefiltration
Por this method of terminal sterilisation the reference through a bacteria-retentative filter may be advisable in cases
absorbed dose is 25 kGy. Other doses may be used provided where a low bioburden cannot be ensured by other means.
that it has satisfactorily been demonstrated that the dos e
chosen delivers an adequate and reproducible level of lethality ASEPTIC PREPARATION
when the process is operated routinely within the established The obj ective of aseptic processing is to maintain the sterility
tolerances. The procedures and precautions employed are of a product that is assembled from components, each of
such as to give an SAL of 10- 6 or better. which has been sterilised by one of the aboye methods. This is
achieved by using conditions and facilities designed to prevent
During the sterilisation procedure the radiation absorbed by
microbial contamination. Aseptic processing may in dude
the product is monitored regularly by means of established
aseptic filling of products into container / dosure systems,
dosimetry procedures that are independent of dose rateo
aseptic blending of formulations followed by aseptic filling
Dosimeters are calibrated against a standard source at a
and aseptic packaging.
reference radiation plant on receipt from the supplier and at
suitable interyals of not longer than one year thereafter. In order to maintain the sterility of the components and the
Where a biological assessment is carried out, this is obtained product during processing, careful attention needs to be given
using a suitable biological indicator (5.1.2). to:
- environment,
Gas sterilisation. This method of sterilisation is only to be
used where there is no suitable alternative. It is essential - personnel,
that penetration by gas and moisture into the material to - critical surfaces,
be sterilised is ensured and that it is followed by a process - container / dosure sterilisation and transfer procedures,
of elimination of the gas under conditions that have been - maximum holding period of the product before filling into
previously established to ensure that any residue of gas or its the final container.
transformation products in the sterilised product is below the
Process validation indudes appropriate checks on al! the
concentration that could give rise to toxic effects during use of
aboye and checks on the process are regularly carried out by
the producto Guidance on this aspect with respect to the use
means of process simulation tests using microbial growth
of ethylene oxide is provided, for example, in the appropriate
media which are then incubated and examined for microbial
European Community Notes for Guidance.
contamination (media fi11 tests). In addition, a suitable sample
Wherever possible, the gas concentration, relative humidity, of each batch of any product that is sterilised by filtration
temperature and duration of the process are measured and/or aseptically processed is tested for sterility (2.6.1) befare
and recorded. Measurements are made where sterilisation the batch is released.
conditions are least likely to be achieved, as determined at
validation.
01/2011:50102
The effectiveness of the process applied to each sterilisation
load is checked using a suitable biological indicator (5.1.2).
A suitable sample of each batch is tested for sterility (2.6.1)
5.1.2. BIOLOGICAL INDICATORS OF
before the batch is released. STERILISATION
FILTRATION Biological indicators are standardised preparatiol1s of
Certain active ingredients and products that cannot be selected micro-organisms used to assess the effectiyeness
terminally sterilised may be subjected to a filtration procedure of a sterilisation procedure. They usually consist of a
using a filter of a type that has been demonstrated to be population of bacterial spores placed on a suitable inert

556 See the information section on general mOl1ographs (cover pages)


EUROPEAN PHARMACOPOEIA 8.0 5.1.3. Efficacy of antimicrobial preservation

carrier. The inoculated carrier is covered in such a way that it radiation energy, especially in the case of accelerated electron
is protected from any deterioration or contamination, while sterilisatiol1. The spores of Bacillus pumilus (for example,
allowing the sterilising agent to enter into contact with the ATCC 27142, NCTC 10327, NCIMB 10692 or CIP 77.25)
micro-organisms. Spore suspensions may be presented in or other strains of micro-organisms having demonstrated
sealed ampoules. Biological indicators are prepared in such equivalent performance are recommended. The number of
a way that they can be sto red under defined conditions; an viable spores exceeds 1 x 10 7 per carrier. The D-value is not
expiry date is seto less than 1.9 kGy. It is verified that there is no growth of the
Micro-organisms of the same bacterial species as the bacteria reference micro-organisms after the biological indicators have
used to manufacture the biological indicators may be been exposed to 25 kGy (minimum absorbed dosel.
inoculated directly into a liquid product to be sterilised or Gas steriHsation. The use of biological indicators is necessary
into a liquid product similar to that to be sterilised. In this fol' al! gas sterilisatiol1 procedures, both for the validation
case, it must be demonstrated that the liquid product has no of the cydes and fol' routine operations. Gas sterilisation
inhibiting effect on the spores used, especially as regards their is widely used for medical devices, isolators, chambers, etc.
germination. Use for such purposes is outside the scope of the European
A biological indicator is characterised by the name of the Pharmacopoeia. The use of spores of Bacillus atrophaeus (for
species of bacterium used as the reference micro-organism, example, ATCC 9372, NCIMB 8058 01' CIP 77.18) or other
the number of the strain in the original collection, the number strains of micro-organisms having demonstrated equivalent
of viable spores per carrier and the D-value. The D-value is performance is recommended for ethylene oxide. The number
the value of a parameter of sterilisation (duration or absorbed ofviable spores exceeds 1 x 10 6 per carrier. The parameters
dosel required to reduce the number of viable organisms to of resistance are the following: the D-value is 110t less than
10 per cent of the original number. It is of significance only 2.5 min for a test cycle involving 600 mg/L of ethylene oxide,
under precisely defined experimental conditions. Only the at 54 oC and at 60 per cent relative humidity. It is verified that
stated micro-organisms are present. Biological indicators there is no growth of the reference micro-organisms after
consisting of more than one species of bacteria on the same the biological indicatol's have been exposed to the test cyde
carrier may be used. Information on the culture medium and described aboye for 25 min and that exposing the indicators
the incubation conditions is supplied. to a reduced temperature cyde (600 mg/L, 30 oC and 60 per
It is recommended that the indicator organisms are placed cent relative humidity) for 50 min leaves revivable spores.
at the locations presumed, or wherever possible, found by
previous physical measurement to be least accessible to the
sterilising agent. After exposure to the sterilising agent,
aseptic technique is used to transfer carriers of spores to the 01/2011:50103
culture media, so that no contamination is present at the time
of examination. Biological indicators that in dude an ampoule 5.1.3. EFFICACY OF ANTIMICROBIAL
of culture medium placed directly in the packaging protecting
the inoculated carrier may be used.
PRESERVATION
A choice of indicator organisms is made such that: If a pharmaceutical preparation does not itself have adequate
a) the resistance of the test strain is suitable for the particular antimicrobial activity, antimicrobial preservatives may be
sterilisation method and is great compared to the resistance of added, particularly to aqueous preparations, to prevent
micro-organisms potentially contaminating the product; proliferation or to limit microbial contamination which,
b) the test strain is non -pathogenic; during normal conditions of storage and use, particularly fol'
multidose containers, could occur in a product and present
c) the test strain is easy to culture. a hazard to the patient from infection and spoilage of the
After incubation, growth of the reference micro-organisms preparation. Antimicrobial preservatives must not be used as
subjected to a sterilisation procedure indicates that the a substitute for good manufacturing practice.
procedure has been unsatisfactory. The efficacy of an antimicrobial preservative may be enhanced
Steam sterilisation. The use ofbiological indicators intended or diminished by the active constituent of the preparation
for steam sterilisation is recommended for the validation of or by the formulation in which it is incorporated or by the
sterilisation cydes. Spores of Geobacillus stearothermophilus container and closure used. The antimicrobial activity of the
(for example, ATCC 7953, NCTC 10007, NCIMB 8157 preparation in its final container is investigated over the period
or CIP 52.81) or other strains of micro-organisms having of validity to ensure that such activity has not been impaired
demonstrated equivalent performance are recommended. by storage. Such investigations may be carried out on samples
The number of viable spores exceeds 5 x 10 5 per carrier. The removed from the final container immediately prior to testing.
D-value at 121°C is not less than 1.5 mino It is verified that During development of a pharmaceutical preparation, it
exposing the biological indicators to steam at 121 ± 1 oC for shall be demonstrated that the antimicrobial activity of the
6 min leaves revivable spores, and that there is no growth of preparation as such or, if necessary, with the addition of
the reference micro-organisms after the biological indicators a suitable preservative 01' preservatives provides adequate
have been exposed to steam at 121 ± 1 oC for 15 min. protection from adverse effects that may arise from microbial
Dry-heat sterilisation. Spores of Bacillus atrophaeus (for contamination or proliferation during storage and use of the
example, ATCC 9372, NCIMB 8058 or CIP 77.18) or other preparation.
strains of micro-organisms having demonstrated equivalent The efficacy of the antimicrobial activity may be demonstrated
performance are recommended for the preparation of by the test described below. The test is not intended to be used
biological indicators. The number of viable spores exceeds for routine control purposes.
1 x 10 6 per carrier and the D-value at 160 oC is not les s
than 2.5 mino Dry heat at temperatures greater than 220 oC TEST FOR EFFICACY OF ANTIMICROBIAL
is frequently used for sterilisation and depyrogenation of PRESERVATION
glassware. In this case, demonstration of a 3 loglo reduction in The test consists of challenging the preparation, wherever
heat-resistant bacterial endotoxin can be used as a replacement possible in its final container, with a prescribed inoculum of
for biological indicators. suitable micro-organisms, storing the inoculated preparation
Ionising radiation sterilisation. Biological indicators may at a prescribed temperature, withdrawing samples from the
be used to monitor routine operations, as an additional container at specified intervals of time and counting the
possibility to assess the effectiveness of the set dose of organisms in the samples so removed.

General Notices (1) apply to all monographs and other texts 557
5.1.3. Efficacy of anHmkrobial preservation EUROPEAN PHARMACOPOEIA 8.0

The preservative properties of the preparation are adequate if, filtration (2.6.12). Ensure that any residual antimicrobial
in the conditions of the test, there is a significant faH or no activity of the product is eliminated by dilution, by filtration or
increase, as appropriate, in the number of micro-organisms by the use of a specific inactivator. When dilution procedures
in the inoculated preparation after the times and at the are used, due allowance is made for the reduced sensitivity
temperatures prescribed. The acceptance criteria, in terms of in the recovery of small numbers of viable micro-organisms.
decrease in the number of micro-organisms with time, vary When a specific inactivator is used, the ability of the system to
for different types of preparations according to the degree of support the growth of the test organisms is conflrmed by the
protection intended (see Tables 5.1.3.-1/2/3). use of appropriate controls.

Test micro-organisms The procedure is validated to verify its ability to demonstrate


the required reduction in count of viable micro-organisms.
Pseudomonas aeruginosa ATCC 9027; NCIMB 8626; CIP 82.118.

Staphylococcus aureus ATCC 6538; NCTC 10788;


NCIMB 9518; CIP 4.83. ACCEPTANCE CRITERIA
Candida albicans ATCC 10231; NCPF 3179; IP 48.72. The criteria for evaluation of antimicrobial activity are given
Aspergillus brasiliel1Sis ATCC 16404; IMI 149007; IP 1431.83. in Tables 5.1.3.-1/2/3 in terms ofthe 10glO reduction in the
number of viable micro-organisms against the value obtained
for the inoculum.
Single-strain challenges are used and the designated
micro-organisms are supplemented, where appropriate, by Table 5.1.3.-1. - Parenteral preparations, eye preparations,
other strains or species that may represent likely contaminants intrauterine preparations and intramammary preparations
to the preparation. It is recommended, for example, that
Escherichia coli (ATCC 8739; NCIMB 8545; CIP 53.126) is Logro reduction
used for all oral preparations and Zygosaccharomyces rouxii
(NCYC 381; IP 2021.92) for oral preparations containing a 611 2411 7a 14 a 28 a
high concentration of sugar. Bacteria A 2 NR
Preparation of inoculum B NI

Preparatory to the test, inoculate the surface of casein soya Fungi A 2 NI


bean digest agar (2.6.12) for bacteria or Sabouraud-dextrose B NI
agar without the addition of antibiotics (2.6.12) for fungi,
with the recently grown stock culture of each of the specified NR: no recovery.
micro-organisms. Incubate the bacterial cultures at 30-35 oC NI: no increase in number of viable micro-organisms comparea to
for 18-24 h, the culture of C. albicans at 20-25 oC for 48 h, and the previous reading.
the culture of A. brasiliensis at 20-25 oC for 1 week or until
good sporulation is obtained. Sub cultures may be needed after The A criteria express the recommended efficacy to be
revival before the micro-organism is in its optimal state, but it achieved. In justifled cases where the A criteria cannot be
is recommended that their number be kept to a minimum. attained, for example for reasons of an increased risk of
adverse reactions, the B criteria must be satisfied.
To harvest the bacterial and C. albicans cultures, use a sterile
suspending fluid, containing 9 giL of sodium chloride R, for Table 5.1.3.-2. - Ear preparations, nasal preparations,
dispersal and transfer of the surface growth into a suitable preparations for cutaneous application and preparations for
vessel. Add sufficient suspending fluid to reduce the microbial inhalation
count to about 10 8 micro-organisms per millilitre. To
Log,o reouction
harvest the A. brasiliensis culture, use a sterile suspending
fluid containing 9 giL of sodium chloride R and 0.5 giL of 2d 70 14 d 28 d
polysorbate 80 R and adjust the spore count to about 10 8 per
Bacteria A 2 NI
millilitre by adding the same solution.
B 3 NI
Remove immediately a suitable sample from each suspension
and determine the number of colony-forming units per Fungi A 2 NI
millilitre in each suspension by plate count or membrane B NI
filtration (2.6.12). This value serves to determine the inoculum
and the baseline to use in the test. The suspensions shall be NI: 110 increase in number of viable micro-organisms compared to
used immediately. the previous reading.

The A criteria express the recommended efficacy to be


METHOD achieved. In justified cases where the A criteria cannot be
attained, for example for reasons of an increased risk of
To count the viable micro-organisms in the inoculated adverse reactions, the B criteria must be satisfied.
products, use the agar medium used for the initial cultivation
of the respective micro-organisms. Table 5.1.3.-3. - Oral preparations, oromucosal preparations
and rectal preparations
Inoculate a series of containers of the product to be examined,
each with a suspension of one of the test organisms to give Log lO reductio!1
an inoculum of 105 to 10 6 micro-organisms per millilitre or 28 d
14 d
per gram of the preparation. The volume of the suspension
of inoculum do es not exceed 1 per cent of the volume of the Bacteria NI
product. Mix thoroughly to ensure homogeneous distribution. NI
Pungí
Maintain the inoculated product at 20-25 oC, protected from NI: no íncrease in number of viable micro-organisms compared to
light. Remove a suitable sample from each container, typically the previous reading.
1 mL or 1 g, at zero hour and at appropriate intervals according
to the type of the product and determine the number The aboye criteria express the recommended efficacy to be
of viable micro-organisms by pi ate COU11t or membrane achieved.

558 See the information section on general monographs (cover pages)


EUROPEAN PHARMACOPOEIA 8.0 5.1.4. Microbiologkal quality of non-sterile producís for pharmaceutical use

0112014:50104 If it has been shown that none of the prescribed tests will
allow valid enumeration of micro-organisms at the leve!
5.1.4. MICROBIOLOGICAL QUALITY prescribed, a validated method with a limit of detection as
close as possible to the indicated acceptance criterion is used.
OF NON-STERILE PHARMACEUTICAL
In addition to the micro-organisms listed in Table 5.1.4.-1, the
PREPARATIONS AND SUBSTANCES significance of other micro-organisms recovered is evaluated
FOR PHARMACEUTICAL USEcJ) in terms of:
The presence of certain micro-organisms in non-sterile - use of the product: hazard varies according to the route of
preparations may have the potential to reduce or even administration (eye, nose, respiratory tract);
inactivate the therapeutic activity of the product and has - nature of the product: its ability to support growth, the
a potential to adversely affect the health of the patient. presence of adequate antimicrobial preservation;
Manufacturers therefore have to ensure a low bioburden of - method of application;
finished dosage forms by implementing current guidelines
- intended recipient: risk may differ for neonates, infants,
on Good Manufacturing Practice during the manufacture,
the debilitated;
storage and distribution of pharmaceutical preparations.
Microbial examination of non-sterile products is performed - use of immunosuppressive agents, corticosteroids;
according to the methods given in general chapters 2.6.12 and - presence of disease, wounds, organ damage.
2.6.13. Acceptance criteria for non-sterile pharmaceutical Where warranted, a risk-based assessment of the relevant
products based upon the total aerobic microbial count factors is conducted by personnel with specialised training in
(TAMC) and the total combined yeasts/moulds count (TYMC) microbiology and the interpretation of microbiological data.
are given in Tables 5.1.4.-1 and 5.1.4.-2. Acceptance criteria For raw materials, the assessment takes account of processing
are based on individual results or on the average of replicate to which the product is subjected, the current technology of
counts when replicate counts are performed (e.g. direct testing and the availability of materials of the desired quality.
plating methods).
When an acceptance criterio n for microbiological quality is Table 5.1.4.-2. - Acceptance criteria for microbiological quality
prescribed it is interpreted as follows: of non-sterile substances for pharmaceutical use
- lO' CFU: maximum acceptable count '" 20; TAMC TYMC
- 10 2 CFU: maximum acceptable count '" 200; (CFU/g or CFU/mL) (CFU/g or CFU/mL)
- 10 3 CFU: maximum acceptable count '" 2000, and so forth.
Substances for
Table 5.1.4.-1 includes a list of specified micro-organisms for lO'
pharmaceutical use
which acceptance criteria are set. The list is not necessarily
exhaustive and for a given preparation it may be necessary to • Recommended acceptance criteria for microbiological quality
test for other micro-organisms depending on the nature of the of herbal medicinal products for oral use and extracts used in
starting materials and the manufacturing process. their preparation are given in general chapter 5.1.8 .•

Table 5.1.4.-1. - Acceptance criteria for microbiological quality of non-sterile dosage forms
TAMC TYMC
Route of administration (CFU/g or (CFU/g or Specified micro-organisms
CFU/mL) CFU/mL)
Non-aqueous preparations for oral use lO' 102 Absence of Escherichia coli (1 g or 1 mL)

Aqueous preparations for oral use lO' 10 1 Absence of Escherichia coli (1 g or 1 mL)

Rectal use 10 3 102 -

Oromucosal use
Gingival use
Absence of Staphylococcus aureus (1 g or 1 mL)
Cutaneous use lO' 10 1
Absence of Pseudomonas aeruginosa (1 g or 1 mL)
Nasal use
Auricular use
Absence of Pseudo monas aeruginosa (1 g or 1 mL)
Vaginal use lO' 10 1 Absence of Staphylococcus aureus (1 g or 1 mL)
Absence of Candida albicans (1 g or 1 mL)
Transdermal patches (limils for one patch Absence of Staphylococcus aureus (1 patch)
102 10'
including adhesive layer and backing) Absence of Pseudomonas aeruainosa (I patch)
Absence of Staphylococcus aureus (1 g or 1 mL)
Inhalation use (special requirements apply to Absence of PseudomolJas aeruginosa (1 g or 1 mL)
lO' 10 1
liquid preparations for nebulisation) Absence of bile-tolerant gram-negative
bacteria (1 g or 1 mL)
• Special Ph. Eur. provision for oral dosage
forms containing raw material s of natural Not more than 102 CFU ofbile-tolerant gram-negative
(animal, vegetal or mineral) origin for which bacteria (1 g or 1 mL)
antimicrobial pretreatment is not feasible and 10' 102 Absence of Salmonella (10 g or 10 mL)
for which the competent authority accepts Absence of Escherichia coli (1 g or 1 mL)
TAMC of the raw material exceeding 103 CFU/g
Absence of Staphylococcus aureus (1 g or 1 mL) +
or CFU/mL.
• Special Ph. Eur. provision for premixes for Not more than 104 CFU ofbile-tolerant gram-negative
medicated feeding stuffs for veterinary use bacteria (1 g or 1 mL)
10 5 !O'
using excipients of plant origin for which Absence of Escherichia coli (1 g or 1 mL)
antimicrobial treatment is not [easible. Absence of Salmonella (25 g or 25 mL) +

(1) This chapler has undergone pharmacopoeiaI harmonisalion. See chapter 5.8. Pharmacopoeial harmonisation.

General Notices (1) apply to all monographs and other texts 559
5.1.5. Application ofthe Fo concept lo steam sterilisation EUROPEAN PHARMACOPOEIA 8.0

Ol/2009:50105 assurance for the quality of pharmaceutical products. These


alternative methods may also find a place in environmental
5.1.5. APPLICATION OF THE Po monitoring.
CONCEPT TO STEAM STERILISATION The microbiological methods described in the European
Pharmacopoeia have been used for almost a century and these
OF AQUEOUS PREPARATIONS methods - for enumerating and identifying micro-organisms
The following chapter is published for information. - still serve microbiologists well. Over the years, these
The Fo value of a saturated steam sterilisation process is methods have been invaluable to help control and secure the
the lethality expressed in terms of the equivalent time production of microbiologically-safe pharmaceutical products.
in minutes at a temperature of 121°C delivered by the Nevertheless conventional microbiological methods are slow,
process to the product in its final container with reference to and results are not available before an incubation period of
micro-organisms possessing a theoretical Z-value of 10. typically up to 14 days. Thus the results from the conventional
microbiological methods seldom enable pro active, corrective
The total Fo of a process takes account of the heating up action to be taken.
and cooling down phases of the cycle and can be calculated
by integration of lethal rates with respect to time at discrete Alternative methods for control of microbiological quality
temperature intervals. have been introduced in recent years, and sorne of these
When a steam sterilisation cycle is chosen on the basis of the methods have shown potential for real-time or near-real-time
Fo concept, great care must be taken to ensure that an adequate results with the possibility of earlier corrective action. These
assurance of sterility is consistently achieved. In addition to new methods can also offer significant improvements in the
validating the process, it may also be necessary to perform quality of testing.
continuous, rigorous microbiological monitoring during In this informational chapter new microbiological methods
routine production to demonstrate that the microbiological offering pharmaceutical applications are described. For each
parameters are within the established toleran ces so as to give method the basic principIe is stated and the benefits and
an SAL of 10- (, or better. disadvantages of the method are then discussed. Potential
In connection with sterilisation by steam, the Z-value uses describe applications that may be envisaged in view of the
relates the heat resistance of a micro-organism to changes principIes on which the method is based: it is not intended
in temperature. The Z-value is the change in temperature to suggest that actual application has been made. Finally,
required to alter the D-value by a factor of 10. general considerations for the validation of the method are
The D-value (or decimal reduction value) is the value of outlined. These are illustrated by specific examples for each
a parameter of sterilisation (duration or absorbed dose) type of method. A detailed validation protocol is given for
required to reduce the number of viable organisms to 10 per information at the end of this chapter.
cent of the original number. It is only of significance under
It is not the intention of this chapter to recommend one method
precisely defined experimental conditions.
over another, nor is it the intention to provide an exclusive
The following mathematical relationships apply: or exhaustive list of alternative methods that can be used for
Fa = D l21 (logNo - 10gN) = D l21 10gIF
pharmaceutical microbiological control. This informational
chapter however may be used in the process of choosing an
alternative microbiological method as a supplement 01' as an
D 121 D-value ofthe reference spores (5.1.2) at 121°C;
alternative to conventional microbiological approaches and to
No initial number of viable micro-organisms; give guidance in the process of validating the chosen method.
In this rapidly developing field, further methods are likely to
N final number of viable micro-organisms;
appear. The guidance offered in this chapter may be equally
lF inactivation factor. applicable to these methods.

z= T2 - TI Thel'e are 3 major types of determinations specific to


10gD I - logD 2 microbiological tests. These include:
- qualitative tests for the presence or absence of
D-value of the micro-organism at temperature TI; micro-organisms;
D-value of the micro-organism at temperature T2 • quantitative tests for enumeration of micro-organisms;

IF = No = 10tlD - identification tests.


N 1-l. QUALITATIVE TESTS FOR THE PRESENCE OR
ABSENCE OF MlCRO-ORGANlSMS
exposure time; In conventional microbiological analysis this type of test is
D D-value of micro-organism in the exposure characterised by the use of turbidity or other growth-related
conditions. changes in a culture medium as evidence of the presence of
viable micro-organisms in the test sample. The most common
example of this test is the sterility test. Other examples of this
01/2008:50106 type of testing are those tests designed to evaluate the presence
or absence of a particular type of viable micro-organism in
5.1.6. ALTERNATIVE METHODS FOR a sample.
CONTROL OF MICROBIOLOGICAL 1-2. QUANTITATIVE TESTS FOR ENUMERATION OF
MICRO-ORGANISMS
QUALITY Membrane filtration and plate count methods are
The following chapter is published for information. conventional methods used to estímate the number of viable
mícro-organísms present in a sample. The Most Probable
1. GENERAL INTRODUCTION Number (MPN) method is another example of these methods.
The objective of this chapter is to facilitate the implementation MPN was developed as a means to estímate the number of
and use of alternative microbiological methods where this can viable micro-organisms present in a sample not amenable to
lead to cost-effective microbiological control and improved direct plating.

560 See the information section on general monographs (cover pages)


EUROPEAN PHARMACOPOEIA 800 5.1.6. Ahemative methods for control of microbiologkal qualUy

1-30 IDENTIFICATION TESTS Potential useso Microbiological assay of antibiotics, efficacy


Biochemical and morphological characterisation of an of antimicrobial preservation and presence/absence in the
unknown micro-organism is the classical method of quantity of sample tested when performing total viable aerobic
identification used in pharmacopoeia! tests o Recently count
developed methods have streamlined and automated aspects 2-1-1-30 Measurement 01 consumption or production 01 gas
of this identification, especially in the areas of data handling, Principies 01 measuremento Active!y multiplying and
analysis, and storageo Several new approaches that have been metabolising organisms utilise appropriate growth media,
integrated into these methods include biochemical reactions, leading to the production of metabolites or elimination
carbon substrate utilisation, charaeterisation of fatty acid of specific nutrientso In one approach, changes in gaseous
composition, restriction endonuclease banding patterns and head-space composition may be monitored in closed culture
use of 16S rDNA sequenee analysiso vessels by pressure transducers responding to gas production
(eogo CO 2 ) or gas consumption (eogo 02)0 Other indicators may
20 GENERAL PRINCIPLES OF ALTERNATIVE METHODS be employed including colorimetric detection of C0 2 0
Alternative microbiological methods employ direct and Critical aspectso For slow-growing micro-organisms such as
indirect methods of detection; in sorne instances amplification mycobacteria, the method offers more rapid detectiono There
of the signal is achieved by enriehment methodso In is no direct relationship between original microbial burden
recognition of these differences, and for eonvenienee and detectable end-point The incubation temperature and the
within this ehapter, alternative methods for the control of algorithm for data processing significantly affect the resultso
microbiological quality are divided into 3 categories: Potential useso Products where slow-growing micro-organisms
- growth-based methods, where a detectable signal is usually may be present
achieved by a period of subeulture; 2-1-1-40 Bioluminescence
- direct measurement, where individual cells are Principies 01 measurement. Adenosine triphosphate (ATP) is
differentiated and visualised; a well-documented marker of ceH viabilitr. In this method,
- cell component analysis, where the expression of specific ATP needs first to be released from micro-organisms using
ceH components offers an indireet measure of microbial an appropriate extractant, followed by quantitative assay
presenceo using the luciferin/luciferase enzyme system, which emits
light in proportion to the ATP present The emitted light is
In sorne instan ces, these distinetions are artificial but they do measured with a bioluminometer and is expressed in relative
enable a working classification to be createdo light units (RLU) for bioluminescence in liquid mediao The
2-1. GROWTH-BASED METHODS RLU obtained from the sample is compared with a threshold
value determined at 2 or 3 times the RLU of the medium used
2-1- L Early detection of growth
for cultivation or sample suspensiono The result is positive
2-1-1- L General critical aspects 01 methods based on early if the RLU obtained with the analysed sample exceeds the
detection 01 growth threshold valueo A modification to the method using growth
Such methods are critically dependent upon microbial growth of micro-organisms captured on a membrane by incubation
in order to achieve a detectable number of micro-organismso on agar medium employs a charge coupled device (CCD)
For the typically low levels of microbial contamination seen camera to deteet the micro-colonies, and results are expressed
in pharmaceutical products, detection may take 24 h or even as microCFU This method is quantitative but has a narrow
more, especially in the case of yeasts and mouldso Increased range of linearityo
sensitivity can be achieved with filtered productso In this case, Critical aspectso If the product sampled has a high leve! of
after filtration, the membrane is incubated in the medium bacterial contamination (about 500-1000 CFU per sample
and the result is expressed as presence or absence in the quantity tested), the detection is rapid (1 h)o For low
quantity corresponding to the filtered volumeo These systems, levels of contamination (less than 100 CFU per quantity
because they use an incubation step in liquid media, do of sample tested), it is necessary to increase the llumber of
not offer quantitative information but a presence/absence micro-organisms by an incubation step in culture media
determination in the quantity analysedo Analysis of more than (liquid or solid agár) for 12-48 h according to the method
one sample quantity may offer a semi-quantitative estimation employedo After this time, in liquid media, one single cell
(limit test)o The major benefit of such methods compared to capable of growth will increase from 1 to 1000 and will be
classical methods frequently resides in the capacity to process detectedo The yield of ATP varies from one micro-organism to
simultaneously a large number of samples and potentially to another, bacteria containing 1-10 fg per cel! and fungi around
obtain a result in a shorter time o 100 fg per ceH and many other factors including the species,
2-1-1-20 Electrochemical methods
the growth phase of the ceU, the nutritional status, the cellular
stress or the cellular age could affect the ATP content of the
Principies 01 measuremento Micro-organisms multiplying and celL Therefore, it is not possible to obtain a count directly
metabolising in appropriate growth media produce highly from the RLU valueo In addition, turbidity and sample colour
charged iOl1ic metabolites from weakly charged organic can affect the reaction by either enhancing the reaction and
nutrients leading to the modification of electrical properties increasing the leve! of light output or acting as a quenching
in those mediao These changes in impedance (measured by agent and lowering the level of light output Since the reaction
conductance or capacitance) are monitored with e!ectrodes is enzymatically based, products which could inhibit or
included in the culture vessels and in contact with the culture decrease the enzyme activity may interfereo In practice, such
mediumo The measurable end-point is the time taken to detect interference is rare but must be thoroughly investigated
a pre-determined impedance change; the detection time is during the validation processo The reaction is also sensitive to
inversely proportional to the initial inoculum sizeo For yeasts the presence of phosphate nucleotides such as ADP or GTP,
and moulds, which produce only small changes in electrical which interfere by producing ATP in the presence of adenylate
impedance, an indirect measurement of conductance using kinaseo This enzyme is used to increase the sensitivity of
a potassium hydroxide reservoir is commonly usedo Direct sorne bioluminescence methods: here a 3,od reagent is added
measurement of capacitance can also be carried out containing ADP and new ATP is produced in the presence of
Critical aspectso Automated detection with electronic data adenylate kinase released from micro-organismso
generation, mapping of the variation of impedance reflecting Potential useso Testing for efficacy of antimicrobial
the growth curve of the micro-organisms, and reduction of preservation, presence/absence in the quantity of sample
the duration of the test to 48 ho tested when performing total viable aerobic count

General Notices (1) apply to all monographs and other texts 561
5.1.6. Altemative methods for control of microbiological quality EUROPEAN PHARMACOPOEIA 8.0

(bioluminescence in tube or microtitre plate), total ability to deteet the presence of specific enzymes using suitable
viable aerobic count (bioluminescence on membrane), substrates has led to the development of a large number of
environmental and water monitoring. The method finds methods for the identification of micro-organis!11s employing
applications in filterable and non-filterable products. manual or automated methods. The incorporation of such
2-1-1-5. Microcalorimetry substrates into a selective or non -selective primary isolation
Principies of measurement. Microbial catabolism generates
medium can eliminate the need for further sub culture
heat which can be accurately measured by microcalorimetry. and biochemical tests to identify certain micro-organisms.
Consequently, chromogenic liquid or solid culture media are
Heat production can be detected by placing the contaminated
sample in a sealed ampoule containing a growth medium designed to produce specific enzymatic activities for detection
and enclosing within a calorimeter. Using sensitive and differentiation of micro-organisms. In these particular
media, defined substrates are introduced into the formulation
instrumentation microbial growth curves can be established.
High bioburdens may be detectable by flow calorimetry. and are hydrolysed by the specific cell enzyme of a given
bacteria or fungi during growth. These substrates are chosen
Critical aspects. Theoretically, this method does not require according to the diagnostic enzymatic aetivity sought and are
microbial growth but simply catabolic activity. Nevertheless, linked to coloured indicators.
a minimum number of micro-organisms are required to give
heat output measures aboye base-line and this is usually Critical aspects. The use of innovative media presents several
achieved by use of an enrichment method. advantages: improved discrimination of colonies in mixed
culture, ease of use and ease of interpretation. In addition,
Potential uses. Test for efficacy of antimicrobial preservation.
response times are shorter because growth and identification
2-1-1-6. Turbidimetry of the micro-organism are simultaneous. However, validation
Principies of measurement. Microbial growth willlead to of the media must be undertaken carefully to ensure a
detectable changes in medium opacity. This can be accurately combinatiol1 of specificity, selectivity and robustness. The
quantified by optical density measurement at a specified quality of the signal is based not only on the careful choice
wavelength. In its simplest form such measurements are of the enzymes used as the basis of detection, as these
performed in a standard spectrophotometer over a wavelength enzymes may be present in different genera, but also on
range generally of 420-615 nm. Alternative automated systems physico-chemical characteristics of the medium such as pH.
employ microtitre plate readers offering continuous readout Potential uses. Detection of specified micro-organisms such as
with early detection of optical density change. E. coli, coliforms, Salmonella, Staphylocaccus and Streptococcus
Critical aspects. Attempts have been made to extrapolate the spp.; particular benefit may be found in presence/absence
initial bioburden from the time for detection but this may be testing. Yeasts can also be detected using chromogenic culture
Iimited to healthy micro-organisms with reproducible growth media.
characteristics. The methods cannot distinguish between 2-2. DIRECT MEASUREMENT
viable and non-viable micro-organisms.
Potential uses. By means of calibration graphs, determination 2-2-l. Solid phase cytometry
of the inoculum size of microbial suspensions for use in Principies of measurement. A membrane filter is used to
pharmacopoeial tests. In automated mode, establishment of retain microbial contaminants. Micro-organisms are stained
the preservative sensitivity of test micro-organisms recovered by labelling using a fluorophore as a viability indicator,
from formulated products. either before or after filtration. The fluorophore is initially
2-1-1-7. Phage-based methods a non-fluorogenic, conjugated substrate that requires
intracellular enzymatic activity to cleave the substrate and
Principies of measurement. Bacterial viruses (bacteriophage,
release the fluorescent moiety. An intact cellular membrane
phage) can infect host cells causing either lysis or incorporation
is required to retain fluorophore within the cytoplasm. Laser
of their genetic material and expression of novel proteins.
excitation and automated scanning allows the detection of
Their high leve! of host specificity can be employed in
single, viable fluorescent micro-organisms. Appropriate
detection methods which exploit the consequences of infection
software permits differentiation of viable micro-organisms
as an end-point. Such end-points include: plaque formation
from auto-fluorescent particles. The high sensitivity and
on a solid lawn of reporter bacteria; detection of intracellular
rapidity of deteetion permits near-real-time detection of
contents released from lysed bacteria (possibly by colorimetric
microbial contaminants. Total ceH counts can be obtained
method); or phage-induced effects such as ice nucleation or
using a permanently fluorescing stain.
bioluminescence following infection by genetically modified
phage. Fluorescently labelled coliphages can be used for the Critical aspects. Metabolically active, fastidious and viable
selective detection of viable E. cali in combination with DEFT non-culturable micro-organisms can be detected. This may
(see 2-3-3.). result in reappraisal of the microbiallimits established for
the samples under evaluation. Spores require initiation of
Critical aspects. Phage-based detection can be used in both
germination to enable detection. Single cell detection may
single and mixed cultures where host specificity allows
be achievable, but identification is not currently part of the
both detection and identification. Detectable end-points
routine test protocol. The use of fluorescent antibody may
often require a minimum number of target cells to ensure a
offer a route to selective detection. False positives may occur
measurable signal, necessitating enrichment in situations of
from auto-fluorescent particles, which can be difficult to
low bioburden. The viral infection process can be adversely
differentiate from micro-organisms.
affected by sample composition. In most casesthere is a
narrow host range which makes it difficult to detect a broad Potential uses. Rapid and sensitive method for the non-specific
spectrum of microbial contaminants. evaluation of bioburden. 1t has found applications in testing
Potential uses. These methods are used mainly for research pharmaceutical-grade waters.
purposes with commercial development aimed principally 2-2-2. Flow cytometry
towards uses in clinical and food microbiology. These Principies of measurement. Fluorophore-labelled
methods are likely to be employed for presence/absence micro-organisms can be detected in suspension as they pass
determinations of specified micro-organisms. through a flow ceH cytometer. Where a viability-indicating
2-1-2. Media development to improve detection fluorophore substrate is employed, viable micro-organisms
Principies of measurement. Culture media have existed for can be differentiated from non-viable particles (see 2-2-1.).
many years and have been constantly improved. A recent Critical aspects. Flow cytometry may be applied for the
innovation is the appearance of chromogenic substrates which microbiological analysis ofboth filterable and non-filterable
are increasingly used in clinical and food microbiology. The materials. Flow cytometric analysis gives near-real-time

562 See the information section 011 general monographs (caver pages)
EUROPEAN PHARMACOPOEIA 8.0 5.1.6. Alternative methods for control of microbiological quaIity

detection, but it is not as sensitive as solid phase cytometry. 2-3-1-2. Fatty acid profiles
To increase sensitivity for use in the pharmaceutical field, it
Principies of measurement. The fatty acid composition of
ofien becomes necessary to add an incubation step in culture
micro-organisms is stable, well conserved and shows a high
media and in that case the method becomes a growth-based
degree of homogeneity within different taxonomic groups.
method. Analysis of non-filterable samples may require serial
The isolate is grown on a standard medium and harvested.
dilution to optimise performance, and particulate size can The fatty acids are saponified, methylated and extracted and
have a significant effect on performance. With the exception
the occurrence and amount of the resulting fatty acid methyl
of filterability, similar considerations apply to those of solid
esters are measured by high resolution gas chromatography.
phase cytometry. Clumping of bacteria can be a problem (e.g.
The fatty acid composition of an unknown isolate is compared
S. aureus).
with a database of known isolates for a possible match and
Potential uses. In contrast with solid phase cytometry, this identification.
method offers the potential to detect and enumerate the Crítical aspects. The use of fatty acid pro files for microbial
microbial bioburden in materials containing significant levels identification requires a high degree of standardisation. It
of particulate matter. If a pre-incubation step is needed, the is critical for the fatty acid composition of microbial cells
method becomes a qualitative determination.
that isolates are grown using standard media and standard
2-2-3. Direct epifluorescent filtration technique (DEFT) incubation conditions. Standard conditions for operation
Principies of measurement. This technique may be considered of the gas chromatograph must be employed, with frequent
to be a forerunner of solid phase cytometry. Micro-organisms runs of calibration standards and known isolates being very
concentrated by filtration from the sample are stained with important.
a fluorescent dye, formerly acridine orange and now more Potential uses. Identification or characterisation of
commonly 4',6-diamidino-2-phenylindole (DAPI), that may environmental and product flora for contaminant tracing and
be detected by epifluorescent illumination. Fluorescent vital detection of specified micro-organisms.
staining techniques as employed in solid phase cytometry
2-3-1-3. Fourier transform infrared (FTIR) spectroscopy
(see 2-2-1.) are amenable to DEFT and fluorescent redox dyes
such as 5-cyano-2,3-ditolyltetrazolium chloride (CTC) can be Principies of measurement. A Fourier transformation of the
used to highlight respiring cells. Coupled with microscopy, infrared spectrum of whole micro-organisms gives a stable,
the method allows rapid detection of micro-organisms, recognisable pattern typical of the taxonomic groups of
the absolute sensitivity depending on the volume of micro-organisms. The analysis of the FTIR pattern can be
product filtered and the number of fields of view examined. performed in instruments available on the market. The isolate
Semi-automated auto-focusing systems coupled to image is grown on a standard medium and harvested. Cell mass is
analysis have served to improve the utility of this method. transferred to a carrier, and the infrared spectrum is recorded.
A modification to the principIe employs sampling using an The Fourier transformation is calculated and the pattern is
adhesive sheet which permits collection of cells from surfaces, compared with a database of known isolates for a possible
staining on the sheet and subsequent direct observation under match and identification.
the epifluorescence microscope. Critical aspects. The use of FTIR-patterns for microbial
Critical aspects. The distribution of micro-organisms on identification requires a high degree of standardisation. It is
the membrane affects method robustness. The intensity critical for the FTIR-pattern of microbial cells that isolates
of fluorescence can be influenced by the staining pro ces s are grown using standard media and standard incubation
and the metabolic status of the micro-organisms. A brief conditions. The cells must be in the same state of the growth
period of culture on the filter surface prior to staining cycle when analysed. Particular attention needs to be paid to
allows microcolony formation; these microcolonies stain the validation process.
readily, can be easily enumerated and are demonstrable Potential uses. Identification or characterisation of
evidence of viability. Developments using fluorescence in environmental and product flora for contaminant tracing and
situ hybridisation (FISH) arising from the complementary detection of specified micro-organisms.
interaction of a fluorescently-Iabelled oligonucleotide probe
with a specific rRNA sequence offer a route to selective 2-3-1-4. Mass spectrometry
detection. Principies of measurement. Gaseous breakdown products
Potential uses. DEFT is generally limited to low viscosity released by heating microbial isolates in a vacuum can be
fluids although pre-dilution or pre-filtration has occasionally analysed by mass spectrometry, providing characteristic
been applied to viscous or particulate products. Bioburden spectra. Similarly, intact microbial cells, when subject to
monitoring has been successfully achieved in aqueous intense ionisation under matrix-assisted laser desorption
pharmaceuticals. ionisation-time offlight (MALDI-TOF) mass spectrometry,
release a distinctive pattern of charged species. Such spectra
2-3. CELL COMPONENT ANALYSIS
can be compared with known profiles as a rapid aid to
2-3-1. Phenotypic identification.
2-3-1-1. Immunological methods Critical aspects. Isolates require culture prior to analysis.
Principies of measurement. Antibody-antigen reactions can be Potential uses. Identification or characterisation of
employed to detect unique cellular determinants of specific environmental and product flora for contaminant tracing and
organisms. These reactions can be linked to agglutination detection of specified micro-organisms.
phenomena, colorimetric or fluorimetric end-points offering
2-3-1-5. Biochemical assays based on physiological reactions
both quantitative and qualitative detection. Enzyme-linked
immunosorbent assays (ELISA) offer simple solid phase Principies of measurement. These assays are usually preceded
methodologies. by a Gram stain or other early differentiation test to decide on
the appropriate testing protocol. Microbial cell suspensions are
Critical aspects. Immunological detection methods depend
tested using biochemical test kits. Micro-organisms are known
upon the unique expression of specific identifiers but
to have particular reactions to these biochemical substances,
do not necessarily demonstrate the presence of viable
e.g. utilisation of specific carbon sources. The identification
micro-organisms. of the culture is done by comparing the biochemical reaction
Potential uses. Detection and identification of specified profile with a database. These methods can be performed
micro-organisms. manually or by automated instruments.

General Notices (1) apply to all monographs and other texts 563
5.1.6. Alternative methods for control of microbiological quality EUROPEAN PHARMACOPOEIA 8.0

Critical aspects. Apure colony is needed which must not be Critical aspects of RNA amplification techniques. These
older than 3 days. The handling of the system is easy but the methods have proven to be very valuable for specific
interpretation of the results can be subjective. Depending on (quantitative) RNA detection. However, they may be more
the system used and the micro-organism under investigation, difficult to implement routinely.
the results can be available quickly. Critical aspects of (semi-) quantitative detection (real-time
Potential uses. Identification of environmental and product PCR). Classical PCR techniques are based on end-point
flora for contaminant tracing and detection of specified detection. In general fragment analysis is carried out using
micro-organisms. agarose gel s and specific size markers. However, there is no
correlation between the amount of PCR product at the end
2-3-2. Genotypic of the reaction and the original amount of target molecule.
2-3-2-1. Nucleic acid amplification techniques (NAAT) In contrast the amount of PCR product detected at the
beginning of the exponential phase of the reaction correIates
General principIes of measurement. NAAT rely on the
very well with the initial starting amount of nucleic acid.
reiteration of the process of DNA polymerisation, leading to Modern real-time PCR techniques are developed to measure
an exponential increase of a specific fragment of the nucleic this exponential phase of the reaction. These techniques
acid, Le. the use of the polymerase chain reaction (PCR). In generate amplification data from which the original amount
this thermophilic cyclic process a specific DNA fragment is of target molecule can be deduced. A specific labelled probe
amplified using oligonucleotide primers (see also general detects in real time the PCR product formed, allowing direcÍ
method 2.6.21). RNA can also be amplified by PCR after visualisation of the exponential part of the PCR reaction.
transcription into cDNA using a reverse transcriptase. This By comparison with amplification plots of a standard
technique is known as reverse transcriptase PCR (RT -PCR). dilution series, a quantification of the target molecule can be
Alternatively, specific RNA -based amplification techniques, obtained. Automated real-time PCR systems are available
for example nucleic acid sequence-based amplification on the market. An additional advantage is that the chance
(NASBA) or transcription-mediated amplification (TMA) of cross-contamination is minimised, as PCR products are
are available to amplify multiple antisense copies of the RNA scanned with a laser while the tubes remain closed. However,
target. Amplified nucleic acid fragments can be analysed by generation of standards will be difficult to accomplish.
several methods: fragment size analysis; specific sequen ce
analysis; reamplification with a second primer pair; or Critical aspects of amplification of genes codingfor 165 or 235
specific detection by hybridisation with a fluorescent labelled rRNA. A powerful application of PCR is the amplification
probe. Depending on the choice of analysis the amplification and subsequent sequence analysis of specific parts of the
technique can be qualitative, semi-quantitative or quantitative. genes coding for 16S or 23S rRNA. Analysis of these specific
For identification/characterisation purposes sequence analysis DNA sequences allows in most cases the identification of
of specific parts of the genome can be used (Le. 16S or 23S a micro-organism at species leve!. Selection of appropriate
rRNA targets). universal primers, or even species-specific primer pairs, from
international databases allows a high specificity in fragment
General critical aspects. NAAT have many advantages over amplification. Modern systematic classification is based on
classical methods for the detection of micro-organisms: comparative sequence analysis.
- the methods are highly specific, provided that the primers Potential uses. Owing to the high specificity of the
chosen are specific for a particular micro-organism or amplificatiol1 techniques, they are very suitable for
group of micro-organisms; identification purposes. NAAT are suitable for the detection
the procedures are rapid, overcoming the problem of of specified micro-organisms or certain groups such as
prolonged incubation times; mycoplasmas. Real-time quantitative PCR is needed for
enumeration.
the methods are highly sensitive allowing ideally the 2-3-2-2. Genetic fingerprinting
detection and amplification of one single nucleic acid
fragment in the reaction mix. Principies of measurement. This technique characterises
and identifies micro-organisms using restriction fragments
However, there are numerous practical restrictions to its use: of l1ucleic acids from bacterial and fungal genomes. DNA
- the sensitivity of the methods is highly dependent on how is extracted from apure microbial celllysate and cut into
successfully the target fragments can be concentrated in fragments by restriction enzymes. DNA fragments are
the sample; size-separated by electrophoresis, visualised, and the pattern is
compared with other known patterns of microbial isolates. The
- the presence of inhibitors of the enzymatic process result gene tic fingerprint is a stable marker that provides definitive
in false negative reactions; species discrimination or even characterisation below species
- the starting volume of the sample tested is small; leve!. Ribotyping is a typical example of this technique. There
are also fingerprinting methods based on PCR with primers
- the procedures are prone to cross-contamination from that bind to several sites in the microbial genome, creating
previously amplified fragments resulting in false positive amplicons with a characteristic size distribution.
results.
Critical aspects. There is a need for apure colony, but
Depending on the aim, a choice must be made for amplification no preliminary cultivation step is necessary. The growth
of an RNA or DNA target. The target choice affects the conditions (temperature, type of media,) do not affect the
correlation with viability. The use of DNA as a marker has the outcome of the analysis. For the identification of bacteria
disadvantage that dead micro-organisms also contain DNA, semi-automated systems are 011 the market.
whereas mRNA is rapidly degraded in dead bacteria and is Potential uses. Genetic fingerprinting is more valuable for
considered a better marker for viability. strain discrimination (characterisation below species level)
Critical aspects of RT-PCR. Reverse transcriptase-PCR is than for identification of species.
characterised by the synthesis of cDNA using RNA as a
template. Reverse transcriptase is used for this step. A 3. GENERAL VALIDATION REQUIREMENTS
specific part of the cDNA is subsequently amplified by The purpose of this section is to provide guidance on the
PCR. Depending on the quality of the RNA isolation, the validation of methods for use as alternatives to microbiological
cDNA synthesis efficiency can vary. RT -PCR can be used to methods of the Pharmacopoeia. Por microbial recovery and
specifically detect RNA if the DNA contamination of the RNA identification, microbiological testing laboratories sometimes
sample is minima!. use alternative test methods to those described in the

564 See the information section on general monographs (cover pages)


EUROPEAN PHARMACOPOEIA 8.0 5.1.6. Altemative methods for control of microbiological quaH.ty

general chapters for a variety of reasons, such as economics, of this could be the sterility test where this would translate
throughput, and convenience. Validation of these methods into a eomparison of the rate of positive and negative
is required. Sorne guidance on validation is provided in the results produced by the alternative method versus the
General Notices section 1.1 on the use of alternative methods. pharmacopoeial method for identical samples. However, in
Validation of alternative microbiological methods must take a case sueh as the sterility test, the low number of failures
into account the large degree of variability associated with would required thousands of comparison tests to establish
conventional methods. When conducting microbiological equivalency and thus would be problematic.
testing by conventional plate count, for example, one A more fe asible method for evaluating the precision
frequently encounters a range of results that is broader than of an alternative qualitative method compared with a
ranges in commonly used chemical tests. pharmacopoeial method might be to observe the degree
Where specific equipment is critical for the application of of agreement between the two when the procedures are
the alternative method, the equipment, including computer performed repeatedly on different lots of the same producto
hardware and software, must be fully qualified as follows: The aceuracy and precision of the alternative method may be
- design qualification (DQ) to provide documented evidence expressed as the relative rates of false positive and false negative
that the design of the equipment is suitable for correct results between the new method and the pharmacopoeial
performance of the method; to be provided by the supplier; method using a standardised, low-level inoculum.
- installation qualification (IQ) to provide documented The rate of occurrence of false negative results in the presenee
evidence that the equipment has been provided and of the sample for the 2 methods can be estimated using low
installed in accordance with its specification; levels of test micro-organisms. This design is similar to the
- operational qualification (OQ) to provide documented standard bacteriostasis/fungistasis test; however, the level of
evidence that the installed equipment operates within micro-organisms inoculated must be very low, for example
pre-determined limits when used in accordance with its about 5 CPU per unit. The level of inoculum should ensure a
operational pro ce dures ; frequency of Íailure rates high enough to provide a means to
compare the 2 methods. The alternative method must provide
- performance qualification (PQ) to provide documented
at least as high a frequency of recovery as the pharmacopoeial
evidence that the equipment, as installed and operated
method.
in accordance with operational procedures, consistently
performs in accordance with predetermined criteria 3-2-2. Specificity
and thereby yields correct results for the method. The specificity of an alternative qualitative method is its
This is typically done with a 'model' system (with test ability to detect the required range of micro-organisms that
micro-organisms) to make sure that the conditions used by may be present in the sample under test. This coneern is
the user laboratory make it possible to satisfy the criteria adequately addressed by growth promotion of the media for
described by the supplier of the method in the laboratory. qualitative methods that rely upon growth to demonstrate
Sorne alternative methods depend on the use of databas es. presence or absence of micro-organisms. Por those methods
The extent of coverage of the database with respect to the that do not require growth as an indicator of microbial
range of micro-organisms of interest must be taken into presenee, the specificity assures that extraneous matter in the
account for validation purposes. test system does not interfere with the test. Where relevant
The value of a new or modified method must be demonstrated for the purpose of the test, mixtures of micro-organisms are
in a comparative study between the official method and the used during validation.
alternative method. The characteristics defined in this chapter 3-2-3. Limit of detection
must be used to establish this comparison.
The limit of deteetion of an alternative qualitative method
3-1. TYPES OF MICROBIOLOGICAL TESTS is the lowest number of micro-organisms in a sample that
1t is critical to the validation effort to identify the portion of can be detected under the stated experimental conditions. A
the test addressed by the alternative method. Por example, microbiologicallimit test determines the presence or absence
there are a variety of methods available to deteet the presence of micro-organisms. Due to the nature of microbiology, the
of viable cells. These methods may have applieations in limit of detection refers to the number of micro-organisms
a variety of tests (e.g. bioburden, sterility tests,) but may present inthe original sample befo re any dilution or ineubatiol1
not, in faet, replaee the critical aspects of the test entirely. steps; it does not refer to the number of micro-organisms
Por example, a sterility test by membrane filtration may be present at the time of testing.
perÍormed according to the pharmacopoeial pro ce dure up to The 2 methods (alternative and pharmacopoeial) must be
the point of eombining the processed filter with the reeovery assessed by using an inoculum containing a low number of test
media, and after that the presenee of viable cells might then micro-organisms, for example about 5 CPU per unit, followed
be demonstrated by use of sorne of the available methods. by a measurement of recovery. The level of inoculation
Validation of this application would, therefore, require must be adjusted until at least 50 per cent of the samples
validation of the recovery system employed rather than the show growth in the pharmacopoeial method. It is necessary
entire test. to repeat this determination several times, as the limit of
General concerns. Validation of a microbiological method detection of a test is determined from an appropriate number
is the process by whieh it is experimentally established of replicates (for example not less than 5). The ability of the
that the performance eharacteristics of the method meet 2 methods to detect the presence of single organisms can be
the requirements for the intended application. Since demonstrated using the X2 test.
microbiologieal tests have 3 basic applications, 3 separate
3-2-4. Robustness
sets of validation criteria are required. These concerns are
described below. The robustness of an alternative qualitative method is a
measure of its capacity to remain unaffeeted by small but
3-2. VALIDATION OF ALTERNATIVE QUALITATIVE
deliberate variations in method parameters, and provides an
TESTS FOR THE PRESENCE OR ABSENCE OF
indication of the method's reliability under a variety of normal
MICRO-ORGANISMS
test conditions, such as different analysts, instruments, batches
3-2-1. Accuracy and precision of reagents and laboratories. Robustness can be defined as the
A direct method to show the equivalence of 2 qualitative intrinsic resistanee to the influenees exerted by operational and
methods would be to run them side by side and determine environmental variables on the results of the microbiological
the degree to which the method under evaluation shows method. Robustness is a validation parameter best suited to
equivalence to the pharmacopoeial method. An example determination by the supplier of the method, but if critical

General Notices (1) apply ta all monographs and ather texts 565
5.1.6. Alternative methods fOl" control of microbiological quality EUROPEAN PHARMACOPOEIA 8.0

parameters are modified by the user their effects on robustness sample containing a known number of micro-organisms, it is
have to be evaluated. Robustness of a qualitative method is essential that the quantification limit is determined from a
judged by its ability to detect the test micro-organisms under number of replicates, for example at least 5. The results of
the deliberate variations to the method parameters. the Iinearity and accuracy studies can also be used. Here,
3-3. VALIDATION OF ALTERNATIVE QUANTITATIVE the lowest concentration in the linear range is considered to
TESTS FOR ENUMERATION OF MICRO-ORGANISMS be the limit of quantification of the method. The limit of
quantification must not be a l1umber greater than that of the
3-3-1. Accuracy pharmacopoeial method.
The accuracy of an alternative quantitative method is the
3-3-5. Linearity
doseness of the test results obtained by the alternative
method to the value obtained by the pharmacopoeial method. The linearity of an alternative quantitative method is its ability
Accuracy must be demonstrated across the practica! range of to produce results that are proportional to the concentration
the test. Accuracy is usually expressed as the percentage of of micro-organisms present in the sample within a given
recovery of micro-organisms by the method. range. The Iinearity must be determined over the range
Accuracy may be shown by preparing a suspension of corresponding to the purpose of the alternative method.
A method to determine this would be to select different
micro-organisms at the upper end of the range of the test,
concentrations of each test micro-organism and conduct
serially diluted down to the lower end of the range of the test.
For example, if the alternative method is meant to replace several replicates of each concentration. The number of
the traditional plate count method for viable counts, then a replicates is chosen so that the entire test can be carried out
reasonable range might be 10°-106 CFU per mL. If it is, instead, during the same working session. 2 more working sessions
are then completed under conditions of maximum variability
a replacement for the MPN method, a much more narrow
(different reagents, different operators, different days, etc.).
range may be used. At least 5 suspensions across the range
of the test must be analysed for each test micro-organismo If After checking the homogeneity of the variances of the
results obtained for each concentration, the regression line is
the alternative method is meant to replace the conventional
calculated. Linearity is demonstrated if the estimated slope
method, it must provide an estimate of viable micro-organisms
is significant and if the test for deviation from linearity is
of not less than 70 per cent of the estimate provided by the
non -significant.
pharmacopoeial method.
The protocol used to check the linearity (see 3-3-5.) of 3-3-6. Range
the method may also be used to check the accuracy: The range of an alternative quantitative method is the interval
the suspensions of micro-organisms prepared for the between the upper and lower levels of micro-organisms that
alternative method are counted at the same time using the have been determined with precision, accuracy, and linearity
pharmacopoeial method. Accuracy is demonstrated if the using the method as written. The range is determined from
suitability tests show that the slope of the regression line do es studies of precision, accuracy and linearity.
not differ significantly from 1 and if the y-intercept is not 3-3-7. Robustness
significantly different from O.
The robustness of an alternative quantitative method is a
3-3-2. Precision measure of its capacity to remain unaffected by small but
The precision of an alternative quantitative method is the deliberate variations in method parameters and provides an
degree of agreement among individual test results when the indication of its reliability under a variety of normal test
procedure is applied repeatedly to multiple samplings of conditions, such as different analysts, instruments, batches of
homogeneous suspensions of micro-organisms under the reagents and laboratories. Robustness can be defined as the
prescribed conditions. The precision is usually expressed as intrinsic resistance to the influences exerted by operational and
the variance, standard deviation or coefficient of variation of a environmental variables on the results of the microbiological
series of measurements. method. Robustness is a validation parameter best suited to
At the very least, a suspension of micro-organisms with a determination by the supplier of the method, but if critical
concentration usually in the middle of the range is counted parameters are modified by the user their effects on robustness
several times. The number of replicates is chosen so that have to be evaluated. Robustness of a quantitative method is
the entire test can be carried out during the same working judged by its ability to enumerate with statistical relevance
session, i.e. under the same operating conditions and without the test micro-organisms under the deliberate variations to
any change in the suspension of micro-organisms. Other the method parameters.
working sessions are then carried out under conditions of 3-4. VALIDATION OF ALTERNA TIVE IDENTIFICATION
maximum variability (different reagents, different operators, TESTS
different days, ete.). The variance of the results observed in There is a large body of evidence that different methods vary
each of the working sessions ('groups') is calculated. If the considerably in their ability to identify micro-organisms in
variances are homogeneous, the variance of the repeatability pharmacopoeial products. It must be accepted that a method
can be calculated. The inter-group variance of the results is of systematics needs to be internally consistent, but may
calculated. The variance of the intermediate precision is the differ from others in identification of isolates. In other words,
sum of the variance of the repeatability and the inter-group identification of an iso late based on biochemical activity may
variance. The coefficients of variation are then calculated. lead to one conclusion, identification by fatty acid analysis
Generally, a coefficient of variation in the 10-15 per cent range to another, identification by DNA analysis may lead to a
is acceptable. Irrespective of the specific results, the alternative third, and other methods may lead to alternative conclusions.
method must have a coefficient of variation that is not larger Microbiological identifications by a particular system flow
than that of the pharmacopoeial method. directly from previous experience with that system, and
3-3-3. Spedficity therefore may well differ from identifications by another
The specificity of an alternative quantitative method is system. It is critical that each system provides a consistent
demonstrated using a range of appropriate micro-organisms. identification of isolates from pharmacopoeial products.
Where relevant for the purpose of the test, mixtures of 3-4-1. Accuracy
micro-organisms are used during validation.
The accuracy of an alternative identification method is
3-3-4. Limit of quantification its ability to identify the desired micro-organism to the
The limit of quantification of an alternative quantitative required taxonomic leve! and to differentiate it from
method is the lowest number of micro-organisms that can be other micro-organisms present in the sample. 1t must be
accurately counted. As it is not possible to obtain a reliable demonstrated with a series of test micro-organisms or

566 See the information section on general monographs (caver pages)


EUROPEAN PHARMACOPOEIA 8.0 5.1.6. Ahernative methods for control of micmbiologi.cal quality

micro-organisms obtained from a typical sample previously construction of a correct phylogenetic correlation tree, another
identified by another method. answer may be more useful in the context of pathogenicity or
3-4-2. Pred§ion other properties of the differentiated micro-organisms.
The precision of an alternative identification method is the 4-1-1. Primary validation
degree of agreement among individual test results when the In order to characterise a specific microbiological method,
procedure is applied repeatedly to multiple samplings of the principIe of detectiol1 must be clearly described by the
suspensions of test micro-organisms across the range of the supplier. The method must be fully detailed with respect to
test. the conditions required for application, the materials and
equipment needed, and the expected signal. The application
3-4-3. Robustness
principIe should be described in a peer-reviewed journal.
The robustness of an alternative identification method is a The principIe of detection must be characterised in a model
measure of its capacity to remain unaffected by small but system and/or with a panel of test micro-organisms, by at least:
deliberate variations in method parameters, and provides
an indication of its reliability under a variety of normal - prerequisite treatment of sampIe or micro-organisms;
test conditions such as different analysts, instruments, - type of response;
batches of reagents and laboratories. Robustness can be - specificity of the response;
defined as the intrinsic resistance to the influences exerted - limit of detection;
by operational and environmental variables on the results
- range;
of the microbiological method. Robustness is a validation
parameter best suited to determination by the supplier of the - linearity of the response;
method, but if critical parameters are modified by the user - accuracy and precision of the response;
their effects on robustness have to be evaluated. Robustness - robustness of the method in a model system;
of an identification method is judged by its ability to identify - limits of suitability.
consistently the test micro-organisms under the deliberate
variations to the method parameters. Once the method has been characterised in this way by the
supplier, the principIe of detectiol1 need not be verified by
4. SPECIFIC VALIDATION REQUIREMENTS each user.
4-1. BACKGROUND 4-1-2. Validation of alternative mkrobiological method
Validation is defined in various contexts with sorne differences, 4-1-2-1. Risk-benefit analysis
but a consensus definition is to establish documented evidence For validation of specific alternative microbiological methods
that a process will consistentIy achieve what it is intended it is critical that the purpose of the procedure is precisely
to do. Hence, in order to perform correct validation of a outlined. Based on the purpose, the type and depth of
new method it is critical to understand and define what the information needed must be defined. The information
procedure is intended to achieve. obtained by, and the limitations of, the conventional method
2 levels of validation must be envisaged for the application and the alternative method must be considered and compared
of conventional or alternative microbiological methods. in a risk-benefit analysis.
Primary validation of a method is typically performed by An alternative method can be justified as being applicable if
the supplier of the new method, whereas validation for the the information obtained gives a scientifically sound answer
actual intended use, which is a verification of the suitability to the questions asked in the procedure, and if the limitatiol1s
or applicability of the method in a given situation, must be of the method are not more severe than the limitations of the
seen as the responsibility of the user. Before validation for the conventional method.
actual intended use, performance qualification is carried out 4-1-2-2. Validation for the actual in tended use
by the user as described in 3. General validation requirements. The alternative method must be applied in the procedure used
Typically, microbiological methods use specific characteristics and with the samples to be analysed under the responsibility
of micro-organisms as indicators or detection principIes for of the user, and must be shown to give comparable results
more general questions. The information needed is presence, as characterised in a model system by the supplier. Specific
number, viability, resistance or identity of micro-organisms in questions to be asked where applicable are:
a given product or environment. - compatibility of the response with the sample preparation
A given method will usually give an indirect and conditional needed for product testing;
answer to the questions. For example, the total number and - limit and range of detection of the method with regard to
viability of micro-organisms is indicated by the number of sample size and sample availability;
micro-organisms able to reproduce under a certain set of
- specificity of the response with regard to the influence of
conditions for sample preparation, cultivation and incubation.
the ingredients of the product;
Reproduction in classical microbiology is hence taken as the
general indicator for viability. There are other parameters, - linearity of the response with regard to all types of samples
however, that can be used as an indication of viability. The to be analysed;
level of ATP or accumulation or metabolism of substrates - accuracy and precision of the response with regard to aH
in living cells can also be taken as an indicator for viability. types of samples to be analysed;
The results of different indication methods for viability may - robustness of the method with regard to all types of samples
not always be identical. Micro-organisms may not be able to to be analysed.
reproduce on a given medium, but may still accumulate and Acceptance criteria for the method in routine use will need
metabolise a substrate. Micro-organisms may be unable at a to be defined as a function of the application and of the
given state of damage to accumulate a substrate, but may still validation data.
be able to recover and to reproduce.
4-2. BIOLUMINESCENCE FOR ENUMERATION OF
Another example is the various methods used for identification MICRO-ORGANISMS
of micro-organisms. Characterisation of the metabolic
pattern of micro-organisms is frequently used for species 4-2-1. Risk-benefit analysis
identification, whereas another method consists of the Extensive scientific evidence and use for years supports the
comparison of DNA sequences. Again, the answer obtained capability of the ATP viability marker to detect the same
may not be fully coincident for the different identification range of micro-organisms as is encountered using standard
methods, and while one answer may be appropriate for the plating methods. Since this method is growth-dependent,

General Notices (1) apply to all monographs and other texts 567
5.1.6. Altematlve methods Íor control oí microbiological quality EUROPEAN PHARMACOPOEIA 8.0

the improvement comparing to the plating methods is the with those obtained using standard plate count. Multiple
rapidity to obtain a result (from 5 days with the plating replicates (at least 5) from overnight cultures diluted across a
methods to 24 h for bioluminescence). It is possible to identify concentration range (e.g. 100 per cent, 75 per cel1t, 50 per cent,
the bioluminescence-detected micro-organisms from the 25 per cent and 10 per cent) must be used to evaluate linearity,
incubation step medium, but it has to be remembered that accuracy, precision, range, specificity, limit of quantification
in a mixed culture sorne micro-organisms may out-compete (quantitative method) and limit of detection (flow cytometry
others during incubation. This method provides evaluation of with pre-incubation step). Since cytometry has high sensitivity
samples within 24 h for filterable and non-filterable products (solid phase cytometry can detect single cells, whereas flow
(water, in-process control, environmental samples, solid and cytometry is sensitive to a leve! of around 10-50 cells per
liquid raw materials, solid and liquid finished products, etc.) millilitre), and detection is not growth based, the linearity of
and for a large number of samples, when the detection step the instrumentation can be tested by comparison of the actual
is automated. resuIts with the expected value.
4-2-2. Vali.dation for the actual intended use Following this step, validation proceeds in 2 phases: validation
The method relies upon the detection of ATP from viable with respect to the product to be examined and comparative
micro-organisms. Performance qualification is carried out testing. Results of each phase must be evaluated against
with test micro-organisms to make sure that under the pre-determined acceptance criteria using positive and negative
conditions applied by the user laboratory it is possible to controls:
satisfy the criteria described by the supplier for precision, - phase 1: individual materials to be evaluated by cytometry
accuracy and linearity (quantitative method), or limit of must be 'spiked' with a defined leve! of micro-organisms to
detection (qualitative and semi-quantitative method) over ensure that the sample preparation process and the samples
the range required for the intended use. Following this step, themselves do 110t have an impact upon the performance
validation proceeds in 3 phases: of the detection system; specifically, the sample matrix
- phase 1: fertility of the medium in the presence of the must not affect detection (i.e. contain endogenous
product (if an incubation step is performed); chromophores, auto-fluorescent particles), and in the case
of flow cytometry, sample size/dilution and flow rate must
- phase 2: search for interferences that may increase or inhibit
be determined for optimal performance;
the ATP production (by addition of an ATP standard
solution to the product to test); - phase 2: testing must be performed in which the results
- phase 3: comparative testing with the pharmacopoeial
obtained by cytometry and the pharmacopoeial method are
compared; the number of samples and the testing period
method.
must be defined in a comparability protocol; the number
A detailed example of validation of the bioluminescence of samples required will vary, but must be representative of
method is given at the end of this chapter. the material evaluation process (i.e. time/number), and
4-3. CYTOMETRY (SOLID AND FLOW) FOR must allow for statistical evaluatiol1; al! samples must be
ENUMERATION OF MICRO-ORGANISMS prepared according to defined procedures and evaluated
4-3-l. Risk-benefit analysis against selected validation and acceptance criteria, similar
to those used for pure culture evaluation.
Extensive scientific evidence supports the capability of
this fluorescence viability marker to detect and/or count 4-4. FATTY A CID PROFILES POR IDENTIFICATION
a wider range of micro-organisms than are encountered 4-4-1. Risk-benefit analysis
using standard plating methods. Cytometry will detect all Idel1tification by fatty acid profiles may be more precise
viable micro-organisms including sorne that may not be than the identification methods based 011 metabolic profiles
discernable by growth-based methods. Whilst being rapid, the in conventional microbiological culture methods. The
recovery of micro-organisms post-analysis is limited. Thus database is broader than for conventional culture methods.
the further processing of analysed samples for identification Pre-incubation is needed, but extraction and identification is
would require alternative fluorescent stains or an alternative faster than in biochemical methods and hence, the result is
method. Currently it is not possible to use this method for obtained faster. Other modern methods, such as 16S rRNA
routine identification of micro-organisms, although basic sequence analysis or genetic fingerprinting, have a similar
morphology is readily discernable in solid phase cytometry broad differentiation range and give a result as fast as this
under fluorescent microscopes. This method provides rapid method.
evaluation of samples and hence allows for a proactive
approach to pharmaceutical manufacturing, facilitating Separation of closely related micro-organisms (e.g. E. coli
building quality into pharmaceutical operations. This method and Salmonella spp.) can be difficult by fatty acid profiles.
is not growth-dependent and hence all metabolically active Where the identification of closely related micro-organisms
micro-organisms will be detected. However, the limit of is especially important, other systems may give more precise
detection for flow cytometry is currently such that it cannot results. For a given application it is important to specify which
be used for enumeration by direet examination for most types of micro-organisms are most important to be identified.
pharmaceutical samples. If pre-incubation is necessary, the If it is most critical to characterise the carrect phylogenetic
estimation becomes semi-quantitative (limit test). species of the isolate, DNA sequence-based identification
methods will give more reliable results.
4-3-2. Validation f01" the actual intended use
Limitations of identification by fatty acid profiles are also seen
The method relies upon the detection of a fluorescent signal in the necessity to grow micro-organisms on standardised
from labelled micro-organisms. media under standard temperature conditions and durations
Performance qualification is carried out to ensure that of incubation. Micro-organisms that cannot be cultivated on
the instruments perform within their defined operational such media cannot be identified.
parameters. This involves the use of fluorescent standards of
4-4-2. Validation for the actual ¡ntended use
prescribed intensity and cultures of known type and number
of micro-orgal1isms. These tests challenge the quantitative Using a rauge of test micro-organisms and at least 3 replicate
detection system. Reagents and consumables (negative determinations in each case, it must be demonstrated that the
cOl1trols) must also be utilised to ensure that the routine test method yields consistent results.
protocol is applicable, and that the quality of the materials A significant number of iso lates from typical samples to be
used in the test do not contribute to the final result. Pure analysed by the user must be identified, at least 3 times each.
culture experiments involving test micro-organisms are used The results in each case should be consistent and in accord
to challenge the detection system, and to compare test results with those obtained using alternative identification methods.

568 See the information section on general monographs (ca ver pages)
EUROPEAN PHARMACOPOEIA 8.0 5.1.6. Altemative methods fOí control of microbiological quality

Where a different identification result is found in another determination in the quantity analysed. Using more than a
identification system, the reason for the difference must be single sample quantity, the system may offer semi-quantitative
investigated. Where a scientifically plausible explanation exists determination (limit test). For example, the classical tested
for the recognition of a different species, a difference between quantity for viable aerobic count on non-sterile products is
identiflcation systems may be acceptable. In such a case it 0.1 g or 0.1 mL leading to absence in 0.1 g or 0.1 mL, i.e.
must be assured that the recognition of the identifled species less than 10 micro-organisms in 1 g or 1 mL for a negative
is robust. It must also be assured that the system does not result and more than or equal to 10 micro-organisms in 1 g
group poorly recognised iso lates under one 'species' thereby or 1 mL in case of a positive result. rf 0.01 g or 0.01 mL is
simulating the repeated isolation of a single species. tested simultaneously, a negative result corresponds to a
4-5. NUCLEIC A CID AMPLIFICATION TECHNIQUES number of micro-organisms less than 100 in 1 g or 1 mL.
The combination between negative for 0.01 g or 0.01 mL
4-5-1. Risk-benefit analysis and positive for 0.1 g or 0.1 mL permits an estimate ofthe
NAAT are widely used in diagnostics for their precision and contamination level of the product to be less than 100 but more
rapidityat a relatively low cost (for the analysis, but not for the than or equal to 10 micro-organisms in 1 g or 1 mL.
instruments,) when compared with the traditional methods. As mentioned in section 2., bioluminescence can be used as
Provided that speciflc validations have been performed, when a quantitative method if micro-organisl11s are captured on a
NAAT are appropriately used, they may offer advantages filtration membrane and later incubated in culture medium
in sorne flelds in comparison to classical methods; on the (bioluminescence 011 membrane).
other hand classical methods are generally more easily
The protocol below describes validation aspects for qualitative,
standardisable, need a lower level of technical competence and
semi-quantitative and quantitative methods.
may have lower costs. Even when NAAT are not more difflcult
to perform than traditional methods, the interpretation of the PERFORMANCE QUALIFICATION OF THE
results generally needs a high degree of scientific competence. ALTERNATIVE METHOD
When used for identification, DNA-based methods cannot Spedfidty
discriminate between dead and live micro-organisms. That
means that they cannot be directly used on the product Screen the method with test micro-organisms appropriate to
but only after passage on a traditional culture medium, the method. For example, for microbial aerobic viable eount
thereby losing part of the advantage in rapidity. Moreover, on non -sterile products, use at least the micro-organisms
if used directly on the product at the end of the analysis, described in chapter 2.6.12 for the fertility ofthe media in the
these methods do not result in a strain to be used in presence of product. This determination is performed at least
further experiments and may not give advantages when the 3 times with each micro-organismo Acceptance criteríon: al!
micro-organisms to be detected are poorly cultivable or test micro-organisms are successfully detected.
stressed. RNA amplification techniques (e.g. RT -PCR) may Limit of detection (only f01" semi-quantitative Ol" qualitative
identify living micro-organisms (but not spores) directly in methods)
the products, but in comparison to traditional methods are Prepare a low inoculum (abont 5 CFU in the initial sample)
much more difficult to use routinely. On the other hand, of each test micro-organismo Perform the analysis in at
where specific primers are used, identification (or typing) by least 5 replicates with the pharmacopoeial method and
NAAT is more precise than the traditional methods and in with the bioluminescence method concerned. Acceptance
sorne cases may have other advantages: for instance for the criterio n : the ability of the 2 methods to deteet the presence
identification of sorne vaccines (e.g. cholera vaccine, whole of a single micro-organism can be demonstrated using the
ceH pertussis vaccine,) their use may substitute for that of X2 test. Alternative procedure: prepare a series of dilutions
specific sera and contribute to reducing the use of animals, or of micro-organisms to have a count in the next dilution
may give a very speciflc identification where this is presently of about 5 CFU per il10culum (e.g.: 10 CFU/inoculum,
lacking (e.g. BCG vaccine). 5 CFU/inoculum, 2.5 CFU/inoculum, 1.25 CFU/inoculum,
These methods are in general non-quantitative (PCR) or 0.75 CFU/inoculum). Perform the test on 5 independent series
semi-quantitative (real-time PCR), meaning that their results of dilutions with the pharmacopoeial method and with the
cannot be compared with those of a colony count where an bioluminescence method concerned. Determine the limit of
exact enumeration of the micro-organisms present in the detection for each method. It corresponds to the last dilution
sample is requested, but even if colony count has a valen ce where the result is positive for the 5 series. Acceptance
consolidated in time this dogma may not be verified for criterion: the limit of detection of the bioluminescence
bacteria which have a tendency to clump (mycobacteria) or are method is equal to or lower than that of the pharmacopoeial
organised in chains or in clusters (streptococci, staphylococci), method.
therefore an accurate standardisation of the semi-quantitative Limit of quantification (quantitative method)
methods may give results of comparable reliability.
This can be performed at the same time as the linearity
4-5-2. Validation for the actual intended use determination. It corresponds to the lowest concentration
The method is validated according to chapter 2.6.21. of the chosen range that satisfies the criteria for linearity,
Comparison of conventional and PCR-based methodologies, accuracy and precision. Acceptance criterion: the limit of
which differ in sensitivity and specificity, is particularly quantification of the bioluminescence method is equal to or
difficult and may lead to divergent conclusions. lower than that of the pharmacopoeial method.
The following example is published for information and not Fredsion
for general application. Quantitative evaluatíon. For each test micro-organism,
perform at least 5 replicates during the same series including
Example validation of an alternative method: at least the concentration of micro-organisms corresponding
to the middle of the range. Perform 3 independent tests.
detailed protocol followed by a laboratory Carry out a statistical analysis to compare the precision of
for the implementation of bioluminescence the 2 methods or calculate the coefficient ofvariation (CV).
Acceptance criterion: CV 15 per cent to 30 per cent or
BACKGROUND precision not different with the risk alpha equal to 5 per
Methods using a pre-incubation step inliquid medium cent between the 2 methods. lf precision is different, the
(bioluminescence in tube or microtitre platel do not bioluminescence method is better than the pharmacopoeial
offer quantitative information but a presence/absence method, indicated by a smaller standard deviation.

General Nofices (1) apply to all monographs and other texts 569
5.1.6. Alternative methods for control of microbiological quality EUROPEAN PHARMACOPOElA 8.0

Qualitative or semi-quantitative evaluation. Use the alternative - phase 2: the absence of interference from the product that
procedure described for setting the limit of detection and may increase or inhibit ATP production;
report the frequency of positive results in parallel with the - phase 3: the testing of the product in parallel with the
pharmacopoeial method. Acceptance criterion: the frequency pharmacopoeial method.
of positive results at the detection limit is 100 per cent and
this frequency is better than or equal to the pharmacopoeial These 3 parts of validation are performed on 3 independent
method. tests using for example at least 2 different batches of producto
Linearity Phase 1: fertility of the medium in the presence of the
For each test micro-organism, prepare 5 concentrations in product
the range of the bioluminescence method (range is normally If the product has a known high contamination level (more
indicated by the supplier). Perform the pharmacopoeial and than 500 micro-organisms per gram or millilitre) the
the bioluminescence methods in parallel. Repeat this test incubation step is unnecessary, the micro-organisms can
2 further times to have results on 3 independent tests. Test be detected direcdy. In this case testing the fertility of the
for linear regression, presence of a slope, and lack of fit with medium in the presence of the product is not necessary.
the F test at alpha equal to 5 per cent. If statistical analysis is However, pharmaceutical products are generally contaminated
not possible, calculate the correlation coefficient (r') and the at a much lower level and growth of the micro-organism is
slope between the 2 methods. Acceptance criterio n : statistical necessary to obtain detection with bioluminescence. It must
analysis may show linear regression, the presence of a slope therefore be proven that the product does not inhibit the
and no lack of fit with a risk of 5 per cent. Equation y = a + bx growth of micro-organisms under the conditions of the test.
is determined where b is the slope and a the intercepto If In order to do so, separately add inoculum at not more than
no statistical analysis is avai1able, r2 is at least 0.9 and the 100 CFU for each test micro-organism into the portion of
slope does not diverge by more than 20 per cent from 1 (b medium containing the product. For bioluminescence in
between 0.8 and 1.2). If the linearity is not demonstrated in tube 01' microtitre plate, perform the bioluminescence test.
such a large range, the range can be decreased and linearity For bio1uminescence on membrane, incubate at 30-35 oC or
demonstrated with only 3 concentrations in place of 5. 20-25 oC for 5 days and count the bioluminescent colonies
Accuracy on the membrane. Acceptance criterion: the test is positive
(bioluminescence in tube or microtitre plate); the quantitative
Quantitative evaluation. Accuracy can be determined with recovery of the micro-organism is at least 70 per cent
data obtained in linearity. For each micro-organism use (bioluminescence on membrane).
3 to 5 concentrations within the linear range of the method.
Perform statistical ana1ysis (Student's t test at risk 5 per cent) Phase 2: search for interference of the product
to test the conformity of the estimated slope (value = 1) The objective is to show that the product do es not add stray
versus the obtained slope and to test the conformity of light or non-microbial ATP (does not lead to false positive
the estimated intercept (value = O) versus the obtained result: criterio n A) or does not decrease the ATP detection
intercept. For examp1e, if the estimated slope is b with a (does not lead to a false negative resnlt: criterion B).
standard deviation 5(0) of 0.090 with 5 concentrations of
Bioluminescence in tube or microtitre plate
micro-organisms, calculate t = (b - l)/s O,)' For intercept a,
with standard deviation equal to sra)' t = (a - O)/s(a)' Compare A. Perform the bioluminescence test with the culture broth
these values to the Student's t at 5 per cent, for 13 degrees of alone and with the culture broth in the presence of the
freedom (3 tests, 5 concentrations). Acceptance criterion: if producto Determine the RLU value for culture broth alone
the t values obtained are less than the Student's t, the method and the RLU value for culture broth in the presence of
is exact in the applied range. In the case that there is no product.
conformity for the s10pe (slope different from 1) or for the B. Perform the bioluminescence test with the culture broth
intercept (intercept different from O) the method is not exact alone and the culture broth in the presence of ATP.
over the applied range. Determine the response coefficient for ATP concentration
Qualitative or semi-quantitative evaluations. Use the in per cent.
alternative pro ce dure described for setting the limit of
Acceptance criterion:
detection. Calculate the proportion of false negatives for
bioluminescence and for the pharmacopoeial method over - criterion A: the RLU value of culture broth in the presence
all tested dilutions. Compare the extent of false negatives of product is less than twice the RLU value of culture
for the 2 or 3 concentrations of micro-organisms just broth alone (if criterion A is not satisfied, it is necessary to
under the detection limit (for example 5 CFU/inoculum, determine a specific threshold for this product);
2.5 CFU/inoculum or l.25 CFU/inoculum) giving a positive - criterion B: the RLU value of culture broth in the presence
resulto By definition, the detection limit corresponds to O per of product and ATP is within the interval 25 per cent
cent of false negatives. Acceptance criterio n : the percentage to 200 per cent of the RLU value of culture broth in the
of false negatives for the bioluminescence method at sample presence of ATP.
concentrations below the detection limit must be equal to or
lower than that of the pharmacopoeial method. Bioluminescence on membrane: perform the complete
bioluminescence test to search for interference. Acceptance
Range criterio n : the recovery of micro-organisms is greater than or
This is the interval between the lowest and the highest equal to 70 per cent and not more than 200 per cent.
concentrations of micro-organisms where linearity, precision
Phase 3: analysis of the product in parallel with the
and accuracy have been demonstrated.
pharmacopoeial method
Robustness
Perform the test according to the validated method for the
The information is given by the supplier. product concerned in paralle1 with the pharmacopoeial
method to show the relationship between the 2 methods for
VALIDATION FOR THE ACTUAL INTENDED USE
the product concerned, on 3 independent tests and using
In the example given, there was no need to determine the at least 2 different batches. Express the result as positive or
accuracy and detection limit in the presence of the product. negative in a certain quantity (bioluminescence in tube or
The validation consists of 3 parts, verifying: microtitre plate) or express the count per filtered quantity
- phase 1 : the fertility of the medium in the presence of the (bioluminescence on membrane). Acceptance criterion:
product; results must be correlated with the pharmacopoeial method.

570 See the information section on general monographs (cover pages)


EUROPEAN PHARMACOPOEIA 8.0 5.1.8. Microbiological quality ofherbal medicinal products and extracts

01/2008:50107 The risk assessment can be based mainly on the manufacturing


conditions if these indude rigorous inactivation steps (for
example, for gelatin etc., and products terminally sterilised by
5.1.7. VIRAL SAFETY steam or dry heat as described in the general texts on sterility
(5.1)).
This chapter provides general requirements concerning the
viral safety of medicinal products whose manufacture has
involved the use of materials of human or animal origino 0112014:50108
Since viral safety is a complex issue, it is important that a
risk assessment is carried out. Requirements to be applied to 5.1.8. MICROBIOLOGICAL QUALITY
a specific medicinal product are decided by the competent
authority. OF HERBAL MEDICINAL PRODUCTS
Where the risk of viral contamination exists, complementary FOR ORAL USE AND EXTRACTS USED
measures are used as appropriate to assure the viral safety of IN THEIR PREPARATION
medicinal products, based on:
- selection of source materials and testing for viral This general chapter presents recommended acceptance criteria
contaminants; for the microbiological quality of both herbal medicinal
products for oral use and the extracts that are used in their
- testing the capacity of the production process to remove preparation.
and/or inactivate viruses;
- testing for viral contamination at appropriate stages of Microbial examination of non-sterile products is performed
production. according to the methods given in general chapters 2.6.12,
Where appropriate, one or more validated procedures for 2.6.13 and 2.6.31. Acceptance criteria based upon the total
removal or inactivation of virus es are applied. aerobic microbial count (TAMC) and the total combined
yeasts/moulds count (TYMC) are given below.
Further detailed recommendations on viral safety, induding
Acceptance criteria are based on individual results or on
validation studies, are provided, in particular, by the Note for
the average of replicate counts when replicate counts are
guidance on virus validation studies: the design, contribution
performed (e.g. direct plating methods).
and interpretation of studies validating the inactivation and
removal of viruses (CPMPlBWPI268195) of the Committee for A list of specified micro-organisms for which acceptance
Proprietary Medicinal Products, and the IeH guideline Q5A: criteria are set can be found below. The list is not necessarily
Viral safety evaluation of biotechnology products derived from exhaustive and for a given preparation it may be necessary
celllines of human or animal origin (induding any subsequent to test for other micro-organisms depending on the nature
revisions of these documents). of the starting materials, the manufacturing process and the
intended use.
Requirements concerning immunological products for
veterinary use are dealt with in the monographs Vaccines HERBAL MEDICINAL PRODUCTS
for veterinary use (0062) and Immunosera for veterinary use A. Herbal medicinal producís containing herbal drugs,
(0030) and related general chapters. with or without excipients, intended for the preparation of
Risk assessment infusions and decoctions using boiling water (for example
herbal teas, with or without added flavourings)
A risk assessment with respect to viral safety is carried out
where materials of human or animal origin are used as TAMC (2.6.12) Acceptance criterian: 10 7 CFU/g
ingredients of medicinal products or in the manufacture of Maximum acceptable caunt: 50000000 CFU/g
active substances, excipients or medicinal products. TYMC (2.6.12) Acceptance criterian: 10 5 CFU / g
The principIe of the risk assessment is to consider various Maximum acceptable caunt: 500000 CFU/g
factors that may influence the potentiallevel of infectious Escherichia colí
Acceptance criterian: 10 3 CFU / g
partides in the medicinal product and factors related to the (2.6.31)
use of the medicinal product that determine or influence the Salmonella (2.6.31) Absence (25 g)
viral risk to the recipients.
B. Herbal medicinal products containing, for example,
The risk assessment takes into consideration relevant factors, extracts and/or herbal drugs, with or without exdpients,
for example: where the method of processing (for example, extraction)
- the species of origin; or, where appropriate, in the case of herbal drugs, of
pre-treatment reduces the levels of organisms to below
- the organ, tissue, fluid of origin;
those stated for this category
- the potential contaminants in view of the origin of the
TAMC (2.6.12) Acceptance criterian: 104 CFU / g ar CFU /mL
raw material and the history of the donor(s), preferably
Maximum acceptable caunt: 50 000 CFU/g
induding epidemiological data; ar CFU/mL
- the potential contaminants from the manufacturing TYMC (2.6.12) Acceptance criterian: 102 CFU/g or CFU/mL
process (for example, from risk materials used during Maximum acceptable caunt: 500 CFU/g
manufacture) ; ar CFU/mL

- the infectivity and pathogenicity of the potential Bile-talerant


gram-negative Acceptance criterian: 102 CFU/g or CFU/mL
contaminants for the intended recipients of the medicinal bacteria (2.6.31)
product, taking account of the route of administration of Escherichia colí
the medicinal product; Absence (1 g ar 1 mL)
(2.6.31)
- the amount of material used to produce adose of medicinal Salmonella (2.6.31) Absence (25 g or 25 mL)
product;
C. Herbal medicinal products containing, for example,
- controls carried out on the donor(s), on the raw material, extracts and/or herbal drugs, with or without excipients,
during production and on the final product; where it can be demonstrated that the method of processing
- the manufacturing process of the product and its capacity (for example, exíraction with low-strength ethanol or water
to remove and/or inactivate viruses. that is not boilil1g, or low-temperature concentration)

General Notices (1) apply to all monographs and other texts 571
5.1.9. Guidelines for using the test for sterility EUROPEAN PHARMACOPOEIA 8.0

or, in the case of herbal drugs, of pre-treatment, would and the efficiency of the adopted sampling plan. Hence for
not reduce the leve! of organisms sufficiendy to reach the the purpose of this text a batch is defined as a homogeneous
criteria required under B collection of sealed containers prepared in such a manner that
the risk of contamination is the same for each of the units
TAMC (2.6.12) Acceptance criterion: 105 CFU/g or CFU/mL
contained therein.
Maximum acceptable count: 500 000 CFU/g
or CFU/mL In the case of terminally sterilised products, physical proofs,
TYMC (2.6.12) Acceptance criterion: lO' CFU/g or CFU/mL biologically based and automatically documented, showing
Maximum acceptable count: 50 000 CFU/g correct treatment throughout the batch during sterilisation are
or CFU/mL of greater assurance than the sterility test. The circumstances
Rile-tolerant in which parametric release may be considered appropriate
gram -negative Acceptance criterion: 10 4 CFU/g or CFU/mL are described under 5.1.1. Methods of preparation of sterile
bacteria (2.6.31) products. The method of media-fill runs may be used to
Escherichia coli
Absence (1 g or 1 mL)
evaluate the process of aseptic production. Apart from that,
(2.6.31) the sterility test is the only analytical method available for
Salman ella (2.6.31) Absence (25 g or 25 mL) products prepared under aseptic eonditions and furthermore
it is, in all cases, the only analytical method available to the
authorities who have to examine a specimen of a product for
EXTRACTS sterility.
Extracts should fulfill the acceptance criteria for category B The probability of detecting micro-organisms by the test for
herbal medicinal products. However, where it can be sterility increases with their l1umber present in the sample
demonstrated that the method of processing would not tested and varies according to the readiness of growth of
reduce the level of micro-organisms sufficiently to reach the micro-organism present. The probability of detecting very
category B criteria, the extracts shall meet the requirements low levels of contamination even when it is homogenous
for category C herbal medicinal products. throughout the batch is very low. The interpretation of the
The recommended acceptance criteria apply to extracts that results of the test for sterility rests on the assumption that
are to be incorporated into herbal medicinal products for the contents of every container in the batch, had they been
oral use. More-stringent acceptance criteria may be required tested, would have given the same result. Since it is manifest
for extracts that are to be incorporated into pharmaceutical that every container cannot be tested, an appropriate sampling
preparations to be administered by other routes in order plan should be adopted. In the case of aseptic production, it is
to satisfy the acceptance criteria for the intended route of recommended to indude samples filled at the beginning and
administration (5.1.4). at the end of the batch and after significant intervention.
It is recognised that for some herbal medicinal products and
OBSERVATION AND INTERPRETATION OF RESULTS
extracts used in their preparation the criteria given above for
TAMC, TYMC and bile-tolerant gram-negative bacteria cannot Conventional microbiological/biochemical techniques are
be met because of the typicallevel of microbial contamination. generally satisfactory for identification of micro-organisms
Less-stringent acceptance criteria may be applied on the basis recovered from a sterility test. However, if a manufacturer
of a risk assessment that takes account of qualitative and wishes to use condition (d) as the sole criterion for invalidating
quantitative characterisation of the microbial eontamination a sterility test, it may be neeessary to employ sensitive typing
(¡nd the in tended use of the herbal medicinal product or extracto techniques to demonstrate that a micro-organism isolated
Jf it has been shown that none of the prescribed tests for a herbal from the product test is identical to a micro-organism isolated
medicinal product or extraet will allow valid enumeration of
from the test materials and/or the testing environment. While
micro-organisms at the leve! prescribed, a validated method
routine microbiological/biochemical identification techniques
with a limit of deteetion as close as possible to the indicated
can demonstrate that 2 isolates are not identical, these
aeceptance criterion is used. methods may not be sufficiently sensitive or reliable enough to
provide unequivocal evidence that 2 isolates are from the same
source. More sensitive tests, for example molecular typing
0112009:50109 with RNA/DNA homology, may be necessary to determine
that micro-organisms are clonally related and have a common
5.1.9. GUIDELINES FOR USING THE origino

TEST FOR STERILITY


0112010:50110
The purpose of the test for sterility (2.6.1), as that of aH
pharmacopoeial tests, is to provide an independent control
analyst with the means of verifying that a particular material
5.1.10. GUIDELINES FOR USING THE
meets the requirements of the European Pharmacopoeia. A TEST FOR BACTERIAL ENDOTOXINS
manufacturer is neither obliged to carry out such tests nor
precluded from using modifications of, or alternatives to, the 1. INTRODUCTION
stated method, provided he is satisfied that, if tested by the Endotoxins from gram-negative bacteria are the most
official method, the material in question would comply with common cause of toxic reactions resulting from contamination
the requirements of the European Pharmacopoeia. of pharmaceutical products with pyrogens; their pyrogenic
activity is much higher than that of m05t other pyrogenic
PRECAUTIONS AGAINST MICROBIAL substances. These endotoxins are lipo-polysaccharides.
CONTAMINATION Although there are a small number of pyrogens which possess
Aseptic conditions for performance of the test can be achieved a different structure, the conclusion is generally justified that
using, for example, a class A laminar-air-flow cabinet located the absence of bacterial endotoxins in a product implies the
within a class B clean room, or an isolator absence of pyrogenic components, provided the presence of
non-endotoxin pyrogenic substances can be ruled out.
GUIDANCE TO MANUFACTURERS The presence of endotoxins in a product may be masked
The level of assurance provided by a satisfactory result of a by factors interfering with the reactiol1 between the
test for sterility (the absence of contaminated units in the endotoxins and the amoebocyte lysate. Hence, the analyst
sample) as applied to the quality of the batch is a function of who wishes to replace the rabbit pyrogen test required in a
the homogeneity of the batch, the conditions of manufacture pharmacopoeial monograph by a test for bacterial endotoxins

572 See the informatíon section on general (caver pages)


EUROPEAN PHARMACOPOEIA 8.0 5.LIO. Guidelines for using the test for bacterial endotoxins

has to demonstrate that a valid test can be carried out on the In setting a threshold concentration of endotoxin for the
product concerned; this may entail a procedure for removing product to be tested, due attention should be paid to the dose
interfering factors. of the product: the threshold should be set so as to ensure that
as long as the endotoxin concentration in the product remains
As indicated in the test for bacterial endotoxins (2.6.14),
below this threshold even the maximal dose administered
information must be available on the 2 following aspects
by the intended route per hour do es not contain sufficient
before a test on a sample can be regarded as valido
endotoxin to cause a toxic reaction.
- The suitability of the material to be used for the test has When the endotoxin concentration in the product exactly
to be established. The absence of endotoxins in the water equals the threshold value, gelation will occur, as is the case
for BET and in the other reagents must be assured and the when the endotoxin concentration is much higher, and the
sensitivity of the amoebocyte lysate must be checked to product will faH the test, because the all-or-none character
confirm the sensitivity dedared by the manufacturer. of the test makes it impossible to differentiate between a
- As the product to be examined may interfere with the concentration exactly equal to the threshold concentration
test, the sensitivity of the amoebocyte lysate is determined and one that is higher. It is only when no gelation occurs that
in the presence and in the absence of the product under the analyst may condude that the endotoxin concentration is
examination. There must be no significant difference below the threshold concentration.
between the 2 sensitivity values. For products in the solid state, this threshold concentration
The text 2.6.14. Bacterial endotoxins indicates methods for of endotoxin per mass unit or per International Unit (IU) of
removing interfering factors; in the case of interference, product has to be translated into a concentration of endotoxin
another test must be carried out after such a method has been per millilitre of solution to be tested, as the test can only be
applied to check whether the interference has indeed been carried out 011 a solution. The case of products that already
neutralised or removed. exist in the liquid state (such as infusion fluid s) is discussed
below.
This general chapter explains the reasons for the requirements
Endotoxin limit: the endotoxin limit for active substances
in the test for bacterial endotoxins, then deals with the reading
administered parenterally, defined on the basis of dose, is
and interpretation of the resuIts.
equal to:
Substitution of the rabbit pyrogen test required in a K
pharmacopoeial monograph by an amoebocyte lysate test
constitutes the use of an alternative method of analysis and NI
hence requires validation; some guidance on how to proceed
is given in section 11. K threshold pyrogenic dose of endotoxin per
kilogram of body mass;
The reference method for bacterial endotoxins is stated in the
IV[ maximum recommended bolus dose of product
monograph on a given product; where no method is stated,
per kilogram of body mass.
method A is the reference method. If a method other than the
reference method is to be used, the analyst must demonstrate When the product is to be injected at frequent intervals
that the method is appropriate for this product and gives a or infused continuously, NI is the maximum total dose
result consistent with that obtained with the reference method administered in a single hour periodo
(see also Section 13). The endotoxin limit depends on the product and its route of
administration and is stated in the monograph. Values for K
2. METHOD are suggested in Table 5.1.1 0.-1.
The addition of endotoxins to amoebocyte lysate may result in For other routes, the acceptance criterio n for bacterial
turbidity, precipitation or gelation (gel-dot); only the gel-dot endotoxins is generally determined on the basis of results
method was used in the Pharmacopoeia as an evaluation obtained during the development of the preparation.
criterion in the first type of test for bacterial endotoxins. The
Table 5.1.10.-1
advantage was the simplicity of basing the decision to pass or
fail the product under examination 011 the absence or prese11ce Route of administratiol1 K (IU of endotoxin per kilogram
of a gel-dot, visible with the naked eye. The quantitative of body mass)
methods described as methods C, D, E and F were developed lntravenons 5.0
later: they require more instrumentation, but they are easier lntravenous, for radiopharmaceuticals 2.5
to automate for the regular testing oflarge numbers of samples
of the same product. lntrathecal 0.2

Endotoxins may be adsorbed onto the surface of tubes Which dilution of the product is to be used in the test to
or pipettes made from certain plastics or types of glass. obtain maximal assurance that a negative result means that
Interference may appear due to the releas e of substances the endotoxin concentration of the product is less than the
from pi as tic materials. Hence, the materials used should be endotoxin limit and that a positive result means that the
checked; subsequent batches of tubes or pipettes may have lysate detected an endotoxin concentration equal to or greater
a slightly different composition, and therefore the analyst is than the endotoxin limit? This dilution depends 011 the
advised to repeat such tests 011 starting with new batches of endotoxin limit and on the sensitivity of the lysate: it is called
materials. the Maximum Valid Dilution (MVD) and its value may be
The decision to use the test for bacterial endotoxins as a limit calculated using the following expression:
test implies first that a threshold endotoxin concentration endotoxin limit x concentration of test solution
must be defined for the product to be tested, and second that
the objective of the test is to know whether the endotoxin
concentration in the product under examination is below or Concentratiol1 of test solution:
aboye this threshold. The quantitative methods C, D, E and F - mg/mL if the endotoxin limit is specified by mass (IV /mg) ;
make it possible to determine the endotoxin concentration
in the sample under examination, but for compliance with - Units/mL if the endotoxin limit is specified by unit of
the Pharmacopoeia and in routine quality control the final biological activity (IV /Unit);
question is whether or not this concentration exceeds a - mLlmL if the endotoxin limit is specified by volume
defined limit. (IU/mL).

General Notices (1) apply to al! monographs and other texts 573
5.1.10. GuideHnes for using tlle test for bacterial endotoxius EUROPEAN PHARMACOPOEIA 8.0

A the labelled lysate sensitivity in the gel-dot technique analysts may prefer to distil the water more than 3 times.
(IU/mL) or the lowest concentration used in the Whatever method is used, the resultant product must be free
standard curve of the turbidimetric or chromogenic of detectable endotoxins.
techniques.
5. pH OF THE MIXTURE
When the value 01' the maximum valid dilution is not a whole
l1umber, a convenient whole number smaller than the MVD In the test for bacterial endotoxins, optimum gel-dot occurs
may be used for routine purposes (which means preparing a for a mixture at pH 6.0-8.0. However, the addition of the lysate
solutiol1 of the product which is less diluted than the MVD to the sample may result in a lowering of the pH.
indicates). In this case, a negative result indicates that the
endotoxin concentration 01' the product líes below the limit 6. VALIDATION OF THE LYSATE
value. However, when the endotoxin concentration of the 1t is important to follow the manufacturer's instructions for
product in such a test is less than the endotoxin limit but the preparation of the solutions of the lysate.
high enough to make the reaction with the lysate result in a
The positive end-point dilution factors in gel-dot methods A
dot, the test may be positive under these conditions. Hence,
and B are converted to logarithms. The reason is that if the
when a test with this 'convenient' dilution factor is positive,
frequency distribution 01' these logarithmic values is plotted, it
the product should be diluted to the MVD and the test should
usually approaches a normal distribution curve much more
be repeated. In any case of doubt or dispute the MVD must
dosely than the frequency distribution of the dilution factors
be used.
themselves; in fact it is so similar that it is acceptable to use
This stresses the importance of the confirmation of the the normal frequency distribution as a mathematical model
sensitivity of the iysate. and to calculate confidence limits with Student's t-test.
Example
A 50 mg/mL solution of phenytoin sodium (intended for 7. PRELIMINARY TEST FOR INTERFERING FACTORS
intravenous injection) has to be tested. Determine the MVD, Some products cannot be tested directly for the presence of
given the following variables: endotoxins because they are not miscible with the reagents,
M maximum human dose = 15 mg per kilogram 01' they cannot be adjusted to pH 6.0-8.0 01' they inhibit or
body mass; activate gel formation. Therefore a preliminary test is required
to check for the presence of il1terfering factors; when these
e 50 mg/mL; are found the analyst must demonstrate that the pro ce dure to
K 5 IU of endotoxin per kilogram of body mass; remove them has be en effective.

A 0.4 IU of endotoxin per millilitre. The object of the preliminary test is to test the null hypothesis
that the sensitivity of the lysate in the presence of the product
under examination do es not differ significantly from the
MVD = 5 x 50 x ~ = 41.67 sensitivity 01' the lysate in the absence of the product. A simple
15 0.4
criterion is used in methods A and B: the null hypothesis is
For routine tests on this product, it may be expedient to dilute accepted when the sensitivity of the lysate in the presence of
1 mL of the solution to be tested to 20 mL (MVD/2 rounded the product is at least 0.5 times and not more than twice the
to the next lower whole number). However, if this test result is sensitivity of the lysate by itself.
positive the analyst will have to dilute 1 mL to 41.67 mL and
A classical approach wouJd have been to calculate the means
repeat the test. A dilution to 41.67 mL is also necessary when
of the log dilution factor fol' the lysate sensitivity with and
the test is performed to settle a dispute.
without the product and to test the difference between the
3. REFERENCE MATERIAL 2 means witlí. Student's t-test.
Endotoxin standard BRP is intended for use as the reference The test for interfering factors in gel-dot methods A and B
preparation. It has been assayed against the WHO requires the use of a sample of the product in which no
International Standard for Endotoxin and its potency is endotoxins are detectable. This presents a theoretical problem
expressed in International Units of endotoxin per ampoule. when an entirely new product has to be tested. Hence, a
The International Unit of endotoxin is defined as the specific different approach was designed for quantitative methods C,
activity of a defined mass of the International Standard. D, E and F.
For routine purposes, another preparation of endotoxin may
be used, provided it has been assayed against the International 8. REMOVAL OF INTERFERING FACTORS
Standard for Endotoxin or the BRP and its potency is The procedures to remove interfering factors must not
expressed in International Units 01' endotoxin. increase or decrease (for example, by adsorption) the amount
NOTE: 1 International Unit (IU) of endotoxin is equal to of endotoxin in the product under examination. The correct
1 Endotoxin Unit (E. u.). way of checking this is to apply the procedures to a spiked
sample of the product, that is, a sample to which a known
4. WATER FOR BET amount of endotoxin has been added, and then to measure
Testing the absence of endotoxin in this reagent by a technique the recovery of the endotoxin.
derived from the rabbit pyrogen test was rejected for practical Methods C and D. If the nature of the product to be analysed
and theoretical reasons: shows interference which cannot be removed by dassical
- the rabbit test is not sensitive enough to detect endotoxin methods, it may be possible to determine the standard curve in
in water for BET intended for tests on products with a very the same type of product freed from endotoxins by appropriate
low endotoxin limit; treatment or by diJution 01' the product. The endotoxins test is
- the relatively low precision of the rising temperature then carried out by comparison with this standard curve.
response in rabbits would caH for many replications in Ultrafiltration with cellulose triacetate asymmetric membrane
rabbits; filters has been found to be suitable in most cases. The
- the terms 'pyrogens' and 'endotoxins' denote groups of filters should be properly validated, because under sorne
entities that do 110t coincide completely. circumstances cellulose derivatives (~-D-glucans) can cause
The text 2.6.14. Bacterial endotoxins indicates that methods false positive results.
other than triple distillation may be used to prepare water for Polysulfone filters have been found to be unsuitable because
BET. Reverse osmosis has been used with good results; sorne false positive results had been obtained by sorne users.

574 See the information section on general monographs (cover pages)


EUROPEAN PHARMACOPOEIA 8.0 5.l.1O. Guidelines for using the test for bacterial en do toxin s

9. THE PURPOSE OF THE CONTROLS interest; such data indudes details of sample preparation
The purpose of the controlmade up with water for BET and of any procedures necessary to eliminate interfering
and the reference preparation of endotoxin at twice the factors; in addition, any available parallel data for rabbit
concentration of the labelled lysate sensitivity is to verify the pyrogen testing that would contribute to an assurance that
activity of the Iysate at the time and under the conditions of the replacement of a rabbit pyrogen test by the test for
the test. The purpose of the negative control is to verify the bacterial endotoxin is appropriate, must be provided.
absence of a detectable concentration of endotoxin in water Additional requirements are defined in the following sections.
for BET.
12. USE OF A DIFFERENT BACTERlAL ENDOTOXIN
The positive control, which contains the product to be TEST FROM THAT PRESCRIBED IN THE MONOGRAPH
examined at the concentration used in the test, is intended to
show the absence of inhibiting factors at the time and under When a test for bacterial en do toxin s is prescribed in a
the conditions of the test. monograph and none of the 6 methods (A to F) described in
chapter 2.6.14 is specified, then method A, the gel-dot method
10. READING AND INTERPRETATION OF THE RESULTS limit test, has been validated for this product. lf one of the
Minute amounts of endotoxin in the water for BET, or in other methods (B to F) is specified, this is the one which has
any other reagent or material to which the lysate is exposed been validated for this producto
during the test, may escape detection as long as they do not B. VALIDATlON OF ALTERNATIVE METHODS
reach the sensitivity limit of the lysate. However, they may
raise the amount of endotoxin in the solution containing the Replacement of a rabbit pyrogen test by a bacterial endotoxin
product under examination to just aboye the sensitivity limit test, or replacement of a stated or implied method for bacterial
endotoxins by another method, is to be regarded as the use of
and cause a positive reaction.
an alternative method in the replacement of a pharmacopoeial
The risk of this happening may be reduced by testing the test, as described in the General N otices :
water for BET and the other reagents and materials with the
"The test and assays described are the official methods
most sensitive lysate available, or at least one that is more
upon which the standards of the Pharmacopoeia are based.
sensitive than the one used in the test on the producto Even
With the agreement of the competent authority, alternative
then, the risk of such a 'false positive result' cannot be ruled
methods of analysis may be used for control purposes,
out completely. 1t should be realised, however, that in this
provided that the methods used enable an unequivocal
respect the test design is 'fail-safe' in contrast to a test design
decision to be made as to whether compliance with the
permitting a false negative result, which could lead to the
standards of the monographs would be achieved if the
reIease of an unsatisfactory product, thus endangering the
officialmethods were used. In the event of doubt or
patient's health.
dispute, the methods of anaIysis of the Pharmacopoeia are
11. REPLACEMENT OF THE RABBIT PYROGEN TEST BY alone authoritative:'
A TEST FOR BACTERIAL ENDOTOXINS The following pro ce dures are suggested for validating a
Monographs on pharmaceutical products intended for method for bacterial endotoxins other than the one implied or
parenteral administration that may contain toxic amounts indicated in the monograph.
of bacterial endotoxins require either a test for bacterial 13-1. The procedure and the materials and reagents used
en do toxin s or a rabbit pyrogen test. As a general policy: in the method should be validated as described for the test
- in any individual monograph, when a test is required, only concerned.
one test is induded, either that for pyrogens or that for 13-2. The presence of interfering factor s (and, if needed, the
bacterial endotoxins; procedure for removing them) should be tested on samples
in the absence of evidence to the contrary, the test for of at least 3 production batches. It should be borne in mind
bacterial endotoxins is preferred over the test for pyrogens, that methods D and E, using a chromogenic peptide, require
since it is usually considered to provide equal or better reagents that are absent in methods A, B, C and F, and hence
protection to the patient; compliance of methods A, B, C or F with the requirements
for interfering factors cannot be extrapolated to method D or
- before induding a test for bacterial endotoxins in a method E without further testing.
monograph, evidence is required that one of the tests
described in chapter 2.6.14 can be applied satisfactorily to 14. VALIDATION OF THE TEST FOR NEW PRODUCTS
the product in question; The procedures described under 13-1 and 13-2 should
- the necessary information is sought from manufacturers; be applied to all new products intended for parenteral
companies are invited to provide any validation data administration that have to be tested for the presence of
that they have concerning the applicability of the test for bacterial endotoxins according to the requirements of the
bacterial endotoxins to the substances and formulations of Pharmacopoeia.

General Notices (1) apply to all monographs and other texts 575
EUROPEAN PHARMACOPOEIA 8,0

576 See ¡he information section on general monographs (caver pages)


EUROPEAN PHARMACOPOEIA 8.0

5.2. General texts on biological


products
5.2. General texts on biological products ...... 0....•• 0........ 0...... 0579 5.2.6. Evaluation of safety of veterinary vaccines and
5.2.1. Terminology used in monographs on biological immunosera ........ 0.. 000 .. 0........ 0.................•............................... 588
products .............. 0................ 00 .............•.•....................•..•.......•.. 579 5.2.7. Evaluation of efficacy of veterinary vaccines and
5.2.2. Chicken flocks free from specified pathogens for the immunosera ................................................. 0 ..........................591
production and quality control of vaccines ......... o.......... o.... 579 5.2.8. Minimising the risk of transmitting animal spongiform
5.2.3. Cell substrates for the production of vaccines for human encephalopathy agents via human and veterinary medicinal
use ................. o...................... o........ o........................................... 582 products .............................................. 000000000000.000000.00000.00000000000592
5.2.4. Cell cultures for the production of veterinary 502090 Evaluation of safety of each batch of immunosera for
vaccines .. 0.•....•............... 0.....••..•...•.....................• 0.....• 0... 0.... 0..... 585 veterinary useoo 00000000000000000000000000.0000.000000 000000000000000 00000 00000000000 .. 0604
5.2.5. Substances of animal origin for the production of
immunological veterinary medicinal products .................. 587

General Notices (1) apply ta all monographs and other texts 577
EUROPEAN PHARMACOPOEIA 8.0

578 See the information section on general monographs (cover pages)


EUROPEAN PHARMACOPOEIA 8.0 5.2.2. SPF chicken flocks for vacdnes

5.2. GENERAL TEXTS ON Single harvest. Material derived on one or more occasions
from a single production ceH culture inoculated with the same
BIOLOGICAL PRODUCTS working seed 10t or a suspensiol1 derived from the working
seed lot, incubated, and harvested in a single production runo
01/2008:50201 Monovalent pooled harvest. Pooled material containing a
correctea 6.0 single strain or type of micro-organism or antigen and derived
from a number of eggs, ceH culture containers etc. that are
5.2.1. TERMINOLOGY USED IN processed at the same time.
MONOGRAPHS ON BIOLOGICAL Final bulk vaccine. Material that has undergone al! the steps
PRODUCTS of production except fol' the final filling. It consists of one or
more monovalent pooled harvests, from cultures of one or
For sorne items, alternative terms commonly used in more species or types of micro-organism, after clarification,
connection with veterinary vaccines are shown in parenthesis. dilution or addition of any adjuvant or other auxiliary
Seea-lot system. A seed-Iot system is a system according to substance. 1t is treated to ensure its homogeneity and is used
which successive batches of a product are derived from the for filling the containers of one or more finallots (batches).
same master seed 101. For routine production, a working seed Finallot (Batch). A collection of closed, final containers or
lot may be prepared from the master seed 101. The origin and other final closage units that are expected to be homogeneous
the passage history of the master seed lot and the working and equivalent with respect to risk of contamination during
seed lot are recorded. filling or preparation of the final product. The dosage units
Master seea lot. A culture of a micro-organism distributed are filled, or otherwise prepared, from the same final bulk
from a single bulk into containers and processed together in vaccine, freeze-dried together (if applicable) and closed in
a single operation in such a manner as to ensure uniformity one continuous working session. They bear a distinctive
and stability and to prevent contamination. A master seed number or code identifying the finallot (batch). Where a final
lot in liquid form is usually stored at or below - 70 oc. A bulk vaccine is filled and/or freeze-dried on severa! separate
freeze-dried master seed lot is sto red at a temperature known sessions, there results a related set of finallots (batches) that
to ensure stability. are usually identified by the use of a common part in the
distinctive number or code; these related finallots (batches)
Working seed loí. A culture of a micro-organism derived are sometimes refen-ed to as sub-batches, sub-lots or filling
from the master seed 10t and intended for use in production. lots.
Working seed 10ts are distributed into containers and stored
as described aboye for master seed lots. Combined vacdne. A multicomponent preparation
formulated so that different antigens are administered
CeH-bank system (CeH-seed system). A system whereby simultaneously. The different antigenic components are
successive finallots (batches) of a product are manufactured intended to protect against different strains or types of the
by culture in cells derived from the same master ceH bank same organism and/or different organisms. A combined
(master cel! seed). A number of containers from the master vaccine may be supplied by the manufacturer either as a single
cell bank (master ceH seed) are used to prepare a working liquid 01' freeze-dried preparation or as several constituents
ceH bank (working cell seed). The cell-bank system (cell-seed with directions for admixture before use.
system) is validated for the highest passage level achieved
during routine production.
Master (eH bank (Master (eH seed). A culture of cells 07/2010:50202
distributed into containers in a single operation, processed
together and stored in such a manner as to ensure uniformity 5.2.2, CHICKEN FLOCKS FREE
and stability and to prevent contamination. A master ceH bank
(master ceH seed) is usually stored at - 70 oC 01' lower.
FROM SPECIFIED PATHOGENS FOR
Working ceH bank (Working (ell seed). A culture of cells THE PRODUCTION AND QUALITY
derived from the master cell bank (master cell seed) and CONTROL OF VACCINES
intended for use in the preparation of production cell cultures.
The working cell bank (working cel! seed) is distributed into Where specified, chickens, embryos or cel! cultures used for
containers, processed and sto red as described for the master the production or quality control of vaccines are derived from
ceH bank (master cel! seed). eggs produced by chicken flocks free from specified pathogens
(SPF). The SPF status of a flock is ensured by means of the
Primary cen cultures. Cultures of cells obtained by system described below. The list of micro-organisms given is
trypsination of a suitable tissue or organ. The cells are based 011 current knowledge and will be updated as necessary.
essentially identical to those of the tissue of origin and are
A flock is defined as a group of birds sharing a common
no more than 5 in vitro passages from the initial preparation
environment and having their own caretakers who have no
from the animal tissue.
contact with non-SPF flocks. Once a flock is defined, no
CeH Hnes. Cultures of cells that have a high capacity for non-SPF birds are added to it.
multiplication in vitro. In diploid cel!lines, the cells have Each flock is housed so as to minimise the risk of
essentially the same characteristics as those of the tissue of
contamination. The facility in which the flock is housed must
origino In continuous celllines, the cel!s are able to multiply not be sited near to any non-SPF flocks of birds with the
indefinitely in culture and may be obtained from healthy or exception of flocks that are in the process of being established
tumoral tissue. Sorne continuous celllines have oncogenic as SPF flocks and that are housed in facilities and conditions
potential under certain conditions.
appropriate to SPF flocks. The SPF flock is housed within
Production ceU culture. A culture of cells intended for use in an isolator or in a building with filtered air under positive
production; it may be derived from one or more container s pressure. Appropriate measures are taken to prevent entry of
of the working ceH bank (working cel! seed) or it may be a rodents, wild birds, insects and unauthorised personnel.
primary cell culture. Personnel authorised to enter the facility must have no contact
Control censo A quantity of cells set aside, at the time of virus with other birds or with agents potentially capable of infecting
inoculation, as uninfected cel! cultures. The uninfected cells the flock. It is advisable for personnel to shower and change
are incubated under similar conditions to those used for the clothing or to wear protective clothing before entering the
production ceU cultures. controlled facility.

General Notices (1) apply ta all monographs and other texts 579
5.2.2. SPF chicken flocks for vaccines EUROPEAN PHARMACOPOEIA 8.0

Wherever possible, items taken into the facility are sterilised. required. Tests are performed on two 5 per cent samples of
In particular it is recommended that the feed is suitably the flock (minimum 10, maximum 200 birds) taken with an
treated to avoid introduction of undesirable micro-organisms interval of at least 4 weeks between the ages of 12-16 weeks
and that water is at least of potable quality, for example from and 16-20 weeks.
a chlorinated supply. No medication is administered to birds
within the flock that might interfere with detection of any AH samples are collected and tested individuaily. Blood
disease. samples for antibody tests and suitable samples for testing for
leucosis antigen are collected. The test methods to be used are
A permanent record is kept of the general health of the flod: as described under Routine testing of designated SPF flocks.
and any abnormality is investigated. Factors to be monitored Only when all tests have conflrmed the absence of infection
include morbidity, mortality, general physical condition, feed may the new generation be designated as SPE
consumption, daily egg production and egg quality, fertility
and hatchability. Records are maintained for a period of at
least 5 years. Details of any deviation from normal in these ROUTINE TESTING OF DESIGNATED SPF FLOCKS
performance parameters or detection of any infection are General examination and necropsy. Clinical examination
notifled to the users of the eggs as soon as practicable. is carried out at least once per week throughout the life
The tests or combination of tests described below must have of the flock in order to verify that the birds are free from
suitable speciflcity and sensitivity with respect to relevant fowl-pox virus and signs of any other infection. In the event
serotypes of the viruses. Samples for testing are taken at of mortality exceeding 0.2 per cent per week, necropsy is
random. performed on al! available carcasses to verify that there is
no sign of infection. Where appropriate, histopathological
A positive result for chicken anaemia virus (CAV) do es and/or microbiological/virological studies are performed to
not necessarily exclude use of material derived from the confirm diagnosis. Speciflc examination for tuberculosis
flock, but live vaccines for use in birds less than 7 days old lesions is carried out and histological samples from any
shall be produced using material from CAV-negative flocks. suspected lesions are speciflcally stained to verify freedom
Inactivated vaccines for use in birds less than 7 days old may from Mycobacterium avium. Caecal contents of all available
be produced using material from flocks that have not been carcasses are examined microbiologically for the presence of
shown to be free from CAV, provided it has been demonstrated Salmonella spp. using the techniques described below. Where
that the inactivation pracess inactivates CAV. appropriate, caecal samples from up to 5 birds may be pooled.

ESTABLISHMENT OF AN SPF FLOCK Cultural testing for Salmonella spp. Cultural testing for
Salmonella spp. is performed either by testing samples of
A designated SPF flock is derived fram chickens shown to be droppings or cloacal swabs or by testing of drag swabs. Where
free from vertical!y-transmissible agents listed in Table 5.2.2- L droppings or cloacal swabs are tested, a total of 60 samples
This is achieved by testing of 2 generations prior to the within each 4-week period is tested throughout the entire
designated SPF flock A general scheme for the procedure life of the flock Tests may be performed on pools of up to
to be followed in establishing and maintaining an SPF flock 10 samples. Where drag swabs are tested, a minimum of
is shown diagrammatically in Table 5.2.2.-2. In order to 2 drag swabs are tested during each 4-week period throughout
establish a new SPF flock, a series of tests must be conducted the entire life of the flock Detection of Salmonella spp. in
on 3 generations of birds. AH birds in the 1st generation must these samples is performed by pre-enrichment of the samples
be tested at least once before the age of 20 weeks for freedom followed by culture using Salmonella-selective media.
from avian leucosis group-antigen and tested by all enzyme
immunoassay (EIA) or by virus neutralisation (VN) for Tests for avian leucosis antigen. Prior to the commencement
freedom of antibodies to avian leucosis virus subtypes A, B and of laying, cloacal swabs or blood samples (using buffy coat
J. Al! birds must also be tested for freedom from antibodies cultivation) are tested for the presence of group-specific
to the vertically-transmissible agents listed in Table 5.2.2-1. leucosis antigen. A total of 5 per cent (minimum 10, maximum
From the age of 8 weeks the flock is tested for freedom from 200) of the flock is sampled during each 4-week periodo
Salmonella. Clinical examination is carried out on the flock During lay, albumen samples from 5 per cent (minimum 10,
from 8 weeks of age and the birds must not exhibít any signs maximum 200) of the eggs are tested in each 4-week periodo
of infectious disease. The test methods to be used for these Tests are performed by EIA for group-speciflc antigen using
tests are given in the table and further guidance is also given methods that are capable of detecting antigen from subgroups
in the section below on routine testing of designated SPF A, B and J.
flocks. From 20 weeks of age, the flock is tested as described
Test for antibodies to other agents. Tests for antibodies to al!
under Routine testing of designated SPF flocks. AH stages
agel1ts listed in Table 5.2.2.-1 are performed throughout the
of this testing regime are also applied to the subsequent
laying period of the flock In each 4-week period, samples are
2 generations, except the testing of every bird before lay for
taken from 5 per cent (mínimum 10, maximum 200) ofthe
vertically-transmissible agents. AH test results must indicate
flock. It is recommended that 1.25 per cent of the flock is
freedom from pathogens in al! 3 generations for the flock
sampled each week since some test methods for some agents
consisting of the 3rd generation to be designated as SPE
must be conducted on a weekly basis. Table 5.2.2.-1 classifles
SPF embryos derived from another designated SPF flock the agents into those that spread rapidly through the flock and
contained within a separate facility on the same site may be those that spread slowly or may not infect the entire flock For
introduced. From 8 weeks of age, these replacement birds those agents listed as slowly spreading, each sample is tested
are regarded as a flock and are tested in accordance with test individually. For those agents listed as rapidly spreading,
procedures described aboye. at least 20 per cent of the samples collected in each 4-week
period are tested individually or, where serum neutralisation
INITIAL TESTING REQUIREMENTS FOR SUBSEQUENT or ELISA tests are employed, al! of the samples may be tested
GENERATIONS DERIVED FROM A DESIGNATED SPF individually or by preparing pools of 5 samples, collected at
FLOCK the same time.
Where a replacement flock is derived exclusively fram a fully Suitable methods to be used for detection of the agents
established SPF flock the new generation is tested prior to are shown in Table 5.2.2.-1. Subject to agreement by the
being designated as SPE In addition to the tests for Salmonella competent authority, other test methods may be used provided
and monitoring of the general health and performance of they are shown to be at least as sensitive as those indicated
the flock, further specíflc testing from the age of 8 weeks is and of appropriate speciflcity.

580 See the information section on general monographs (cover pages)


EUROPEAN PHARMACOPOEIA 8.0 5.2.2. SPF chicken flocks fOI" vacdlles

Table 5.2.2.-1
Agent Test Vertical Rapid/slow
to be used** transmission spread
Avian adenoviruses, group 1 AGP, EIA yes slow

Avian encephalon1yelitis virus AGP, EIA yes rapid

Avian infectious bronchitis virus HI, EIA no rapid

Avian infeetious laryngotracheitis virus VN, ErA 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 retieuloendotheliosis virus AGP, IS, EIA yes slow

Chicken anaen1ia 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
Salman ella pullorum Agg yes slow

Agg: agglutination HI: haemagglutination inhibition


AGP: agar gel precipitation; the technique is suitable where testing is carried IS: immunostaining
out weekly VN: virus neutralisation
ErA: 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 speeificity.

Table 5.2.2-2. - Schematic description of the establishment and maintenance of SPF flocks
NEW STOCK Establish freedom from vertieaHy-transmissible agents

Test aH 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

20d GENERATION Test aH birds for avian leucosis antigen and antibodies prior to 20 weeks of age

Test for Salmanella spp. and perform general c1inical observation from 8 weeks of age

Carry out routine testing for specified agents from 20 weeks of age

3'" 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

DESIGNATE FLOCK AS SPF IF ALL TESTS ARE SATISFACTORY

3,d GENERATION Carry out routine testing for specified agents from 20 weeks of age

Carry out post-lay testing for verticaHy-transmissible agents

SUBSEQUENT GENERATIONS Test two 5 per eent samples for avian leucosis antigen and for antibodies against
specified agents between 12 and 20 weeks of age
Test for Salmonella spp. and perform general clinieal observation from 8 weeks of age

Carry out routine testing for specified agents from 20 weeks of age

Carry out post-lay testing for verticaHy-transmissible agents

TESTS TO BE CONDUCTED AT THE END OF THE minimum of 5 per cent of the flock (minimum 10, maximum
LAYING PERIOD 200) is retained for at least 4 weeks. Blood samples are
Following the last egg coHection, final testing to confirm collected from every bird in the group during the 4-week
the absence of verticalIy -transmissible agents indicated in period with at least 1.25 per cent of the birds (25 per cent
Table 5.2.2.-1 is performed. After the last egg collection, a of the sample) being bled not earlier than 4 weeks after

General Notices (1) apply to all monographs and other texts 581
5.23. Cell substrates for the production of vaccines far human use EUROPEAN PHARMACOPOEIA 8.0

the final egg col!ection. Serum samples are tested for working cell bank is prepared from one or more containers of
vertically-transmissible agents (as defined by Table 5.2.2.-1) the master cel! bank. The use, identity and inventory control
using the methods indicated. Where sampling is performed of the containers is carefully documented.
on a weekly basis, at least 1.25 per cent of the birds (25 per Media and substances of human or animal origino The
cent of the sample) are tested each week during this periodo compositiol1 of media used for isolation and all subsequent
Alternatively, within 4 weeks of the final egg col!ection blood culture is recorded in detail, and if substances of human or
and/or other suitable sample materials are col!ected from at animal origin are used they must be free from extraneous
least 5 per cent of the flock and tested for the presence of agents (2.6.16) and must comply with the general chapter on
vertically-transmissible agents using validated nucleic acid 5.1.7. Viralsafety.
amplification techniques (2.6.21).
If human albumin is used, it complies with the monograph
ACTION TO BE TAKEN IN THE EVENT OF DETECTION Human albumin solution (0255).
OF A SPECIFIED AGENT If bovine serum is used, it complies with the monograph
Bovine serum (2262).
If evidence is found of contamination of the flock by an
agent listed as slowly spreading in Table 5.2.2.-1, al! materials Trypsin used for the preparation of cel! cultures is examined
derived from the flock during the 4-week period immediately by suitable methods and shown to be sterile and free from
preceding the date on which the positive sample was col!ected mycoplasmas and virus es, notably pestiviruses, circoviruses
are considered unsatisfactory. Similarly, if evidence is found and parvoviruses.
of contamination of the flock by an agent listed as rapidly Cell seed. The data used to assess the suitability of the cel!
spreading in Table 5.2.2.-1, allmaterials derived from the flock seed comprises information, where available, on source,
during the 2-week period immediately preceding the date history and charaeterisation.
on which the positive sample was collected are considered Source of the cell seed. For human celllines, the following
unsatisfactory. Any product manufactured with such information concerning the donor is recorded: ethnic and
materials, and for which the use of SPF materials is required, is geographical origin, age, sex, general physiological condition,
considered unsatisfactory and must be discarded; any quality tissue or organ used, results of any tests for pathogens.
control tests conducted using the materials are invalid.
For animal celllines, the fol!owing informatiol1 is recorded
Producers must notify users of all eggs of the evidence of concerning the source of the cells: species, strain, breeding
contamination as soon as possible following the outbreak. conditions, geographical origin, age, sex, general physiological
Any flock in which an outbreak of any specified agent is condition, tissue or organ used, results of any tests for
confirmed may not be redesignated as an SPF flock. Any pathogens.
progeny derived from that flock during or after the 4-week Cells of neural origin, such as neuroblastoma and P12 cel!
period prior to the last negative sample being collected may lines, may contain substances that concentrate agents of
not be designated as SPF. spongiform encephalopathies and such cells are not used for
vaccine production.
History of the cell seed. The following information is recorded:
the method used to iso late the cell seed, culture methods,
01/2011:50203 any other procedures used to establish the master cel! bank,
notably any that might expose the cells to extral1eous agents.
5.2.3. CELL SUBSTRATES FOR THE Full information may not be available on the ingrediel1ts of
media used in the past for cultivation of cells, for example on
PRODUCTION OF VACCINES FOR the source of substances of animal origin; where justified and
HUMAN USE authorised, cel! banks already established using such media
may be used for vaccine production.
This general chapter deals with diploid celllines and Characterisation of the cell seed. The following properties are
continuous celllines used as cell substrates for the production investigated:
of vaccines for human use; specific issues relating to vaccines
prepared by recombinant DNA technology are covered by the (1) the identity of the cells (for example, isoenzymes, serology,
monograph Products of recombinant DNA technology (0784). nucleic acid fingerprinting);
Testing to be carried out at various stages (ceH seed, master (2) the growth characteristics of the cells and their
cell bank, working cel! bank, cells at or beyond the maximum morphological properties (optical and electro n microscopes);
population doubling level used for production) is indicated in (3) for diploid celllines, karyotype;
Table 5.2.3.-1. General provisions for the use of celllines and (4) for diploid cellhnes, the in vitro life span in terms of
test methods are given below. Where primary cells or cells population doubling leve!.
that have undergone a few passages without constitution of a
cell bank are used for vaccine production, requirements are Cell substrate stability. Suitable viability of the cellline in
given in the individual monograph for the vaccine concerned. the intended storage conditions must be demonstrated. For
a given product to be prepared in the cellline, it is necessary
DipIoid ceH Hnes. A diploid cellline has a high but fmite to demonstrate that consistent production can be obtail1ed
capacity for multiplication in vitro. with cells at passage levels at the beginning and end of the
Continuous celllines. A continuous cellline has the capacity intended span of use.
to multiply indefinitely in vitro; the cel!s often have differences Infectious extraneous agents. Celllines for vaccine
in karyotype compared to the original cells; they may be production shall be free from infectious extraneous agents.
obtained from healthy or tumoral tissue either from mammals Tests for extraneous agents are carried out as shown in
or from insects. Table 5.2.3.-1 using the methods described below.
For injectable vaccines produced in continuous celllines, For eelllines of inseet origin, tests for specific viruses relevant
the purification process is validated to demonstrate removal to the species of origin of the insect cells and for arboviruses
of substrate-cell DNA to a level equivalent to not more than (arthropod - borne virus es) are applied. The panel of viruses
lOng per single human dose, unless otherwise prescribed. tested is chosen according to the current state of scientific
CeH-bank system. Production of vaccines in diploid or knowledge.
continuous celllines is based on a cel!-bank system. The in Celllines that show the presence of retroviruses capable of
vitro age of the cells is counted from the master cel! banlc Each replieation are 110t aceeptable for production of vaccines.

582 See the information section on general monographs (cover pages)


EUROPEAN PHARMACOPOEIA 8.0 5.2.3. Cell substrate§ for the production of vaccines for human use

Table 5.2.3.-1 - Testing of celllines


Test Cell seed Master cell bank Working cell bank Cells at or beyond the
(MCB) (WCB) maximum population
doubling leve! used for
production
1. IDENTITY AND PURITY

Morphology + + + +

Identification: nucleic acid fingerprinting and a relevan!


selection of the following tests: biochemical (e.g.
+ + + +
isoenzyrnes), immunological (e.g. histocompatibility),
cytogenetic markers
+(1)
Karyotype (diploid celllines) + +

Life span (diploid celllines) + +

2. EXTRANEOUS AGENTS

Bacterial and fungal contamination + +

Mycoplasmas + +

Spiroplasmas (insect celllines) + +

Electron microscopy (insect celllines) + (3) + (3)

Tests for extraneous agents in cell cultures +


+(2)
Co-cultivation
+(2)
Tests in animals and eggs

Specific tests for possible contaminants depending on +(2)


the origin of the cells
+(3)
Retroviruses

3. TUMORIGENICITY
+(5)
Tumorigenicity

(1) The diploid character is established for each working cell bank but using cells at or beyond the rnaxirnum population doubling leve! used
for production.
(2) Testing is carried out for each working cell bank, but using cells at or beyond the maxirnurn population doubling leve! used for production.

(3) Testing is carried out for the master cell bank, but using cells at or beyond the maxirnurn population doubling level used for production.

(4) The MRC-S, WI-38 and FRhL-2 celllines are recognised as being non-tumorigenic and they need not be tested. Tests are not carried out on ceH
lines that are known or assumed to be tumorigenic, for example CHO and BHK-21.

(5) Testing is carried out on the cell seed, bul using cells al or beyond the maximurn population doubling leve! used for production.

of 200 cells in metaphase are examined for exact count of


Tumorigenidty. For the preparation of live vaccines, the cell chromosomes and for frequency ofhyperploidy, hypoploidy,
line must not be tumorigenic at any population doubling polyploidy, breaks and structural abnormalities.
level used for vaccine production. Where a tumorigenic cell The MRC-5, the WI-38 and the FRhL-2 celllines are
hne is used for the production of other types of vaccine, the recognised as being diploid and well characterised; where
purification process is vahdated to demonstrate that residual they are not genetically modified, further characterisation is
substrate-cell DNA is reduced to a level equivalent to not not necessary.
more than 10 ng per single human dose of the vaccine,
unless otherwise prescribed, and that substrate-cell protein is TEST METHODS FOR CELL CULTURES
reduced to an acceptable level. Morphology: the morphology of the cells is adequately
A cellline that is known to have tumorigenic potential described and documented.
does not have to be tested further. If a cellline is of Identiflcation. Nucleic acid fingerprint analysis and a relevant
unknown tumorigenic potential, it is either regarded as being selection of the following are used to establish the identity
tumorigenic or it is tested for tumorigenicity using an in of the cells:
vivo test as described below and, optionally, an in vitro test if
additional information is needed. The tests are carried out (1) biochemical characteristics (isoenzyme analysis);
using cells at or beyond the maximum population doubling (2) immunological characteristics (histocompatibility
level that will be used for vaccine production. antigens) ;
The MRC-5, WI-38 and FRhL-2 cellHnes are recognised as (3) cytogenetic markers.
being non-tumorigenic and further testing is not necessary. Contaminating cells. The nucleic acid fingerprint analysis
carried out for identification also serves to demonstrate
Chromosomal characterisation. Diploid celllines shall freedom from contaminating cells.
be shown to be dipIoid. More extensive characterisation of Bacteria! and fungal contamination. The master ceH
a diploid cellline by karyotype analysis is required if the bank and each working cel! bank comply with the test for
removal of intact cells during processing after harvest has not sterility (2.6.1), carried out using for each medium 10 mL of
been validated. Samples from 4 passage levels evenly spaced supernatant fluid from ceH cultures. Carry out the test on
over the life-span of the cellline are examined. A minimum 1 per cent of the containers, with a minimum of 2 containers.

General Notices (1) apply to all monographs and other texts 583
5.2.3. CeH substrates for the production of vacdnes for human use EUROPEAN PHARMACOPOEIA 8.0

Mycoplasmas (2.6.7). The master cell bank and each working Tests in eggs. Using an inoculum of 10 6 viable cells per
cell bank comply with the test for mycoplasmas. Use one or egg, inoculate the cells into the allantoic cavity of ten 9- to
more containers for the test. ll-day-oId SPF embryonated hens' eggs (5.2.2) and into the
Spiroplasmas (insect celllines). The master cel! bank and yolk sac of ten 5- to 6-day-old SPF embryonated hens' eggs.
each working cel! bank of insect cells are demonstrated to be Incubate for not less than 5 days. Test the allantoic fluids for
free of spiroplasmas by a validated method approved by the the presence oí haemagglutinins using mammalian and avian
competent authority. Use one or more containers for the test. red blood cells; carry out the test at 5 ± 3 oC and 20-25 oC
and read the results after 30-60 mino The cells comply with
Electron microscopy (insect ceUHnes). The master cell the test if no evidence of any extraneous agent is found. The
bank is examined by electron microscopy for the presence test is invalid if fewer than 80 per cent of the embryos remain
of adventitious agents. Celllines are maintained at the healthy and survive to the end of the observation periodo
temperature routinely used for production and taken at or
Spedfic tests for possible contaminanís depending on the
beyond the maximum population doubling leve!. In addition,
celllines are maintained at temperatures aboye and below origin of the cdls. Tests for specific pathogens are carried out
using nucleic acid amplification techniques (NAT) (2.6.21)
that routinely used for production and may also be subjected
with or without prior amplification in ceUs. Alternatively,
to other treatments such as exposure to chemical stressors.
suitable serological techniques such as enzyme-linked
The maintenance temperatures and treatments used are
immunosorbent assay, serum neutralisation and anti-body
agreed with the competent authority along with the number
production tests in suitable permissive animals may be used.
of sectioned cells to be examined.
For celllines of rodent origin, use either antibody production
Test for extraneous agents in ceH cultures. The cells comply tests in mice, rats or hamsters or nucleic acid amplification
with the test for haemadsorbing viruses and with the tests in techniques (2.6.21) to detect species-specific viruses. Testing
cel! cultures for other extraneous agents given in chapter 2.6.16 must take account of the origin and culture history of the ceH
under Production ceH culture: control cells. lf the cells are of lineo The tests are designed to detect potential contaminants,
simian origin, they are also inoculated into rabbit kidney cell particularly those that are known to infect latently the species
cultures to test for herpesvirus B (cercopithecid herpesvirus 1). of origin, for example simian virus 40 in rhesus monkeys or
Co-cultivation. For mammalian and avian celllines, Flock house virus in insect cells.
co-cultivate intact and/or disrupted cells separately with other Tests for tumorigenidty in vivo. The test consists in
ceH systems including human cells and simian cells. For insect establishing a comparisol1 between the continuous cellline
celllines, extracts of disrupted cells are incubated with other and a suitable positive control (for exampIe, HeLa or Hep2
cel! systems, including human, simian, and at least 1 cellline cells).
that is different from that used in production, is permissible to Animal systems that have been shown to be suitable for this
insect virus es and allows detection ofhuman arboviruses (for test include:
example BHK-21). Carry out examinations to detect possible
(1) athymic mice (Nu/Nu genotype);
morphological changes. Carry out tests on the cell culture
fluids to detect haemagglutinating viruses, or 011 cells to (2) newborn mice, rats or hamsters that have been treated
detect haemadsorbing viruses. The test for haemagglutinating with antithymocyte serum or globulin;
viruses does not apply for arboviruses to be detected in insect (3) thymectomised and irradiated mice that have been
ceUs. The cells comply with the test if no evidence of any reconstituted (T-, W) with bone marrow from healthy mice.
extraneous agent is found. Whichever animal system is selected, the cellline and the
Retroviruses. Examine for the presence of retroviruses using: reference cells are injected into separate groups of 10 animal s
each. In both cases, the inoculum for each animal is 10 7 cells
(1) product-enhanced reverse transcriptase (PERT) assay suspended in a volume of 0.2 mL, and the injection may be by
(2.6.21) carried out for cell bank supernatants using cells at either the intramuscular 01' the subcutaneous route. Newborn
or beyond the maximum population doubling leve! that will animals are treated with 0.1 I11L of antithymocyte serum or
be used for production; globulin on days 0, 2, 7 and 14 after birth. A potent serum or
(2) transmission electron microscopy. globulin is one that suppresses the immune mechanisms of
growing animals to the extent that the subsequent inoculum
lf test (1) and/or test (2) gives a positive result, test (3) is
of 10 7 positive reference cells regularly produces tumours
carried out:
and metastases. Severely affected animals showing evident,
(3) infectivity assays carried out on human cells with an progressively growing tumours are euthanised before the end
endpoint PERT assay on the supernatant. of the test to avoid unnecessary suffering.
Since the sensitivity of PERT assays is very high, interpretation At the end of the observation period al! animals, including the
of a positive signal may be equivocal and a decision on the reference group(s), are euthanised and examined for gross and
acceptability of a cell substrate is based on aH available data. microscopic evidence of the proliferation of inoculated cells at
the site of injectiol1 and in other organs (for example, lymph
Tests in animals. Inject intramuscularly (or, for suckling nodes, lungs, kidneys and liver).
mice, subcutaneously) into each of the following groups of
animals 10 viable cells divided equally between the animals
7 In all test systems, the animals are observed and palpated
in each group: at regular intervals for the formation of nodules at the
sites of injection. Any nodules formed are measured in
(1) 2 litters of suckling mice less than 24 h old, comprising not 2 perpendicular directions, the measurements being recorded
fewer than 10 animals; regularly to determine whether there is progressive growth of
(2) 10 adult mice. the nodule. Animals showing nodules that begin to regress
during the period of observation are euthanised before the
Inject intracerebraHy into each of 10 adult mice 10 6 nodules are no longer palpable, and processed for histological
viable cells to detect the possible presence of lymphocytic examination. Animals with progressively growing nodules
choriomeningitis virus. are observed for 1-2 weeks. Among those without nodule
Observe the animals for at least 4 weeks. Investigate animals formation, half are observed for 3 weeks and half for 12 weeks
that become sick or show any abnormality to establish the before they are euthanised and processed for histological
cause oí illness. The cells comply with the test if no evidence examination. A necropsy is performed 011 each animal and
of any extraneous agent is found. The test is invalid if fewer includes examination for gross evidence oí tumour formation
than 80 per cent of the animal s in each group remain healthy at the site of injection and in other organs such as lymph
and survive to the end of the observation periodo no des, lungs, brain, spleen, kidneys and liver.

584 See the information section on general monographs (caver pages)


EUROPEAN PHARMACOPOEIA 8.0 5.2.4. CeU cultures for [he production of veterinary vacdnes

AH tumour-like lesions and the site of injection are examined Table 5.2.4.-1. - Cell culture stage at which tests
histologically. In addition, since sorne celllines may give rise are carried out
to metastases without evidence of local tumour growth, any Master cel! Working Cell from working
detectable regionallymph nodes and the lungs of al! animals seee! cell ,eee! cell seed al highest
are examined histologically. passage leve!
General microscopy + + +
The test is invalid iffewer than 9 of the 10 animals injected
with the positive reference cells show progressively growing Bacteria and fungi + +
tumours. +
Mycoplasmas
Tests for tumorigenidty in vitro. The following test systems
Viruses + +
may be used:
Identification of species +
(1) colony formation in soft agar gels;
Karyotype + +
(2) production of invasive cel! growth following inoculation
into organ cultures; Tumorigenicity +
(3) study of transformation activity using, for example, the
3T3 assay system for active oncogenes. Characteristics of culture. The appearance of cel!
monolayers, before and after histological staining, is described.
Information, if possible numerical data, is provided especially
on the speed and rate of grawth. Similarly, the presence or
absence of contact inhibition, polynucleated cells and any
other cellular abnormalities are specified.
0112008:50204
Karyotype. A chromosomal examination is made of not fewer
than fifty cells undergoing mitosis in the master cell seed
5.2.4. CELL CULTURES FOR THE and at a passage level at least as high as that to be used in
PRODUCTION OF VETERINARY production. Any chromosomal marker present in the master
ceU seed must also be found in the high passage cells and the
VACCINES modal number of chromosomes in these ceUs must 110t be
more than 15 per cent higher than of cells of the master cel!
Cell cultures for the production of vaccines for veterinary seed. The karyotypes must be identical. If the modal number
use comply with the requirements of this section. It may also exceeds the level stated, if the chromosomal markers are not
be necessary that ceH cultures used for testing of vaccines found in the working cell seed at the highest level used for
for veterinary use also comply with sorne or al! of these production or if the karyotype differs, the cellline shall not be
requirements. used for manufacture.
Por most mammalian viruses, propagation in celllines is Identification of the spedes. It shall be shown, by one
possible and the use of primary cells is then not acceptable. validated method, that the master ceH seed and the ceHs from
Permanently infected cells used for production of veterinary the working ceH seed at the highest passage leve! used for
vaccines comply with the appropriate requirements described production come from the species of origin specified. When a
below. The ceUs shall be shown to be infected only with the fluorescence test is carried out and the corresponding serum
agent stated. to the species of origin of cells is used and shows that all the
tested cells are fluorescent, it is not necessary to carry out
other tests with reagents able to detect contamination by cells
CELL UNES of other species.
Celllines are normally handled according to a cell-seed Bacterial and fnngal contamination. The cells comply with
system. Each master cell seed is assigned a specific code for the test for sterility (2.6.1). The sample of cells to be examined
identification purposes. The master cell seed is stored in consists of not less than the number of cells in a monolayer
aliquots at - 70 oC or lower. Production of vaccine is not with an area of 70 cm 2 or, for cells grown in suspension,
normally undertaken on cells more than twenty passages an approximately equivalent number of ceUs. The cells are
from the master ceH seed. Where suspension cultures are maintained in culture for at least 15 days without antibiotics
used, an increase in cel! numbers equivalent to approximately before carrying out the test.
three population doublings is considered equivalent to one
passage. If ceHs beyond twenty passage levels are to be used Mycoplasmas (2.6.7). The cells comply with the test for
for production, it shal! be demonstrated, by validation or mycoplasmas. The cells are maintained in culture for at least
further testing, that the production cel! cultures are essentially 15 days without antibiotics before carrying out the test.
similar to the master ceH seed with regard to their biological Absence of contaminating viruses. The ceUs must not be
characteristics and purity and that the use of such cells has no contaminated by virus es; suitably sensitive tests, including
deleterious effect on vaccine production. those prescribed below, are carried out.
The history of the cellline shall be known and recorded in The monoJayers tested shall have an area of at least 70 cm 2, and
detail (for example, origin, number of passages and media shall be prepared and maintained using medium and additives,
used for multiplication, storage conditions). and grown under similar conditions to those used for the
preparation of the vaccine. The monolayers are maintained in
The method of storing and using the ceUs, including details culture for a total of at least 28 days. Sub cultures are made at
of how it is ensured that the maximum number of passages 7-day intervals, unless the cells do not survive for this length
permitted is 110t exceeded during product manufacture, are of time, when the sub cultures are made on the latest day
recorded. A sufficient quantity of the master ceH seed and possible. Sufficient cells, in suitable containers, are produced
each working cel! seed are kept for analytical purposes. for the final sub culture to carry out the tests specified below.
The tests described below are carried out (as prescribed in The monolayers are examined regularly throughout the
Table 5.2.4.-1) on a culture of the master cel! seed and the incubation period for the possible presence of cytopathic
working cel! seed or 011 ceH cultures from the working cel! effects and at the end of the observation period for cytopathic
seed at the highest passage level used for production and effects, haemadsorbent viruses and specific virus es by
derived from a homogeneous sample demonstrated to be immuno-fluorescence and other suitable tests as indicated
representative. below.

General Notices (1) apply ta all monographs and other texts 585
5.2.4. Cell cultures fur the production ofveterinary vacdnes EUROPEAN PHARMACOPOEIA 8.0

Deíection of cytopathic viruses. Two monolayers of at least Wherever possible, particularly for mammalian ceUs, a
6 cm 2 each are stained with an appropriate cytological stain. seed-lot system is used with, for example, a master ceH seed
The entire area of each stained monolayer is examined for formed after less than five passages, the working cel! seed
any inclusion bodies, abnormal numbers of giant cells or any being no more than five passages from the initial preparation
other lesion indicative of a cellular abnormality which might of the cel! suspension from the animal tissues.
be attributable to a contaminant.
Each master ceH seed, working cel! seed and cells of the
Detection of haemadsorbent viruses. Monolayers totalling highest passage of primary cells are checked in accordance
at least 70 cm 2 are washed several times with an appropriate with Table 5.2.4.-2 and the procedure described below. The
buffer and a sufficient volume of a suspension of suitable red sample tested shal! cover al! the sources of cells used for the
blood cells added to cover the surface of the monolayer evenly. manufacture of the batch. N o batches of vaccine manufactured
After different incubation times cells are examined for the using the cells may be released if any one of the checks
presence of haemadsorption. performed produces unsatisfactory results.
Detection of specified viruses. Tests are carried out for Table 5.2.4.-2. - Cell culture stage at which tests
freedom from contaminants specific for the species of origin are carried out
of the cellline and for the species for which the product is
intended. Sufficient cells on suitable supports are prepared Master Working Highest passage
to carry out tests for the agents specified. Suitable positive cel! seed cell seed leve!
controls are included in each test. The cells are subjected General D1icroscopy + + +
to suitable tests, for example using fluorescein-conjugated Bacteria and fungi + +
antibodies or similar reagents.
Mycoplasmas + +
Tests in other ceH cultures. Monolayers totalling at least
140 cm 2 are required. The cells are frozen and thawed at least Viruses + +
three times and then centrifuged to remove cellular debris. Identification of species +
Inoculate aliquots onto the following cells at any time up to
70 per cent confluency: Characteristics of cultures. The appearance of ceH
- primary cells of the source species; monolayers, before and after histological staining, is described.
Information, if possible numerical data, is recorded, especially
- cells sensitive to virus es pathogenic for the species for on the speed and rate of growth. Similarly, the presence or
which the vaccine is intended; absence of contact inhibition, polynucleated cells and any
- cells sensitive to pestiviruses. other ceHular abnormalities are specified.
The inoculated cells are maintained in culture for at least Identification of spedes. It shall be demonstrated by one
7 days, after which freeze-thawed extracts are prepared as validated test that the master cel! seed comes from the
aboye and inoculated onto sufficient fresh cultures of the same specified species of origino
cel! types to allow for the testing as described below. The cells When a fluorescence test is carried out and the corresponding
are incubated for at least a further 7 days. The cultures are serum to the species of origin of cells is used and shows that
examined regularly for the presence of any cytopathic changes al! the tested cells are fluorescent, it is not necessary to carry
indicative of living organisms. out other tests with reagents able to deteet contaminatiol1 by
At the end of this period of 14 days, the inoculated cells are cells of other species.
subjected to the following checks: Bacterial and fungal sterility. The cells comply with the
- freedom from cytopathic and haemadsorbent organisms, test for sterility (2.6.1). The sample of cells to be examined
using the methods specified in the relevant paragraphs cOl1sists of not less than the number of cells in a monolayer
aboye, with an area of 70 cm 2 or for cells grown in suspension an
approximately equivalent number of cells. The cells are
- absence of pestiviruses and other specific contaminants maintained in culture for at least 15 days without antibiotics
by immunofluorescence or other validated methods as before carrying out the test.
indicated in the paragraph aboye on Detection of Specified
Mycoplasmas (2.6.7). The cells comply with the test for
Viruses.
mycoplasmas. The cells are maintained in culture for at least
Tumorigenidty. The risk of a cellline for the target species 15 days without antibiotics before carrying out the test.
must be evaluated and, if necessary, tests are carried out.
Absence of contaminating viruses. The cells must not be
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 be at least 70 cm 2 , and shall be
acceptable for the manufacture of vaccines since celllines can prepared and maintained in culture using the same medium
be used. If there is no alternative to the use of primary cells, and additives, and under similar conditions to those used for
the cells are obtained from a herd or flock free from specified the preparation of the vaccine.
pathogens, with complete protection from introduction of
diseases (for example, disease barriers, filters on air inlets, The monolayers are maintained in culture for a total of at
suitable quarantine before introduction of animals). Chicken least 28 days 01' for the longest period possible if culture for
28 days is impossible. Sub cultures are made at 7-day intervals,
flocks comply with the requirements prescribed in general
chapter 5.2.2. Chicken Flocks Free from Specified Pathogens for unless the cells do 110t survive for this length of time when the
the Production and Quality Control of Vaccines. Por all other
sub cultures are made on the latest day possible. Sufficient cells,
species, the herd or flock is shown to be free from relevant in suitable containers are produced for the final sub culture to
specified pathogens. Al! the breeding stock in the herd or carry out the tests specified below.
flock intended to be used to produce primary cells for vaccine The monolayers are examined regularly throughout the
manufacture is subject to a suitable monitoring procedure incubation period for the possible presence of cytopathic
including regular serological checks carried out at least twice effects and at the end of the observation period for cytopathic
ayear and two supplementary serological examinations effects, haemadsorbent viruses and specific viruses by
performed in 15 per cent of the breeding stock in the herd immunofluorescence and other suitable tests as indicated
between the two checks mentioned aboye. below.

586 See the information section on general monographs (cover pages)


EUROPEAN PHARMACOPOEIA 8.0 5.2.5. Substances (Jf animal origin for immunological veterinary products

Deíection of cytopathic viruses. Two monolayers of at least 2. GENERAL PRINCIPLES AND REQUIREMENTS
6 cm 2 each are stained with an appropriate cytological stain. Substances of animal origin comply with the requirements of the
Examine the entire area of each stained monolayer for any European Pharmacopoeia (where a relevant monograph exists).
inclusion bodies, abnormal numbers of giant ceUs or any
other lesion indicative of a ceUular abnormality that might be Restrictions are placed 011 the use of substances of animal
attributable to a contaminant. origin because of safety C011cerns associated with pathogens
that may be present in them and epidemiological and/or
Detection of haemadsorbent viruses. Monolayers totalling regulatory concerns associated with the presence of particular
at least 70 cm 2 are washed several times with a suitable buffer antigens (either live or inactivated).
solution and a sufficient volume of a suspension of suitable General principIes:
red blood cells added to cover the surface of the monolayer
evenly. After different incubation times, examine ceUs for the - it is recommended to minimise, wherever practicable, the
presence of haemadsorption. use of substances of animal origin;
- unless otherwise justified, the use of substances of animal
Detection of spedfied viruses. Tests are be carried out for
origin as constituents in the formulation of medicinal
freedom of contaminants specific for the species of origin of
products is not acceptable except where such substances
the cells and for the species for which the product is intended.
are subject to a treatment validated for the inactivation of
Sufficient cells on suitable supports are prepared to carry out live extraneous agents.
tests for the agents specified. Suitable positive controls are
General requirements:
included in each test. The ceUs are subjected to suitable tests
using fluorescein-conjugated antibodies or similar reagents. - any batch of substance (after inactivation and/or processing,
if relevant) found to contain or suspected of containing any
Tests in oíher ceH cultures. Monolayers totalling at least living extraneous agent shall be discarded or used only in
140 cm 2 are required. The cells are frozen and thawed at least exceptional and justified circumstances; to be accepted for
three times and then centrifuged to remove cellular debris. use, further processing must be applied that will ensure
Aliquots are inoculated onto the following ceUs at any time elimination and/or inactivation of the extraneous agent,
up to 70 per cent confluency: and it shall then be demonstrated that the elimination
- primary cells of the source species; and/or inactivation has been satisfactory;
cells sensitive to viruses pathogenic for the species for - any batch of substance that, as concluded from the risk
which the vaccine is intended; assessment, may induce an unacceptable detectable
- cells sensitive to pestiviruses. immune response in the target species as a consequence
of contamination with inactivated extraneous agents,
The inoculated cells are maintained in culture for at least must 110t be used for the manufacture of that particular
7 days, after which freeze-thawed extracts are prepared as immunological veterinary medicinal product.
aboye, and inoculated onto sufficient fresh cultures of the
same cel! types to allow for the testing as described below. The 3. RISK MANAGEMENT
cells are incubated for at least a further 7 days. AH cultures are No single measure or combination of measures can guarantee
regularly examined for the presence of any cytopathic changes the safety of the use of substances of animal origin, but they
indicative of living organisms. can reduce the risk from such use. It is therefore necessary
At the end of this period of 14 days, the inoculated cells are for the manufacturer of immunological veterinary medicinal
subjected to the following checks: products to take account of this when choosing a substance
- freedom from cytopathic and haemadsorbent organisms is of animal origin to use in manufacture, and to conduct a risk
demonstrated using the methods specifled in the relevant assessment, taking into account the origin of the substance
paragraphs above; and the manufacturing steps applied to it.
- relevant substrates are tested for the absence of pestiviruses In addition, risk management procedures must be applied.
and other specific contaminants by immunofluorescence Any residual risk must be evaluated in relation to the
or other validated methods as indicated in the paragraph potential benefits derived from the use of the substance for
aboye 011 Detection of Specified Viruses. the manufacture of the immunological veterinary medicinal
producto
3-1. RISK ASSESSMENT
The risk assessment must take account of the animal diseases
07/2009:50205 occurring in the country of origin of the animals used as
a source of the substance, the potential infectious diseases
occurring in the source species and the likely infectivity in
5.2.5, SUBSTANCES OF ANIMAL the source organ or tissue. Prom this information, as part of
ORIGIN FOR THE PRODUCTION OF the risk assessment, a list can be prepared of the extraneous
IMMUNOLOGICAL VETERINARY agents that may be present in the substance.
The risk of contamination of the substance and the
MEDICINAL PRODUCTS resultant immunological veterinary medicinal product
with living extraneous agents needs to be assessed. The
1. SCOPE risk of contamination of the substance and the resultant
Substances of animal origin (for example serum, trypsin and immunological veterinary medicinal product with inactivated
serum albumin) may be used during the manufacture of extraneous agents may also need to be taken into account.
immunological veterinary medicinal products. This would be the case if, for example, the contaminant was
The requirements set out in this chapter apply to substances of one from which a European country is officially free and/or is
animal origin produced on a batch basis, for use at al! stages the subject of a specific disease control program in a European
of manufacture, for example in culture media or as added country and where the presence of the inactivated agent could
constituents of products during blending. These requirements lead to the stimulation of a detectable immune response in
are not intended for the control of seed material s or substrates recipient animals.
of animal origin that are covered by requirements in other As part of the risk assessment, the presence in the substance
pharmacopoeial texts such as the monograph Vaccines for of antibodies that can interfere with the detectiol1 and/or
veterinary use (0062) and chapter 5.2.4. Cell cultures for the inactivation of living extraneous agents must also be taken
production of veterinary vaccines. into account.

General Notices (1) apply to all monographs and other texts 587
5.2.6. Evaluation oí safety of veterinary vaccines and immunosera EUROPEAN PHARMACOPOEIA 8.0

The risk assessment may need to be repeated and the risk For inactivated immunological veterinary medicinal products,
management steps described below re-evaluated and revised the method used for inactivation of the active ingredient may
in order to take account of changes: also be validated for inactivation of possible contaminants
from substances of animal origin used in the manufacture of
- in the incidence of diseases occurring in the country or
this active ingredient.
countries of origin of animals used as the source for the
substance, including emerging diseases (new pathogens); 4-4. TESTS
Depending on the outcome of the risk assessment and the
- in the inciden ce of diseases and of disease control measures
validation data available for any procedure applied, tests for
applied in the European countries in which immunological
extraneous agents may be conducted on each batch before
veterinary medicinal products manufactured with the
and/or after the application of an inactivation/processing
substance are used.
step. For examination of the substance for freedom from
3-2. RISK CONTROL extraneous agents, any solids are dissolved or suspended in a
For each of the potential extraneous agents identified by the suitable medium to provide a suitable preparation for testing.
risk assessment, and taking into account the proposed use of A sufficient quantity of the preparation is tested to give a
the substance, the risk must be controlled by the use of one or suitably sensitive test, as established in the validation studies.
a combination of the followings measures: As well as tests for living extraneous agents, tests may need
to be conducted for the presence of inactivated extraneous
- placing restrictions on the source of the material and agents, depending 011 the risks identified.
auditing this;
Freedom fmm living extraneous viruses. A sample from
- using validated inactivation pro ce dures ; each batch of the substance is tested for extraneous viruses by
- demonstrating the ability of a production step to remove or general and specific tests. These tests are validated with respect
inactivate extraneous agents; to sensitivity and specificity for detectiol1 of a suitable range
of potential extraneous viruses. Suitably sensitive cel! cultures
- testing for extraneous agents. are used for the tests for extraneous virus es, including primary
cells from the same species as the substance to be examined.
4. CONTROL MEASURES General test. The inoculated cel! cultures are observed
4-1. SOURCE regularly for 21 days for cytopathic effects. At the end of each
All substances of animal origin used in the manufacture 7 -day period, a proportion of the original cultures is fixed,
(including blending) of immunological veterinary medicinal stained and examined for cytopathic effects, and a proportion
products must be from a known and documented source is tested for haemadsorbing agents.
(including species of origin and country of origin of source Specific tests. A proportion of the cells available at the end
animals and tissues). of the general test is tested for specific viruses. The specific
viruses to be tested for are potential extraneous viruses that
4-2. PREPARATION
are identified through the risk assessment and that would
Substances of animal origin are prepared from a homogeneous not be detected by the general test. A test for pestiviruses is
bulk designated with a batch number. A batch may contain conducted if the source species is susceptible to these.
substances derived from as many animals as desired but once
defined and given a batch number, the batch is not added to Bacteria and fungi. Before use, substances are tested for
or contaminated in any way. sterility (2.6.1), or sterilised to inactivate any bacterial or
The production method used to prepare the substance of fungal contaminants.
animal origin from the raw material may contribute to Mycoplasma. Before use, substal1ces are tested for freedom
the removal and/or inactivation of extraneous agents (see from mycoplasma (2.6.7), or sterilised to inactivate any
section 4- 3). mycoplasmal contaminants.
4-3. INACTIVATION AND/OR OTHER PROCESSING STEPS
POR REMO VAL OP EXTRANEOUS AGENTS 04/2013:50206
The inactivation procedure and/or other processing steps
chosen shall have been validated and shown to be capable of 5.2.6. EVALUATION OF SAFETY
reducing the titre of potential extraneous agents described OF VETERINARY VACCINES AND
below in the substance concerned by a factor of at least 10 6 •
If this reduction in titre cannot be shown experimentally, a IMMUNOSERA
maximum pre-treatment titre of the extraneous agent must be The term 'producf means either a vaccine or an immunoserum
set, taking into account the reduction in titre afforded by the throughout the texto
inactivation/processing step and including a safety margin During development, safety tests are carried out in the target
factor of 100; each batch of substance must be tested to
species to show the risks from use of the product.
determine the pre-treatment starting titre and confirm it is no
greater than the specified limit, unless proper risk assessment,Immune status for tests on vaccines. The immune status of
based on valid and suitable data, shows that titres will always animals to be used for the safety test is specified in the specific
be at least lOO-fold below the titre that can effectively be monograph. For most monographs, 1 of the 3 following
inactivated. categories is specified:
1) the animals must be free from antibodies against the
The validation of the procedure(s) is conducted with a suitable virus/bacterium/toxin etc. contained in the vaccine;
representative range of viruses covering different types
and sizes (enveloped and non-enveloped, DNA and RNA, 2) the animals are preferably free from antibodies against
single- and double-stranded, temperature- and pH-resistant), the virus/bacterium/toxin etc. contained in the vaccine, but
including test viruses with different degrees of resistance, animal s with a low level of antibody may be used as long as
taking into account the type of procedure(s) to be applied the animals have 110t been vaccinated and the administration
and the viruses that may be present in the material. The of the vaccine do es not cause an anamnestic response;
evidence for the efficacy of the procedure may take the form 3) the animals must not have been vaccinated against the
of references to published literature and/or experimental data disease that the vaccine is intended to prevent.
generated by the manufacturer, but must be relevant to the As a general rule, category 1 is specified for live vaccines.
conditions that will be present during the production and For other vaccines, category 2 is usually specified, but where
inactivation/processing of the substance. most animals available for use in tests would comply with

588 See the information section on general monographs (ca ver pages)
EUROPEAN PHARMACOPOEIA 8.0 5.2.6. Evaluation of safety oí veterinary vacdnes and irnrnunosera

category 1, this may be specified for inactivated vaccines also. Unless otherwise prescribed in a specific monograph or, in the
Category 3 is specified for sorne inactivated vaccines where absence of a specific monograph, unless otherwise justified
determination of antibodies prior to testing is unnecessary or and authorised, the vaccine complies with the test if no animal
impractical. For poultry vaccines, as a general rule the use of shows abnormallocal or systemic reactions or signs of disease,
specified-pathogen-free (SPF) birds is specified. or dies from causes attributable to the vaccine.
For avian vaccines, the safety test is generally carried out 1-2. SAFETY OF 1 ADMINISTRATION OF AN OVERDOSE
using SPF chickens (5.2.2), except that for vaccines not Overdose testing is required only for live vaccines. An
recommended for use in chickens it is carried out using birds overdose of the product is administered by each recommended
of one of the species for which the vaccine is recommended, route of administration to animals of the categories of the
the birds being free from antibodies against the disease agent target species that are expected to be the most sensitive, such
for which the vaccine is intended to provide protection. as animals of the youngest age. If multiple routes and methods
Vaccines. In laboratory tests, 'dose' means that quantity of of administration are specified for the product con cerned,
the product to be recommended for use and containing the administration by all routes is recommended. lf 1 route of
maximum titre or potency like!y to be contained in production administration has been shown to cause the most severe
batches. Live vaccines are prepared only from strains of effects, this single route may be selected as the only one for use
organisms that have been shown to be safe. For live vaccines, in the study. The overdose normally consists of 10 doses of a
use a batch or batches of vaccine containing virus/bacteria live vaccine. For freeze-dried live vaccines, the 10 doses shall
at the least attenuated passage leve! that will be present in a be reconstituted in a suitable volume of diluent for the test. For
batch of vaccine. vaccines intended for use in mammals, in general 8 animals
For combined vaccines, the safety shall be demonstrated; for per group are used unless otherwise justified or specified in a
live components of combined vaccines, compliance with the specific monograph. For vaccines intended for use in fish, in
special requirements for live vaccines stated be!ow shal! be general 50 fish per group are used unless otherwise justified or
demonstrated separately for each vaccine strain. specified in a specific monograph. For vaccines intended for
For inactivated vaccines, safety tests carried out on the use in birds older than 3 weeks, in general 8 birds per group
combined vaccine may be regarded as sufficient to demonstrate are used unless otherwise justified or specified in a specific
the safety of the individual components. monograph. For vaccines intended for use in birds younger
than 3 weeks, in general 10 birds per group are used unless
Immunasera. In the tests, 'dose' means the maximum quantity
otherwise justified or specified in a specific monograph. The
of the product to be recommended for use and containing
animals are observed and examined at least daily for signs
the maximum potency and maximum total protein like!y
of local and systemic reactions. Other objeetive criteria are
to be contained in production batches. In addition, if
recorded, su eh as body temperature (for mammals) and
appropriate, the dos e tested also contains maximum quantities
performance measurements. The animals are observed and
of immunoglobulin or gammaglobulin.
examined for at least 14 days after administration.
The tests described below, modified or supplemented by tests Unless otherwise prescribed in a specific monograph or, in the
described in the Production section of a monograph, may be absence of a specific monograph, unless otherwise justified
carried out as part of the tests necessary during development and authorised, the vaccine complies with the test if no animal
to demonstrate the safety of the product. shows abnormallocal or systemic reactions or signs of disease,
1. LABORATORY TESTS or die s from causes attributable to the vaccine.
1-1. SAFETY OF THE ADMINISTRATION OF 1 DOSE 1-3. SAFETY OF THE REPEATED ADMINISTRATION OF
For each of the recommended routes of administration, 1 DOSE
administer 1 dose of product to animals of each species and Repeated administration of 1 dose may be required to
category for which use of the product is to be recommended. reveal any adverse effects induced by such administration.
This must in elude animals of the youngest recommended age These tests are particularly important where the produet,
and pregnant animals, if appropriate. notably an immunoserum, may be administered on several
For vaccines intended for use in mammals, in general occasions over a relatively short period of time. These tests
8 animal s per group are used unless otherwise justified or are carried out on the most sensitive categories of the target
specified in a specific monograph. species, using each recommended route of administration. If
For fish vaccines administered by immersion, bathe the fish multiple routes and methods of administration are specified
for twice the recommended time using a bath at twice the for the product concerned, administration by al! routes is
recommended. If 1 route of administration has been shown
recommended concentration.
to cause the most severe effects, this single route may be
For vaccines intended for use in fish, in general 50 fish per selected as the only one for use in the study. The number of
group are used unless otherwise justified or specified in a administrations must be not less than the maximum number
specific monograph. reeommended; for vaccines, this shall take account of the
For vaccines intended for use in birds older than 3 weeks, in number of administrations for primary vaccination and the
general 8 birds per group are used unless otherwise justified 1't re-vaccination; for immunosera, it shall take account of
or specified in a specific monograph. For vaccines intended the number of administrations required for treatment. The
for use in birds younger than 3 weeks, in general 10 birds interval between administrations shall be suitable (e.g. period
per group are used unless otherwise justified or specified in of risk or required for treatment) and appropriate to the
a specific monograph. recommendations of use. Although, for convenience, as far
The animals are observed and examined at least daily for signs as vaccines are con cerned, a shorter interval may be used in
of abnormallocal and systemic reactions. Where appropriate, the study than that recommended in the field, an interval
these studies shall indude detailed post-mortem macroscopic of at least 14 days must be allowed between administrations
and microscopic examinations of the injection site. Other for the development of any hypersensitivity reaction. For
objective criteria are recorded, such as body temperature immunosera, however, administration shall follow the
(for mammals) and performance measurements. The body recommended schedule. For vaccines intended for use in
temperatures are recorded on at least the day before and at the mammals, in general 8 animals per group are used unless
time of administration of the product, 4 h later and on the otherwise justified or specified in a specific monograph. For
following 4 days. The animals are observed and examined at vaccines intended for use in fish, in general 50 fish per group
least daily until reactions may no longer be expected but, in are used unless otherwise justified or specified in a specific
al! cases, the observation and examination period extends at monograph. For vaccines intended for use in birds older than
least until 14 days after administration. 3 weeks, in general 8 birds per group are used unless otherwise

General Natices (1) apply ta all mónagraphs and other texts 589
5.2.6. Evaluation of safety of veterinary vaccines and immunosera EUROPEAN PHARMACOPOEIA 8.0

justified or specified in a specific monograph. For vaccines 1-7. ADVERSE EFFECTS FROM INTERACTIONS
intended for use in birds younger than 3 weeks, in general Studies are undertaken to show a lack of adverse effect on
10 birds per group are used unless otherwise justified or the safety of the product when simultaneous administration
specified in a specific monograph. The animals are observed is recommended or where administration of the product
and examined at least daily for at least 14 days after the last is recommended as part of a schedule of administration of
administration for signs of systemic and local reactions. Other products within a short period of time.
objective criteria are recorded, such as body temperature and
1-8. SPECIAL REQUIREMENTS FOR LIVE VACCINES
performance measurements.
The following laboratory tests must also be carried out with
Unless otherwise prescribed in a specific monograph or, in the
live vaccines.
absence of a specific monograph, unless otherwise justified
For the following tests except for the test for increase in
and authorised, the product complies with the test if no
virulence (section 1-8-3), use the vaccine strain at the least
animal shows abriormallocal or systemic reactions or signs of
attenuated passage level that will be present between the
disease, or die s from causes attributable to the product.
master seed lot and a batch of vaccine.
1-4. EXAMINATION OF REPRODUCTIVE PERFORMANCE
1-8-1. Spread of the vacdne strain. Spread of the vaccine
When the vaccine is recommended for use or may be used strain from vaccinated to unvaccinated target animals is
in pregnant animals or laying birds, carry out a test for investigated using the recommended route of administration
safety in this category of animals. lf the reproductive safety most likely to result in spread. Moreover, it may be necessary to
studies are not performed, an exclusion statement appears investigate the safety of spread to non-target species that couId
on the labe!, unless a scientific justification for absence of be highly susceptible to a live vaccine strain. An assessment
risk is provided. Examination of reproductive performance must be made of how many animal-to-animal passages are
must also be considered when data suggest that the starting likely to be sustainable under normal circumstances together
material from which the product is derived may be a risk with an assessment of the likely consequences.
factor. Where appropriate, reproductive performance of males
and females and harmful effects on the progeny, including 1-8-2. Dissemination in vacdnated animal. Faeces, urine,
teratogenic or abortifacient effects, are investigated by each of milk, eggs, and oral, nasal and other secretions shall be tested
the recommended routes of administration. lf multiple routes for the presence of the organism as appropriate. Moreover,
and methods of administration are specified for the product studies may be required of the dissemination of the vaccine
con cerned, administration by an routes is recommended. If strain in the body, with particular attention being paid to
1 route of administration has been shown to cause the most the predilection sites for replication of the organismo In the
severe effects, this single route may be selected as the only case of live vaccines for well-established zoonotic diseases for
one for use in the study. food-producing animals, these studies are obligatory and shall
For vaccines intended for use in mammals, in general 8 particularly take into account the persistence of the strain at
animals per group are used unless otherwise justified or the injection site.
specified in a specific monograph. Vaccines recommended 1-8-3. Increase in vindence. Unless otherwise prescribed in a
for use or that may be used in pregnant animals, are tested in specific monograph or, in the absence of a specific monograph,
each of the specific periods of gestation recommended for use unless otherwise justified and authorised, the following
on the ¡abe!' An exclusion statement will be required for those applies. This test is carried out using the master seed 101. rf
gestation periods not tested. the quantity of the master seed lot sufficient for performing
the test is not available, the lowest passage material used for
The observation period is extended to parturition, to examine
the production that is available in sufficient quantity may be
any harmful effects during gestation or 011 progeny, unless
used. At the time of inoculation, the animals in aH groups are
otherwise justified or specified in a specific monograph.
of an age suitable for recovery of the strain. Serial passages are
The following protocol is given as an example of an appropriate carried out in target animals using 5 groups of animals, unless
test for vaccines. there is justification to carry out more passages or unless
Safety in pregnant anima/s. Use not fewer than 8 animals the strain disappears from the test animal sooner. In vitro
per group, at the recommended stage of gestation or at a propagation may not be used to expand the passage inoculum.
range of stages of gestation according to the recommended The passages are carried out using animals most appropriate
schedule. Not fewer than 8 animals are used for each stage of to the potential risk being assessed.
pregnancy (i.e. 24 animals for 3 trimesters of pregnancy in The initial administration is carried out using the
cattle). Administer to each animal a recommended dose of recommended route of administration most likely to lead to
the vaccine. lf the recommended schedule requires a 2nd dose, reversion to virulence, using an initial inoculum containing
administer another dose after an interval of at least 14 days. the maximum release titre. After this, 110t fewer than 4 further
Unless otherwise prescribed in a specific monograph, observe serial passages through animals of the target species are
the animal s at least daily until 1 day after parturition. Unless undertaken. The passages are undertaken by the route of
otherwise prescribed in a specific monograph, or, in the administration most likely to lead to reversion to virulence.
absence of a speciflc monograph, unless otherwise justifled lf the properties of the strain allow sequential passage via
and authorised, the vaccine complies with the test if no animal natural spreading, this method may be used, otherwise
shows abnormallocal or systemic reactions or signs of disease, passage as described in each specific monograph is carried
or dies from causes attributable to the vaccine, and if no out and the micro-organisms that have been recovered at
adverse effects on the pregnancy or the offspring are noted. the final passage are tested for increase in virulence. For
1-5. RESIDUES the first 4 groups, a minimum of 2 animals is used for
mammalian vaccines, and a minimum of 5 birds is used
In the case oflive vaccines for well-established zoonotic
for avian vaccines. The last group consist of a minimum
diseases, the determination of residual vaccine organisms at
the injection site may be required, in addition to the studies of of 8 mammals or 10 birds. At each passage, the presence
of living vaccine-derived micro-organisms in the material
dissemination described below.
used for passage is demonstrated. Care must be taken to
1-6. ADVERSE EFFECTS ON IMMUNOLOGICAL avoid contamination by micro-organisms from previous
FUNCTIONS passages. When the micro-organism is not recovered from any
Where the product might adversely affect the immune intermediate in vivo passage, repeat the passage in 10 animals
response of the animal to which the product is administered or using in vivo passaged material from the last passage in which
of its progeny, suitable tests 011 the immunological functions the micro-organism was recovered. The micro-organism
are carried out. recovered is used as the inoculum for the next passage. lf the

590 See the information section on general mrJnIWflUin" (cover pages)


EUROPEAN PHARMACOPOEIA 8,0 5.2.7. Evaluation oí efficacy ofveterínary vacdnes and irnrnunosera

target micro-organism is not recovered, the experiment is significant exposure of the environment to the product, the
considered to be completed with the conclusion that the target potential ecotoxicity is evaluated, taking into account the
micro-organism do es not show an increase in virulence. properties of the product
General clinical observations are made during the study.
Animals in the Iast group are observed for 21 days unless 04/2008:50207
otherwise justified, These observations include all relevant
parameters typical for the disease that could indicate increase
in virulence. Compare the clinical signs and other relevant 5.2.7. EVALUATION OF EFFICACY
parameters with those observed in the animals used in the OF VETERINARY VACCINES AND
test for safety of the administration of 1 dose (section 1-1). lf
the last groupof animals shows no evidence of an increase in
IMMUNOSERA
virulence, further testing is not required. Otherwise, material The term 'product' means either a vaccine or an immunoserum
used for the 1st passage and the microorganisms recovered at throughout the text
the final passage level are used in a separate experiment using During development of the product, tests are carried out to
at least 8 animals per group for mammal vaccines and at least demonstrate that the product ís efficacious when administered
10 birds per group for avian vaccines, to compare directly the by each of the recommended routes and methods of
clinical signs and other relevant parameters, This study is administration and using the recommended schedule to
carried out using the route of administration that was used animals of each species and category for which use of the
for previous passages, An alternative route of administration product is to be recommended, The type of efficacy testing to
may be used if justified, be carried out varies considerably depending on the particular
Unless otherwise justified and authorised, the product type of product
complíes with the test if no animal dies or shows signs As part of tests carried out during development to establish
attributable to the vaccine strain and no indication of increased efficacy, the tests described in the Production section of a
virulence is observed in the animals of the last group, monograph may be carried out; the following must be taken
1-8-4, Biological properties of the vacdne strain, Other into account
tests may be necessary to determine as precisely as possible The dose to be used is that quantity of the product to be
the intrinsic biological properties of the vaccine strain (for recommended for use and containing the minimum titre or
example, neurotropism), For vector vaccines, evaluation is potency expected at the end of the period of validity,
made of the risk of changing the tropism or virulence of the For live vaccines, use vaccine containing virus/bacteria at the
strain and where necessary specific tests are carried out Such most attenuated passage level that will be present in a batch
tests are systematically carried out where the product of a ofvaccine,
foreign gene is incorporated into the strain as a structural
For immunosera, if appropriate, the dose tested also contains
protein,
minimum quantities of immunoglobulin or gammaglobulin
1-8-5, Recombination or genomic reassortment of strain. and/or total protein,
The probability of recombination or genomic reassortment The efficacy evidence must support all the claims being made,
with field or other strains shall be considered, For example, claims for protection against respiratory disease
must be supported at least by evidence of protection from
2, FIELD STUDIES clinical signs of respiratory disease, Where it is claimed that
Results from laboratory studies shall normally be there is protection from infection this must be demonstrated
supplemented with supportive data from field studies, using re-isolation techniques, lf more than one claim is made,
Provided that laboratory tests have adequately assessed the supporting evidence for each claim is required,
safety and efficacy of a product under experimental conditions Vaccines, The influence of passively acquired and maternally
using vaccines of maximum and mínimum titre or potency derived antibodies 011 the efficacy of a vaccine is adequately
respectively, a single batch of product may be used to assess evaluated, Any daims, stated or implied, regarding onset and
both safety and efficacy under field conditions, In these cases, duration of protection shall be supported by data from triaIs,
a typical routine batch of intermediate titre or potency may Claims related to duration of immunity are supported by
be used, evidence of protection, The test model described under
For food-producing mammals, the studies in dude Immunogenicity and/or Potency is not necessarily used to
measurement of the body temperatures of a sufficient number support daims regarding the duration of immunity afforded
of animals, before and after administration of the product; by a vaccine,
for other mammals, such measurements are carried out if the The efficacy of each of the components of multivalent and
laboratory studies indicate that there might be a problem, The combined vaccines shall be demonstrated using the combined
size and persistence of any local reaction and the proportion vaccine,
of animals showing local or systemic reactions are recorded,
Immunosera, Particular attention must be paid to providing
Performance measurements are made, where appropriate,
supporting data for the efficacy of the re gime that is to be
Performance measures for broilers indude weeldy mortality, recommended, For example, if it is recommended that
feed conversion ratios, age at slaughter and weight, down the immunoserum needs only to be administered once to
grading and rejects at the processing plant For vaccines achieve a prophylactic or therapeutic effect then this must
for use in laying birds or in birds that may be maintained be demonstrated, Any claims, stated or implied, regarding
to lay, the effect of the vaccine on laying performance and onset and duration of vrotection or therapeutic effect must be
hatchability is investigated, as appropriate, supported by data fro~ trials, For examp-le, the duration of
the protection afforded by a prophylactic dose of an antiserum
3, ECOTOXICITY must be studied so that appropriate guidance for the user can
An assessment is made of the potential harmful effects of the be given on the labe!'
product for the environment and any necessary precautionary Studies of immunological compatibility are undertaken when
measures to reduce such risks are identified, The likely simultaneous administration is recommended or where it is a
degree of exposure of the environment to the product is part of a usual administration schedule, Wherever a product
assessed, taking into account: the target species and mode is recommended as part of an administration scheme, the
of administration; excretion of the product; and disposal of priming or booster effect or the contribution of the product to
unused product If these factors indicate that there will be the efficacy of the scheme as a whole is demonstrated,

General Notices (1) apply to all monographs and other texts 591
5.2.8. Minimising the risk of transmitting TSE via medicinal products EUROPEAN PHARMACOPOEIA 8.0

LABORATORY TESTS 4. RISK ASSESSMENT OF MATERIALS OR SUBSTANCES


In principie, demonstration of efficacy is undertaken under USED IN THE MANUFACTURE AND PREPARATION
well-controlled laboratory conditions by challenge of the OF A MEDICINAL PRODUCT IN THE CONTEXT OF
target animal under the recommended conditions of use. REGULATORY COMPLIANCE
In so far as possible, the conditions under which the challenge 5. BENEFIT/RISK EVALUATION
is carried out shall mimic the natural conditions for infection, 6. SPECIFIC CONSIDERATIONS
for example with regard to the amount of challenge organism 6-1. Collagen
and the route of administration of the challenge.
6-2. Gelatin
Vaccines. Unless otherwise justified, challenge is carried out
6-3. Bovine blood and blood derivatives
using a strain different from the one used in the production
of the vaccine. 6-4. Tallow derivatives
If possible, the immune mechanism (cell-mediated/humoral, 6-5. Animal charco al
local/general, classes of immunoglobulin) that is initiated 6-6. Milk and milk derivatives
after the administration of the vaccine to target animals shall 6-7. Wool derivatives
be determined. 6-8. Amino acids
Immunosera. Data are provided from measurements of
6-9. Peptones
the antibody levels achieved in the target species after
administration of the product, as recommended. Where 1. INTRODUCTION
suitable published data exist, references are provided to
1-1. SCIENTIFIC BACKGROUND
relevant published literature on protective antibody levels and
challenge studies are avoided. Transmissible Spongiform Encephalopathies (TSEs) are
chronic degenerative nervous diseases charaeterised by
Where challenges are required, these can be given before or the accumulation of an abnormal isoform of a cellular
after administration of the product, in accordance with the glycoprotein (known as PrP or prion protein). The abnormal
indications and speeific claims to be made. isoform of PrP (Prp TSE ) differs from normal PrP (PrPe) in
FIELD TRIALS being highly resistant to pro tease and heat denaturation
treatments. Prp TSE is eonsidered to be the infective agent
In general, results from laboratory tests are supplemented with
responsible for transmitting TSE disease.
data from field trials, carried out, unless otherwise justified,
with untreated control animals. Provided that laboratory tests TSE diseases in animals include:
have adequately assessed the safety and efficacy of a produet - bovine spongiform encephalopathy (BSE) in cattle,
under experimental conditions using vaccines of maximum - scrapie in sheep and goats,
and minimum titre or potency respectively, a single batch of - chronic wasting disease (CWD) in cervids (deer and elk),
product could be used to assess both safety and efficacy under
field conditions. In these cases, a typical routine batch of - transmissible mink encephalopathy (TME) in farmed mink,
intermediate titre or potency may be used. Where laboratory - feline spongiform encephalopathy (FSE) in felids
trials cannot be supportive of efficacy, the performance of field (specifically domestic cats and captive large cats), and
trials alone may be aceeptable. - spongiform encephalopathy of exotic ungulates in zoos.
In humans, spongiform encephalopathies include
07/2011:50208 different forms of Creutzfeldt-Jakob Disease (CJD), Kuru,
Gerstmann-Straussler-Scheinker Syndrome (GSS), and Fatal
5.2.8. MINIMISING THE RISK Familial Insomnia (FFI).
OF TRANSMITTING ANIMAL Iatrogenic transmission of spongiform encephalopathies
has been reported. In sheep, scrapie has been accidentally
SPONGIFORM ENCEPHALOPATHY transmitted by the use of Louping III vaccine prepared from
AGENTS VIA HUMAN AND pooled, formaldehyde treated ovine brain and spleen in which
VETERINARY MEDICINAL PRODUCTS material from scrapie-infected sheep had been inadvertently
incorporated. AIso, transmission of scrapie to sheep and goats
This chapter is identical with the Note for Guidance on occurred following use of a formol-inactivated vaccine against
Minimising the Risk of Transmitting Animal Spongiform contagious agalactia, prepared with brain and mammary gland
Encephalopathy Agents vía Human and Veterínary Medicinal homogenates of sheep infeeted with Mycaplasma agalactiae.
Products - Revision 3, (EMA/410/01 rev. 3). In man, cases of transmission of CJD have been reported
which have been attributed to the parenteral administration
Contents of growth hormone and gonadotropin derived from human
cadaveric pituitary glands. Cases of CJD have also been
1. INTRODUCTION attributed to the use of contaminated instruments in brain
1-1. Scientific background surgery and with the transplantation of human dura mater
1-2. Regulatory compliance and cornea.
2. SCOPE Interspecies TSE transmission is restricted by a number of
natural barriers, transmissibility being affected by the species
3. GENERAL CONSIDERATIONS
of origin, the prion strain, dose, route of exposure and, in
3-1. Scientific principies for minimising risk some species, the host allele of the PRNP gene. Species
3-2. Animal source barriers can be crossed under appropriate conditions.
3 -2-1. Geographical sourcing BSE was first diagnosed in the United Kingdom in 1986 and a
3-2-1-1. Bovine materials large number of cattle and individual herds have been affected.
3-2-1-2. Sheep and goats (small ruminants) 1t is clear that BSE is a food borne disease associated with feed
3-2-2. BSE negligible risk (closed) bovine herds (e.g. meat and bone mea!) derived from TSE affected animals.
Other countries have experienced cases of BSE, either in
3-3. Animal parts, body fluids and secretions as starting animals imported from the United Kingdom or in indigenous
material animals. There is convincing evidence to show that the
3-4. Age of animals variant form of CJD (vCJD) is caused by the agent which is
3-5. Manufacturing Process responsible for ESE in cattle. Therefore, a cautious approach

592 See the ínformation sectíon on general monographs (ca ver pages)
EUROPEAN PHARMACOPOEIA 8.0 5.2.8. Minimising th" risk oí transmitting TSE via medicinal products

continues to be warranted if biological materials from species must demonstrate that medicinal products are manufactured
naturally affected by TSE diseases, especially bovine species, in accordance with the latest version of this note for guidance
are used for the manufacture of medicinal products. published in the Official Journal of the European Union. This
In the course of active surveillance programs, two previously is a continuing obligation after the marketing authorisation
unrecognized forms of atypical BSE (BSE-L, also named has been granted.
BASE, and BSE-H) have been identified in rare sporadic By definition, the principIe of Specified Risk Materials as
cases from Europe, North America, and Japan. The 'L' and defined in Regulation (EC) No 999/2001 ofthe European
'H' identify the higher and lower electrophoretic positions Parliament and of the Council!5) does not apply to medicinal
oftheir protease-resistant Prp TSE isoforms. It is noteworthy products. However, Regulation (EC) No 1774/2002 ofthe
that atypical cases have been found in countries that did not European Parliament and of the Counci](6), which applies
experience c1assical BSE so far, like Sweden, or in which only since 1st May 2003, lays down health rules concerning
few c1assical BSE cases have been found like Canada or USA. animal by-products not intended for human consumption.
The atypical BSE agent has been experimentally transmitted As a general rule, and unless properly justified, all animal
to transgenic mice expressing the human prion protein and to by-products used as starting materials in the manufacture of
a cynomolgus monkey. medicinal products should be 'Category 3 (i.e. safe) materials
Scrapie occurs worldwide and has been reported in most or equivalent: as defined in Regulation (EC) No 1774/2002.
European countries. It has the highest incidence in Cyprus. Justification for the use of substances derived from other, high
While human s have been exposed to naturally occurring infectivity materials must follow an appropriate benefitlrisk
scrapie for over 250 years, there is no epidemiological evidence evaluation (see further below).
directly linking scrapie to spongiform encephalopathies in The note for guidance should be read in conjunction with the
humansOJ. However, there remains a theoretical and currently various EU legal instruments including Commission decisions
unquantifiable risk that some BSE-contaminated protein progressively implemented since 1991. Where appropriate,
supplement may have been fed to sheep. Further, it should references to these decisions are given in the texto Position
also be assumed that any ESE agent introduced into the small statements and explanatory notes made by the Committee for
ruminant population via contaminated feed is likely to be Medicinal Products for Human Use (CHMP) and Committee
recycled and amplified!2J. for Medicinal Products for Veterinary Use (CVMP) are still
There is interest in infecting ceUs with TSE agents to develop applicable for the purpose of regulatory compliance unless
assays and for basic scientific reasons. Some success has been otherwise superseded by the note for guidance.
reported, usually but not always with neural celllines. The The general monograph Products with risk of transmitting
conditions needed to infect a ceH are not well understood and agents of animal spongiform encephalopathies of the European
the process is difficult requiring particular combinations of Pharmacopoeia refers to this chapter, which is identical
agent and cell. It is not considered appropriate to make specific with the note for guidance. The monograph forms the basis
recommendations in terms of cell substrates to be used for for issuing Certificates of Suitability as a procedure for
production of biological/biotechnology-derived substances. demonstrating TSE compliance for substances and materials
Nevertheless, the possibility of infection of celllines with TSE used in the manufacture of human and veterinary medicinal
agents should be taken into account in risk assessments. products.
1-2. REGULA TORY COMPLIANCE Clarification of note for guidance. As the scientific
understanding of TSEs, especially the pathogenesis of the
Risk assessment. Since the use of animal-derived materials diseases, is evolving, from time to time CHMP and its
is unavoidable for the production of some medicinal Biologics Working Party in collaboration with CVMP and
products and that complete elimination of risk at source is its Immunologicals Working Party may be required in the
rarely possible, the measures taken to manage the risk of future to develop supplementary guidance in the form of
transmitting animal TSEs vía medicinal products represent position statements or explanatory notes for the purpose
risk minimisation rather than risk elimination. Consequently, of clarifying the note for guidance. The supplementary
the basis for regulatory compliance should be based on a risk guidance shall be published by the Commission and on the
assessment, taking into consideration al! pertinent factors as website of the European Medicines Agency and taken into
identified in this chapter (see below). consideration accordingly in the scope of the certification
Legal basis. The note for guidance is published by the of the European Directorate for the Quality of Medicines &
European Commission following HealthCare (EDQM).
- Annex 1, part 1, module 3, section 3.2: Content:
2. SCOPE
basic principIes and requirements, point (9) of
Directive 200 l/83/EC of the European Parliament and TSE-RELEVANT ANIMAL SPECIES
of the Council of 6 November 2001 on the Community Cattle, sheep, goats and animals that are naturally susceptible
code relating to medicinal products for human use(3), as to infection with transmissible spongiform encephalopathy
amended, and agents or susceptible to infection through the oral route other
- Annex 1, Title 1, part 2, section C Production and control of than humans(7) and non-human primates are defined as
starting material of Directive 200 l/82/EC of the European "TSE-relevant animal species"W
Parliament and of the Council of 6 N ovember 2001 on MATERIALS
the Community code relating to veterinary medicinal This chapter is concerned with materials derived from
products(4), as amended. "TSE-relevant animal species" that are used for the preparation
These directives require that applicants for marketing of:
authorisation for human and veterinary medicinal products - active substances,

(1) This is currently being assessed by EFSA ancl ECDC. Por updated informatiol1, pIease refer to the following link: http://registerofqucstions.efsa.europa.eu/roqFrontend/
q uestionsListLoader?mandate== M -2009-0221
(2) In January 2005, after confirmation of BSE in a goat in additionallegislative measures were taken related to monitoring and an increased testing of srnall ruminants. The
increased surveillance did 110t identify additional cases of BSE in and goats in the EU.
(3) 01 L 311, 28.11.2001. p. 67.
(4) 01 L 311. 28.11.2001, p. 1.
(5) 01 L 147. 31.5.2001, p. 1.
(6) 01 L 273,10.10.2002, p.!. Regulation (Ee) 1774/2002 has been repealed by Regulation (Ee) 106912009 that will apply from4 March 2011 (01 L 300,14.11.2009, p. 1).
(7) Regulatory guidance and position papers have been issued by the Committee [or Medicinal Producls for Human Use and its Biologics vVorking Parly on human tissue derived
medicinal producls in relatian to CID and vCTD. Such guidancc can be found 011 hltp://www.ema.europa.eu
(8) Pigs and birds, which are animal of particular intercst for the production of medicinal products, are not naLurally susceptible to infection via the oral route. Therefore lhey are
not TSE-relevant animal species within meaning of thi$ chapter. Also dogs, rabbits and fish are non TSE-relevant animal species within the meaning of thi5 charter.

General NoNces (1) apply io all monographs and other texts 593
5.2.8. Minimising the r:isk of transmitting TSE via medicinal products EUROPEAI'.J PHARMACOPOEIA 8.0

- excipients and adjuvants, and However, where materials derived from the "TSE-relevant
- raw and starting materials and reagents used in production animal species" are used in fermentation/routine production
(e.g. bovine serum albumin, enzymes, culture media processes or in the establishment of working seeds and
induding those used to prepare working ceU banks, or new working cel! banks, the applicant must demonstrate that they
master cell banks for medicinal products which are subject fulfil the requirements of the note for guidance.
to a new marketing authorisation). 3. GENERAL CONSIDERATIONS
This chapter is also applicable to materials that come into
3-1. SCIENTIFIC PRINCIPLES POR MINIMISING RISK
direct contact with the equipment used in manufacture of the
medicinal product or that come in contact with the medicinal vVhen manufacturers have a choice, the use of material s from
product and therefore have the potential for contamination. "non TSE-relevant animal species" or non-animal origin is
preferred. The rationale for using materials derived from
Materials used in the qualification of plant and equipment, "TSE-relevant animal species" instead of materials from
such as culture media used in media fill experiments to validate "non-TSE-relevant species" or of non -animal origin should be
the aseptic filling process, shall be considered in compliance given. rf materials from "TSE-relevant animal species" have
with this chapter provided that the constituent or constituents to be used, consideration should be given to al! the necessary
are derived from tissues with no detectable infectivity measures to minimise the risk of transmission of TSE.
(category rc tissues), where the risk of cross-contamination
with potentially infective tissues has been considered (see Readily applicable diagnostic tests for TSE infectivity in vivo
section 3-3) and where the materials are sourced from are not yet available. Diagnosis is based on post-mortem
countries with negligible BSE risk or controlled BSE risk confirmation of characteristic brain lesions by histopathology
(Categories A and B, respectively - see section 3-2). Such and/or detection of PrpTSE by Western blot or immunoassay.
information shall be provided in the dossier for a marketing The demonstration of infectivity by the inoculation of suspect
authorisation and verified during routine inspection for tissue into target species or laboratory animals is also used for
compliance with Good Manufacturing Practice (GMP). confirmation. However, due to the long incubation periods of
al! TSEs, results of in vivo tests are available only after months
Other materials such as deaning agents, softeners and or years.
lubricants that come into contact with the medicinal product
Several immunochemical tests have been developed for the
during its routine manufacture or in the finishing stage or in
detection of Pr p TSE in post -mortem samples and sorne are
the primary packaging are considered in compliance with
now considered to be extremely sensitive. However, their
this chapter if they are tallow derivatives prepared using the
ability to detect an infected animal depends on the timing
rigorous physicochemical processes as described in section 6.
of sample collection in relation to timing of exposure, the
SEED LOTS, CELL BANKS AND ROUTINE type of tissue collected and infectious dose acquired, together
PERMENTATION/PRODUCTION!9) with consequential timing of onset of clinical disease. There
For the purpose of regulatory compliance, master seeds or is currently insufficient information on how this might be
master ceU banks in marketing authorisation applications affected by strain variations.
lodged after 1 July 2000 (for human medicinal products) or Although screening of source animals by in vitro tests
1 October 2000 (for veterinary medicinal products) shall be may prevent the use of animals at late stages of incubation
covered by the note for guidance. of the disease and may provide information about the
Master seeds and master ceH banks, epidemiological status of a given country or region, none of
- for vaccine antigens, the tests are considered suitable to unambiguously confirm
- for a biotechnology-derived medicinal product as described the negative status of an animal.
in the Annex to Regulation (EC) No 726/2004 of the Minimising the risks of transmission of TSE is based upon
European Parliament and of the Council(lO), and three complementary parameters :
for other medicinal products using seed lots or cell banking - the source animals and their geographical origin,
systems in their manufacture, - nature of animal material used in manufacture and any
that have already been approved for the manufacture of procedures in place to avoid cross-contamination with
a constituent of an authorised medicinal product shall be higher risk materials,
considered in compliance with the note for guidance even if - production process(es) including the quality assurance
they are incorporated in marketing authorisation applications system in place to ensure product consistency and
lodged after 1 JuIy 2000 (for human medicinal products) or traceability.
1 October 2000 (for veterinary medicinal products). 3-2. ANIMAL SOURCE
Master cel! banks and master seeds established before The source materials used for the production of materials
1 July 2000 (for human medicinal products) or 1 October 2000 for the manufacture of medicinal products shall be derived
(for veterinary medicinal products), but not yet approved from animals fit for human consumption following ante- and
as a constituent of an authorised medicinal product shaU post-mortem inspection in accordance with EU or equivalent
demonstrate that they fulfil the requirements of the note for (third country) conditions, except for materials derived from
guidance. lf, for sorne raw or starting materials or reagents live animals, which should be found healthy after clinical
used for the establishment of these cel! banks or seeds, ful! examination.
documentary evidence is no longer available, the applicant 3-2-l. Geographical sourdng
should present a risk assessment as described in Section 4 of
the note for guidance. 3-2-1-1. Bovine materials
The World Organisation for Animal Health (OIE)(1j) lays
Established working seeds or cel! banks used in the
manufacture of medicinal products authorised before down the criteria for the assessment of the status of countries
1 July 2000 (human medicines) or 1 October 2000 (veterinary in the chapter of the International Animal Health Code on
medicines), which have been subjected to a properly bovine spongiform encephalopathy. Countries or regions are
conducted risk assessment by a Competent Authority of classified as follows:
the Member States or the European Medicines Agency and A. countries or regions with a negligible BSE risk;
declared to be acceptable, shal! also be considered compliant. B. countries or regions with a control!ed ESE fisk;

(9) See also: Position paper 011 the assessment of the risk oE transmission of animal spongiform encephalopathy agents by master seed materials used in the production o[ veterinary
vaccines (EMEA/CVMP/019/01-Febrnary 2001 adopted by lhe Commiltee for Medicinal Products for Veterinary Use (CVMP) in )uly 2001, (O) e 286.12.10.2001, p. 12)).
(0) O) L 136, 30.4.2004, p. 1.
(11) hllp:! /www.oie.int/eng/Status/BSE/en_BSE_free.htm

594 See the information section on general monographs (ca ver pages)
EUROPEAN PHARMACOPOEIA 8.0 5.2.8. Minimising the risk of transmitti.ng TSE via medicinal producís

C. countries or regions with an undetermined BSE risk. in sheep, the use of a genotype(s) showing resistance to
BSE/scrapie infection could be con si de red in establishing TSE
As stipulated in Commission Regulation (EC) No 999/2001, free flocks(17), However, the possibility that genotypes resistant
as amended (12), the classification of countries or regions io scrapie could be susceptible to BSE (experimental oral
thereof according to their ESE risk, based on the rules laid exposure) or atypical scrapie (natural cases) shou1d also be
down by OlE, is lega11y binding in the EU since 1 July 2007. taken into account. Goats have not been studied sufficiently
Commission Decision 2007/453/EC(13) as amended, provides with regard to a genotype specific sensitivity.
the classification of countries or regions according to their
ESE risk. Material of small ruminant origin should preferably be
sourced from countries with a long history of absence of
Previously, the European Commission Scientific Steering
scrapie, Justification shall be required if the material is sourced
Committee (SSC)(14) had established a temporary system for
from some other origino
classifying the countries according to their geographical ESE
risk (GBR)(J5). 3-2-2. ESE negligible risk (dosed) bovine herds. The safest
sourcing is from countries or regions with a negligible risk
For the purposes of this chapter the ESE classification based (Category A countries). Other countries may have or have
on the OlE rules should be used. If a country, which was had cases of BSE at sorne point in time and the practical
previously classified in accordance to the SSC GBR criteria, concept of "Negligible risk (closed) bovine herds" has been
has not been classified yet according to the OlE rules, the GBR developed by the SSC and endorsed by the CHMP and CVMP.
elassification can be used until OlE classification has taken Criteria for establishing and maintaining a "BSE negligible
place, provided that there is no evidence of significant change risk (closed) bovine herd" can be found in the SSC opinion
in its BSE risk(J6). of 22-23 July 1999(18),

Where there is a choice, animals should be sourced from


countries with the lowest possible BSE risk (negligible ESE For the time being it is not possible to quantify the reduction
risk countries (Category A)) unless the use of material from of the geographical BSE risk for cattle from BSE 'negligible
countries with a higher BSE risk is justified. Some of the risk (elosed) bovine herds', However, it is expected that this
materials identified in Section 6, "Specific Conditions" can be risk reduction is substantial. Therefore, sourcing from such
sourced from countries with controlled BSE risk (Category B) closed bovine herds sha11 be considered in the risk assessment
and, in sorne cases, from countries with undetermined BSE in conjunction with the OlE classification of the country.
risk (Category C), provided that the contro15 and requirements 3-3, ANIMAL PARTS, BODY FLUIDS AND SECRETIONS
as specified in the relevant sections below are applied. Apart AS STARTING MATERIAL
from these exceptions, animals must not be sourced from In a TSE infected animal, different organs and secretions have
countries with undetermined BSE risk (Category C), and different levels of infectivity. If materials from 'TSE-re1evant
justification5 for the use of animal s from countries with animal species' have to be used, consideration should be given
undetermined BSE risk (Category C) must always be provided. to use materials of the lowest category of risk. The tables given
in the Annex of this chapter(19! sumrnarise current data about
3-2-1-2, Sheep and goats (small ruminants)
the distribution of infectivity and PrpTSE in cattle with BSE,
and in sheep and goats with scrapie(20).
Naturally occurring clinical scrapie cases have been reported
in a number of countries worldwide, As BSE in sheep and
goats could possibly be mistaken for scrapie, as a precautionary The information in the tables is based exclusively upon
measure, sourcing of materials derived from small ruminants observatio11s of naturally occurring disease or primary
shall take into account the prevalence of both BSE and scrapie experimental infection by the oral route (in cattle) but
in the country and the tissues from which the materials are do es not inelude data 011 models using strains of TSE
derived. that have been adapted to experimental animals, because
passaged strain phenotypes can differ significantly and
The principIes reIated to "BSE negligible risk (elosed) bovine unpredictably from those of naturally occurring disease.
herds" (see section 3-2-2) could equally be applied in the Because immunohistochemical and/or Western blot detection
context of small ruminants in order to deveIop a framework of misfolded host protein (PrP TSE ) have proven to be a
to define the TSE status of a flock of sma11 ruminants. For surrogate marker of infectivity, PrpTSE testing results have been
sheep, because of the concern over the possibility of BSE presented in parallel with bioassay data. Tissues are grouped

(12) Regulation (EC) No 722/2007 (o¡ L 164, 26,6,2007, p. 7)


(13) o¡ L 172, 30,6,2007, p, 84
(14) Tbe Scicntific Steering Committee established by Commi'ssion Decision 97/404/EC (OJ L 169,27.6.1997, p. 85) shall assist the Commission to obtain the best scientific advice available
011 matters relating to consumer health. Since May 2003, its tasks have been taken ayer by the European Food Safety Authority (EFSA): http://vvww.efsa.europa.eu
(15) Tlle European Scientific Steering Committee classification for geographical BSE risk (GBR) gives an indication of the Ievel of likelihood of the presence of one ar more cattJe clinically
or pre-clinically infected with BSE in a given country or region. A definition of the four categories is provided in the following Table.

GBR level Presence of one or more cattle dinically or pre-dinically infected with ESE in a geographical region/ cOlllntry

Highly unlikely

JI Unlikely but not excluded

1II Likely but not confirmed or confinned at a lower level

IV Confirmed at a higher level (:2: 100 cases/1 Million adlllt cattle per year)

Reports of the GBR assessment of the countries are available on the SSC website (http://ec.europa.ell/food!fs/sc/ssc/outcome_en.html)
(16) Experts consider that the GBR classification system is stable enough, so that it can continue to be used, during the interim period, for the demonstration of compliance with this chapter.
(17) Opinion ofthe Scientific Panel on Biological Hazards on 'the breeding programme for TSE resistance in sheep': http://www.efsa.europa.eu/EFSA/efsa_locale-
1178620753812 1178620775678,htm
(18) SSC Scientific Opinion on the conditions related to "BSE Negligible Risk (Closed) Bovine Herds" adopted at the meeting of 22-23 JuIy 1999. http://ec.europa.eu/
food!fs/sc/ssc/out56 en.htrnl
(19) The tissue classification tables are based llpon the most recent WHO Guidelines on Tissue Infectivity Distribution in Transmissible Spongiform Encephalopathies (2010)
http://www.who.int/bloddproducts/tablestissueinfectivity.pdf
(20) A Scientific opinion on BSE/TSE infectivity in small ruminant tissucs is currently being reviewed by EFSA (Question No EFSA-Q-2010-052). For updated information pIease follow
this link: hltp://registerofquestions.efsa.europa.en/roqFrontend/ q uestionsListLoader?mandate=M -2010-0041

General Notices (1) apply to all monographs and other texts 595
5.2.8. Minimising the risk of transmitting TSE via medicinal products EUROPEAN PHARMACOPOEIA 8.0

into three major infectivity categories, irrespective of the stage - the extent of brain damage,
of disease: - the dissemination of brain particles in the animal body.
Category lA High-infectivity tissues These factors must be considered in conjunction with the
central nervous system (CNS) tissues that atlain a high
OIE/GBR classification of the source animals, the age of the
titre of infectivity in the later stages of al! TSEs, and animals in the case of cattle and the post-mortem testing of
certain tissues that are anatomical!y associated with the the cattle using a validated method.
CNS
Category lB Lower- infectivity tissues
The underIying principIes indicated aboye would be equally
applicable to sheep and goats.
peripheral tissues that have tested positive for infectivity
and/or Pr pTSE in at least one form of TSE
The risk posed by cross-contamination will be dependent on
Category IC Tissnes with no detectable infectivity several complementary factors including:
- measures adopted to avoid contamination during collection
tissues that have been exarnined for infectivity, without
any infectivity detected, and/or PrPTSE, with negative of tissues (see aboye),
results - level of contamination (amount of the contaminating
Category lA tissues and substances derived from them shall tissue),
not be used in the manufacture of medicinal products, unless - amount and type of materials collected at the same time.
justified (see Section 5). Manufacturers or the marketing authorisation
Although the category of lower risk tissues (category lB holders/applicants should take into account the risk
tissues) almost certainly includes sorne (e.g. blood) with with respect to cross-contamination.
a lower risk than others (e.g. lymphoreticular tissues), the 3-4. AGE OF ANIMALS
data about infectivity levels in these tissues are too limited to As the TSE infectivity accumulates in bovine animals over
subdivide the category into different levels of risk. It is also an incubation period of several years, it is prudent to source
evident that the placement of a given tissue in one or another from young animals.
category can be disease and species specific, and subject to
revision as new data emerge. Presence of infectious material has essentially been reported
in the centralnervous system and related tissues, as well as
For the risk assessment (see section 4), manufacturers and/or in the lymphoreticular system, depending on the TSE agent
marketing authorisation holders/applicants shall take into (BSE in cattle or scrapie in sheep and goat). The exact time
account the tissue classification tables in the Annex to this course of infectivity in the respective body parts and tissues,
chapter. from the date of infection, is not known in both species and,
The categories in the tables are only indicative and it is as such, it is difficult to give clear guidance on the age aboye
important to note the following points. which the various tissues may be infected and should not be
collected. The initial recommendation to collect tissues in the
- In certain situations there couId be cross-contamination youngest age is still valido In addition, it is noteworthy that the
of tissues of different categories of infectivity. The potential age criteria depend also on the geographical origino Age is a
risk will be influenced by the circumstances in which more important parameter for materials from countries where
tissues were removed, especially by contact of tissues the risk is higher (Category B and C countries), than from
with lower-infectivity tissues or no detectable infectivity countries with a negligible BSE risk (Category A countries).
(categories lB and lC tissues) with high-infectivity tissues
(category lA tissues). Thus, cross-contamination of sorne 3-5. MANUFACTURING PROCESS
tissues may be increased if infected animals are slaughtered The assessment of the overall TSE risk reduction of a medicinal
by brain stunning (penetrative or non penetrative) or product shall take into account the control measures instituted
if the brain and/or spinal cord is sawed. The risk of with respect to:
cross-contamination will be decreased if body fluids are - sourcing of the raw/starting materials, and
collected with minimal damage to tissue and cellular - the manufacturing process.
components are removed, and if foetal blood is collected
without contamination from other maternal or foetal Controlled sourcing is a very important criterion in achieving
tissues including placenta, amniotic and allantoic fluids. acceptable safety of the product, due to the documented
For certain tissues, it is very difficult or impossible to resistance of TSE agents to most inactivation procedures.
prevent cross-contamination with category lA tissues (e.g. A quality assurance system, such as ISO 9000 certification,
skull). This has to be considered in the risk assessment. HACCP(22) or GMP, must be put in place for monitoring the
production process and for batch delineation (i.e. definition
- For certain classes of substances the stunning/slaughtering
of batch, separation of batches, cleaning between batches).
techniques used may be important in determining the
potential risk(21) because of the likelihood of disseminating Procedures shall be put in place to ensure traceability as well
as self-auditing and to auditing suppliers of raw/starting
the brain particles into the peripheral organs, particularly
materials.
to the lungs. Stunning/slaughtering techniques should
be described as well as the procedures to remove high Certain production procedures may contribute considerably
infectivity tissues. The procedures to collect the animal to the reduction of the risk of TSE contamination, e.g.
tissues/organs to be used and the measures in place to procedures used in the manufacture of tallow derivatives (see
avoid cross-contamination with a higher risk material must section 6). As such rigorous processing cannot be applied to
also be described in detail. many products, processes involving physical removal, such as
precipitation and filtration to remove prion-rich material, are
- The risk of contamination of tissues and organs with likely to be more appropriate than chemical treatments. A
BSE-infectivity potentially harboured in central nervous description of the manufacturing process, including in -process
material as a consequence of the stunning method used for contr01s applied, shall be presented and the steps that might
cattle slaughtering depends on the following factors : contribute to reduction or elimination of TSE contamination
- the amount of BSE-infectivity in the brain of the should be discussed. Whenever different manufacturing
slaughtered animal, sites are involved, the steps performed at each site shall be

(21) SSC opinion OIl stunning methods and BSE risk (The risk of dissemination ofbrain particles into the bIoad and carcas s when applying certain stunning methods), adopted at the
meeting of 10-11 January 2002. http://ec.europa.eu/food/fs/sc!ssc/out245_en.pdf. Report of the EFSA Working group on BSE risk from dissemination ofbrain particles in bIoad and
carcass. Question No EFSA-Q-2003-122, adopted on 21 October 2004, http://www.efsa.europa.eu/EFSA/efsa_locale-1178620753812_1178620777397.htm
(22) Hazard Analysis Critical Control PoinL

596 See the information section on general monographs (caver pages)


EUROPEAN PHARMACOPOEIA 8.0 5.2.8. Minimising the risk of transmitting TSE via medi.cinal products

clearly identified. The measures in place in order to ensure As indicated in the introduction to this chapter, regulatory
traceability of every production batch to the source material compliance is based on a favourable outcome from a
should be described. risk assessment. The risk assessments, conducted by the
Cleaning process. Cleaning of process equipment may be manufacturers and/or the marketing authorisation holders
difficult to validate for the elimination of TSE agents. It is or applicants for the different materials or substances from
reported that after exposure to high titre preparations of TSE "TSE-relevant animal species" used in the manufacture of a
agent, detectable infectivity can remain bound to the surface of medicinal product shall show that al! TSE risk factors have
stainless steel. The removal of all adsorbed protein by the use been taken into account and, where possible, risk has been
of 1 M sodium hydroxide or chlorine releasing disinfectants minimised by application of the principIes described in this
(e.g. 20000 ppm chlorine for 1 h) have been considered chapter. TSE Certificates of suitability issued by the EDQM
acceptable approaches where equipment that cannot be may be used by the marketing authorisation holders or
replaced has been exposed to potentially contaminated applicants as the basis of the risk assessments.
material. Milder treatments with limited concentrations of An overall risk assessment for the medicinal product,
alkali or stabilized bleach, when properly fonnulated with conducted by the marketing authorisation holder s or
detergents and used at specified temperatures, have been applicants, shall take into account the risk assessments for all
shown to exhibit similar efficiency for removing prions as did the different material s from "TSE-relevant animal species"
classical NaOH or chlorine treatments. A system based on and, where appropriate, TSE reduction or inactivation by the
vaporised hydrogen peroxide also appeared to be efficient manufacturing steps of the active substance and/or finished
for inactivating TSE agents. These new treatments are more producto
compatible with delicate materials and may be suitable for
The final determination of regulatory compliance rests with
practical use(23).
the competent authority.
If risk materials are used in the manufacture of a product,
cleaning procedures, including control measures, shall be put It is incumbent upon the manufacturers and/or the marketing
in place in order to minimise the risk of cross-contamination authorisation holders or applicants for both human and
between production batches. This is especially important veterinary medicinal products to select and justify the control
if materials from different risk categories are handled in measures for a given "TSE-relevant animal species" derivative,
the same plant with the same equipment. In the case of taking into account the latest scientific and technical progress.
using category lA materials in the manufacture of a product,
dedicated equipment shall be used, unless otherwise justified. 5. BENEFIT/RISK EVALUATION
Further research is needed to develop and validate In addition to the parameters as mentioned in sections 3 (that
new decontamination procedures to lower the risk of may be covered by a TSE Certificate of Suitability issued by
cross-contamination for material and devices which are not the EDQM) and 4, the acceptability of a particular medicinal
compatible with WHO-recommended procedures. product containing materials derived from a "TSE-relevant
animal species", or which as a result of manufacture could
Removal/Inactivation validation. Validation studies of
contain these materials, shall take into account the following
removal/inactivation procedures for TSEs can be difficult
factors:
to interpret. It is necessary to take into consideration the
nature of the spiked material and its relevan ce to the natural - route of administration of the medicinal product,
situatiol1, the design of the study (including scaling-down of - quantity of animal material used in the medicinal product,
processes) and the method of detection of the agent (in vitra
or in vivo assay). Further research is needed to develop an - maximum therapeutic dosage (daily dose and duration of
understanding of the most appropriate "spike preparation" for treatment),
validation studies. Therefore, validation studies are currently - intended use of the medicinal product and its clinical
not generally required. However, if claims are made for benefit,
the safety of the product with respect to TSEs based on the
ability of manufacturing processes to remove or inactivate - presence of a species barrier.
TSE agents, they must be substantiated by appropriate High -infectivity tissues (category lA tissues) and substances
investigational studies(24). derived thereof shallnot be used in manufacture of medicinal
In addition to appropriate sourcing, manufacturers are products, their starting materials and intermediate products
encouraged to continue their investigations into removal and (including active substances, excipients and reagents), unless
inactivation methods to identify steps/processes that would justified. A justification why no other materials can be
have benefit in assuring the removal or inactivation of TSE used shall be provided. In these exceptional and justified
agents. In any event, a production process wherever possible circumstances, the use ofhigh-infectivity tissues could be
shall be designed taking account of available information envisaged for the manufacture of active substances, when,
on methods which are thought to inactivate or remove TSE after performing the risk assessment as described in Section 4
agents. of this chapter, and taking into account the intended clinical
use, a positive beneflt/risk assessment can be presented by
For certain types of products (see section 6-3 Bovine blood
the marketing authorisation applicant. Substances from
and blood derivatives), where validated removal/inactivation
category lA materials, if their use is justified, must be
is not readily applicable, process evaluation might be required.
produced from animals of countries with negligible BSE risk
This should be based on the starting material and any
(Category A).
published data on TSE risk.
4. RISK ASSESSMENT OF MATERIALS OR SUBSTANCES 6. SPECIFIC CONSIDERATIONS
USED IN THE MANUFACTURE AND PREPARATION The following materials prepared from "TSE-relevant animal
OF A MEDICINAL PRODUCT IN THE CONTEXT OF species" are considered in compliance with this chapter
REGULATORY COMPLIANCE provided that they meet at least the conditions specifled
The assessment of the risk associated with TSE needs careful below. The relevant information or a certificate of suitability
consideration of al! of the parameters as outlined in section granted by the EDQM shall be provided by the marketing
3-1 (Scientific PrincipIes for Minimising Risk). authorisation applicant/holder.

(23) WHO Guidelines 011 Tissue Infectivity Distribution in Transmissible Spongiform Encephalopathies (2006) http://wvvw-.who.int/bloodproducts/tse/WHO%20TSE%20Guide-
lines%20FINAL-22%20JunellpdatedNL.pdf
(24) Guideline on the investigaban ofmanufacturing process fOI plasma-derived medicinal products with regard lo vCTD risk CPMP/BWP/5136/03

General Natices (1) apply ta all managraphs and ather texts 597
5.2.8. Minimising the risk of transmitting TSE via medicinal products EUROPEAN PHARMACOPOEIA 8.0

6-1. COLLA GEN 2. Vertebrae shall be removed from the raw/starting


Collagen is a fibrous protein component of mammalian materials from cattle over 30 months from countries with
connective tissue. a controlled or an undetermined BSE risk (Categories B
or C).
For collagen, documentation to demonstrate compliance with
this chapter needs to be provided taking into account the 3. Gelatin for parenteral use should only be manufactured
provisiol1s listed in sections 3 to 5. In addition, consideration from bones coming from countries with a negligible or
should be given to the following. a controlled BSE risk (Category A and B, respectively).
Gelatin for oral use can be manufactured from bones from
- For collagen produced from bones, the conditions specified
countries with a negligible, a controlled or an undetermined
for gelatin are applicable (see below). Lower inactivation
BSE risk (Category A, B and C, respectively).
capacity is expected from the collagen manufacturing
process than from that of gelatin. Therefore, sourcing 4. Gelatin shall be manufactured using one of the
becomes a more critical aspect to considero manufacturing methods described below.
- Collagen produced from tissues such as hides, skins, Manufacturing methods
tendons and sinews do not usually present a measurable Hides. No specific measures with regard to the processing
TSE risk provided that contamination with potentially conditions are required for gelatin produced from hides
infected material s, for example spillage of blood and/or provided that control measures are put in place to avoid
central nervous tissues, is avoided during procurement. cross-contamination both during the procurement of the
Therefore, hides represent a safer raw material for hides and during the manufacturing process.
human implants derived from collagen. However,
cross-contamination with brain material released during Bones. Where bones are used as the starting material, the
the slaughtering process that may have dried on the surface mode of manufacture will be the second parameter that will
of hides would be difficult to eliminate. This is another ensure the safety of gelatin.
aspect to consider in the evaluation of safety of this source - Gelatin can be manufactured from bones from countries
material. with a negligible, a controlled or an undetermined BSE
The collagen manufacturing process can have some steps in risk (Categories A, B or C) sourced in accordance with
common with the manufacture of gelatin such as alkaline the conditions described in section 6-2 under The source
and sodium sulphate treatment, calcium hydroxide and material used, using the acid, alkaline or heat/pressure
sodium hydroxide treatments or enzyme treatment. However, manufacturing process.
even these common steps can differ in duration and pH - The manufacturing process shall be taken into
condition which can result in significant differences in their consideration when performing the risk assessment as
inactivation capacity. Manufacturers should at least conduct described in Section 4 of this chapter. Both the acid
a process evaluation based on the similarities of the collagen and the alkaline manufacturing methods have shown
processing steps, as compared to known inactivation steps similar overall inactivationlremoval of TSE infectivity
in the manufacture of geJatin, in order to support the safety in the ge!atin validation experiments. Studies have
of the product. In addition to processing, differences also shown that an additional alkaline treatment (pH 13,
exist in the final use of the material and, consequently, in 2 h) of the bones/ossein further increases the TSE
their risk assessment, while gelatin is widely used for oral inactivation/removal capacity of the manufacturing
administration, many collagen applications are in the form process. Other processing steps such as filtration,
of surgical implants. This aspect should also be considered ion-exchange chromatography and UHT sterilisation also
in the final risk assessment. contributes to the safety of gelatin.
6-2. GELATIN - For a typical alkaline manufacturing process, bones are
Gelatin is a natural, soluble protein, gelling or non-gelling, finely crushed, degreased with hot water and demineralised
obtained by the partial hydrolysis of collagen produced from with dilute hydrochloric acid (at a minimum of 4 per cent
bones, hides and skins of animals. and pH < 1.5) over a period of at least 2 days to produce
For gelatin, documentation to demonstrate compliance with the ossein. This is followed by an alkaline treatment with
this chapter needs to be provided taking into account the saturated lime solution (pH at least 12.5) for a period of
provisions listed in sections 3 to 5. In addition, consideration at least 20 days.
should be given to the following(25J. - Bovine bones may also be treated by an acid process. The
The source material used liming step is then replaced by an acid pre-treatment where
the ossein is treated at pH < 3.5 for a mínimum of 10 hours.
Gelatin used in medicinal products can be manufactured from
bones or hides. - A "flash" heat treatment (sterilisation) step at 138 oC
minimum for 4 s at least is applied to both acid and alkaline
Hídes as the starting material. On the basis of current manufacturing process.
knowledge, hides used for gelatin production represent a
safer source material as compared to bones. However, it is - In the heat/pressure process, the dried degreased crushed
highly recommended that measures should be put in place to bones are autoclaved with saturated steam at a pressure
avoid cross-contamination with potentially infected materials greater than 3 bar and a minimum temperature of 133 oC,
during procurement. for at least 20 min, followed by extraction of the protein
with hot water.
Bones as the starting material. Where bones are used to
manufacture ge!atin, the quality of the starting materials The finishing steps are similar for the alkaline, acid and
needs to be controlled as an additional parameter to ensure heat/pressure process and in dude extraction of the gelatine,
the safety of the final product. Therefore, the following should washing, filtration and concentration.
be applied. 6-3. BOVINE BLOOD AND BLOOD DERIVATIVES
l. Skulls and spinal cord shall be removed from the Foetal bovine serum is commonly used in cel! cultures. Foetal
collected bones (raw/starting material) independent of the bovine serum should be obtained from foetuses harvested in
age or the country of origin of the cattle. abattoirs from healthy dams fit for human consumption and

Ba::.ed 011 the Opinion of the Scientific Panel on Bio!ogical Hazards of the European Fand Safety Authority on the 'Quantitative assessment of the human ESE risk pased by gelatine
respect to residual ESE risk', The EFSA Joumal, 312, (1-28). http://www.efsa.europa.eu/EFSAlefsa_locale-1178620í53812_1178620776107.htm
The requirements far source material selection and manufacture are appropriate fOl' oral or parcnteral gelatín far use in human and velerinary medicinal products.

598 See the informatian section on general monagraphs (ca ver pages)
EUROPEAN PHARMACOPOEIA 8.0 5.2.8. Minimising the risk of transmitting TSE vía medicinal products

the womb should be completely removed and the foetal blood Whenever a risk of cross-contamination ofblood with brain
harvested in dedicated space or are a by cardiac puncture into cannot be avoided at routine slaughtering in countries with
a dosed coHection system using aseptic technique. a controlled ESE risk (Category E), safety measures such
Newborn calf serum is obtained from calves under 20 days old as restriction of the age of the cattle and/or reduction of
and calf serum from animals under the age of 12 months. In infectious agents during manufacture have to be applied.
the case of donor bovine serum, given that it may be derived Age
from animals less than 36 months old, the TSE negative status
For countries with a controlled ESE risk (Category E), a
of the donor herd shall be well defined and documented.
precautionary age limit of 21 months shall apply for bovine
In all cases, serum shall be collected according to specified
blood or blood derivatives where no significant reduction of
protocols by personnel trained in these procedures to avoid
TSE agents can be assumed from manufacture. An age limit of
cross-contamination with higher risk tissues.
30 months is considered sufficient for blood derivatives where
For bovine blood and blood derivatives, documentation significant reduction of TSE agents can be demonstrated as
to demonstrate compliance with this chapter needs to be described below.
provided taking into account the provisions listed in sections 3
to 5. In addition, consideration should be given to the Reduction of TSE agents during manufacture
following. For blood derivatives, the capacity of the manufacturing
Traceability process to reduce/eliminate TSE agents should be estimated
from investigational studies. The estimation may be based on
Traceability to the slaughterhouse must be assured for each published data or in house data whenever it can be shown that
batch of serum or plasma. Slaughterhouses must have available such data is relevant to the specific manufacturing process.
lists of farms from which the animals are originated. lf serum If it cannot be conduded that the reduction capacity is
is produced from living animals, records must be available for comparable, it is recommended that manufacturers undertake
each serum batch which as sures the traceability to the farms. product-specific investigational studies. Investigations using
Geographical origin biochemical assay may be sufficient if there is scientific
Whilst tissue infectivity of BSE in cattle is more restricted evidence that this assay correlates with infectivity data.
than scrapie, as a precautionary measure bovine blood should General guidance for investigational studies on reduction
be sourced from Category A countries. Eovine blood from ofTSE agents has been outlined(29). Erain-derived spike
Category E countries is also acceptable provided that there is preparations are appropriate for studies investigating the risk
no risk for cross-contamination of blood with brain material from brain-contaminated blood.
from the slaughter of animal s over 21 months(26) of age. 6-4. TALLOW DERIVATIVES
Stunning methods Tallow is fat obtained from tissues including subcutaneous,
lf it is sampled from slaughtered animals, the method of abdominal and inter-muscular are as and bones. Tallow
slaughter is of importance to assure the safety of the material. used as the starting material for the manufacture of tallow
It has been demonstrated that stunning by captive boh stunner
derivatives shall be 'Category 3 material or equivalent', as
with or without pithing as weH as by pneumatic stunner, defined in Regulation (EC) No 1774/2002 of the European
especially if it injects air, can destroy the brain and disseminate Parliament and of the Council of 3 October 2002 laying down
brain material into the blood stream. Non-penetrative health rules concerning animal by-products not intended for
stunning is no more considered as an alternative to penetrative human consumption.
stunning because contamination of blood with brain material Tallow derivatives, such as glycerol and fatty acids,
has been demonstrated(27). Negligible risk can be expected manufactured from tallow by rigorous processes are thought
from electro-narcosis(28), but this even do es not provide strict unlikely to be infectious and they have been the subject of
safety because, when unsuccessful, animals may have to be specific consideration by CHMP and CVMP. For this reason,
additionally stunned. The stunning methods must therefore such materials manufactured under the conditions at least as
be described for the bovine blood collection process. rigorous as those given below shall be considered in

Table 5.2.8.-1. - Concept for acceptance of bovine blood/sera and derivatives


Product Foetal bovine Donor calf Adult bovine Calf serum Adult bovine Adult bovine Adult bovine Adult bovine
serum senllll donor serum serum I plasma serum I serum sentm
plasma I serum derivative derivative
derivative
Geographical origin Cal. A and B Cal. A and B Cal. A and B1 Cal. A and B Cal. A Cal. B Cal. A Cal. B
of cattle
Age of cattle unborn < 1 year < 36 months < 1 year Nolimit < 21 months 2 No hmit < 30 months

Slaughtering/cross- No risk of cross-contamination Risk of cross-contamination


contamination of blood
with CNS material
Demonstration of No No Yes 3
Prion reduction during
manufacture
1. When sourced in Category B countries, cattle should be from well-defined and documented herds.

2. A higher age may be allowed if cross-contamination ofblood with CNS material can be clearly ruled out (e.g. halal slaughter).

3. Demonstration of prion reduction may not be required if cross-contamination of blood with CNS material can be clearly ruled out (e.g. halal
slaughter).

(26) Opinion of the Scientific Panel on Biological Hazards on the assessment of the age limit in cattle far the removal of certain Specified Risk Materials (SRM). Question No
EFSA-Q-2004-146, adopted 011 28 April2005
(27) The tissue classificatiol1 tables are based upan the mast recent WHO Guidelines on Tissue Infectivity Distribution in Transmissible Spongiform Encephalopathies (2010)
http://www. who.int/bloodproducts/tablestissueinfectivity.pdf
(28) Report of the EFSA Working Group Oil BSE risk from dissemination ofbrain particles in bIood and carcass. Question No EFSA-Q-2003-112, adopted 011 21 October 2004,
http://www.efsa.europa.eu/en/sciencebiohaz/biohaz_opinions/opinioD_annexes/733.html
(29) Guideline on the investigatian of manufacturing process for plasma-derived medicinal products with regard to vCJD risk CPMP/B\,VP/5136/03.

General Notices (1) apply to al! monographs and other texts 599
5.2.8. Minimising the risk of transmitting TSE via medicinal products EUROPEAN PHARMACOPOEIA 8.0

compliance for this chapter, irrespective of the geographical 6-7. WOOL DERIVATIVES
origin and the nature of the tissues from which tallow Derivatives of wool and hair of ruminants, such as lanolin
derivatives are derived. Examples of rigorous processes are: and wool aleohols derived from hair shall be considered in
- trans-esterification or hydrolysis at not less than 200 oC for compliance with this chapter, provided the wool and hair are
not less than 20 min under pressure (glycerol, fatty acids sourced from live animals.
and fatty acid esters production), Wool derivatives produced from wool which is sourced from
- saponification with 12 M NaOH (glycerol and soap slaughtered animals declared "fit for human consumption"
production) : and the manufacturing process in relation to pH, temperature
- batch process: at not less than 95 oC for not les s than 3 h, and duration of treatment meets at least one of the stipulated
processing conditions listed below are unlikely to present any
- continuous process: at not less than 140 oc, under TSE risk and shall therefore be considered complíant with this
pressure for not less than 8 min, or equivalent, chapter.
- distillation at 200 oc. Treatment at pH ;:: 13 (initial; corresponding to a NaOH
Tallow derivatives manufactured according to these conditions concentration of at least 0.1 M NaOH) at 60 oC for at least
are unlikely to present any TSE risk and shall therefore be 1 h. This occurs normally during the reflux stage of the
considered compliant with this chapter. organic-alkaline treatment.
Tallow derivatives produced using other conditions must - Molecular distillation at ;:: 220 oC under reduced pressure.
demonstrate compliance with this chapter.
Wool derivatives produced using other conditions must
6-5. ANIMAL CHARCOAL demonstrate compliance with this chapter.
Animal charco al is prepared by carbonisation of animal 6-8. AMINO ACIDS
tissues, such as bones, using temperatures higher than 800 oc.
Amino acids can be obtained by hydrolysis of materials from
Unless otherwise justified, the starting material for the
various sources.
manufacture of animal charcoal shall be Category 3 material
or equivalent, as defined in Regulation (EC) No 1774/2002 of Unless otherwise justified, the starting material for the
the European Parliament and of the Council of 3 October 2002 manufacture of amino acids shall be 'Category 3 material or
laying down health rules concerning animal by-products equivalent: as defined in Regulation (EC) No 177412002 ofthe
not intended for human consumption. Irrespective of the European Parliament and of the Council of 3 October 2002
geographical origin and the nature of the tissue, for the laying down health rules concerning animal by-products not
purpose of regulatory compliance, animal charco al shall be intended for human consumption.
considered in compliance with this chapter. Amino acids prepared using the following processing
Charco al manufactured according to these conditions is conditions are unlikely to present any TSE risk and shall be
unlikely to present any TSE risk and shall therefore be considered compliant with this chapter:
considered compliant with this chapter. Charco al produced - amino acids produced from hides and skins by a process
using other conditions must demonstrate compliance with which il1volves exposure of the material to a pH of 1 to 2,
this chapter. followed by a pH of> 11, followed by heat treatment at
6-6. MILK AND MILK DERIVATIVES 140 oC for 30 min at 3 bar,
In the light of the current scientific knowledge and irrespective - the resuIting amino acids or peptides must be filtered after
of the geographical origin, bovine milI< is unlikely to present production, and
any risk of TSE contamination(30).
- analysis is performed using a validated and sensitive
Certain materials, including lactose, are extracted from whey, method to control any residual intact macromolecules,
the spent liquid from che ese production following coagulation. with an appropriate limit set.
Coagulation can involve the use of calf rennet, an extract from
abomasum, or rennet derived from other ruminants. The Amino acids prepared using other conditions must
CHMP/CVMP have performed a risl< assessment for lactose demonstrate compliance with this chapter.
and other whey derivatives produced using calf rennet and 6-9 PEPTONES
concluded that the TSE risk is negligible if the calf rennet is Peptones are partial hydrolysates of protein, achieved by
produced in accordance with the process described in the risk enzymic or acid digestion. They are used in microbiological
assessment report(31). The conclusion was endorsed by the culture media to support the nutritional requirements of
SS032) which has also performed an assessment of the TSE micro-organisms, which might be used as seed stocks or in
risk of rennet in general(33). industrial scale fermentations for the production of human
Bovine milk derivatives manufactured according to the and veterinary medicinal products, including vaccines. There
conditions described below are unlikely to present any TSE is considerable interest in the use of vegetable protein as an
risk and shall therefore be considered complíant with this alternative to animal sourced protein. However:
chapter. - where gelatin is used as the protein source material,
- The milk is sourced from healthy animals in the same reference is made to section 6-2 Gelatin, of this chapter,
conditions as milk collected for human consumption, and - where casein is used as the protein source material,
- no other ruminant materials, with the exception of calf reference is made to section 6-6 Milk and milk derivatives,
rennet, are used in the preparation of such derivatives (e.g. of this chapter,
pancreatic enzyme digests of casein). - where tissue of TSE-relevant animal species is the protein
Milk derivatives produced using other processes or rennet source material, the tissue must be sourced from animals
derived from other ruminant species must demonstrate fit for consumption (see section 3-2 Source animals, of this
compliance with this chapter. chapter) with a maximum age of 30 months old for cattle

(30) For milk and milk derivatives from small ruminants, pIease see EFSA opinion 011 Question No EFSA-Q-2008-310, adopted on 22 October 2008,
http://www.efsa.europa.eu!en!scdocs/scdoc!849.htm
(31) Committee for Medicinal Products for Human Use and its Biologics Working Party conducted a risk and regulatory assessment oflactose prepared using calfrennet. The risk
assessment included the source of the animals, the excision of the abomasums and the availability of well-defined quality assurance procedures. The quality of any milk replacers used as
feed for the animals from which abomasums are obtained is particularly important. The report can be found on http://www.ema.europa.eu/pdfs/human/press/pus/057102.pdf
(32) Provisional statement on the safety of calf-derived rennet for the manufacture oflactose, adopted by the SSC at its meeting of 4-5 April2002 (http://ec.europa.eu/
food/fs/se/sse/out255_en.pdO
(33) The SSC issued an opinion on the safety of animal rennet in regard to risks from animal TSE and BSE in particular, adopted at its meeting of 16 May 2002
(http://ec.europa.eulfoodlfs/ sel ssel out265 _en.pdO

600 See the information section on general monographs (caver pages)


EUROPEAN PHARMACOPOEIA 8.0 5.2.8. Minimising the l"isk of tnmsmitting TSE via medicinal products

from countries with a controlled BSE risk (Category B). Data entries are shown as follows:
The age of animals is of minimal concern for animals from + Presence of infectivity or Prp TSE
countries with a negligible BSE risk (Category A).
Absence of detectable infectivity or Prp TSE
Annex: major categories of infectivity NT Not tested

The tables below are taken from the WHO Guidelines on NA Not applicable
Tissue Infectivity Distribution in Transmissible Spongiform Uncertain interpretation
Encephalopathies (2010).
() Limited or preliminary data
[] Infectivity or Prp TSE data based exclusively on
bioassays in transgenic (Tg) mice over-expressing
the PrP-encoding gene or PRp TSE amplification
methods
Category IA: High -infectivity tissues
Tissue Cattle Sheep and goats Elk and deer
BSE Serapie CWD
Infectivity' PrPTSE Infeetivity' PrpTSE Infeetivity' PrpTSE

Brain + + + + + +

Spinal cord + + + NT +

Retina + NT NT + NT +

Optic nerve' + NT NT + NT +

Spinal ganglia + + + + NT +

Trigeminal ganglia + + NT + NT

Pituitary gland ' NT + + NT +

Dura lnater3 NT NT NT NT NT NT

Category lB: Lower-infectivity tissues


Tissue Caule Sheep and goats EJk and deer
BSE Scrapie CWD
Infectivity' PrP TSE Infectivity' PrpTSE InfectivityI PrpTSE

Peripheral nervous system

Peripheral nerves [+] + + + NT +

Autonomic ganglia·J NT + NT + NT +

Lymphoreticular tissues

Spleen + + NT +

Lymph nodes + + NT +

Tonsil + + + NT +

Nictitating membrane + [+] + NT +

Thymus NT + + NT

Alimentary traet S

Oesophagus NT [+] + NT +

Fore-stomach' (ruminants NT [+] + NT +


only)
Stol11ach/ abol11asum NT [+] + NT +

Duodenllm [+] + NT +

Jejllnllm7 + [+] + NT NT

I1eU111 7 + + + NT +

Appendix NA NA NA NA NA NA

Colon/caeCllm 7 + + NT +

Rectlll11 NT NT NT + NT +

Reproductive tisslles

Placenta3 NT + + NT

Ovari NT NT

NT NT

General Notices (1) apply to all monographs and other texts 601
5.2.8. Minimising the risk of tnmsmitting TSE via medicinal products EUROPEAN PHARMACOPOEIA 8.0

Tissue Cattle Sheep aud goals Elk aud deer


BSE Scra12ie CWD
Infectivity' PrpTSE Infectivity' PrPTSE Infectívity' PrPTSE

Other tissues

Mammary gland/udder' NT + NT NT

Skin 3,'O NT [+1 [+1

Adipose tissue NT NT NT [+1 NT

Heart/pericardium NT NT NT +

Lung NT NT +

Liver 3 NT + NT

Kidney',ll [+1 + NT +

Adrenal [+1 + + NT +

Pancreas 3 NT + NT NT +

Bone rnarrow 12 [+1 NT + NT NT

Skeletal muscle 13 l+l NT [+1 + [+]

Tongue'" NT [+1 + NT

Blood vessels NT NT NT

Nasal 111ucosa1 5 NT + + NT +

Salivary gland NT + NT

Cornea 16 NT NT NT NT NT NT

Body fluids, secretion and excretions

CSF NT + NT NT

Blood 17 + +

Saliva NT NT NT + H
Milk lS + [+1 NT NT

Urine 19 NT -[+1 [+1

Feces l9 NT NT -[+1 NT

Category lC: Tissues with no detectable infectivity

Tissue Caule Sheep and goats EIk ami deer


BSE Scrapie CWD

Infectivity' PrpTSE Infectivity' PrpTSE Illfectivity' PrPTSE

Reproductive tissues

Testis NT NT

Prostate/Epididymis/Seminal NT NT
vesicle
Semen NT NT NT

Placenta fluids NT NT NT NT NT

Foetus 2U NT NT (-)

Embryos20 NT NT NT NT

Musculo-skeletal tissues

Bone NT NT NT NT NT

Tendon NT NT NT NT NT

Olher tissues

Gingival tissues NT NT NT NI' NT NT

Dental pulp NT NT NI' NT NT NI'

Trachea NT NT NI' NT

Thyroid gland NI' NI' NI' NT

Body f1uids, secretions and excretions

Colostrum 21 (-) (1) NT NT NT

602 See the information section on general monographs (cover pages)


EUROPEAN PHARMACOPOEIA 8.0 5.2.8. Minimising the risk of transmitting TSE vía medicinal prodncts

Cord blood 2! NT NT NT NT NT
Sweat NT NT NT NT NT NT
Tears NT NT NT NT NT NT
Nasal mucus NT NT NT NT NT NT
Bile NT NT NT NT NT NT

l. Infectivity bioassays ofhuman tissues have been conducted in either primates or mice (or both), bioassays of cattle tissues have been conducted in
either cattle or mice (or both), and most bioassays of sheep and/or goat tissues have been conducted only in mice. In regard to sheep and goats not aU
results are consistent for both spedes. for example, two goats (but no sheep) have contracted BSE naturaUy [Eurosurveillance, 2005, Jeffrey et al.,
2006]. Similarly, most of the results described for CWD were derived from studies in deer, and findings may not be identical in elk or other cervids.
2. In experimental models of TSE, the optic nerve has been shown to be a route of neuroinvasion, and contains high titres of infectivity.

3. No experimental data about infectivity in pituitary gland or dura mater in humans with all forms ofhuman TSE have been reported, but cadaveric
dura mater patches, and growth hormone derived from cadaveric pituitaries have transmitted disease to hundreds of people and therefore must
be included in the category of high-risk tissues. Prp TSE was detected by immunoblot in the dura mater of a vCJD patient who died in the US
after an unusuaUy long incubation period (see also Table lB for other positive tissues: skin, kidney, liver, pancreas, ovary and uterus) [Notari et
al., 2010]. It must be mentioned that earlier studies ofnumerous cases examined in the UK reported aU ofthese tissues to be negative [Ironside
et al., 2002, Head et al., 2004].
4. In cattle, Prp TSE is reported to be inconsistently present in the enteric plexus in the distal ileum, but immunohistochemical examination of tissues
from a single 'fallen stock' case of BSE in Japan suggested (albeit equivocally) involvement of myenteric plexuses throughout the smal! and large
intestine [Kimura and Haritani, 2008].
5. In vCJD, PrP TSE is limited to gut-associated Iymphoid and nervous tissue (mucosa, muscle, and serosa are negative).

6. Ruminant fore stomachs (reticulum, rumen, and omasum) are widely consumed, as is the true stomach (abomasum). The abomasum of cattle
(and sometimes sheep) is also a source of rennet.
7. When a large BSE oral dose was used to infect cattle experimentally, infectivity was detected in the jejunum and the ileo-caecum junction in Tg
mice overexpressing PrP [courtesy of Dr M Groschup]. Prp TSE was detected at low incidence in Iymphoid tissue of ileum [Terry et al., 2003] and has
been detected at an even lower frequency in jejunallymphoid tissue of cattle similarly infected by the oral route [EFSA, 2009].
8. A single report of transmission of sporadic CJD infectivity from human placenta has never been confirmed and is considered improbable.
9. PrP TSE has been detected in scrapie-infected sheep with chronic mastitis, but not from infected sheep without mastitis [Ligios et al., 2005].

10. Studies in hamsters oral!y infected with scrapie revealed that Prp TSE deposition in skin was primarily located within smal! nerve fibres. Also,
apical skin 'velvet' from the antlers of CWD-infected deer is reported to contain PrpTSE and infectivity [Angers et al., 2009].
11. Prp TSE detected by immunocytochemistry in the renal pelvis of scrapie-infected sheep [Siso et al., 2006], and in Iymphoid follicles within
connective tissue adjacent to the renal pelvis in CWD-infected mule deer [Fox et al., 2006].
12. A single positive marrow in multiple transmission attempts from cattle orally dosed with BSE-infected brain [Wells et al., 1999, Wells el al.,
2005, Sohn et al., 2009].
13. Muscle homogenates have not transmitted disease to primates from humans with sporadic CJD, or to cattle from cattle with BSE. However,
intra-cerebral inoculation of a semitendinosus muscle homogenate (including nervous and lymphatic elements) from a single caw with clinical BSE
has transmitted disease to transgenic mice that overexpress PrP at arate indicative of trace levels of infectivity [Buschmann and Groschup, 2005].
Also, recent published and unpublished studies have reported the presence of PrP TSE in skeletalmuscle in experimental rodent models of scrapie and
vqD [Beekes et al., 2005], in experimental and natural scrapie infections of sheep and goats [Andreoletti et al., 2004], in sheep orally dosed with BSE
[Andreoletti, unpublished data], and in humans with sporadic, iatrogenic, and variant forms of CJD [Glatzel et al., 2003, Kovacs et al., 2004, Peden et
al., 2006]. Bioassays ofmuscle in transgenic mice expressing cervid PrP have documented infectivity in CWD-infected mule deer [Angers et al.,
2006], and experiments are underway to determine whether detectable Prp TSE in other forms of TSE is also associated with infectivity.
14. In cattle, bioassay of infectivity in the tongue was negative, but the presence of infectivity in palatine ton sil has raised concern about possible
infectivity in lingual tonsillar tissue at the base ofthe tonpue that may not be removed at slaughter [Wells et al., 2005, EFSA, 2008]. In sheep naturally
infected with scrapie, 7 of 10 animals had detectable PrP SE in the tongue [Casalone et al., 2005, Corona et al., 2006].
15. Limited chiefly to regions involved in olfactory sensory reception.

16. Because only one case of iatrogenic CJD has been certainly attributed to a corneal transplant among hundreds of thousands of recipients (one
additional case is considered probable, and another case only possible), cornea has been categorized as a lower-risk tissue, other anterior chamber
tissues (lens, aqueous humour, iris, conjunctiva) have been tested with a negative result both in vCJD and other human TSEs, and there is no
epidemiological evidence that they have been associated with iatrogenic disease transmission.
17. A wealth of data from studies of blood infectivity in experimental rodent models of TSE have been extended by recent studies documenting
infectivity in the blood of sheep with naturally occurring scrapie and in sheep transfused with blood from BSE-infected cattle [Houston et al., 2008],
of deer with naturally occurring CWD [Mathiason et al., 2006], and (from epidemiological observations) in the red cell fraction (which includes
significant amounts ofboth plasma and leukocytes) offour blood donors in the pre-clinical phase ofvCJD infections [reviewed in Brown, 2006,
Hewitt et al., 2006]. Plasma Factor VIII administration has also been potentially implicated in a subclinical case ofvCJD in a haemophilia patient
[Peden et al., 2010]. Blood has not been shown to transmit disease from humans with any form of 'classica!' TSE [Dorse~ et al., 2009], or from
cattle with BSE (including fetal calf blood). A number oflaboratories using new, highly sensitive methods to detect PrP TE are reporting success in a
variety of animal and human TSEs. However, several have experienced difficulty obtaining reproducible results in plasma, and it is not yet clear that
positive results imply a ¡otential for disease transmissibility, either because of false positives, or of 'true' positives that are due to sub-transmissible
concentratians ofPrp IS ·. Because ofthese considerations (and the fact that no data are yet available on blinded testing of specimens from natural!y
infected humans or animals) the expert group felt that it was still too early to evaluate the validity of these tests with sufficient confidence to permit
either a negative or positive conclusion.
18. Evidence that infectivity is not present in milk from BSE-infected bovines includes temporo-spatial epidemiologic observations failing to detect
maternal transmission to calves suckled for long periods, clinical observations of over a hundred calves suckled by infected cows that have not
developed BSE, and experimental observations that milk from infected cows reared to an age exceeding the minimum incubation period has not
transmitted disease when administered intra-cerebrally or orally to mice[Middleton and Barlow, 1993, Taylor et al., 1995]. AIso, PrP TSE has not been
detected in milk from cattle incubating BSE following experimental oral challenge [SEAC, 2005]. However, low levels (¡.tg to ng/L) of normal PrP have
been detected in milk from both animals and humans [Franscini et al., 2006]. PrP TSE has been detected in the mammary glands of scrapie-infected
sheep with chronic mastitis [Ligios et al., 2005], and very recently it has been reported that milk (which in some cases also contained colostrum) from
scrapie-infected sheep transmitted disease to healthy animals [Konold et al., 2008, Lacroux et al., 2008].

General Notices (1) apply to all monographs and other texts 603
5.2.9. Safety of batches of immunosera for veterinary use EUROPEAN PHARMACOPOEIA 8.0

19. A mixed inoculum of urine and faeces from naturally infected CWD deer did not transmit disease during an 18-month observation period
after inoculation ofhealthy deer with a heterozygous (96 G/S) PRNP genotype [Mathiason et al., 2006]. However, recent bioassays in Tg mi ce have
transmitted disease from both urine [Haley et al., 2009] and faeces [Tamgüney et al., 2009]. In addition, mice with lymphocytic nephritis that were
experimentally infected with scrapie shed both PrpTSE and infectivity in urine, when bioassayed in Tg mice [Seegeret al., 2005]. Very low levels
of infectivity have also been detected in the urine (and histologically normal kidneys) of hamsters experimentally infected with scrapie [Gregori
and Rohwer, 2007, Gonzalez-Romero et al., 2008]. Finally, in an experimental scrapie-hamster model, oral dosing resulted in inÍectious faeces
when bioassayed in Tg mice over-expressing PrP [Safar et al., 2008].
20. Embryos from BSE-aftected cattle have not transmitted disease to mice, but no infectivity measurements have been made on fetal calf tissues
other than blood (negative mOllse bioassay) [Fraser and Foster, 1994]. Calves born of dams that received embryos from BSE- affected cattle have
sllrvived for observations periods of up to seven years, and examination of the brains of both the unaffected dams and their offspring revealed no
spongiform encephalopathy or Prp LSE [Wrathall et al., 2002].
21. Early reports of transmission of sporadic CJD infectivity from human cord blood and colostrum have never been confirmed and are considered
improbable. A bioassay from a cow with BSE in transgenic mice over-expressing bovine PrP gave a negative result [Buschmann and Groschup, 2005],
and PrP TSE has not been detected in colostrum from cattle incubating BSE following experimental oral challenge [SEAC, 2005].

0412013:50209 other criteria are recorded, such as body temperature, body


mass, other performance measurements and food intake.
5.2.9. EVALUATION OF SAFETY OF Local reactions. As far as appropriate and possible, the size
and persistence of any local reaction (induding inciden ce of
EACH BATCH OF IMMUNOSERA FOR painful reactions) and the proportion of animals showing
VETERINARY USE local reactions are recorded.
Systemic reactions. Body temperature and, if appropriate,
Dennition of abnormal reactions. During development body l11ass are documented as general indicators of systemic
studies, the type and degree of reactions expected after effects of administration of the immunoserum. In addition,
administration of the immunoserum are defined in the light al! clinical signs are recorded.
of safety testing. This definition of normal or abnormallocal
and systemic reactions is then used as part of the operation Body temperature. For mammals, the studies include
procedure for the batch safety test to evaluate acceptable and measurement of body temperature during the observation
unacceptable reactions. period. The body temperatures are recorded beginning at least
3 days before administration of the immunoserum, at the time
Amount to be administered in the test. In the tests, 'dose' of administration, 4 h after and at suitable intervals. The body
means the quantity of the immunoserum to be recommended temperature before administration of the immunoserum has
for use and containing the titre or potency within the to be within the physiological range. At leasi for immunosera
limits specified for production batches. The amount to be where a significant increase in body temperature may be
administered in the test is usual!y defined in a number of expected or where an increase in body temperature is specified
doses. in an individual monograph, it is recommended to use the
Route of administration. The immunoserum is administered mean temperature of the days before administration of
by a recommended route. In principie, preference should be the immunoserum (e.g. day - 3 to day O) as the baseline
given to the application route with the higher possibility to temperature to have clear guidance for evaluation of the test.
detect reactions. Body mass and food intake. Where it is known to be a reliable
Target animal spedes and category of animals. Use and useful indicator of safety, for example in young growing
animals of the most sensitive species and of the minimum animals, the body mass is measured and documented shortly
age recommended for administration of the immunoserum, before administration of the immunoserum and during
unless otherwise justified and authorised. the observation period. The food intake is monitored and
documented as an indicator of the effect of administering the
Animal numbers. The number of animals to be used for the
immunoserum. In most cases, it will be sufficient to record the
test is prescribed in the general monograph Immunosera for
daily ration has been consumed ar partIy or wholly rejected
veterinary use (0030).
but, in some cases it may be necessary to record the actual
Ident.ification of animals. Unless otherwise justified and weight of food consumed, if this is a relevant indicator of the
authorised, al! animal s are marked in a suitable way to ensure safety of the immunoserum.
individual documentation of data for the whole observation
Clinicalsigns. Al! expected and unexpected clinical signs of a
period.
general nature are recorded, including changes in health status
Observation periodo Where objective criteria such as body and behaviour changes.
temperature are to be recorded as described below, the Scare sheets. The score sheets are prepared for each
animal s are examined and observed for at least 3 days prior to immunoserum in the light of expected signs. All parameters
administration of the immunoserum. Aftel' administration of and data are recorded in score sheets. The score sheets contain
the immunoserum, the animals are observed and examined at general parameters but are also adapted for each kind of
least once every day for a period of at least 14 days fol' signs of immunoserum to list clinical signs that might be more evident
local and systemic reactions. On the day of administration of for a given immunoserum.
the immunoserum, at least 1 additional inspection is necessary
after 4 h ol' at intervals as specified in the monograph. Where Criteria for repeating the test. If an abnormal sign
there is a 2nd administration of the immunoserum the period occurs, the responsible veterinarian determines, based on
usual!yends 14 days after the 2nd administration. post-mortel11 exal11ination if necessary, whether this was due
to the immunoserum or not. If it is not clear what caused the
Local and systemic reactions. Animals showing severe abnormal sign or where an animal is withdrawn for reasons
abnormallocal or systemic reactions are euthanised. Al! unrelated to the immunoserum, the test may be repeated.
dead animals undergo a post-mortem with macroscopic If in the 2nd test there is the same abnormal sign as in the
examination. Additional microscopic and microbiological 1st test, the immunoserum do es not comply with the test. Any
investigations may be indicated. treatment administered to an animal during the observation
The animals are observed and examined for signs of local and period is recorded. If the treatment may interfere with the
systemic reactions. Where it is known to be a useful indicator, test, the test is invalido

604 See the information section on general monographs (ca ver pages)
EUROPEAN PHARMACOPOEIA 8.0

5.3. Statistical analysis of results of


biological assays and tests
5.3. Statistical analysis of results of biological assays and 5. Examples" "."."."".""""".".".""""."".".""."""."".""."" ... 618
tests"". """ "". """ "" " .. "". " ... "" "" """ "". """ " .. ",," .. " .... ". "". 607 6. Combination of assay results" """"".""."".""""""""""".628
l. Introduction" ".""""""."."""." .. """" ... ""." .. """"." .. """".607 7. Beyond this annex" """"""".""""."""""""""""""""""".,630
2. Randomisation and independence of individual 8. Tables and generating procedures".".""""""""""".".".". 631
treatments ........................................... " ....... " .......................... 607 9. Glossary of symbols" "."""""."" ..... ".""""".""""""""",,,.634
3. Assays depending upon quantitative responses" """.""" 608 10. Literature" '"'''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''''' 635
4. Assays depending upon quantal responses" """"".""".".615

General Natices (1) apply ta al/ managraphs and ather texts 605
EUROPEAN PHARMACOPOEIA 8.0

606 See the information section on general monographs (cover pages)


EUROPEAN PHARMACOPOEIA 8.0 5.3. Statistical analysis

01/2008:50300 design and the sample size. The 95 per cent confidence
interval is usually chosen in biological assays. Mathematical
statistical methods are used to calculate these limits so as to
warrant the statement that there is a 95 per cent probability
5.3. STATISTICAL ANALYSIS that these limits indude the true potency. Whether this
OF RESULTS OF BIOLOGICAL precision is acceptable to the European Pharmacopoeia
depends on the requirements set in the monograph for the
ASSAYS AND TESTS preparation concerned.
The terms "mean" and "standard deviation" are used here as
defined in most current textbooks of biometry.
The terms "stated potency" or "labelled potency': "assigned
potency", "assumed potency", "potency ratio" and "estimated
1. INTRODUCTION potency" are used in this section to indicate the following
concepts:
This chapter provides guidance for the design of bioassays - "stated potency" or "labelled potency": in the case of
prescribed in the European Pharmacopoeia (Ph. Eur.) and for a formulated product a nominal value assigned from
analysis of their results. It is intended for use by those whose knowledge of the potency of the bulk material; in the case of
primary training and responsibilities are not in statistics, but bulk material the potency estimated by the manufacturer;
who have responsibility for analysis or interpretation of the
results of these assays, often without the help and advice of - "assigned potency": the potency of the standard
a statistician. The methods of calculation described in this preparation;
annex are not mandatory for the bioassays which themselves - "assumed potency": the provisionally assigned potency
constitute a mandatory part of the Ph. Eur. Alternative of a preparation to be examined which forms the basis of
methods can be used and may be accepted by the competent calculating the doses that would be equipotent with the
authorities, provided that they are supported by relevant data doses to be used of the standard preparation;
and justified during the assay validation process. A wide range
of computer software is available and may be useful depending - "potency ratio" of an unknown preparation; the ratio of
on the facilities available to, and the expertise of, the analyst. equipotent doses of the standard preparation and the
unknown preparation under the conditions of the assay;
Professional advice should be obtained in situations where: a
comprehensive treatment of design and analysis suitable for - "estimated potency" : the potency calculated from assay
research or development of new products is required; the data.
restrictions imposed on the assay design by this chapter are Section 9 (Glossary of symbols) is a tabulation of the more
not satisfied, for example particular laboratory constraints important uses of symbols throughout this annex. Where
may require customized assay designs, or equal numbers of the text refers to a symbol not shown in this section or uses
equally spaced doses may not be suitable; analysis is required a symbol to denote a different concept, this is defined in that
for extended non-linear dose-response curves, for example as part of the text.
may be encountered in immunoassays. An outline of extended
dose-response curve analysis for one widely used model is
nevertheless included in Section 3.4 and a simple example is
given in Sectiol1 5.4.
2. RANDOMISATION AND
1.1. GENERAL DESIGN AND PRECISION
INDEPENDENCE OF INDIVIDUAL
Biological methods are described for the assay of certain
substances and preparations whose potency cannot be TREATMENTS
adequately assured by chemical or physical analysis. The
principIe applied wherever possible throughout these assays The allocation of the different treatments to different
is that of comparison with a standard preparation so as experimental units (animals, tubes, etc.) should be made
to determine how much of the substance to be examined by sorne strictly random process. Any other choice of
produces the same biological effect as a given quantity, the experimental conditions that is not deliberately allowed for
Unit, of the standard preparation. It is an essential condition in the experimental design should also be made randomly.
of such methods of biological assay that the tests on the Examples are the choice of positions for cages in a laboratory
standard preparation and on the substance to be examined be and the order in which treatments are administered. In
carried out at the same time and under identical conditions. particular, a group of animal s receiving the same dos e of any
preparation should not be treated together (at the same time or
For certain assays (determination of virus titre for example) in the same position) unless there is strong evidence that the
the potency of the test sample is not expressed relative to a relevant source of variation (for example, between times, or
standard. This type of assay is dealt with in Section 4.5. between positions) is negligible. Random allocations may be
obtained from computers by using the built-in randomisation
Any estimate of potency derived from a biological assay is
functiol1. The analyst must check whether a different series of
subject to random error due to the inherent variability of
numbers is produced every time the function is started.
biological responses and calculations of error should be
made, if possible, from the results of each assay, even when The preparations allocated to each experimental unit should
the official method of assay is llsed. Methods for the design be as independent as possible. Within each experimental
of assays and the calculation of their errors are, therefore, group, the dilutions allocated to each treatment are not
described below. In every case, before a statistical method normally divisions of the same dose, but should be prepared
is adopted, a preliminary test is to be carried out with an individually. Without this precaution, the variability
appropriate number of assays, in order to ascertain the inherent in the preparation will not be fully represented
applicability of this method. in the experimental error variance. The result will be an
under-estimation of the residual error leading to:
The confidence interval for the potency gives an indication of
the precision with which the potency has been estimated in 1) an unjustified in crease in the stringency of the test for the
the assay. It is calculated with due regard to tbe experimental analysis of variance (see Sections 3.2.3 and 3.2.4),

General Notices (1) apply to all monographs and other texts 607
5.3. Statistical analysis EUROPEAN PHARMACOPOElA 8.0

2) an under-estimation of the true confidence limits for the - The square transformation of y to y2 can be useful if, for
test, which, as shown in Section 3.2.5, are calculated from the example, the dos e is more likely to be proportional to
estímate of 52, the residual error mean square. the area of an inhibition zone rather than the measured
diameter of that zone.
For sorne assays depending on quantitative responses, such as
3. ASSAYS DEPENDING UPON immunoassays or cell-based in vitro assays, a large number
of doses is used. These doses give responses that completely
QUANTITATIVE RESPONSES span the possible response range and produce an extended
non-linear dose-response curve. Such curves are typical for
3.1. STATISTICAL MODELS
all bioassays, but for many assays the use of a large number of
3.1.1. GENERAL PRINCIPLES doses is not ethical (for example, in vivo assays) or practical,
The bioassays induded in the Ph. Eur. have been conceived as and the aims of the assay may be achieved with a limited
"dilution assays", which means that the unknown preparation number of doses. It is therefore customary to restrict doses
to be assayed is supposed to contain the same active to that part of the dose-response range which is linear under
principie as the standard preparation, but in a different suitable transformation, so that the methods of Sections 3.2
ratio of active and inert components. In such a case the or 3.3 apply. However, in sorne cases analysis of extended
unknown preparation may in theory be derived from the dose-response curves may be desirable. An outline of one
standard preparation by dilution with inert components. To model which may be used for such analysis is given in
check whether any particular assay may be regarded as a Section 3.4 and a simple example is shown in Section 5.4.
dilution assay, it is necessary to compare the dose-response There is another category of assays in which the response
relationships of the standard and unknown preparations. If cannot be measured in each experimental unit, but in which
these dose-response relationships differ significantly, then only the fraction of units responding to each treatment can be
the theoretical dilution assay model is not valido Significant counted. This category is dealt with in Section 4.
differences in the dose-response relationships for the standard
3.1.2. ROUTINE ASSAYS
and unknown preparations may suggest that one of the
preparations contains, in addition to the active principIe, When an assay ís in routine use, it is seldom possible to
other components which are not inert but which influence check systematically for conditions 1 to 3, because the limited
the measured responses. number of observations per assay is likely to influence the
sensitivity of the statistical tests. Fortunately, statisticians have
To make the effect of dilution in the theoretical model shown that, in symmetrical balanced assays, small deviations
apparent, it is useful to transform the dose-response from homogeneity of variance and normality do not seriously
relationship to a linear function on the widest possible range affect the assay results. The applicability of the statistical
of doses. 2 statistical models are of interest as models for model needs to be questioned only if a series of assays shows
the bioassays prescribed: the parallel-line model and the doubtful validity. It may then be necessary to perform a
slope-ratio model. new series of preliminary investigations as discussed in
The application of either is dependent on the fulfilment of the Section 3.1.1.
following conditions:
2 other necessary conditions depend on the statistical model
1) the different treatments have been randomly assigned to to be used:
the experimental units,
- for the parallel-line model:
2) the responses to each treatment are normally distributed,
4A) the relationship between the logarithm of the dose and
3) the standard deviations of the responses within each
the response can be represented by a straight line over the
treatment group of both standard and unknown preparations
range of doses used,
do not differ significantly from one another.
When an assay is being developed for use, the analyst has to 5A) for any unknown preparation in the assay the straight
determine that the data collected from many assays meet these line is parallel to that for the standard.
theoretical conditions. - for the slope-ratio model:
- Condition 1 can be fulfilled by an efficient use of Section 2. 4B) the relationship between the dose and the response can
- Condition 2 is an assumption which in practice is almost be represented by a straight line for each preparation in the
always fulfilled. Minor deviations from this assumption assay over the range of doses used,
will in general not introduce serious flaws in the analysis 5B) for any unknown preparation in the assay the straight
as long as several replicates per treatment are included. In line intersects the y-axis (at zero dose) at the same point
case of doubt, a test for deviations from normality (e.g. the as the straight line of the standard preparation (Le. the
Shapiro-Wilk(J) test) may be performed. response functions of all preparations in the assay must
- Condition 3 can be checked with a test for homogeneity of have the same intercept as the response function of the
variances (e.g. Bartlett's(2) test, Cochran's(3) test). Inspection standard).
of graphical representations of the data can also be very Conditions 4A and 4B can be verified only in assays in which
instructive for this purpose (see examples in Section 5). at least 3 dilutions of each preparation have been tested. The
When conditions 2 and/or 3 are not met, a transformation of use of an assay with only 1 or 2 dilutions may be justified when
the responses may bring a better fulfilment of these conditions. experience has shown that linearity and parallelism or equal
Examples are In y, Vfj, y2. intercept are regularly fulfilled.
- Logarithmic transformation of the responses y to In y After having collected the results of an assay, and before
can be useful when the homogeneity of variances is not calculating the relative potency of each test sample, an
satisfactory. It can also improve the normality if the analysis of variance is performed, in order to check whether
distribution is skewed to the right. conditions 4A and 5A (or 4B and 5B) are fulfilled. For this,
- The transformation of y to Vfj is useful when the the total sum of squares is subdivided into a certain number
observations follow a Poisson distribution i.e. when they of sum of squares corresponding to each condition which has
are obtained by counting. to be fulfilled. The remaining sum of squares represents the

(1) Wilk, lV1.B. and Shapiro, s.s. (1968). The joint assessment of normality of several independent samples, Technometrics 10, 825-839.
(2) Bartlett, M.S. (1937). Properties of sufficiency and statistical tests, Prac. Roy. Soc. Londan, Series A 160, 280-282.
(3) Cochran, \tV.G. (1951). Testing a linear relatian among variances, Biomefrics 7, 17-32.

608 See the information section on general monographs (ca ver pages)
EUROPEAN PHARMACOPOEIA 8.0 5.3. Statistkal analysis

residual experimental error to which the absence or existence b) In the parallel-line model, the ratio of adjacent doses must
of the relevant sources of variation can be compared by a be constant for all treatments in the assay; in the slope-ratio
series of F-ratios. model, the interval between adjacent doses must be constant
When validity is established, the potency of each unknown for al! treatments in the assay.
relative to the standard may be calculated and expressed as e) There must be a11 equal number of experimental units to
a potency ratio or converted to some unit relevant to the each treatment.
preparation under test e.g. an International Unit. Confidence If a design is used which meets these restrictions, the
limits may also be estimated from each set of assay data. calculations are simple. The formulae are given in Sections 3.2
Assays based on the parallel-line model are discussed in and 3.3. It is recommended to use software which has been
Section 3.2 and those based on the slope-ratio model in developed for this special purpose. There are several programs
Section 3.3. in existen ce which can easily deal with all assay-designs
lf any of the 5 conditions (1, 2, 3, 4A, SA or 1, 2, 3, 4B, SB) described in the monographs. Not all programs may use the
are not fulfilled, the methods of calculation described here same formulae and algorithms, but they should alllead to the
are invalid and an investigation of the assay technique should same results.
be made. Assay designs not meeting the aboye mentioned restrictions
The analyst should not adopt another transformation unless may be both possible and correct, but the necessary formulae
it is shown that non -fulfilment of the requirements is not are too complicated to describe in this text. A brief description
incidental but is due to a systematic change in the experimental of methods for calculation is given in Section 7.1. These
conditions. In this case, testing as described in Section 3.1.1 methods can also be used for the restricted designs, in which
should be repeated before a new transformation is adopted case they are equivalent with the simple formulae.
for the routine assays. The formulae for the restricted designs given in this text
Excess numbers of invalid assays due to non-parallelism may be used, for example, to create ad hoc programs in a
or non-linearity, in a routine assay carried out to compare spreadsheet. The examples in Section 5 can be used to clarify
similar materials, are likely to reflect assay designs with the statistics and to check whether such a program gives
inadequate replication. This inadequacy commonly results correct results.
from in complete recognition of aH sources of variability
affecting the assay, which can result in underestimation of the 3.2. THE PARALLEL-LINE MODEL
residual error leading to large F-ratios. 3.2.1.INTRODUCTION
It is not always feasible to take account of all possible sources The parallel-line model is illustrated in Figure 3.2.1.-I. The
ofvariation within one single assay (e.g. day-to-dayvariation). logarithm of the doses are represented on the horizontal axis
In such a case, the confidence intervals from repeated assays with the lowest concentration on the left and the highest
on the same sample may not satisfactorily overlap, and care concentration on the right. The responses are indicated on
should be exercised in the interpretation of the individual the vertical axis. The individual responses to each treatment
confidence intervals. In order to obtain a more reliable are indicated with black dots. The 2 lines are the calculated
estimate of the confidence interval it may be necessary to ln(dose)-response relationship for the standard and the
perform several independent assays and to combine these unknown.
into one single potency estimate and confidence interval (see
Section 6).
For the purpose of quality control of routine assays it is
recommended to keep record of the estimates of the slope of
regression and of the estimate of the residual error in control • Standard
e
charts.
An exceptionally high residual error may indicate some • Test
technical problem. This should be investigated and, $sample
if it can be made evident that something went wrong
during the assay procedure, the assay should be repeated.
An unusually high residual error may also indicate the
presence of an occasional outlying or aberrant observation.
A response that is questionable because of failure to
comply with the procedure during the course of an assay
is rejected. If an aberrant value is discovered after the
responses have been recorded, but can then be traced
to assay irregularities, omission may be justified. The
arbitrary rejection or retention of an apparently aberrant
response can be a serious source of bias. In general, the
rejection of observations solely because a test for outliers is
significant, is discouraged.
- An exceptionally low residual error may once in a while In dose (x)
occur and cause the F-ratios to exceed the critical values.
In such a case it may be justified to replace the residual Figure 3.2.1.-1. - The parallel-line model for a 3 + 3 assay
error estimated from the individual assay, by an average Note: the naturallogarithm (In or log,) is used throughout
residual error based on historical data recorded in the this text. Wherever the tenn "antilogarithm" is used, the
control charts. quantity eX is meant. However, the Briggs or "common"
3.1.3. CALCULATIONS AND RESTRICTIONS logarithm (log or log¡o) can equally well be used. In this case
According to general principIes of good design the following the corresponding antilogarithm is lQx.
3 restrictions are normally imposed on the assay designo For a satisfactory assay the assumed potency of the test sample
They have advantages both for ease of computation and for must be close to the true potency. On the basis of this assumed
precision. potency and the assigned potency of the standard, equipotent
a) Each preparation in the assay must be tested with the same dilutions (iffeasible) are prepared, i.e. corresponding doses
number of dilutions. of standard and unknown are expected to give the same

General Notices (1) apply to all monographs and other texts 609
5.3. Stadstical analysis EUROPEAN PHARMACOPOElA 8.0

response. If no information on the assumed potency is 3.2.2.4. Cross-over design


available, preliminar y assays are carried out over a wide range This design is useful when the experiment can be sub-divided
of doses to determine the range where the curve is linear. into blocks but it is possible to apply only 2 treatments to
The more nearly correct the assumed potency of the unknown, each block. Por example, a block may be a single unit that can
the closer the 2lines will be together, for they should give equal be tested on 2 occasions. The design is intended to increase
responses at equal doses. The horizontal distance between the precision by eliminating the effects of differences between
lines represents the "true" potency of the unknown, relative to units while balancing the effect of any difference between
its assumed potency. The greater the distance between the 2 generallevels of response at the 2 occasions. If 2 doses of a
lines, the poorer the assumed potency of the unknown. If the standard and of an unknown preparation are tested, this is
line of the unknown is situated to the right of the standard, known as a twin cross-over test.
the assumed potency was overestimated, and the calculations The experiment is divided into 2 parts separated by a suitable
will indicate an estimated potency lower than the assumed time interval. Units are divided into 4 groups and each group
potency. Similarly, if the line of the unknown is situated to the receives 1 of the 4 treatments in the first part of the test.
left of the standard, the assumed potency was underestimated, Units that received one preparation in the first part of the
and the calculations will indicate an estimated potency higher test receive the other preparation on the second occasion,
than the assumed potency. and units receiving small doses in one part of the test receive
3.2.2. ASSA y DESIGN large doses in the other. The arrangement of doses is shown in
The fol!owing considerations will be useful in optimising the Table 3.2.2.-1. An example can be found in Section 5.1.5.
precision of the assay design:
Table 3.2.2.-1. - Arrangement of doses in cross-over design
1) the ratio between the slope and the residual error should
be as large as possible, Group of units Time 1 Time n
2) the range of doses should be as large as possible,
S] T2
3) the lines should be as close together as possible, i.e. the
assumed potency should be a good estimate of the true 2 S, T]
potency.
3 T] S2
The allocation of experimental units (animals, tubes, etc.) to
different treatments may be made in various ways. 4 T2 S]
3.2.2.1. Completely randomised design
3.2.3. ANALYSIS OF VARIANCE
If the totality of experimental units appears to be reasonably
This section gives formulae that are required to carry out the
homogeneous with no indication that variability in response
analysis of variance and will be more easily understood by
will be smaller within certain recognisable sub-groups, the
reference to the worked examples in Section 5.1. Reference
allocation of the units to the different treatments should be
should also be made to the glossary of symbols (Section 9).
made randomly.
Jf units in sub-groups such as physical positions or The formulae are appropriate for symmetrical assays where
experimental days are likely to be more homogeneous than the one or more preparations to be examined (T, U, etc.) are
totality of the units, the precision of the assay may be increased compared with a standard preparation (S). It is stressed that
by introducing one or more restrictions into the designo A the formulae can only be used if the doses are equally spaced, if
careful distribution of the units over these restrictions permits equal numbers of treatments per preparation are applied, and
irrelevant sources of variation to be eliminated. each treatment is applied an equal number of times. It should
not be attempted to use the formulae in any other situation.
3.2.2.2. Randomised block design
Apart from some adjustments to the error term, the basic
In this design it is possible to segregate an identifiable source analysis of data derived from an assay is the same for
of variation, 5uch as the sensitivity variation between litters of completely randomised, randomised block and Latin square
experimental animals or the variation between Petri dishes designs. The formulae for cross-over tests do not entirely fit
in a diffusion microbiological assay. The design requires this scheme and these are incorporated into Example 5.1.5.
that every treatment be applied an equal number of times in
every block (Iitter or Petri dish) and is suitable only when the Having considered the points discussed in Section 3.1 and
block is large enough to accommodate all treatments once. transformed the responses, if necessary, the values should
This is illustrated in Section 5.1.3. It is also possible to use a be averaged over each treatment and each preparation, as
randomised design with repetitions. The treatments should be shown in Table 3.2.3.-1. The linear contrasts, which relate
allocated randomly within each block. An algorithm to obtain to the slopes of the In(dose)-response lines, should also
random permutations is given in Section 8.5. be formed. 3 additional formulae, which are necessary for
the construction of the analysis of variance, are shown in
3.2.2.3. Latin square design Table 3.2.3.-II.
This design is appropriate when the response may be affected The total variation in response caused by the different
by two different sources of variation each of which can as sume treatments is now partitioned as shown in Table 3.2.3.-III
k different levels or positions. Por example, in a plate assay the sums of squares being derived from the values obtained
of an antibiotic the treatments may be arranged in a k x k in Tables 3.2.3.-1 and 3.2.3.-1I. The sum of squares due to
array on a large plate, each treatment occurring once in 110n -linearity can only be calculated if at least 3 doses per
each row and each column. The design is suitable when the preparation are included in the assay.
number of rows, the number of columns and the number
of treatments are equal. Responses are recorded in a square The residual error of the assay is obtained by subtracting the
format known as a Latin square. Variations due to differences variations allowed for in the design from the total variation
in response among the k rows and among the k columns may in response (Table 3.2.3.-IV). In this table fj represents the
be segregated, thus reducing the error. An example of a Latin mean of al! responses recorded in the assay. It should be noted
square design is given in Section 5.1.2. An algorithm to obtain that for a Latin square the number of replicate responses (n) is
Latin squares is given in Section 8.6. More complex designs in equal to the number of rows, columns or treatments (dh).
which one or more treatments are replicated within the Latin The analysis of variance is now completed as follows. Each
square may be useful in some circumstances. The simplified sum of squares is divided by the corresponding number of
formulae given in this Chapter are not appropriate for such degrees of freedom to give mean squares. The mean square
designs, and professional advice should be obtained. for each variable to be tested is now expressed as a ratio to the

610 See the ínformation sectíon on general monographs (cover pages)


EUROPEAN PHARMACOPOEIA 8.0 5.3. Statistical analysis

Table 3.2.3.-1. - Formulae for parallel-line assays with d doses of each preparation
Standard 1" Test sample 2nd Test sample
(S) (1) (U, etc)

Mean response lowest


51 T1 U1
dose

Mean response 2nd dose 52 T2 U2

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

Mean response highest


5d Td Ud
dose

Total preparation Ps = SI + S2 + ... + Sd PT = TI + T 2 + ." + T d Pu = ... etc.

~
Linear contras! 1
Ls = lS 1 + 2S2 + ... + dSd - (d + 1) Ps LT = lTl + 2T2 + ... + dTd - 2 (d + 1) PT Lu = ... etc.
2

Table 3.2.3.-II. - Additional formulae for the construction of the analysis of variance
11. 11.(PS+PT+ ... )2
K=
d hd

Table 3.2.3.-III. - Formulae to calculate the sum of squares and degrees offreedom
Source of variation Degrees of freedom (j) Sum of squares

Preparations h - 1 SSprep = Hp (pJ + P~ + ... ) - K

Linear regression 1 1
SSreg = ¡;HL (L s + LT +

Non-parallelism h - 1 SSpar = HL (L~ + L~ + ... ) - SSreg

Non-linearity* h (d - 2) SSlin = SStreat - SSprep - SSreg - SSpar

Trealments hd - 1 SS'reat = 11. (Si + . . + S~ + Ti + ... + TJ + ... ) -K

* Not calculated for two-dose assays

Table 3.2.3.-IV. - Estimation of the residual error


Source of variation Degrees of freedom Sum of squares

Blocks (rows)* 11. - 1 SSblock = hd (Ri + ... + R~) - K

Columns** 1 (e; + ... + e;) -

¡
11. -
SScol = hd K

Completely randomised hd (11. - 1) SSres = SStot - SStreat

Residual error*** Randomised block (hd - 1) (11. - 1) SSres = SStot - SStreat - SSblock

Latin square (hd - 2) (11. - 1) S Sres = S Stot - S Streat - S Sblock - S Seo!

Total nhd - 1 SS'ot =¿ (y _ )1)2

For Latin square designs, lhese formulae are only applicable if n = hd


* Not calculated for completely randomised designs
** Only calculated for Latin square designs
*** Depends 011 the type of desigl1

residual error (S2) and the significance of these values (known 3) The term for non-linearity is not significant, i.e. the
as F-ratios) are assessed by use of Table 8.1 or a suitable calculated probability is not less than 0.05. This indicates that
sub-routine of a computer programo condition 4A, Section 3.1, is satisfied.
3.2.4. TESTS OF VALIDITY A significant deviation from parallelism in a multiple assay
Assay results are said to be "statistically valid" if the outcome may be due to the inclusion in the assay-design of a preparation
of the analysis of variance is as follows. to be examined that gives an ln(dose)-response line with a
slope different from those for the other preparations. Instead
1) The linear regression term is significant, i.e. the calculated of declaring the whole assay invalid, it may then be decided
probability is less than 0.05. If this criterio n is not met, it is to eliminate all data relating to that preparation and to restart
not possible to calculate 95 per cent confidence limits. the analysis from the beginning.
2) The term for non-parallelism is not significant, i.e. the When statistical validity is established, potencies and
calculated probability is not less than 0.05. This indicates that confidence limits may be estimated by the methods described
condition 5A, Section 3.1, is satisfied; in the next section.

General Notices (1) apply to all monographs and other texts 611
5.3. Statistical analysis EUROPEAN PHARMACOPOElA 8.0

3.2.5. ESTIMA TION OF POTENCY AND CONFIDENCE Completely randomised design


LIMITS In a completely randomised assay the missing value can be
rf 1 is the In of the ratio between adjacent doses of any replaced by the arithmetic mean of the other responses to the
preparation, the common slope (b) for assays with d doses of same treatment.
each preparation is obtained from: Randomísed block desígn
b = HL(Ls + LT + ... ) (3.2.5.-1) The missing value is obtained using the equation:
Inh , nB' + kT' - G' (3.2.6.-1)
Y = (n-l)(k-l)
and the logarithm of the potency ratio of a test preparation,
for example T, is: where B' is the sum of the responses in the block containing
the missing value, T' the corresponding treatment total and G'
filI' = PT - Ps (3.2.5.-2)
is the sum of all responses recorded in the assay.
T db
Latín square design
The calculated potency is an estimate of the "true potency" of The missing value y' is obtained from:
each unknown. Confidence limits may be calculated as the
antilogarithms of: k (B' + e' + T') - 2G' (3.2.6.-2)
y' =
(k - 1) (k - 2)
eM!r ± J(e - 1) (eM:; + 2V) (3.2.5.-3)
where B' and C' are the sums of the responses in the row and
SSreg column containing the missing value. In this case k = n.
h
w ere e = ss 2 2
reg - S t Cross-aver desígn
lf an accident leading to loss of values occurs in a cross-over
The value of t may be obtained from Table 8.2 for p = 0.05 design, a book on statistics should be consulted (e.g. D.].
and degrees of freedom equal to the number of the degrees of Finney, see Section 10), because the appropriate formulae
freedom of the residual error. The estimated potency (R T ) and depend upon the particular treatment combinations.
associated confidence limits are obtained by multiplying the 3.3. THE SLOPE-RATIO MODEL
values obtained by Al' after antilogarithms have been taken.
If the stock solutions are not exactly equipotent on the basis 3.3.1.INTRODUCTION
of assigned and assumed potencies, a correction factor is This model is suitable, for example, for some microbiological
necessary (See Examples 5.1.2 and 5.1.3). assays when the independent variable is the concentration of
an essential growth factor below the optimal concentration
3.2.6. MISSING VALUES of the medium. The slope-ratio model is illustrated in
In a balanced assay, an accident totally unconnected with Figure 3.3.1.-1.
the applied treatments may lead to the 10s5 of one or more
responses, for example because an animal dies. lf it is ®
considered that the accident is in no way connected with Standard
the composition of the preparation administered, the exact
calculations can still be performed but the formulae are
necessarily more complicated and can only be given within
the framework of general linear models (see Section 7.1).
However, there exists an approximate method which keeps
the simplicity of the balanced design by replacing the missing
response by a calculated value. The loss of information is G Test
taken into account by diminishing the degrees of freedom " Sample
for the total sum of squares and for the residual error by the
number of missing values and using one of the formulae below
.
for the missing values. It should be borne in mind that this is .
only an approximate method, and that the exact method is to .
be preferred.
"
lf more than one observation is missing, the same formulae
can be used. The procedure is to make a rough guess at al! the
missing values except one, and to use the proper formula for
this one, using all the remaining values including the rough
guesses. FiIl in the calculated value. Continue by similarly
dose x)
calculating a value for the first rough guess. After calculating
al! the missing values in this way the whole cycle is repeated Figure 3.3.1.-I. - The slape-ratío model for a 2 x 3 + 1 assay
from the beginning, each calculation using the most recent The doses are represented on the horizontal axis with zero
guessed or calculated value for every response to which the concentration on the left and the highest concentration on the
formula is being applied. This continues until 2 consecutive
right. The responses are indicated on the vertical axis. The
cycles give the same values; convergence is usually rapid. individual responses to each treatment are indicated with black
Provided that the number of values replaced is small relative dots. The 2 lines are the calculated dose-response relationship
to the total number of observations in the full experiment for the standard and the unknown under the assumption that
(say less than 5 per cent), the approximation implied in they intersect each other at zero-dose. Unlike the parallel-line
this replacement and reduction of degrees of freedom by model, the doses are not transformed to logarithms.
the number of missing values so replaced is usually fairly Just as in the case of an assay based on the parallel-line
satisfactory. The analysis should be interpreted with great care model, it is important that the assumed potency is close to
however, especially if there is a preponderance of missing the true potency, and to prepare equipotent dilutions of the
values in one treatment or block, and a biometrician should be test preparations and the standard (if feasible). The more
consulted if any unusual features are encountered. Replacing nearly correct the assumed potency, the closer the 2 lines
missing values in a test without replication is a particularly will be together. The ratio of the slopes represents the "true"
delicate operation. potency of the unknown, relative to its assumed potency. If

612 See the informatian sectían on general monographs (caver pages)


EUROPEAN PHARMACOPOEIA 8.0 5.3. Statistkal analysis

the slope of the unknown preparation is steeper than that A completely randomised, a randomised block or a Latin
of the standard, the potency was underestimated and the square design may be used, such as described in Section 3.2.2.
calculations will indicate an estimated potency higher than the The use of any of these designs necessitates an adjustment to
assumed potency. Similarly, if the slope of the unknown is less the error sum of squares as described for assays based on the
steep than that of the standard, the potency was overestimated parallel-line model. The analysis of an assay of one or more
and the calculations will result in an estimated potency lower test preparations against a standard is described below.
than the assumed potency. 3.3.3. ANALYSIS OF VARIANCE
In setting up an experiment, all responses should be
3.3.3.1. The (hd + l)-design
examined for the fulfilment of the conditions 1, 2 and 3 in
Section 3.1. The analysis of variance to be performed in The responses are verified as described in Section 3.1 and,
routine is described in Section 3.3.3 so that compliance with if necessary, transformed. The responses are then averaged
conditions 4B and SB of Section 3.1 may be examined. over each treatment and each preparation as shown in
Table 3.3.3.1.-I. Additionally, the mean response fOI blanks (B)
3.3.2. ASSAY DESIGN is calculated.
The use of the statistical analysis presented below imposes the
following restrictions on the assay: The sums of squares in the analysis of variance are calculated
as shown in Tables 3.3.3.1.-I to 3.3.3.1.-III. The sum of
a) the standard and the test preparations must be tested with squares due to non-linearity can only be calculated if at least
the same number of equally spaced dilutions, 3 dos es of each preparation have been included in the assay.
b) an extra group of experimental units receiving no treatment The residual error is obtained by subtracting the variations
may be tested (the blanks), allowed for in the design from the total variation in response
c) there must be an equal number of experimental units to (Table 3.3.3.1.-IV).
each treatment. The analysis of variance is now completed as follows. Each
As already remarked in Section 3.1.3, assay designs not sum of squares is divided by the corresponding number of
meeting these restrictions may be both possible and correct, degrees of freedom to give mean squares. The mean square
but the simple statistical analyses presented here are no longer for each variable to be tested is now expressed as a ratio to the
applicable and either expert advice should be sought or residual error (52) and the significance of these values (known
suitable software should be used. as F-ratios) are assessed by use of Table 8.1 or a suitable
sub-routine of a computer programo
A design with 2 doses per preparation and 1 blank, the
"common zero (2h + l)-design': is usually preferred, since 3.3.3.2. The (hd)-design
it gives the highest precision combined with the possibility The formulae are basically the same as those fOI the
to check validity within the constraints mentioned aboye. (hd + l)-design, but there are some slight differences.
However, a linear relationship cannot always be assumed to
- B is discarded from all formulae.
be valid down to zero-dose. With a slight loss of precision a
design without blanks may be adopted. In this case 3 doses per _ K = n (Ps + PT + ··l
preparation, the "common zero (3h)-design", are preferred to hd
2 doses per preparation. The doses are thus given as follows: - is removed from the analysis of variance.
SSblank

1) the standard is given in a high dose, near to but not - The number of degrees of freedom for treatments becomes
exceeding the highest dose giving a mean response on the hd - 1.
straight portion of the dose- response line, - The number of degrees of freedom of the residual error
2) the other doses are uniformly spaced between the highest and the total variance is calculated as described for the
dose and zero dose, parallel-line model (see Table 3.2.3.-IV).
3) the test preparations are given in corresponding doses Validity of the assay, potency and confidence interval are
based on the assumed potency of the material. found as described in Sections 3.3.4 and 3.3.5.

Table 3.3.3.1.-I. - Formulae for slope-ratio assays with d doses of each preparation and a blank
Standard 1st Test sample 2nd Test sample
(S) (T) (U, etc.)

Mean response lowest dose

Mean response 2"d dose 5, T, u,

Mean response highest dos e

Total preparatioll Fu = ...

Linear product Lu = ...


Intercept value as = (4d+2)Fs - 6Ls au = ...

Slope value bs = 2Ls - (d+l)Fs bu = ...

Treatment value Gs = S~ + ... + S~ Gu =

Non-linearity* Ju = ...

* No! calculated for two-dose assays

General Notices (1) apply to all monographs and other texts 613
5.3. Statistical analysis EUROPEAN PHARMACOPOEIA 8.0

Table 3.3.3.1.-11. - Additional formulae for the construction of the analysis of variance

nhd 2 - nhd
HI =
n
a =
as +aT + ". K =
n (B + Ps + P T + .. )2
~----~--~--~
HB = hd2 _ hd + 4d + 2 4d 3 - 2d 2 - 2d h (d 2 - d) hd +1

Table 3.3.3.1.-III. - Formulae to calculate the sum Of squares and degrees Offreedom

Source of variation Degrees of freedom (fJ Sum of squares

Regression h SSreg = SStreat - SSblank - SSint - SSlin

B1anks 1 SSblank = H B (B - a)2

Intersection h - 1 SSint = HI ((a~ + a~ +. .) _ h (d 2 _ d) 2 a 2 )

Non-Iinearity* h (d - 2) SSlin = n (Js + JT + ".)


Treatments hd SStreat = n (B 2 + Gs + G T + ".) - K

* Not calculated for two-dose assays

Table 3.3.3.1.-IV. - Estimation Of the residual error

Source oi variation Degrees of freedom Sum of squares

Blocks (rows)* n - 1 SSblock = hd (Ri + ". + R~) - K

Columns** n - 1 SScol = hd (c~ + ". + e;;) - K

Residual error***
¡ Completely
randomised

Randomised block

Latin square
(hd + 1) (n

hd (n - 1)

(hd - 1) (n - 1)
- 1) SSres

SSres

S5 res
=

=
SStot -

SStot -
SSt'reat

SStreat - SSblock

= SStot - SStreat - SSblock - SScol

Total nhd +n - 1 SStot = I: (y - y)2

For Latin square designs, these formulae are only applicable if n = hd


* Not calculated for completely randomised designs
** Only calculated for Latin square designs
*** Depends on the type of design

3.3.4. TESTS OF VALIDlTY 3.3.5. ESTIMATION OF POTENCY AND CONFIDENCE


Assay results are said to be "statistically valid" if the outcome LIMlTS
of the analysis of variance is as follows: 3.3.5.1. The (hd + l)-design
1) the variation due to blanks in (hd + l)-designs is not The common intersection a' of the preparations can be
significant, i.e. the calculated probability is not smaller than calculated from:
0.05. This indicates that the responses of the blanks do not
significantly differ from the common intercept and the linear (2d + 1) B + (2d - 3) ha
a' ==
relationship is valid down to zero dose; h (2d - 3) + 2d + 1 (3.3.5.1.-1)

2) the variation due to intersection is not significant, Le. the The slope of the standard, and similarly for each of the other
calculated probability is not less than 0.05. This indicates that preparations, is calculated from:
condition 5B, Section 3.1 is satisfied;
b' _ 6Ls-3d(d+l)a'
3) in assays including at least 3 doses per preparation, the s - 2d3 + 3d2 + d (3.3.5.1.-2)
variation due to non-linearity is not significant, Le. the
calculated probability is not less than 0.05. This indicates that The potency ratio of each of the test preparations can now be
condition 4B, Section 3.1 is satisfied. calculated from:

A significant variation due to blanks indicates that the R' _ b~


hypothesis of linearity is not valid near zero dose. rf this is T - b~ (3.3.5.1.-3)
likely to be systematic rather than incidental for the type of
assay, the (hd-design) is more appropriate. Any response to which has to be multiplied by Ap the assumed potency of the
blanks should then be disregarded. test preparation, in order to find the estimated potency Ry.
rf the step between adjacent doses was not identical for the
When these tests indicate that the assay is valid, the potency standard and the test preparation, the potency has to be
is calculated with its confidence limits as described in multiplied by lsll]. Note that, unlike the parallel-line analysis,
Section 3.3.5. no antilogarithms are calculated.

614 See the information section on general monographs (ca ver pages)
EUROPEAN PHARMACOPOEIA 8.0 5.3. Statistical analysis

The confidence interval for R; is calculated from: The general shape of the curves can usually be described by
a logistic function but other shapes are also possible. Each
CR~ - K' ± J(C - 1) (CR:} + 1) + K' (K' - 2CR~) curve can be characterised by 4 parameters: The upper
asymptote (a), the lower asymptote (o), the slope-factor (~),
(3.3.5.1.-4) and the horizontallocation (y). This model is therefore
often referred to as a four-parameter model. A mathematical
= b,§ - b'§s2t2V1
representation of the ln( dosel-response curve is:
where C and K' = (C
-
1) V
2
0'-6
u = 6+ -,----;;-;--,
V¡ are V 2 are related to the variance and covariance of the 1 + e- f3 (x--y)
numerator and denominator of Rr They can be obtained
from: For a valid assay it is necessary that the curves of the standard
and the test preparations have the same slope-factor, and the
same maximum and minimum response level at the extreme
V1 = n(2:+1)(d(d~1) +2(2d+l)!hd(d-lJ parts. Only the horizontallocation (y) of the curves may
be different. The horizontal distance between the curves is
(3.3.5.1.-5) related to the "true" potency of the unknown. If the assay
is used routinely, it may be sufficient to test the condition
of equal upper and lower response levels when the assay is
3d(d+l) developed, and then to retest this condition directly only at
V2 = ~~~~~~--~~~~
(3d + l)(d + 2) + hd (d - 1) (3.3.5.1.-6)
suitable intervals or when there are changes in materials or
The confidence limits are multiplied by Ay> and if necessary assay conditions.
byIsfIr The maximum-likelihood estimates of the parameters and
their confidence intervals can be obtained with suitable
3.3.5.2. The (hd)-design
computer programs. These computer programs may include
The formulae are the same as for the (hd + l)-design, with sorne statistical tests reflecting validity. For example, if the
the following modifications: maximum likelihood estimation shows significant deviations
from the fitted model under the assumed conditions of equal
a' = a (3.3.5.2.-1)
upper and lower asymptotes and slopes, then one or all of

v _
1-
6
nd(2d+l)
(~1
d+l
3)
+ h(d-l) (3.3.5.2.-2)
these conditions may not be satisfied.
The logistic model raises a number of statistical problems
which may require different solutions for different types of
assays, and no simple summary is possible. A wide variety
3 (d + 1) of possible approaches is described in the relevant literature.
V2 = (3.3.5.2.-3)
3(d+l)+h(d-l) Professional advice is therefore recommended for this type
of analysis. A simple example is nevertheless included in
3.4. EXTENDED SIGMOID DOSE-RESPONSE CURVES Section 5.4 to illustrate a "possible" way to analyse the data
This model is suitable, for example, for some immunoassays presented. A short discussion of alternative approaches and
when analysis is required of extended sigmoid dose-response other statistical considerations is given in Section 7.5.
curves. This model is illustrated in Figure 3.4.-I. lf professional advice or suitable software is not available,
alternative approaches are possible: 1) if "reasonable" estimates
of the upper limit (a) and lower limit (o) are available, select
for all preparations the doses with mean of the responses (u)
. falling between approximately 20 per cent and 80 per cent
. . of the limits, transform responses of the selected doses to
y = In ( u -
a-u
6) and use t~e parallelline model (Section 3.2)
for the analysis; 2) select a range of doses for which the
responses (u) or suitably transformed responses, for example
In u, are approximately linear when plotted against ln( dose) ;
the parallelline model (Section 3.2) may then be used for
analysis.

Test sample 4. ASSAYS DEPENDING UPON


QUANTAL RESPONSES
4.1. INTRODUCTION
.. . In certain assays it is impossible or excessively laborious to
.. . measure the effect on each experimental unit on a quantitative
scale. Instead, an effect such as death or hypoglycaemic
lO
. symptoms may be observed as either occurring or not
occurring in each unit, and the result depends on the number
In dose (x) of units in which it occurs. Such assays are called quantal or
all-or-none.
Figure 3.4.-I. - The four-parameter logistic curve model The situation is very similar to that described for quantitative
The logarithms of the doses are represented on the horizontal assays in Section 3.1, but in place of n separate responses to
axis with the lowest concentration on the left and the highest each treatment a single value is recorded, i.e. the fraction of
concentration on the right. The responses are indicated on units in each treatment group showing a response. When
the vertical axis. The individual responses to each treatment these fractions are plotted against the logarithms of the doses
are indicated with black dots. The 2 curves are the calculated the resulting curve will tend to be sigmoid (S-shaped) rather
ln(dose)-response relationship for the standard and the test than linear. A mathematical function that represents this
preparation. sigmoid curvature is used to estimate the dose-response curve.

General Notices (1) apply to all monographs and other texts 615
5.3. Statistical analysis EUROPEAN PHARMACOPOE1A S.O

The most commonly used function is the cumulative normal the more extreme parts of the curve. This method, the analysis
distribution function. This function has some theoretical of variance, and the estimation of the potency and confidence
merit, and is perhaps the best choice if the response is a interval are described below.
reflection of the tolerance of the units. If the response is 4.2.1. TABULATION OF THE RESULTS
more likely to depend upon a process of growth, the logistic
Table 4.2.1.-1 is used to enter the data into the columns
distribution model is preferred, although the difference in
indicated by numbers:
outcome between the 2 models is usually very smal!.
(1) the dose of the standard or the test preparation,
The maximum likelihood estimators of the slope and location
of the curves can be found only by applying an iterative (2) the number n of units submitted to that treatment,
procedure. There are many procedures which lead to the (3) the number of units r giving a positive response to the
same outcome, but they differ in efficiency due to the speed treatment,
of convergence. One of the most rapid methods is direct (4) the logarithm x of the dose,
optimisation of the maximum-likelihood function (see Section
7.1), which can easily be performed with computer programs (5) the fraction p = rln of positive responses per group.
having a built-in procedure for this purpose. Unfortunately, The first cycle starts here.
most of these procedures do not yield an estimate of the (6) column Y is filled with zeros at the first iteratiol1,
confidence interval, and the technique to obtain it is too
complicated to describe here. The technique described below (7) the corresponding value <D = <D(Y) of the cumulative
is not the most rapid, but has been chosen for its simplicity standard normal distributiol1 function (see also Table 8.4).
compared to the alternatives. It can be used for assays in which The columns (8) to (10) are calculated with the following
one or more test preparations are compared to a standard. formulae:
Furthermore, the fol!owing conditions must be fulfilled: _y2/2
1) the relationship between the logarithm of the dose and (S) z= _e_ _ (4.2.1.-1)
the response can be represented by a cumulative normal ~
distribution curve, p-<l?
(9) y=y+-- (4.2.1.-2)
2) the curves for the standard and the test preparation are z
parallel, i.e. they are identically shaped and may only differ nZ 2
in their horizontallocation, (lO) (4.2.1.-3)
w = <l? _ g,2
3) in theory, there is no natural response to extremely low
The columns (ll) to (15) can easily be calculated from
doses and no natural non-response to extremely high doses.
columns (4), (9) and (lO) as wx, wy, wx 2 , wi and wxy
4.2. THE PROBIT METHOD respectively, and the sum (2:) of each of the columns (lO) to
(15) is calculated separately for each of the preparations.
The sigmoid curve can be made linear by replacing each
The sums calculated in Table 4.2.1.-1 are transferred to
response, i.e. the fraction of positive responses per group, by
columns (1) to (6) ofTable 4.2.1.-II and 6 additional columns
the corresponding value of the cumulative standard normal
(7) to (12) are calculated as follows:
distribution. This value, often referred to as "normit", ranges
theoretically from - to + oo. In the past it was proposed to
00

add 5 to each normit to obtain "pro bits". This facilitated the


(7) (¿wx)" (4.2.1.-4)
hand-performed calculations because negative values were Sxx = ¿wx 2 -
avoided. With the arrival of computers the need to add 5 to
¿w
the normits has disappeared. The term "normit method" (¿wx)(¿wy)
would therefore be better for the method described below. (S) Sxy = ¿wxy- (4.2.1.-5)
¿w
However, since the term "probit analysis" is so widely spread,
the term will, for historical reasons, be maintained in this text. (¿wy)2
(9) Syy = ¿wy2 - ( 4.2.1.-6)
Once the responses have been linearised, it should be possible ¿w
to apply the parallel-line analysis as described in Section 3.2.
Unfortunately, the validity condition of homogeneity of ¿wx
variance for each dose is not fulfilled. The variance is minimal
(10) x ¿w
( 4.2.1.-7)

at normit = O and increases for positive and negative values of


the normit. It is therefore necessary to give more weight to (ll) ¿wy (4.2.1.-8)
y
responses in the middle part of the curve, and less weight to ¿w

Table 4.2.l.-1. - First working table


(1) (2) (3) (4) (5) (6) (7) (8) (9) (10) (11) (12) (13) (14) (15)

dose n r x p y <D Z y w wx wy wx 2 wy' wxy

2:= 2:= 2:= 2:= 2:= 2:=


T

2:= 2:= 2:= 2:= 2:= 2:=


etc.

616 See the information section on general monographs (caver pages)


EUROPEAN PHARMACOPOEIA 8.0 5.3. Statistical anaiysis

Table 4.2.1.-I1. - Second working table


(1) (2) (3) (4) (5) (6) (7) (8) (9) (la) (11) (12)

¿w ¿wx ¿wy ¿wx' ¿wy' ¿wxy Su S,y Sr} x y a

T
etc.

¿~ ¿~

The common slope b can now be obtained as: are slightly modified. t becornes the t-value (p = 0.05) with
the same number of degrees of freedom as used in the check
b _ ~Sxy (4.2.1.-9) for linearity and 52 becomes the X2 value divided by the same
- ~Sxx
number of degrees of freedom (and thus typically is greater
and the intercept a of the standard, and similarly for the test than 1).
preparations is obtained as: The test for parallelism is also slightly modified. The X2 value
(12) a = y - b-x (4.2.1.-10) for non-parallelism is divided by its number of degrees of
freedom. The resulting value is divided by 52 calculated aboye
Column (6) of the first working table can now be replaced to obtain an F-ratio with h - 1 and N - 2h degrees of freedorn,
by y = a + bx and the cycle is repeated until the difference which is evaluated in the usual way at the 0.05 significance
between 2 cycles has become small (e.g. the maximum leve!.
difference of Y between 2 consecutive cycles is smaller
than 10- 8). 4.3. THE LOGIT METHOD
4.2.2. TESTS OF VALIDITY As indicated in Section 4.1 the logit method may sometimes
Before calculating the potencies and confidence intervals, be more appropriate. The name of the method is derived
validity of the assay must be assessed. If at least 3 doses for from the logit function which is the inverse of the logistic
each preparation have been included, the deviations from distribution. The procedure is similar to that described for
linearity can be measured as follows: add a 13 th column to the probit method with the following modifications in the
Table 4.2.1. -II and fill it with: forrnulae for <D and Z.

1 (4.3.-1)
(4.2.2.-1) ¿p =
1 + e-Y

The column total is a measure of deviations from linearity and -y (4.3.-2)


e
is approximately X2 distributed with degrees of freedom equal Z
to N - 2h. Significance of this value may be assessed with (1+e- y )2
the aid of Table 8.3 or a suitable sub-routine in a computer
programo If the value is significant at the 0.05 probability leve!, 4.4. OTHER SHAPES OF THE CURVE
the assay must probably be rejected (see Section 4.2.4).
The probit and logit method are almost always adequate for
When the aboye test gives no indication of significant the analysis of quantal responses called for in the European
deviations from linear regression, the deviations from Pharmacopoeia. However, if it can be made evident that the
parallelism are tested at the 0.05 significance leve! with: ln(dose)-response curve has another shape than the 2 curves
described aboye, another curve <D may be adopted. Z is then
X2 _ ~ S~y _ (~Sxy)2 (4.2.2.-2) taken to be the first derivative of <D.
- Sxx ~Sxx
For example, if it can be shown that the curve is not symmetric,
with h - 1 degrees of freedom. the Gompertz distribution may be a?propriate (the ~ompit
4.2.3. ESTIMATION OF POTENCY AND CONFIDENCE method) in which case ¿p = 1 - e-e and Z = e Y -e .
LIMITS
When there are no indications for a significant departure 4.5. THE MEDIAN EFFECTIVE DOSE
from parallelism and linearity the ln(potency ratio) M~ is In sorne types of assay it is desirable to determine a median
calculated as: effective dose which is the dose that produces a response in
M!r = aT - as 50 per cent of the units. The probit method can be used to
(4.2.3.-1)
b determine this median effective dose (ED so )' but since there
is no need to express this dose relative to a standard, the
and the antilogarithm is taken. Now let t = 1.96 and s = 1. formulae are slightly different.
Confidence limits are calculated as the antilogarithms of:
Note: a standard can optionally be included in order to
CM;'-(C-l)(XS-XT)±V(C-l) (v ~ Sxx+C(M;,-xS+XT)") validate the assay. Usually the assay is considered valid if the
calculated EDso of the standard is close enough to the assigned
(4.2.3.-2) EDso' What "close enough" in this context means depends on
the requirements specifled in the monograph.
b2 ~ Sxx 1 1
where e= b2 '"
L..
S _ 2 2 and V
xx s t
=-
~w
-+-
~w
- The tabulation of the responses to the test samples, and
S T
optionalIya standard, is as described in Section 4.2.1. The
test for linearity is as described in Section 4.2.2. A test for
4.2.4. INVALID ASSA YS parallelisrn is not necessary for this type of assay. The EDso of
If the test for deviations from linearity described in test sarnple T, and similarly for the other samples, is obtained
Section 4.2.2 is significant, the assay should normally be as described in Section 4.2.3, with the following modifications
rejected. lf there are reasons to retain the assay, the formulae in formula e 4.2.3.-1 and 4.2.3.-2).

General Notices (1) apply to all monographs and other texts 617
5.3. Statistical analysis EUROPEAN PHARMACOPOEIA 8.0

400
M' _ -aT (4.5.-1)
T - ~b-

380

cM;-eC-l)XT±veC-l) (v I: Sxx+C(M;-XT )2) 360


"
G

(4.5.-2)
..
'" 3ilO

'Z"" 320
1 3
I: w e is ¡eft unchanged
~ 300
where V = and
""
T e "
~ 280

~
=
o
~260 . "
'"
~ "
240 ..
• "
220 ". "~
200
"
5, EXAMPLES SI S2 TI T2 VI U2
180~----------~~----------~--------------~

This section consists of worked examples illustrating the


application of the formulae. The examples have been selected Figure 5.1.1.- I.
primarily to illustrate the statistical method of calculation.
The formulae in Tables 3.2.3.-1 and 3.2.3.-II lead to:
They are not intended to reflect the most suitable method
of assay, if alternatives are permitted in the individual Ps 580.4 Ls - 41.8
monographs. To increase their value as program checks, more PT 567.9 L¡ - 39.95
decimal places are given than would usually be necessary. It
should also be noted that other, but equivalent methods of Pu 532.2 Lu - 16.1
calculation exist. These methods should lead to exactly the
10 120
same final results as those given in the examples. H p = - =5 HL = - =20
2 6

The analysis of variance can now be completed with the


5.1. PARALLEL-LINE MODEL formulae in Tables 3.2.3-III and 3.2.3.-1Y. This is shown in
Table 5.1.1.-11.
5.1.1. TWO-DOSE MULTIPLE ASSAY WITH COMPLETELY
RANDOMISED DESIGN Table 5.1.1.- n. - Analysis of variance
An assay of corticotrophin by subcutaneous injection in rats
SOUfce of Degrees of Smn of Mean Proba-
F-ratio
The standard preparation is administered at 0.25 and 1.0 units variation freedom squares s<¡uare bility
per 100 g of body mass. 2 preparations to be examined are
both assumed to have a potency of 1 unit per milligram and Preparations 2 6256.6 3128.3
they are administered in the same quantities as the standard. Regression 1 63830.8 63830.8 83.38 0.000
The individual responses and means per treatment are given
in Table 5.1.1.-1. A graphical presentation (Figure 5.1.1.-1) Non-parallelism 2 8218.2 4109.1 5.37 0.007
gives no rise to doubt the homogeneity of variance and TreatInents 5 78 305.7
normality of the data, but suggests problems with parallelism
for preparation U. Residual error 54 41 340.9 765.57

Total 59 119646.6
Table 5.1.1.-1. - Response metameter y: mass of ascorbic acid
(mg) per 100 g of adrenal gland
The analysis confirms a highly significant linear regression.
Departure from parallelism, however, is also significant
Standard S Preparation T Preparation U
(p = 0.0075) which was to be expected from the graphical
SI 52 TI T2 UI U, observation that preparation U is not parallel to the standard.
This preparation is therefore rejected and the analysis repeated
300 289 310 230 250 236
using only preparation T and the standard (Table 5. 1. l.-III).
310 221 290 210 268 213

330 267 360 280 273 283 Table S.Ll.-lII. - Analysis ofvariance without sample U

290 236 341 261 240 269 Source of Degrees of Sum of Mean Proba-
F-ratio
variation freedom s<¡uares s<¡uare bility
364 250 321 241 307 251

328 231 370 290 270 294 Preparations 1 390.6 390.6

390 229 303 223 317 223 Regression 1 66830.6 66830.6 90.5 0.000

360 269 334 254 312 250 Non-parallelism 1 34.2 34.2 0.05 0.831

342 233 295 216 320 216 Treatments 3 67255.5

306 259 315 235 265 265 Residual error 36 26587.3 738.54

Mean 332.0 248.4 323.9 244.0 282.2 250.0 Total 39 93842.8

618 See the information section on general monographs (cover pages)


EUROPEAN PHARMACOPOElA 8.0 5.3. Statistical analysis

The analysis without preparation U results in compliance Table 5.1.2.-I. - Distribution of treatments over the plate
with the requirements with respect to both regression and
parallelism and so the potency can be calculated. The 2 3 4 5 6
formulae in Section 3.2.5 give:
51 TI T2 53 52 T3

- for the common slope: 2 TI T, 51 5, T2 53

3 T2 53 52 51 T3 TI
b= 20(-41.8-39.95) =-58.970
In4 x 10 x 2 4 T3 T2
53 52 TI 51
5 52 T2 53 T3 TI 51
- the ln(potency ratio) is:
6 T3 51 TI T2 53 52
M' - 567.9 - 580.4 = 0.1060
T - 2x (-58.970)
66830.6
e =
66830.6 - 738.54 x 2.028 2
= 1.0476

66830.6 Table 5.1.2.-11. - Measured inhibition zones in mm x 10


V = 2 = 0.9609
(-58.970) x 2 x 10
2 4 5 6 Row mean

- and ln(confidence limits) are: 161 160 178 187 171 194 175.2 = R l

1.0476 x 0.1060 ± VO.0476 x (1.0476 x 0.1060 2 +2 x 0.9609) 2 151 192 150 172 170 192 171.2=R,

= 0.1110 ± 0.3034 162 195 174 161 193 151 172.7 = R3

4 194 184 199 160 163 171 178.5 = R4


By taking the antilogarithms we find a potency ratio of 1.11
5 176 181 201 202 154 151 177.5 = Rs
with 95 per cent confidence limits from 0.82-1.5l.
6 193 166 161 186 198 182 181.0 = R,
Multiplying by the assumed potency of preparation T yields a Col. 172.8 179.7 177.2 178.0 174.8 173.5
potency of 1.11 units/mg with 95 per cent confidence limits Mean = el = e2 = e3 = e4 = es = e,
from 0.82 to 1.51 units/mg.
5.l.2. THREE-DOSE LATIN SQUARE DESIGN
Antibiotic agar diffusion assay using a rectangular tray

The standard has an assigned potency of 4855 IU /mg. The test


preparation has an assumed potency of 5600 IU /mg. For the Table 5.1.2.- m. - Means of the treatments
stock solutions 25.2 mg of the standard is dissolved in 24.5 mL
of solvent and 21.4 mg of the test preparation is dissolved Standard 5 Preparation T
in 23.95 mL of solvent. The final solutions are prepared by
first diluting both stock solutions to 1/20 and further using SI S2 S3 TI T2 T,
a dilution ratio of 1.5. Mean 158.67 176.50 194.50 156.17 174.67 195.50

A Latin square is generated with the method described


in Section 8.6 (see Table 5.1.2.-1). The responses ofthis
routine assay are shown in Table 5.l.2.-U (inhibition zones
in mm x 10). The treatment mean values are shown in
Table 5.l.2.-III. A graphical representation of the data (see
Figure 5.1.2.-I) gives no rise to doubt the normality or
homogeneity of variance of the data. 210

The formulae in Tables 3.2.3.-1 and 3.2.3.-II lead to: . .


P5 = 529.667 = 35.833
200
.. "
L5
= 526.333
S "
Pr Lr = 39.333 ..... 190

6 72
;.<i
El
.
Hp - =2
3
H[. = - =3
24
El
-180
..
o

~
1::
<:)
NI
..
¡s 170 :
The analysis of variance can now be completed with the :E .
formulae in Tables 3.2.3.-III and 3.2.3.-IY. The result is shown ..el
:a1:: 160 ....
in Table 5.1.2.-IY.
- I

..
The analysis shows significant differences between the rows. 150 ~
.
This indicates the increased precision achieved by using a
Latin square design rather than a completely randomised SI 82 83 TI T2 T3
designo A highly significant regression and no signiflcant 140
departure of the individual regression lines from parallelism
and linearity conflrms that the assay is satisfactory for potency
calculations. Figure 5.1.2.-1.

General Notices (1) apply to all monographs and other texts 619
5.3. Statistical analysis EUROPEAN PHARMACOPOEIA 8.0

Table 5.l.2.-IV. - Analysis of variance Table 5.1.3.-1. - Absorbances of the suspensions (x 1000)
Standard S Preparation T
Source of Degrees of Sum of Mean Proba-
F-ratio Mean
variation freedom squares square bility Block 51 52 5, 5, T1 T2 T3 T,

Preparations 1 11.1111 11.1111 1 252 207 168 113 242 206 146 115 181.1

Regression 1 8475.0417 8475.0417 408.1 0.000 2 249 201 187 107 236 197 153 102 179.0

Non-parallelism 1 18.3750 18.3750 0.885 0.358 3 247 193 162 111 246 197 148 104 176.0

Non-linearity 2 5.4722 2.7361 0.132 0.877 4 250 207 155 108 231 191 159 106 175.9

Treatments 5 8510 5 235 207 140 98 232 186 146 95 167.4

Rows 5 412 82.40 3.968 0.012 Mean 246.6 203.0 162.4 107.4 237.4 195.4 150.4 104.4

Columns 5 218.6667 43.73 2.106 0.107 275

Residual error 20 415.3333 20.7667


250
Total 35 9556
••
• i>
225
The formulae in Section 3.2.5 give: •
200
• ••
¡¡¡'"'"
for the common slope:
175
b = 3 x (35.833 + 39.333) = 46.346 ...
,.Q
<:)
".
In (l.5) X 6 X 2 1l 150
...r: $

- the ln(potency ratio) is: 125


526.333 - 529.667 •
M~ = ~-3-x-4-6-.3-4-6-- = -0.023974 100
8475.0417
e = = l.0108
75
8475.0417 - 20.7667 x 2.086 2
8475.0417 SI S2 S3 84 TI T2 T3 T4
v = 46.346 2 x 3 x 6
= 0.2192
50

Figure 5.1.3.-1.
- and ln(confidence limits) are:
The formulae in Tables 3.2.3.-1 and 3.2.3.- II lead to:
1.0108 x (-0.0240) ± - 229.1
PI 719.4 L,
JO.OI08 x (l.0108 x (-0.0240)2 +2 x 0.2192)
= -0.02423 ± 0.06878 P,. 687.6 LT - 222

5 60
Hp 4" = 1.25 HL 60 =1
The potency ratio is found by taking the antilogarithms,
resulting in 0.9763 with 95 per cent confidence limits from The analysis of variance is constructed with the formulae
0.9112-1.0456. in Tables 3.2.3.-III and 3.2.3.-1V. The result is shown in
Table 5.1.3.-11.
A correction factor of 4855 x 25.2/24.5 = 0.99799 is Table 5.1.3.-11. - Analysis of variance
5600 x 21.4 23.95
necessary because the dilutions were not exactly equipotent Source of Degrees oí Sum of Mean square F-ratio Proba-
on the basis of the assumed potency. Multiplying by this variation freedom squares bility
correction factor and the assumed potency of 5600 IU/mg
Preparations 1 632.025 632.025
yields a potency of 5456 IU/mg with 95 per cent confidence
limits from 5092 to 5843 IV /mg. Regression 1 101 745.6 101 745.6 1887.1 0.000

5.1.3. FOUR-D05E RANDOMI5ED BLOCK DE5IGN Non-parallelism 1 25.205 25.205 0.467 0.500
Antibiotic turbidimetric assay 4
Non-linearity 259.14 64.785 1.202 0,332

This assay is designed to assign a potency in international Treatments 7 102662


units per vial. The standard has an assigned potency of 4
Blocks 876.75 219.188 4.065 0.010
670 IU/mg. The test preparation has an assumed potency of
20 000 IU /vial. On the basis of this information the stock Residual error 28 1509.65 53.916
solutions are prepared as follows. 16.7 mg of the standard
Total 39 105 048.4
is dissolved in 25 mL solvent and the contents of one vial
of the test preparation are dissolved in 40 mL solvento The A significant difference is found between the blocks. This
final solutions are prepared by first diluting to 1/40 and indicates the increased precision achieved by using a
further using a dilution ratio of 1.5. The tubes are placed randomised block designo A highly significant regression
in a water-bath in a randomised block arrangement (see and no significant departure from parallelism and linearity
Section 8.5). The responses are listed in Table 5.1.3.-1. confirms that the assay is satisfactory for potency calculations.
The formulae in Section 3.2.5 give:
Inspection ofFigure 5.1.3.-1 gives no rise to doubt the validity - for the common slope:
of the assumptions of normality and homogeneity of variance
of the data. The standard deviation of 53 is somewhat high b = 1 x (-229.1 - 222) = -11l.255
but is no reason for concern. In (l.5) x 5 x 2

620 5ee the information section on general monographs (ca ver pages)
EUROPEAN PHARMACOPOEIA 8.0 5.3. Statistical analysis

- the ln(potency ratio) is: 0.5

M' _ 687.6 - 719.4 = 0.071457


T - 4 x (-111.255) 0.0 "
101745.6
e =
101 745.6 - 53.916 x 2.048 2
= 1.00223
-0.5
101745.6
V = = 0.4110
(-111.255)2 x 4 x 5
~ -1.0
Ci
.
- and ln(confidence limits) are: ..'"5;
..Q

-1.5
1.00223 x 0.0715± ,.Q
$.
JO.00223 x (1.00223 x 0.0715 2 + 2 x 0.4110)
.s -2.0
= 0.07162 ± 0.04293

·2.5
The potency ratio is found by taking the antilogarithms,
resulting in 1.0741 with 95 per cent confidence
·3.0
limits from 1.0291 to 1.1214. A correction factor of $
670 x 16.7/25 . .. S T u v
20000 x 1/40 = 0.89512 IS necessary because the dllutlOns
·3.5.1-.----"------'--------'------1
were not exactly equipotent on the basis of the assumed
potency. Multiplying by this correction factor and the assumed Figure 5.1.4.-1.
potency of 20 000 IU /vial yields a potency of 19 228 IU /vial
with 95 per cent confidence limits from 18423-20075 IU/vial. The formuIae in TabIes 3.2.3.-1 and 3.2.3.-II give:
Ps - 9.108 Ls 6.109
5.1.4. FIVE-DOSE MULTIPLE ASSAY WITH COMPLETELY
RANDOMISED DESIGN Pr - 5.586 LT 6.264
An in-vitro assay of three hepatitis B vaccines against a standard
Pu - 6.544 Lu 6.431

3 independent two-fold dilution series of 5 dilutions were PI' - 6.027 L,. 6.384
prepared from each of the vaccines. After sorne additional
3 36
steps in the assay procedure, absorbances were measured. Hp - =0.6 HL -=0.3
5 120
They are shown in Table 5.1.4.-1.
The analysis of variance is compIeted with the formulae in
Table 5.1.4.-1. - Optical densities TabIes 3.2.3.-III and 3.2.3.-IY. This is shown in TabIe 5.1.4.-IIL

TabIe 5.1A.-III. - Analysis of varíance


Dilution Standard S Preparation T
Sonrce of Degrees of Sum of Mean Proba-
1:16000 0.043 0.045 0.051 0.097 0.097 0.094 F-ratio
variation freedom squares square bility
1:8000 0.093 0.099 0.082 0.167 0.157 0.178
Preparations 3 4.475 l.492
1:4000 0.159 0.154 0.166 0.327 0.355 0.345
Regression 1 47.58 47.58 7126 0.000
1:2000 0.283 0.295 0.362 0.501 0.665 0.576
Non- 3 0.0187 0.006 0.933 0.434
1:1000 0.514 0.531 0.545 1.140 1.386 l.051 parallelisrn

Non-linearity 12 0.0742 0.006 0.926 0.531

Dilution Preparation U Preparation V Treatrnents 19 52.152

1:16000 0.086 0.071 0.073 0.082 0.082 0.086 Residual error 40 0.267 0.0067

1:8000 0.127 0.146 0.133 0.145 0.144 0.173 Total 59 52.42

1:4000 0.277 0.268 0.269 0.318 0.306 0.316


A highly significant regression and a non-significant departure
1:2000 0.586 0.489 0.546 0.552 0.551 0.624 from parallelism and linearity confirm that the potencies can
be safeIy calculated. The formuIae in Section 3.2.5 give:
1:1000 0.957 0.866 l.045 l.037 l.039 l.068
- for the common slope:
The logarithms of the opticaI densities are known to have b = 0.3 x (6.109 + 6.264 + 6.431 + 6.384) = 0.90848
a linear relationship with the logarithms of the doses. The ln2 x 3 x 4
mean responses of the ln-transformed optical densities are - the In(potency ratio) for preparation T is:
listed in TabIe 5.1.4.-11. No unusual features are discovered in
a graphicaI presentation ofthe data (Figure 5.1A.-I). M'T -5.586 - (-9.108) = 0.7752
5 x 0.90848
47.58
TabIe 5.1.4.-11. - Means ofthe ln-transformed absorbances e = 47.58 - 0.0067 x 2.02]2
= 1.00057

47.58
Si - 3.075 T] - 2.344 u] - 2.572 V] - 2.485
V = 0.9085 2 X 5 x 3 = 3.8436
S2 - 2.396 T2 - l.789 U2 - 2.002 V2 - l.874
- and In(confidence limits) for preparation Tare:
S3 - l.835 T3 - l.073 U3 - 1.305 V3 - 1.161
1.00057 x 0.7752±
S, - 1.166 T] - 0.550 U4 - 0.618 V4 - 0.554 JO.00057 x (1.00057 x 0.7752 2 +2 x 3.8436)

Ss - 0.635 Ts 0.169 U, - 0.048 Vs 0.047 = 0.7756 ± 0.0689

General Notíces (1) apply to all monographs and other texts 621
5.3. Statistical analysis EUROPEAf,J PHARMACOPOEIA 8.0

By taking the antilogarithms a potency ratio of 2.171 is 160


found with 95 per cent confidence limits from 2.027 to 2.327. ~

Al! samples have an assigned potency of 20 flg protein/mL 140


$

and so a potency of 43.4 flg protein/mL is found for test


preparation T with 95 per cent confidence limits from ~
40.5-46.5 flg protein/mL. ;:¡- 120 "
S

~
Q
@
.
O
The same procedure is followed to estímate the potency and

~
;; 100
confidence interval of the other test preparations. The results '-'el)
---
are listed in Table 5.1.4.-IV.
! 80

'"¡::'"e
$ $

Table 5.1.4.-IV. - Final potency estimates and 95 per cent


confidence intervals of the test vaccines (in flg protein/mL)
s::>, 60
.
'"
o>
Q:::
@ .o
Lower limit Estimate Upper Iimit 40
¡¡
..
Vaccine T 40.5 43.4 46.5
.
20
Vacdne U 32.9 35.2 37.6
SI S2 TI T2
Vacdne V 36.8 39.4 42.2 o~----------------~--------------~

5.1.5. TWIN CROSS-OVER DESIGN Figure 5.1.5.- I.


Assay of insulin by subcutaneous injection in rabbits The analysis of variance is more complicated for this assay
than for the other designs given because the component of the
The standard preparation was administered at 1 unit and sum of squares due to parallelism is not independent of the
2 units per millilitre. Equivalent doses of the unknown component due to rabbit differences. Testing of the parallelism
preparation were used based 011 an assumed potency of of the regression lines involves a second error-mean-square
40 units per millilitre. The rabbits received subcutaneously term obtained by subtracting the parallelism component and
0.5 mL of the appropriate solutions according to the design 2 "interaction" compol1ents from the component due to rabbit
in Table 5.1.5.-1 and responses obtained are shown in differences.
Table 5.1.5.-II and Figure 5.1.5.-1. The large variance illustrates 3 "interaction" components are present in the analysis of
the variation between rabbits and the need to employ a variance due to replication within each group:
cross-over designo days x preparation; days x regression; days x parallelism.
These terms indicate the tendency for the components
Table 5.1.5.-1, - Arrangements oftreatments (preparations, regression and parallelism) to vary from day
to day. The corresponding F-ratios thus provide checks on
Group of rabbits these aspects of assay validity. If the values of F obtained are
significantly high, care should be exercised in interpreting
1 2 3 4 the results of the assay and, if possible, the assay should be
Day 1 51 S, TI T,
repeated.
The analysis of variance is constructed by applying the
Day 2 T, TI 52 S] formulae given in Tables 3.2.3.-I to 3.2.3.-III separately for
both days and for the pooled set of data. The formulae in
Tables 3.2.3.-1 and 3.2.3.-II give:
Day 1: Po 165.25 Lo -13

PT 162.25 L,. - 8.75

Hp 8 HL 96
Table 5.1.5.-11. - Response y: sum of blood glucose readings -=4 - = 16
2 6
(mg1100 mL) at 1 hour and 2~ hours
Day2: Ps 173.38 Ls - 20.06
Group 1 Group 2 Group 3 Group 4 Pr 176.00 LT - 5.25

S] T, S, TI T 52 T, 51 8 96
]
Hp -=4 H, - = 16
112 104 65 72 105 91 118 144
2 6
Pooled: Ps 169.31 Ls - 16.53
126 112 116 160 83 67 119 149
P,. 169.13 Ly - 7.00
62 58 73 72 125 67 42 51
Hp 16 = 8 HL 192 = 32
86 63 47 93 56 45 64 107 2 6
52 53 88 113 92 84 93 117 and with the formulae in Table 3.2.3.-III this leads to:
110 113 63 71 101 56 73 128 Day 1 Day 2 Po oled

116 91 50 65 66 55 39 87 SSp", 18.000 SSprep 13.781 SSprep 0.141

101 68 55 100 91 68 31 71 SS"g 3784.5 5125.8 8859.5

Mean 95.6 82.8 69.6 93.3 89.9 66.6 72.4 106.8 SSp", 144.5 SSp", 1755.3 SS", 1453.5

622 See the information section on general monographs (ca ver pages)
EUROPEAN PHARMACOPOEIA 8.0 5.3. Statistkal analysis

The interaction terms are found as Day 1 + Day 2 - Pooled. - and ln(confidence limits) are:

SSdaysxprep = 31.64
1.0695 x 0.00276 ± ,10.0695 x (1.0695 x 0.00276 2 +2 x 0.2402)
= 0.00295 ± 0.18279

= 50.77 By taking the antilogarithms a potency ratio of 1.003 with


95 per cent confidence limits from 0.83S to l.204 is found.
Multiplying by AT = 40 yields a potency of 40.1 units
SSdaysxpar = 446.27 per millilitre with 95 per cent confidence limits from
33.4-48.2 units per millilitre.
In addition the sum of squares due to day-to-day variation is
calculated as:
5.2. SLOPE-RATIO MODEL

SSdays = I (2
2N DI + D 22) - K = 478.52 5.2.1. A COMPLETELY RANDOMISED (Q,3,3)-DESIGN
An assay offactor VIII
and the sum of squares due to blocks (the variation between A laboratory carries out a chromogenic assay of factor VIII
rabbits) as: activity in concentrates. The laboratory has no experience
with the type of assay but is trying to make it operational.
2
SSblock = 2 I: Bi - K = 39794.7 3 equivalent dilutions are prepared ofboth the standard and
the test preparation. In addition a blank is prepared, although
where B¡ is the mean response per rabbit. a linear dose- response relationship is not expected for low
doses. 8 replicates of each dilution are prepared, which is
The analysis of variance can now be completed as shown in more than would be done in a routine assay.
Table S.loS.-III.
A graphical presentation of the data shows clearly that the
Table 5.1.5.-III. - Analysis ofvariance dose-response relationship is indeed not linear at low doses.
The responses to blanks will therefore not be used in the
Degrees calculations (further assays are of course needed to justify this
SOUfce of Snm of Mean Proba-
of F-ratio decision). The formulae in Tables 3.3.3.1.-1 and 3.3.3.1.-II
variation sqnares s<¡nare bility
freedom yield
Non- 1 1453.5 1453.5 1.064 0.311 Ps 0.6524 Pr 0.5651
parallelism
Ls 1.4693 [,) 1.2656
Days x Prep. 1 31.6 31.6 0.023 0.880
as 0.318 aT 0.318
Days x Regr. 1 50.8 50.8 0.037 0.849
b5 0.329 bT 0.271
Residual
error between 28 38258.8 1366.4 Gs 0.1554 G l' 0.1156
rabbits
15 4.17. lO-K Ir 2.84. 10- 6
Rabbits 31 39794.7 1283.7
and
Preparations 1 0.14 0.14 0.001 0.975 ,
H, = 0.09524 a = 0.05298 K = 1.9764
Regression 1 8859.5 8859.5 64.532 0.000
and the analysis of variance is completed with the formula e in
Days 1 478.5 478.5 3.485 0.072
Tables 3.3.3.1.-III and 3.3.3.1.-IV
Days x non- 1 446.3 446.3 3.251 0.082
par. A highly significant regression and no significant deviations
from linearity and intersection indicate that the potency can
Residual error 28 3844.1 137.3 be calculated.
within rabbits
Slope of standard:
Total 63 53423.2
, 6 x l.469 - 36 x 0.0530
bs = 84 = 0.0822
The analysis of variance confirms that the data fulfil the
necessary conditions for a satisfactory assay: a highly Slope of test sample:
significant regression, no significant departures from
, 6 x l.266 - 36 x 0.0530
parallelism, and none of the three interaction components bT = 84 = 0.0677
is significant.
Formula 3.3.5.1.-3 gives:
The formulae in Section 3.2.5 give:
0.0677
R = 0.823
for the common slope: 0.0822

b = 32 x (-16.53 - 7) 0.0822 2
In2 x 16 x 2
-33.95 e = = 1.000083
0.0822 2 - 3.86·10 6 x 2.018 2 x 0.0357

- the ln(potency ratio) is: K' = 0.000083 x 0.75 = 0.000062


169.13 - 169.31
M'T = 0.00276 and the 95 per cent confidence limits are:
2 x (-33.95)

8859.5
0.823 ± ,10.000083 x l.678 + 0.000062 x (-1.646)
e = 8859.5 - 137.3 x 2.048 2
= 1.0695 = 0.823 ± 0.006

8859.5 The potency ratio is thus estimated as 0.823 with 95 per cent
V = 2 = 0.2402
(-33.95) x 2 x 16 confidence limits from 0.817 to 0.829.

General Notices (1) apply lo all monographs and other texts 623
5.3. Statistical analysis EUROPEAN PHARMACOPOEIA 8.0

Table 5.2.1.-1. - Absorbances external and the internal reactant is established, the zone of
the annulus precipitation area is measured. The results are
B1ank Standard S Preparation T shown in Table 5.2.2.-1.
(in IU/rnL) (in lU/rnL)

Conc. B 5, 52 53 T, T2 T3 Table 5.2.2.-1. - Zone of precipítatiol1 area (mm 2)


0.01 0.02 0.03 0.01 0.02 0.03
Cone. Standard S Preparation T Preparation U
0.022 0.133 0.215 0.299 0.120 0.188 0.254
(flg/mL) 1 II 1 IX 1 H
0.024 0.133 0.215 0.299 0.119 0.188 0.253
7.5 18.0 18.0 15.1 16.8 15.4 15.7
0.024 0.131 0.216 0.299 0.118 0.190 0.255
15.0 22.8 24.5 23.1 24.2 20.2 18.6
0.026 0.136 0.218 0.297 0.120 0.190 0.258
22.5 30.4 30.4 28.9 27.4 24.2 23.1
0.023 0.137 0.220 0.297 0.120 0.190 0.257
30.0 35.7 36.6 34.4 37.8 27.4 27.0
0.022 0.136 0.220 0.305 0.121 0.191 0.257

0.022 0.138 0.219 0.299 0.121 0.191 0.255 A graphical presentation of the data shows no unusual features
(see Figure 5.2.2.-l). The formulae in Tables 3.3.3.1.-1 and
0.023 0.137 0.218 0.302 0.121 0.190 0.254
3.3.3.1.- II yield
Mean 0.0235 0.1351 0.2176 0.2996 0.1200 0.1898 0.2554 P5 108.2 Pr 103.85 Pu 85.8

L5 30l.l Lr 292.1 Lu 234.1

as 141.0 aT 116.7 au 139.8


0.350 39.2
bs 61.2 b[ 64.95 bu
S3
Gs 3114.3 Gr 2909.4 Gu 1917.3
0.300
f5 0.223 fr 2.227 fu 0.083

0.250 S2
and
1'3 H¡ 0.0093 a 11.04 K 14785.8
a...
Q,}

0.200
-e SI and the analysis of variance is completed with the formulae
51 in Tables 3.3.3.1.-III and 3.3.3.1.-IV. This is shown in
~ 0.150 1'2
Table 5.2.2.-H.

A highly significant regression and no significant deviations


0.100
TI from Iinearity and intersection indicate that the potency can
be calculated.
0.050
Slope of standard:
B 6 x 301.1 - 60 x 11.04
b's = 6.356
0.000 - ' - - - - - - - - - - - - - - - - - - - - - - - ' 180

Figure 5.2.1.-1. Slope of T is:


6 x 292.1 - 60 x 11.04
Table 5.2.1.-11. - Analysis of variance b~ = 180 = 6.056

Degrees Proba- 510pe of U is:


Sonree of Snrn of Mean
of F-ratio
variation sqnares sqnare bility 6 x 234.1 - 60 x 11.04
freedorn
b~ = - - - - 1 8 - 0 - - - - = 4.123
Regression 2 0.1917 0.0958 24850 0.000

lntersection 1 3.10. 9 3.10- 9 7· 10" 0.978 This leads to a potency ratio of 6.056/6.356 == 0.953 for
vaccine T and 4.123/6.356 == 0.649 for vaccine U.
Non·linearity 2 2.10- 5 1 . 10- 5 2.984 0.061
6.356 2
Treatments 5 0.1917 e = 6.356 2 _ 1.068 X 2.1792 x 0.0444 = 1.0056
., 3.86. 10- 6
Residual error 42 1.62· 10 K' = 0.0056 x 0.625 = 0.0035

Total 47 0.1919
And the confidence limits are found with formula 3.3.5.1.-4.
5.2.2. A COMPLETELY RANDOMISED (O,4,4,4)-DESIGN
For vaccine T:
An in-vitro assay of influenza vaccines
The haemagglutinin antigen (HA) content of 2 influenza
0.955 ± JO.0056 x 1.913 + 0.0035 x (-1.913) = 0.955 ± 0.063

vaccines is determined by single radial immunodiffusion.


Both have a labelled potency of 15 ¡.tg HA per dose, which is For vaccine U:
equivalent with a content of 30 ¡.tg HA/mL. The standard has 0.649 ± JO.0056 x 1.423 + 0.0035 x (-1.301) = 0.649 ± 0.058
an assigned content of 39 ¡.tg HA/mL.
Standard and test vaccines are applied in 4 duplicate The HA content in ¡.tg/ dose can be found by multiplying the
concentrations which are prepared on the basis of the assigned potency ratios and confidence Iimits by the assumed content
and the labelled contents. When the equilibrium between the of 15 ¡.tg/dose. The results are given in Table 5.2.2.-IIL

624 See the informatiol1 section 011 general monographs (caver pages)
EUROPEAN PHARMACOPOEIA 8.0 5.3. Statistkal analysis

40 84 5.3. QUANTAL RESPONSES


" 5.3.1. PROBIT ANALYSIS OF A TEST PREPARATION
35 AGAINST A REFERENCE
An in-vivo assay of a diphtheria vaccine
30 A diphtheria vaccine (assumed potency 140 IU/vial) is assayed

..
<'l
¡; 25
against a standard (assigned potency 132 IU/vial). On the
basis of this information, equivalent doses are prepared and
randomly administered to groups of guinea-pigs. After a
'"
i¡PO given period, the animals are challenged with diphtheria toxin
and the number of surviving animals recorded as shown in
..=
<'l

~ 15
TI
Table 5.3.1.-1.

VI Table 5.3.1.-1. - Raw datafrom a diphtheria assay in


guinea-pigs
10
Standard (S) Test preparation (1)
Assigned potency Assumed potency
5 132 IV/vial 140 IV/vial

dose chal- protected dose chal- protected


O (IU/mL) lenged (LU./mL) lenged

1.0 12 O 1.0 11 O

Figure 5.2.2.-1. 1.6 12 3 1.6 12 4

2.5 12 6 2.5 11 8

4.0 11 10 4.0 11 10

Table 5.2.2.-11. - Analysis of variance The observations are transferred to the first working table and
the subsequent columns are computed as described in Section
4.2.1. Table 5.3.1.-II shows the first cycle ofthis procedure.
Degrees Proba-
Source of Sum of Mean The sums of the last 6 columns are then calculated per
of F-ratio
variation squares square bility
freedom preparation and transferred to the second working table (see
Table 5.3.1.-III). The results in the other columns are found
Regression 3 1087.7 362.6 339.5 0.000
with formulae 4.2.1.-4 to 4.2.1.-10. This yields a common
Intersection 2 3.474 1.737 1.626 0.237 slope b of 1.655.
Non-linearity 6 5.066 0.844 0.791 0.594 The values for Y in the first working table are now replaced by
a + bx and a second cycle is carried out (see Table 5.3.1.-1V).
Treatments 11 1096.2
The cycle is repeated until the difference between 2 consecutive
Residual error 12 12.815 1.068 cycles has become small. The second working table should
then appear as shown in Table 5.3.1.- V.
Total 23 1109.0
Linearity is tested as described in Section 4.2.2. The x2-value
with 4 degrees offreedom is 0.851 + 1.070 = 1.921 representing
a p-value of 0.750 which is not significant.
Since there are no significant deviations from linearity, the
test for parallelism can be carried out as described in the same
section. The l-value with 1 degree of freedom is
(16.71 + 17 14.152 . 1 f
Table 5.2.2.-III. - Estimates of HA content (Ilg/dose) .27) - - - = 0.001 representmg a p-va ue o
5.89
0.974 which is not significant.
Lower Iimit Estimate Upper limit The ln(potency ratio) can now be estimated as described in
Vacdne T 13.4 14.3 15.3
Section 4.2.3.

Vaccine U 8.9 9.7 10.6 M'T -l.721- (-2.050) = 0.137


2.401

Table 5.3.1.-II. - First working table in the first cycle


Vac-
Dose n r x p y <I> Z y w wx wy wx' wy' wxy
cine

S 1.0 12 O 0.000 0.000 O 0.5 0.399 -1.253 7.64 0.00 - 9.57 0.00 12.00 0.00

1.6 12 3 0.470 0.250 O 0.5 0.399 -0.627 7.64 3.59 - 4.79 1.69 3.00 - 2.25

2.5 12 6 0.916 0.500 O 0.5 0.399 0.000 7.64 7.00 0.00 6.41 0.00 0.00

4.0 11 10 1.386 0.909 O 0.5 0.399 1.025 7.00 9.71 7.18 13.46 7.36 9.95

T 1.0 11 O 0.000 0.000 O 0.5 0.399 - 1.253 7.00 0.00 - 8.78 0.00 11.00 0.00

1.6 12 4 0.470 0.333 O 0.5 0.399 - 0.418 7.64 3.59 - 3.19 1.69 1.33 - 1.50

2.5 11 8 0.916 0.727 O 0.5 0.399 0.570 7.00 6.42 3.99 5.88 2.27 3.66

4.0 11 10 1.386 0.909 O 0.5 0.399 1.025 7.00 9.71 7.18 13.46 7.36 9.95

General Notices (1) apply to all monographs and other texts 625
5.3. Statistical analysis EUROPEAN PHARMACOPOEIA 8.0

Table 5.3.1.-III. - Secand working table in the first cycle


Vaccine LW LWX LWy LWx' Lwi LWxy Sxx Sxy Syy
¡j)
11 a

S 29.92 20.30 - 7.18 21.56 22.36 7.70 7.79 12.58 20.64 0.68 - 0.24 - 1.36

T 28.65 19.72 - 0.80 21.03 21.97 12.11 7.46 12.66 21.95 0.69 - 0.03 - 1.17

Table 5.3.1.-IV. - First working table in the second cycle


Vac- y
Dose n r x p <l> Z y W wx wy wx' wi wxy
cine

S 1.0 12 O 0.000 0.000 - l.36 0.086 0.158 - 1.911 3.77 0.00 - 7.21 0.00 13.79 0.00

1.6 12 3 0.470 0.250 - 0.58 0.279 0.336 - 0.672 6.74 3.17 - 4.53 1.49 3.04 - 2.13

2.5 12 6 0.916 0.500 0.15 0.561 0.394 - 0.001 7.57 6.94 - 0.01 6.36 0.00 - 0.01

4.0 11 10 l.386 0.909 0.93 0.824 0.258 l.260 5.07 7.03 6.39 9.75 8.05 8.86

T 1.0 11 O 0.000 0.000 - 1.17 0.122 0.202 - 1.769 4.20 0.00 - 7.43 0.00 13.14 0.00

1.6 12 4 0.470 0.333 - 0.39 0.349 0.370 - 0.430 7.23 3.40 - 3.11 1.60 l.34 - 1.46

2.5 11 8 0.916 0.727 0.35 0.637 0.375 0.591 6.70 6.14 3.96 5.62 2.34 3.63

4.0 11 10 l.386 0.909 1.13 0.870 0.211 l.311 4.35 6.03 5.70 8.36 7.48 7.90

Table 5.3.1.-V. - Secand working table after sufficient cycles


Vaccine LW LWX LWy LWx' Lwi LWXy Sxx Sxy Syy ¡j)
11 a

S 18.37 14.80 - 2.14 14.85 17.81 5.28 2.93 7.00 17.56 0.81 - 0.12 - 2.05

T 17.96 12.64 - 0.55 11.86 18.35 6.76 2.96 7.15 18.34 0.70 - 0.03 -1.72

Further: rather than an alternative to the probit method in this specific


case. Another shape of the curve may be adopted only if this
2.401 2 x 5.893
e = 2.401 2 )( 5.893 - 1 2 X l.960 2
= l.127 is supported by experimental or theoretical evidence. See
Table 5.3.2.- I.
1 1
V = 18.37 + 17.96 = O.llO Table 5.3.2.-1. - Results by using alternative curves
So In confidence limits are: Logit Gompit Angle*
0.155 - 0.013 ± ylO.127 (0.649 + l.127 x 0.036 2 ) = 0.142 ± 0.288
<t> 1 1 - e -e Y 1 1
---
+ e-Y - sin y+-
The potency and confidence limits can now be found by taking 1 2 2
the antilogarithms and multiplying these by the assumed 1
Z e-Y eY-e Y
potency of 140 IU/vial. This yields an estimate of 160.6 IU/vial - cos Y
with 95 per cent confidence limits from 121.0-215.2 IU/vial. (1 + e- y )2 2

slope b 4.101 2.590 1.717


1.0

0.9 tI . x'lin 2.15 3.56 1.50

0.8 t X' par 0.0066 0.168 0.0010

Potency 162.9 158.3 155.8


0.7

:5 0.6 Lower limit 12l.í 118.7 122.6

:ci
J<:; 0.5

¡
Upper ¡imit 221.1 213.3 200.7
..
Q..

,.¡
0.4

0.2
t If Y

* If Y >
<-

~:
1
-71" then

then ip
ip = O and Z

= 1 and Z = O
= O

0.1
5.3.3. THE EDso DETERMINATION OF A SUBSTANCE
S T
USING THE PROBIT METHOD
o.o~~~--------------~------------------~
An in-vitro assay of oral poliomyelitis vaccine
Figure 5.3.1.- I. In an EDso assay of oral poliomyelitis vaccine with 10 different
5.3.2. LOGIT ANALYSIS AND OTHER TYPES OF ANALYSES dilutions in 8 replicates of 50 flL on an ELISA-plate, results
OFA TEST PREPARATION AGAINST A REFERENCE were obtained as shown in Table 5.3.3.-1.
Results will be given for the situation where the logit method The observations are transferred to the first working table and
and other "c1assicaI" methods of this family are applied to the the subsequent columns are computed as described in Section
data in Section 5.3.1. This should be regarded as an exercise 4.2.1. Table 5.3.3.-II shows the first cycle ofthis procedure.

626 See the information section on general monographs (caver pages)


EUROPEAN PHARMACOPOEIA 8.0 5.3. Statistical analysis

Table 5.3.3.-1. - Dilutions (10x [AL of the undiluted vaccine) So In confidence limits are:
-3.5 -4.0 -4.5 - 5.0 - 5.5 -6.0 -6.5 -7.0 -7.5 -8.0 -14.692- (-2.420)± VO.197X (2.882+1.197XO.OO9 2 )

+ + + + =-12.272±O.754

+ + + +

+ + This estimate is still expressed in terms ofthe ln(dilutions). In


order to obtain estimates exoressed in ln(EDso)/mL the values
+ + + +
are trans f;orme d to -MT+1n
I 50 .
(1000)
+ + +
Since it has become common use to express the potency of
+ + + + + this type of vaccine in terms of 10glO(EDso)/mL, the results
+ + + + + + have to be divided by ln(lO). The potency is thus estimated as
6.63 10g¡o(EDso)/mL with 95 per cent confidence limits from
+ + + + + 6.30 to 6.9610g¡o(ED so )/mL.
The sums of the last 6 columns are calculated and transferred
to the second working table (see Tab1e 5.3.3.-III). The results
in the other co1umns are found with formula e 4.2.1.-4 to 1.0
4.2.1.-10. This yields a common slope b of - 0.295.
0.9
The values for Y in the first working table are now replaced by
a + bx and a second cycle is carried out. The cycle is repeated
"
0.8
until the difference between 2 consecutive cycles has become
small. The second working table should then appear as shown 0.7
in Table 5.3.3.-IV.
Linearity is tested as described in Section 4.2.2. The x2-value E 0.6
with 8 degrees of freedom is 2.711 representing a p-value of :sji 0.5
0.951 which is not significant.
The potency ratio can now be estimated as described in
e
Q.. 0.4
Section 4.5.
- (-7.931) 0.3
-12.273
-0.646
0.2
Further:
0.1
(-0.646)2 X 55.883
e =
(-0.646)2 X 55.883 - 12 X l.960 2
= 1.197
0.0 - ' - - - - - - - - - - - - - - - - - - - _ < > _ - = = = 0 - - - '

v = = 0.052
19.39 Figure 5.3.3.-1.

Table 5.3.3.-11. - First working table in the first cycle


Vaccine Dose n r x p y <I> Z y W wx wy wx' wy' wxy

T 10- 35 8 o - 8.06 0.000 0.00 0.5 0.399 - 1.253 5.09 - 41.04 - 6.38 330.8 8.00 51.4

10- '.0 8 o - 9.21 0.000 0.00 0.5 0.399 - 1.253 5.09 - 46.91 - 6.38 432.0 8.00 58.8
10- 45 8 1 - 10.36 0.125 0.00 0.5 0.399 - 0.940 5.09 - 52.77 - 4.79 546.8 4.50 49.6
10- 5.0 8 2 - 11.51 0.250 0.00 0.5 0.399 - 0.627 5.09 - 58.63 - 3.19 675.1 2.00 36.7
10- 5.5 8 6 - 12.66 0.750 0.00 0.5 0.399 0.627 5.09 - 64.50 3.19 816.8 2.00 - 40.4
10- 60 8 7 - 13.82 0.875 0.00 0.5 0.399 0.940 5.09 -70.36 4.79 972.1 4.50 - 66.1
10- 6.5 8 7 - 14.97 0.875 0.00 0.5 0.399 0.940 5.09 -76.23 4.79 1140.8 4.50 - 71.7

10- 70
8 8 -16.12 1.000 0.00 0.5 0.399 1.253 5.09 - 82.09 6.38 1323.1 8.00 -102.9

10- 7.5 8 8 -17.27 1.000 0.00 0.5 0.399 J.253 5.09 - 87.95 6.38 1518.9 8.00 -110.2
10- 80 8 8 -18.42 1.000 0.00 0.5 0.399 1.253 5.09 -93.82 6.38 1728.2 8.00 -117.6

Tab1e 5.3.3.-III. - Second working table in the first cycle


Vacdlle LW LWX LWy LWX2 LWy' :Ewxy S., Sxy Syy ¡¡; y a

T 50.93 - 674.3 11.17 9484.6 57.50 - 312.32 556.92 - 164.43 55.05 - 13.24 0.219 - 3.690

Table 5.3.3.-1V. - Second warking table after sufficient cycles


VacCÍlle :Ew :Ewx LWy LWx' LWy' LWXy Sxx Sxy Sy!, ¡¡; y a

T 19.39 - 238.2 0.11 2981.1 26.05 - 37.45 55.88 - 36.11 26.05 - 12.28 0.006 - 7.931

General Natices (1) apply to all manographs and other texts 627
5.3. Statistical analysis EUROPEAN PHARMACOPOEIA 8.0

5.4. EXTENDED SIGMOID DOSE-RESPONSE CURVES


=y+ ( ~)-'"
a-5
5.4.1. FOUR-PARAMETER LOGISTIC CURVE ANALYSIS y ,6 (x -"'1) Y, Z
A serological assay of tetanus sera Z2 (eY _ 5)2
w
As already stated in Section 3.4, this example is intended to
illustrate a "possible" way to analyse the data presented, but
not necessarily to reflect the "only" or the "most appropriate" z= (1 + e- y )2
way. Many other approaches can be found in the literature,
The resulting weighted analysis of variance of the transformed
but in most cases they should not yield dramatically different
responses (y) using weights (w) is shown in Table 5.4.1.-1I.
outcomes. A short discussion of alternative approaches and
other statistical considerations is given in Section 7.5. Table 5.4.1.-U - Weighted analysis ofvariance
Degrees of X2
A guinea-pig antiserum is assayed against a standard serum Soarce of variatioll
freedom
Probability
(0.4 IU/mL) using an enzyme-linked immunosorbent assay
technique (ELISA). 10 two-fold dilutions of each serum were Preparations 1 0.529653 0.467
applied on a 96-well ELISA plateo Each dilution was applied Regression 1 6599.51 0.000
twice. The observed responses are listed in Table 5.4.1.-1.
Non-parallelism 1 0.0458738 0.830
Table 5.4.1.-1. - Observed responses Non-linearity 16 8.89337 0.918

Standard S Preparation to be examined T Treatments 19 6608.98 0.000

Di!. Obs. 1 Obs. 2 Di!. Obs. 1 Obs.2 Residual error 20 20.0000

l/lO 2.912 2.917 1/10 3.017 2.987 Total 39 6628.98

1/20 2.579 2.654 1/20 2.801 2.808 There are no significant deviations from parallelism and
1/40 2.130 2.212 1/40 2.401 2.450 linearity and thus the assay is satisfactory for potency
calculations. If the condition of equal upper and lower
l/80 1.651 1.638 l/80 1.918 1.963 asymptotes is not fulfilled, significant deviations from linearity
1/160 1.073 0.973 1/160 l.364 1.299
and/or parallelism are likely to occur because the tests for
linearity and parallelism reflect the goodness of fit of the
1/320 0.585 0.666 1/320 0.861 0.854 complete four-parameter model. The residual error in the
analysis of variance is always equal to 1 as a result of the
1/640 0.463 0.356 1/640 0.497 0.496
transformation. However, a heterogeneity factor (analogous
1/1280 0.266 0.234 1/1280 0.340 0.344 to that for the probit model) can be computed.
1/2560 0.228 0.197 1/2560 0.242 0.217
The relative potency of the test preparation can be
obtained as the antilogarithm of Ys - Yr Multiplying by
l/5120 0.176 0.215 1/5120 0.178 0.125 the assigned potency of the standard yields an estímate of
1.459 x 0.4 = 0.584 IU/mL. Formula 4.2.3.-2 gives 95 per cent
For this example, it will be assumed that the laboratory has confidence limits from 0.557-0.612 !U/mL.
validated conditions 1 to 3 in Section 3.1.1 when the assay was
being developed for routine use. In addition, the laboratory
has validated that the upper limit and lower limit of the 6. COMBINATION OF ASSAY RESULTS
samples can be assumed to be equal. 6.1. INTRODUCTION
No unusual features are discovered in a graphical Replication of independent assays and combination of their
representation. A least squares method of a suitable computer results is often needed to fulfil the requirements of the
program is used to fit the parameters of the logistic function, European Pharmacopoeia. The question then arises as to
assuming that the residual error terms are independent and whether it is appropriate to combine the results of such assays
identically distributed normal random variables. In this case, and if so in what way.
3 parameters (ct, ~ and o) are needed to describe the common 2 assays may be regarded as mutually independent when
slope-factor and the common lower and upper asymptotes. the execution of either does not affect the probabilities of
2 additional parameters (Ys and yy) are needed to describe the the possible outcomes of the other. This implies that the
horizontallocation of the 2 curves. random errors in al! essential factors influencing the result
(for example, dilutions of the standard and of the preparation
The following estimates of the parameters are returned by the to be examined, the sensitivity of the biological indicator) in
program: one assay must be independent of the corresponding random
3.196 "'13 -4.307 errors in the other one. Assays on successive days using the
original and retained dilutions of the standard therefore are
1.125 "'IT -4.684 not independent assays.
5 0.145 There are several methods for combining the results of
independent assays, the most theoretically acceptable being
In addition, the estimated residual variance (52) is returned the most difficult to apply. 3 simple, approximate methods are
as 0.001429 with 20 degrees offreedom (within-treatments described below; others may be used provided the necessary
variation). conditions are fulfilled.
Before potencies from assays based on the parallel-iine
In order to obtain confidence limits, and also to check for or probit model are combined they must be expressed in
parallelism and linearity, the observed responses (u) are logarithms; potencies derived from assays based on the
linearised and submitted to a weighted parallel-line analysis slope-ratio model are used as such. As the former models are
by the programo This procedure is very similar to that more common than those based on the slope-ratio model,
described in Section 4.2 for probit analysis with the following the symbol M denoting In potency is used in the formulae
modifications: in this section; by reading R (slope-ratio) for M, the analyst

628 See the information section on general monographs (cover pages)


EUROPEAN PHARMACOPOElA 8.0 5.3. Statistkal analysis

may use the same formulae for potencies derived from assays where the number of degrees of freedom of t equals the sum of
based on the slope-ratio model. AH estimates of potency must the number of degrees of freedom for the error mean squares
be corrected for the potency assigned to each preparation to in the individual assays.
be examined before they are combined. 6.2.4. WEIGHTED MEAN AND CONFIDENCE LIMITS
6.2. WEIGHTED COMBINATION OF ASSAY RESULTS BASED ON THE INTRA- AND INTER-ASSAY VARIATION
When results of several repeated assays are combined, the
This method can be used provided the following conditions (x2-value may be significant. The observed variation is then
are fulfilled:
considered to have two components:
1) the potency estimates are derived from independent assays; - the intra-assay variation s~ = l/W,
2) fol' each assay C is close to 1 (say less than 1.1); ¿(M_M)2
3) the number of degrees of freedom of the individual residual - the inter-assay variation S2M = ( )
ni n ' -1
errors is not smaller than 6, but preferably larger than 15;
where M is the unweighted mean. The former varies from
4) the individual potency estimates form a homogeneous set
assay to assay whereas the latter is common to all M.
(see Section 6.2.2).
For each M a weighting coefficient is then cakulated as:
When these conditions are not fulfilled this method cannot be
applied. The method described in Section 6.3 may then be W' = 1
used to obtain the best estimate of the mean potency to be 2
SM
+ SM
2
adopted in further assays as an assumed potency.
which replaces W in Section 6.2.3. where t is taken to be
6.2.1. CALCULATION OF WEIGHTING COEFFICIENTS approximately 2.
It is assumed that the results of each of the ni assays have been
analysed to give ni values of M with associated confidence 6.3. UNWEIGHTED COMBINATION OF ASSAY RESULTS
limits. For each assay the logarithmic confidence interval L To combine the ni estimates of M from ni assays in the simplest
is obtained by subtracting the lower limit from the upper. A way, the mean is calculated and an estimate of its standard
weight W for each value of M is calculated from equation deviation is obtained by calculating:
6.2.1.-1, where t has the same value as that used in the
calculation of confidence limits. 2 ¿(M_M)2 (6.3.-1)
s--
M
= -~~-~-
ni (ni - 1)
4t 2 (6.2.1.-1)
W=-
L2 and the limits are:
6.2.2. HOMOGENEITY OF POTENCY ESTIMA TES
By squaring the deviation of each value of M from the weighted
M ± tS M (6.3.-2)
mean, multiplying by the appropriate weight and summing where t has (ni - 1) degrees offreedom. The number ni of
over al! assays, a statistic is obtained which is approximately estimates of Mis usually small, and hence the value of t is
distributed as X2 (se e Table 8.3) and which may be used to test quite large.
the homogeneity of a set of In potency estimates:
6.4. EXAMPLE OF A WEIGHTED MEAN POTENCY WITH
~ ¿W (M - M)2 where M _ ¿WM CONFIDENCE LIMlTS
ni - ¿W
Table 6.4.-1 lists 6 independent potency estimates of the same
(6.2.2.-1) preparatiol1 together with their 95 per cent confidence limits
and the number of degrees of freedom of their error variances.
lf the calculated X2 is smaller than the tabulated value Conditions 1, 2 and 3 in Section 6.2. are met. The In potencies
corresponding to (ni - 1) degrees of freedom the potencies are and the weights are calculated as described in Section 6.2.
homogeneous and the mean potency and limits obtained in
Section 6.2.3 will be meaningful. Table 6.4.-1. - Potency estimates and confidence intervals of
6 independent assays
lf the calculated value of this statistic is greater than the
tabulated value, the potencies are heterogeneous. This means Potency Lower Upper Degrees In Weight
that the variation between individual estimates of M is greater estimate limit limit of po- W
than would have been predicted from the estimates of the (IV/vial) (IV/vial) (IV/vial) free- teney
confidence limits, i.e. that there is a significant variability dom M
between the assays. Under these circumstances condition 4 is
18367 17755 19002 20 9.8183 3777.7
not fulfilled and the equations in Section 6.2.3 are no longer
applicable. Instead, the formulae in Section 6.2.4 may be used. 18003 17415 18610 20 9.7983 3951.5
6.2.3. CALCULATION OF THE WEIGHTED MEAN AND
18064 17319 18838 20 9.8017 2462.5
CONFIDENCE LIMITS
The products WM are formed for each assay and their sum 17832 17253 18429 20 9.7887 4003.0
divided by the total weight for all assays to give the logarithm
of the weighted mean potency. 18635 17959 19339 20 9.8328 3175.6

- _ ¿WM (6.2.3.-1) 18269 17722 18834 20 9.8130 4699.5


M - ¿W
Homogeneity of potency estimates is assessed with formula
The standard error of the In (mean potency) is taken to be the 6.2.2.-1 which gives a X2 of 4.42 with 5 degrees of freedom.
square root of the reciprocal of the total weight: This is not significant (p = 0.49) and thus all conditions are
met.
(6.2.3.-2) A weighted mean potency is calculated with formula 6.2.3.-1
which yields 9.8085.
and approximate confidence limits are obtained from the Formula 6.2.3.-2 gives a standard deviation of 0.00673 and
antilogarithms of the value given by approximate 95 per cent confidence limits of 9.7951 and
(6.2.3.-3) 9.8218 are calculated with formula 6.2.3.-3 where t has
120 degrees of freedom.

General Notices (1) apply to all monographs and other texts 629
5.3. Statistical analysis EUROPEAN PHARMACOPOEIA 8.0

By taking the antilogarithms a potency of 18 187 IU/vial 7.3. OUTLIERS AND ROBUST METHODS
is found with 95 per cent confidence limits from The method of least squares described in this annex has the
17 946-18 431 IV/vial. disadvantage of being very sensitive to outliers. A dear outlier
may completely corrupt the calculations. This problem is often
remedied by discarding the outlying result from the dataset.
This policy can lead to arbitrary rejection of data and is not
7. BEYOND THIS ANNEX always without danger. It is not easy to give a general guideline
on how to decide whether or not a specific observation is an
It is impossible to give a comprehensive treatise of statistical outlier and it is for this reason that many robust methods
methods in a pharmacopoeial text. However, the methods have been developed. These methods are less sensitive to
described in this annex should suffice for most pharmacopoeial outliers because they give less weight to observations that are
purposes. This section tries to give a more abstract survey of far away from the predicted value. New problems usually arise
alternative or more general methods that have been developed. in computing confidence intervals or defining a satisfactory
The interested reader is encouraged to further explore the fU11ction to be minimised.
existing literature in this area. The use of more specialised
statistical methods should, in any case, be left to qualified 7.4. CORRELATED ERRORS
personnel. Absolute randomisation is not always fe asible or very
undesirable from a practical point of view. Thus, subsequent
7.1. GENERAL LINEAR MODELS doses within a dilution series often exhibit correlated errors
The methods given in this annex can be described in terms leading to confidence limits that are far too narrow. Sorne
of general linear models (or generalised linear models to methods have been developed that take account of this
indude the probit and logit methods). The principIe is to autocorrelation effect.
define a linear structure matrix X (or design matrix) in which
each row represents an observation and each column a linear 7.5. EXTENDED NON-LINEAR DOSE-RESPONSE CURVES
effect (preparation, block, column, dosel. For example: the Analysis of extended non-linear dose-response curves raises a
Latin square design in example 5.1.2 would involve a matrix number of statistical questions which require consideration,
with 36 rows and 13 columns. 1 column for each of the and for which professional advice is recommended. Sorne of
preparations, 1 column for the doses, 5 columns for each these are indicated below.
block except the first, and 5 columns for each row except the 1) An example using the four-parameter logistic function
first. All columns, except the one for doses, are filled with O or has been shown. However, models based on functions giving
1 depending on whether or not the observation relates to the other sigmoid curves may also be used. Models incorporating
effect. A vector Y is fiUed with the (transformed) observations. additional asymmetry parameters have been suggested.
The effects are estimated with the formula (xtxt iX'Y from 2) Heterogeneity of varianCe is common when responses
which the potency estimate m can easily be derived as a ratio cover a wide range. lf the analysis ignores the heterogeneity,
of relevant effects. Confidence intervals are calculated from interpretation of results may not be correct and estima tes

*)]
Fieller's theorem: may be biased. Use of the reciprocal of the error variances
as weights is unlikely to be reliable with limited numbers of
[m - ~ ± ~ Vl1 - 2mV12 +m 2 v22 - g (Vl1 - replicates. 1t may be appropriate to estimate a function which
relates varia11ce to mean response.
m"m[J (l-g)
3) The statistical curve-fitting procedures may give different
estimates depending on assumptions made about the
homogeneity of the variance and on the range of responses
used.
and Vil' V22' V 12 represent the variance multipliers for the
numerator, the denominator and their covariance multiplier 4) In principIe, equality of upper and lower response limits for
respectively. These are taken directly from (X'xt i or the different preparations induded in an assay can be directly
indirectly by noting that: tested in each assay. However, interpretation of the results of
these tests may not be straightforward. The tests for linearity
Var(a i - a2 ) = Var(a¡) + Var(a 2)-2Cov(al'a 2 ) and parallelism given by the simplified method of analysis
(Example 5.4.1) indirectly incorporate tests for equality and
and Cov(a¡ - a2 ,b) = Cov(a¡ ,b) - Cov(a z ,b) accuracy of upper and lower limits.
5) Many assays indude "controls" which are intended to
A full analysis of variance in which all components are identify the upper and/or lower response limits. However,
partitioned is slightly more complicated as it involves a these values may not be consistent with the statistically fitted
renewed definition of X with more columns to relax the upper and lower response limits based on the extended
assumptions of parallelism and linearity, after which the dose-response curve. .
linear hypotheses can be tested. For assays depending upon 6) The simplified method of analysis given in Example 5.4.1
quantal responses the linear effects (intercepts as' aT etc. and provides approximate confidence intervals. Other methods
the common slope b are faund by maximising the sum over may also be used, for example intervals based 011 lack-of-fit of
treatments of nln <D(a¡ + bx) + (n - r)ln(1 - <D(a¡ + bx)) where the completely specified model. For typical assay data, with
x is the ln(dose), <D denotes the shape ofthe distribution and responses covering the complete range for each preparation
i E {S, T, ... }. tested, all methods give similar results.
7.2. HETEROGENEITY OF VARIANCE 7.6. NON-PARALLELISM OF DOSE-RESPONSE CURVES
Heterogeneity of variance cannot always be solved by simply Similarity of dose-response relationships is a fundamental
transforming the responses. A possible way to cope with this criterion for assessing whether an assay may be regarded
problem is to perform a weighted linear regression. In order as a dilution assay and hence whether the estimation of
to obtain an unbiased estimate, the weight of the observations relative potency is valid (see Section 3.1.1). This criterion
is taken to be proportional to the reciprocal of the error is frequently met by showing that dose-response curves for
variances. Since the true error variance is not always known, standard and test samples do not deviate significantly from
an iterative reweighted linear procedure may be followed. parallelism. Underestimation of the residual error can lead
However, the calculation of the confidence interval involves to excess rejection of assays due to significant deviations
new problems. from parallelism and/or linearity. This is often an artefact of

630 See the infarmatian sectian an general managraphs (caver pages)


EUROPEAN PHARMACOPOEIA 8.0 5.3. Statistical analysis

inappropriate assay design or analysis. Minor modifications extensive tables. Many computer programs indude statistical
to assay designs might in many cases substantially improve functions and their use is recommended instead of the tables
the estimation of the residual error. Analysis allowing for in this section. Alternatively, the generating procedures given
the actuallevel of replication may also improve the situation. below each table can be used to compute the probability
If estimation of the relevant residual error is not fe asible corresponding to a given statistic and number of degrees of
for individual assays, for example because it is impractical freedom.
to create independent doses and/or replicates, it might be
possible to obtain a more correct estímate of the residual error
during the assay validation process. There may also be cases
8.1. THE F-DISTRIBUTION
where the assay system is sufficiently precise to detect slight
but genuine non -parallelism. lf there is true non -parallelism lf an observed value is higher than the value in Table 8.1.-1, it
this needs to be recognised and a suitable solution adopted. A is considered to be significant (upper lines, p = 0.05) or highly
solution might, for example, require a suitable standard that significant (lower lines, p = 0.01). dfl is the number of degrees
is similar in composition to, and therefore parallel to, the test of freedom of the numerator and df2 is the number of degrees
samples. lf the assay system is responding in a non-specific of freedom of the denominator.
manner to extraneous components of the standard or test
samples, then a more specific assay system that does not Generating procedure. Let F be the F-ratio and dfl and df2
respond to the irrelevant components may be the solution. as described aboye. Let pi = 7r = 3.14159265358979 ... The
No simple, generally applicable statistical solution exists to procedure in Table 8.l.-U will then generate the p-value.
overcome these fundamental problems. The appropriate
action has to be decided on a case-by-case basis with the help
of statistical expertise.
8.2. THE t-DISTRIBUTION

lf an observed value is higher than the value in Table 8.2.-1, it


is considered to be significant (p = 0.05) or highly significant
(p = 0.01).

8. TABLES AND GENERATING Generating procedures. The p-value for a gíven t with df
degrees of freedom can be found with the pro ce dures in
PROCEDURES e,
Section 8.1 where F = dfl = 1 and df2 = df.

The tables in this section list the critical values for the most The t-value (p = 0.05) for a given number of degrees of
frequently occurring numbers of degrees of freedom. lf a freedom df can be found with the procedure in Table 8.2.-Ir,
critical value is not listed, reference should be made to more which should be accurate up to 6 decimal places.

Table 8.1.-1 - Critical values of the F-distribution

dfl-i> 2 3 4 5 6 8 10 12 15 20

df2.J,
10 4.965 4.103 3.708 3.478 3.326 3.217 3.072 2.978 2.913 2.845 2.774 2.538

10.044 7.559 6.552 5.994 5.636 5.386 5.057 4.849 4.706 4.558 4.405 3.909

12 4.747 3.885 3.490 3.259 3.106 2.996 2.849 2.753 2.687 2.617 2.544 2.296

9.330 6.927 5.953 5.412 5.064 4.821 4.499 4.296 4.155 4.010 3.858 3.361

15 4.543 3.682 3.287 3.056 2.901 2.790 2.641 2.544 2.475 2.403 2.328 2.066

8.683 6.359 5.417 4.893 4.556 4.318 4.004 3.805 3.666 3.522 3.372 2.868

20 4.351 3.493 3.098 2.866 2.711 2.599 2.447 2.348 2.278 2.203 2.124 1.843

8.096 5.849 4.938 4.431 4.103 3.871 3.564 3.368 3.231 3.088 2.938 2.421

25 4.242 3.385 2.991 2.759 2.603 2.490 2.337 2.236 2.165 2.089 2.007 1.711

7.770 5.568 4.675 4.177 3.855 3.627 3.324 3.129 2.993 2.850 2.699 2.169

30 4.171 3.316 2.922 2.690 2.534 2.421 2.266 2.165 2.092 2.015 1.932 1.622

7.562 5.390 4.510 4.018 3.699 3.473 3.173 2.979 2.843 2.700 2.549 2.006

50 4.034 3.183 2.790 2.557 2.400 2.286 2.130 2.026 1.952 1.871 1.784 1.438

7.171 5.057 4.199 3.720 3.408 3.186 2.890 2.698 2.563 2.419 2.265 1.683

3.841 2.996 2.605 2.372 2.214 2.099 1.938 1.831 1.752 1.666 1.571 1.000

6.635 4.605 3.782 3.319 3.017 2.802 2.511 2.321 2.185 2.039 1.878 1.000

General Notices (1) apply to all rnonographs and other texts 631
5.3. Statistical analysis EUROPEAN PHARMACOPOEIA 8.0

Table 8.1.-II - Generating procedure for the F-distribution


If dfl is even If dfl is odd and df2 is even If dfl and df2 are odd
x=df1/(df1+df2/F) x=df2/(df2+df1*F) x=atn(sqr(df1*F/df2) )

s=l cs=cos(x)

sn=sin(x)

for Í=2 to (dfl-2) step 2 for Í=2 to (df2-2) step 2 x=x/2

t=t*x*(df2+i-2)/i t=t*x*(df1+i-2)/i

s=s+t s=s+t t=sn*cs/2

next i next i v=O

for Í=2 to (df2 1) step 2

s=s+t

t=t*i/(i+1)*cs*cs

next i

for Í=1 to (dfl-2) step 2

v=v+w

w=w*(df2+i)/(i+2)*sn*sn

next i

p=1+(t*df2*v-x-s)/pi*4

Table 8.2.-1 - Critical values of the t-distribution Table 8.2.-II - Generating procedure for the t-distribution
df p ~ 0.05 P = 0.01 df P = 0.05 P = 0.01
t = 1.959964+
12.706 63.656 22 2.074 2.819
2.37228/df+
2 4.303 9.925 24 2.064 2.797
2.82202/dC2+
3.182 5.841 26 2.056 2.779
2.56449/df A3+
4 2.776 4.604 28 2.048 2.763
1.51956/dfA4+
2.571 4.032 30 2.042 2.750
1.02579/df A5+
6 2.447 3.707 35 2.030 2.724
0.44210/dC7
7 2.365 3.499 40 2.021 2.704

8 2.306 3.355 45 2.014 2.690

9 2.262 3.250 50 2.009 2.678

10 2.228 3.169 60 2.000 2.660

12 2.179 3.055 70 1.994 2.648

14 2.145 2.977 80 1.990 2.639

16 2.120 2.921 90 1.987 2.632

18 2.101 2.878 100 1.984 2.626

20 2.086 2.845 1.960 2.576

632 See the information section on general monographs (caver pages)


EUROPEAN PHARMACOPOEIA 8.0 5.3. Statistical analysis

8.3. THE X2- DISTRIBUTION Table 8.4.-1 - Values of the <D-distribution

x <D x <D x <1>


Table 8.3.-1 - Critical values of the X2-distribution
0.00 0.500 l.00 0.841 2.00 0.977
df p = 0.05 P = 0.01 df P = 0.05 P = 0.01
0.05 0.520 l.05 0.853 2.05 0.980
3.841 6.635 11 19.675 24.725
0.10 0.540 l.l0 0.864 2.10 0.982
2 5.991 9.210 12 2l.026 26.217
0.15 0.560 l.l5 0.875 2.15 0.984
3 7.815 11.345 13 22.362 27.688
0.20 0.579 l.20 0.885 2.20 0.986
4 9.488 13.277 14 23.685 29.141
0.25 0.599 l.25 0.894 2.25 0.988
5 1l.070 15.086 15 24.996 30.578
0.30 0.618 1.30 0.903 2.30 0.989
6 12.592 16.812 16 26.296 32.000
0.35 0.637 1.35 0.911 2.35 0.991
7 14.067 18.475 20 3l.41O 37.566
0.40 0.655 l.40 0.919 2.40 0.992
15.507 20.090 25 37.652 44.314
0.45 0.674 1.45 0.926 2.45 0.993
9 16.9l9 2l.666 30 43.773 50.892
0.50 0.691 l.50 0.933 2.50 0.994
10 18.307 23.209 40 55.758 63.691
0.55 0.709 l.55 0.939 2.55 0.995

lf an observed value is higher than the value in Table 8.3.-1, it 0.60 0.726 l.60 0.945 2.60 0.995
is considered to be significant (p = 0.05) or highly significant
(p = 0.01). 0.65 0.742 l.65 0.951 2.65 0.996

0.70 0.758 l.70 0.955 2.70 0.997


Generating procedure. Let X2 be the x2-value and df as
described aboye. The procedure in Table 8.3.-II will then 0.75 0.773 l.75 0.960 2.75 0.997
generate the p-value.
0.80 0.788 l.80 0.964 2.80 0.997

0.85 0.802 l.85 0.968 2.85 0.998

0.90 0.816 l.90 0.971 2.90 0.998

Table 8.3.-II - Generating procedure for the X2 -distribution 0.95 0.829 l.95 0.974 2.95 0.998

If df is even If df is odd Table 8.4.-II - Generating procedure for the cJJ-distribution


s=O x=sqr(x2)
s=O
t=exp(-x2/2) s=O
t=x
for i=2 to df step 2 t=x*exp (-x2/2) / sqr (pi/2)
i=l
s=s+t for i=3 to df step 2
repeat
t=t*x2/i s=s+t s=s+t
next i t=t*x2/i i=i+2
p=l-s next i t=t*x*x/i
p=1-s-2*phi(x) until t<lE-16

phi=0.S+s*exp(-x*x/2)/sqr(2*pi)
In this procedure phi is the cumulative standard normal
distribution function <D (see Section 8.4).

8.5. RANDOM PERMUTATIONS


8.4. THE <D-DISTRIBUTION (THE CUMULATIVE
STANDARD NORMAL DISTRIBUTION) Random permutations are needed in randomised block
designs. The following algorithm shows how the built-in
random generator of a computer can be used to create random
The <D-value for negative x is found from Table 8.4.-1 as
permutations of N treatments.
1 - <D( -x).
Step l. Write the N possible treatments down in a row.
Generating procedure: Let x be the x-value. The procedure
in Table 8.4.-II will generate the corresponding <D-value if Step 2. Obtain a random integer r such that 1~ r ~ N.
O ~ x ~ 8.15. rf x is greater than 8.15 the <D-value can be set to
1. rf x is negative, the formula given aboye can be used. This Step 3. Exchange the r-th treatment with the N-th treatment
procedure assumes that the computer can represent about 15 in the row.
decimal places. Ifless digits or more digits can be represented,
the procedure needs sorne trivial modifications. Step 4. Let N =N - 1 and repeat steps 2 to 4 until N = 1.

General Notices (1) apply to all monographs and other texts 633
5.3. Statistical analysis EUROPEAN PHARMACOPOEIA 8.0

An example with 6 treatments will illustrate this algorithm. 3 4 6 2 5

1. N =6 5, 5, T, T, 2 l' , 5, T2 l' , 5,
2. r = 2
3 52 5, S, 1',
3. T, 5,
6 5, T, 53 T,
4. N= 5
1'2 52 T3 53
2. r= 4
4 T, 5, T3 53
3. 5, T, 5,
4. N=4 5 53 T2 5, T,

2. r= 4

3. T3 5, T, l', 5,
2 3 4 5 6
4. N= 3
1', 5,
2. r=1
2 5, T, T, 53
3. 5, T, T2 52
4. N= 2 3 T] 52 T3 T, 53

2. r=1 4 53 52 1',

3. T, 53 T, 5, 5 T3 T, 5, 5, T,
4. N= 1
6 T, 53 l', 5, 5] T3

8.6. LATIN SQUARES

The following example shows how 3 independent permutations 9. GLOSSARY O F SYMBO LS


can be used to obtain a Latin square.

1) Generate a random permutation of the N possible Symbol Definition


treatments (see Section 8.5):
a Intersection of linear regression of responses
on dose or In (dos e)
53 1', 1'] 5,
b Slope of linear regression of responses on
2) A simple Latin square can now be constructed by "rotating" dose or on ln(dose)
this permutation to the right. This can be done as follows.
Write the permutation found in step 1 down on the first row. d Number of dose levels for each preparation
The second row consists of the same permutation, but with al! (excluding the blank in slope-ratio assays)
treatments shifted to the right. The rightmost treatment is put
on the empty place at the left. This is repeated for al! the rows e Base of natural logarithms
until al! the treatments appear once in each column: (= 2.71828182845905 ... )

g Statistic used in Fieller's theorem:


53 5] T, l' , 5,
C-l
g=--
52 53 T2 1', C
53 1', h Number of preparations in an assay,
T, 5, T, 53
including the standard preparation

T2 5, T3 53 m Potency estimate obtained as a ratio of


53 1', 52 T,
effects in general linear models
n Number of replicates for each treatment
3) Generate 2 índependent random permutations of the
figures 1 to N: p Probability of a given statistic being larger
than the observed value. Also used as the
- one for the rows: ratio r/ n in probit analysis
r The number of responding units per
2 3 6 4 5
treatment group in assays depending upon
quantal responses
- and one for the columns:
Estímate of standard deviation (= H)
3 4 6 2 5
Estimate of residual variance given by error
mean square in analysis of variance
4) The Latin square can now be found by sorting the rows
and columns of the simple Latin square according to the
Student's statistic (Table 8.2.)
2 permutations for the rows and columns:

634 See the information section on general monographs (ca ver pages)
EUROPEAN PHARMACOPOEIA 8.0 5.3. Statistical analysis

Symbol Definition Symbol Definition

u Observed response in four-parameter Mean response to the lowest dose 1 up to the


analysis highest dose d of the standard preparation S

Vll'V12,V22 (Co )variance multipliers for numerator and SS Sum of squares due to a given source of
denominator of ratio m in Fieller's theorem variation

w Weighting coefficient r,u,v; ... Test preparations

x The ln(dose) Mean response to the lowest dose 1 up to the


highest dose d of test preparation T
y Individual response or transformed response
v Variance coefficient in the calculation of
A Assumed potencies of test preparations confidence limits
when making up doses
w Weighting factor in combination of assay
B Mean response to blanks in slope-ratio assays results

C Statistic used in the calculation of x Linear structure or design matrix used in


. 1 general linear models
confidence ll1tervals: e = --
l-g
y Vector representing the (transformed)
Mean response to each column of a Latin responses in general linear models
square design
z The first derivative of <D
Mean response on time 1 or time 2 in the
twin cross-over design a Upper asymptote of the ln( dose) -response
curve in four-parameter analysis
F Ratio of 2 independent estimates of variance
following an F-distribution (Table 8.1.) f3 Slope-factor of the ln(dose)-response curve
in four-parameter analysis
Treatment values used in the analysis of
variance for slope-ratio assays y The ln(dose) giving 50 per cent response in
the four-parameter analysis
Multipliers used in the analysis of variance
for parallel-line assays Lower asymptote ofthe ln(dose)-response
curve in four-parameter analysis
Multipliers used in the analysis of variance
for slope-ratio assays 3.141592653589793238 ...

1 In parallel-line assays, the In of the ratio Cumulative standard normal distribution


between adjacent doses. In slope-ratio function (Table 8.4.)
assays, the interval between adjacent doses
Chi-square statistic (Table 8.3.)
Linearity values used in the analysis of
variance for slope-ratio assays
10. LITERATURE
K Correction term used in the calculation of
sums of squares in the analysis of variance This section lists sorne recommended literature for further study.
Finney, D.J. (1971). Pro bit Analysis, 3rd Ed. Cambridge
L Width of confidence interval in logarithms University Press, Cambridge.
Nelder, J.A. & Wedderburn, R.W.M. (1972). Generalized
Linear contrasts of standard and test linear models, ¡oumal of the Royal Statistical Society, Series A
preparations 135, 370-384.
DeLean, A., Munson, P.l., and Rodbard, D. (1978).
In potency ratio of a given test preparation Simultaneous analysis of families of sigmoidal curves:
Application to bioassay, radioligand assay, and physiological
N Total number of treatments in the dose-response curves, Am. 1. Physiol. 235(2): E97-E102.
assay (= dh)
Finney, D,J. (1978). Statistical Method in Biological Assay,
3,d Ed. Griffin, London.
Sum of standard and test preparations
Sakal, R.R. & Rohlf, F.R. (1981). Biometry: Principies and
R Estimated potency of a given test preparation Practice of Statistics in Biological Research, 2nd Ed. W.H.
Freemann & ca, New York.
Potency ratio of a given test preparation Peace, K.E. (1988). Biopharmaceutical Statistics for Drug
Development, Marce! Dekker Inc., New York/Basel.
Rl' ... , RII Mean response in each of rows 1 to n of a Bowerman, B.L. & O'Connell, R.T. (1990). Linear Statistical
Latin square design, or in each block of a Models an Applied Approach, 2 nd Ed. PWS-KENT Publishing
randomised block design Company, Bostan.
Govindarajulu, Z. (2001). Statistical Techniques in Bioassay,
s Standard preparation
2nd revised and enlarged edition, Karger, New York.

General Notices (1) apply lo all monographs and other texts 635
EUROPEAN PHARMACOPOEIA 8.0

636 See the information section on general monographs (caver pages)


EUROPEAN PHARMACOPOEIA 8.0

5.4. Residual solvents


5.4. Residual solvents ............................................................... 639

General Notices (1) apply to all monographs and other texts 637
EUROPEAN PHARMACOPOEIA 8.0

638 See the information section on general monographs (caver pages)


EUROPEAN PHARMACOPOEIA 8.0 5.4. Residual solvents

0112008:50400 IMPURITIES: GUIDELINES


FOR RESIDUAL SOLVENTS
5.4. RESIDUAL SOLVENTS (CPMP /ICH/283/95)
1. INTRODUCTION
LIMITING RESIDUAL SOLVENT LEVELS
IN ACTIVE SUBSTANCES, EXCIPIENTS 2. SCOPE OF THE GUIDELINE
AND MEDICINAL PRODUCTS 3. GENERAL PRINCIPLES
The International Conference on Harmonisation of Technical 3.1. CLASSIFICATION OP RESIDUAL SOLVENTS BY RISK
Requirements for Registration of Pharmaceuticals for Human ASSESSMENT
Use (ICH) has adopted Impurities Guidelines for Residual 3.2. METHODS POR ESTABLISHING EXPOSURE LIMITS
Solvents which prescribes limits for the content of solvents 3.3. OPTIONS POR DESCRIBING LIMITS OP CLASS 2
which may remain in active substances, excipients and SOLVENTS
medicinal products after processing. This guideline, the text
of which is reproduced below, excludes existing marketed 3.4. ANALYTICAL PROCEDURES
products. The European Pharmacopoeia is, however, 3.5. REPORTING LEVELS OP RESIDUAL SOLVENTS
applying the same principIes enshrined in the guideline to
existing active substances, excipients and medicinal products 4. LIMITS OF RESIDUAL SOLVENTS
whether or not they are the subject of a monograph of the 4.1. SOLVENTS TO BE AVOIDED
Pharmacopoeia. AH substances and products are to be tested 4.2. SOLVENTS TO BE LIMITED
for the content of solvents likely to be present in a substance
or producto 4.3. SOLVENTS WITH LOW TOXIC POTENTIAL
Where the limits to be applied comply with those given below, 4.4. SOLVENTS POR WHICH NO ADEQUATE
tests for residual solvents are not generally mentioned in TOXICOLOGICAL DATA WAS POUND
specific monographs since the solvents employed may vary GLOSSARY
from one manufacturer to another and the requirements of
this general chapter are applied via the general monograph APPENDIX 1. LIST OF SOLVENTS INCLUDED IN THE
on Substances for Pharmaceutical Use (2034). The competent GUIDELINE
authority is to be informed of the solvents employed during
the production process. This information is also given APPENDIX 2. ADDITIONAL BACKGROUND
in the dossier submitted for a certificate of suitability of A2.l: ENVIRONMENTAL REGULATION OP ORGANIC
the monographs of the European Pharmacopoeia and is VOLATILE SOLVENTS
mentioned on the certificate. A2.2: RESIDUAL SOLVENTS IN PHARMACEUTICALS
Where only Class 3 solvents are used, a test for loss on drying
may be applied or a specific determination of the solvent may APPENDIX 3. METHODS FOR ESTABLISHING EXPOSURE
be made. If for a Class 3 solvent a justified and authorised limit LIMITS
higher than 0.5 per cent is applied, a specific determination of 1. INTRODUCTION
the solvent is required.
The objective of this guideline is to recommend acceptable
When Class 1 residual solvents or Class 2 residual solvents amounts of residual solvents in pharmaceuticals for the safety
(or Class 3 residual solvents which exceed the 0.5 per cent) of the patient. The guideline recommends the use of les s toxic
are used, the methodology described in the general method solvents and describes levels considered to be toxicologically
(2.4.24) is to be applied wherever possible. Otherwise an acceptable for sorne residual solvents.
appropriate validated method is to be employed.
Residual solvents in pharmaceuticals are defined here as
When a quantitative determination of a residual solvent is organic volatile chemicals that are used or produced in the
carried out, the result is taken into account for the calculation manufacture of active substances or excipients, or in the
of the content of the substance except where a test for drying preparation of medicinal products. The solvents are not
is carried out. completely removed by practical manufacturing techniques.
Appropriate selection of the solvent for the synthesis of active
substance may enhance the yield, or determine characteristics
such as crystal form, purity, and solubility. Therefore, the
solvent may sometimes be a critical parameter in the synthetic
process. This guideline do es not address solvents deliberately
used as excipients nor does it address solvates. However, the
content of solvents in such products should be evaluated and
justified.
Since there is no therapeutic beneflt from residual solvents, aH
residual solvents should be removed to the extent possible to
meet product specifications, good manufacturing practices,
or other quality-based requirements. Medicinal products
should contain no higher levels of residual solvents than can
be supported by safety data. Sorne solvents that are known
to cause unacceptable toxicities (Class 1, Table 1) should be
avoided in the production of active substances, excipients, or
medicinal products unless their use can be strongly justified
in a risk-benefit assessment. Sorne solvents associated with
less severe toxicity (Class 2, Table 2) should be limited in
order to protect patients from potential adverse effects.
IdeaHy, less toxic solvents (Class 3, Table 3) should be used
where practica!. The complete list of solvents included in this
guideline is given in Appendix 1.

General NoNces (1) apply to all monographs and other texts 639
5.4. Residual solvents EUROPEAN PHARMACOPOEIA 8.0

The lists are not exhaustive and other solvents can be used Class 2 solvents: Solvents to be limited
and later added to the lists. Recommended limits of Class 1 Non-genotoxic animal carcinogens or possible causative
and 2 solvents or classification of solvents may change as new agents of other irreversible toxicity such as neurotoxicity
safety data becomes available. Supporting safety data in a or teratogenicity.
marketing application for a new medicinal product containing
Solvents 5uspected of other significant but reversible
a new solvent may be based on concepts in this guideline
toxicities.
or the concept of qualification of impurities as expressed in
the guideline for active substances (Q3A, 1mpurities in New Class 3 solvents: Solvents with low toxic potential
Active Substances) or medicinal products (Q3B, 1mpurities in Solvents with low toxic potential to man; no health-based
New Medicinal Products), or al! three guidelines. exposure limit is needed. Class 3 solvents have PDEs of
50 mg or more per day.
3.2. METHODS POR ESTABLISHING EXPOSURE LIMITS
2. SCOPE OF THE GUIDELINE The method used to establish permitted daily exposures for
residual solvents is presented in Appendix 3. Summaries of the
Residual solvents in active substances, excipients, and in toxicity data that were used to establish limits are published in
medicinal products are within the scope of this guideline. Pharmeuropa, Vol. 9, No.!, Supplement April1997.
Therefore, testing should be performed for residual solvents 3.3. OPTIONS POR DESCRIBING LIMITS OP CLASS 2
when production or purification processes are known to result SOLVENTS
in the presence of such solvents. 1t is only necessary to test Two options are available when setting limits for Class 2
for solvents that are used or produced in the manufacture solvents.
or purification of active substances, excipients, or medicinal
product. Although manufacturers may choose to test the Option 1 : The concentration Iimits in parts per million stated
medicinal product, a cumulative method may be used to in Table 2 can be used. They were calculated using equation (1)
calculate the residual solvent levels in the medicinal product below by assuming a product mass of 10 g administered daily.
from the levels in the ingredients used to produce the
. 1000 x PDE (1)
medicinal producto If the calculation results in a level equal to ConcentratlOl1 (ppm) = - - d - - -
or below that recommended in this guideline, no testing of the ose
medicinal product for residual solvents need be considered. If
however, the calculated level is aboye the recommended leve!, Here, PDE is given in terms of mg/day and close is given
the medicinal product should be tested to ascertain whether in g/day.
the formulation process has reduced the relevant solvent level These limits are considered acceptable for al! substances,
to within the acceptable amount. Medicinal product should excipients, or products. Therefore this option may be applied
also be tested if a solvent is used during its manufacture. if the daily dose is not known or fixed. If all excipients and
active substances in a formulation meet the limits given
This guideline does not apply to potential new active in Option 1, then these components may be used in any
substances, excipients, or medicinal products used during the proportion. No further calculation is necessary provided
clinical research stages of development, nor does it apply to the daily close does not exceed 10 g. Products that are
existing marketed medicinal products. administered in doses greater than 10 g per day should be
considered under Option 2.
The guideline applies to all dosage forms and routes of Option 2: It is not considered necessary for each component
administration. Higher levels of residual solvents may be of the medicinal product to comply with the limits given
acceptable in certain cases such as 5hort term (30 days or less) in Option 1. The PDE in terms of mg/day as stated in
or topical application. Justification for these levels should be Table 2 can be used with the known maximum daily dose
made 011 a case by case basis. and equation (1) aboye to determine the concentration
of residual solvent allowed in a medicinal product. Such
See Appendix 2 for additional background information related limits are considered acceptable provided that is has been
to residual solvents. demonstrated that the residual solvent has been reduced to the
practical minimum. The limits should be realistic in relation
to analytical precision, manufacturing capability, reasonable
3. GENERAL PRINCIPLES variation in the manufacturing process, and the limits should
reflect contemporary manufacturing standards.
3.1. CLASSIFICATION OP RESIDUAL SOLVENTS BY RISK
Option 2 may be appliecl by adding the amounts of a residual
ASSESSMENT
solvent present in each of the components of the medicinal
The term "tolerable daily intake" (TDI) is used by the producto The sum of the amounts of solvent per day should be
International Program on Chemical Safety (IPCS) to describe less than that given by the PDE.
exposure limits of toxic chemicals and "acceptable daily intake"
Consider an example of the use of Option 1 and Option 2
(ADI) is used by the World Health Organization (WHO)
applied to acetonitrile in a medicinal product. The permitted
and other national and international health authorities and
daily exposure to acetonitrile is 4.1 mg per day; thus, the
institutes. The new term "permitted daily exposure" (PDE)
Option 1 limit is 410 ppm. The maximum administered
is defined in the present guideline as a pharmaceutically
daily mass of a medicinal product is 5.0 g, and the medicinal
acceptable intake of residual solvents to avoid confusion of
product contains two excipients. The composition of the
differing values for ADI's of the same substance.
medicinal product and the calculated maximum content of
residual acetonitrile are given in the fol!owing tableo
Residual solvents assessed in this guideline are listed in
Appendix 1 by common names and structures. They were Component Amonnt in Acetonitrile Daily
evaluated for their possible risk to human health and placed formnlation content exposure
into one of three classes as follows: Active substance 0.3 g 800 ppm 0.24 mg

Excipient 1 0.9 g 400 ppm 0.36 mg


Class 1 solvents: Solvents to be avoided
Excipient 2 3.8 g 800 ppm 3.04 mg
Known human carcinogens, strongly suspected human 5.0 g 728 ppm 3.64 mg
Medicinal product
carcinogens, and environmental hazards.

640 See the information section on general monographs (cover pages)


EUROPEAN PHARMACOPOEIA 8.0 5.4. Residual solvents

Excipient 1 meets the Option llimit, but the drug substance, lf solvents of Class 2 or Class 3 are present at greater than
excipient 2, and medicinal product do not meet the Option 1 their Option 1 limits or 0.5 per cent, respectively, they should
limit. Neverthe!ess, the product meets the Option 2 limit of be identified and quantified.
4.1 mg per day and thus conforms to the recommendations
in this guideline. 4. LIMITS OF RESIDUAL SOLVENTS
Consider another example using acetonitrile as residual 4.1. SOLVENTS TO BE AVOIDED
solvent. The maximum administered daily mass of a medicinal Solvents in Class 1 should not be employed in the
product is 5.0 g, and the medicinal product contains two manufacture of active substances, excipients, and medicinal
excipients. The composition of the medicinal product and the products because of their unacceptable toxicity 01' their
calculated maximum content of residual acetonitrile is given deleterious environmental effect. However, if their use is
in the following table. unavoidable in order to produce a medicinal product with
a significant therapeutic advance, then their leve!s should
Component Ammmtin Acetonitrile Daily
formulation content exposure be restricted as shown in Table 1, unless otherwise justified.
1,1,1-Trichloroethane is included in Table 1 because it is an
Active substance 0.3 g 800 ppm 0.24 mg
environmental hazard. The stated limit of 1500 ppm is based
Excipient 1 0.9 g 2000 ppm 1.80 mg on a review of the safety data.
Excipient 2 3.8 g 800 ppm 3.04 mg Table 1. - Class 1 solvents in pharmaceutical products
Medicinal product 5.0 g 1016 ppm 5.08 mg
(solvents that should be avoided)
Solvent Concentration limit Concern
In this example, the product meets neither the Option 1 (I'I'm)
Benzene 2 Carcinogen
nor the Option 2 limit according to this summation. The
manufacturer could test the medicinal product to determine Carbon tetrachloride 4 Toxic and environmental hazard
if the formulation process reduced the leve! oí acetonitrile. lf
1,2-Dichloroethane 5 Toxic
the leve! of acetonitrile was 110t reduced during formulation
to the allowed limit, then the manufacturer of the medicinal 1.1-Dichloroethene 8 Toxic
product should take other steps to reduce the amount of
acetonitrile in the medicinal product. If all of these steps Íail 1,1,1-Trichloroethane 1500 Environmental hazard
to reduce the level of residual solvent, in exceptional cases
the manufacturer could provide a summary of efforts made 4.2. SOLVENTS TO BE LIMITED
to reduce the solvent level to meet the guideline value, and Solvents in Table 2 should be limited in pharmaceutical
provide a risk-benefit analysis to support allowing the product products because oí their inherent toxicity. PDEs are given to
to be utilised containing residual solvent at a higher level. the nearest 0.1 mg/day, and concentrations are given to the
3.4. ANALYTICAL PROCEDURES
nearest 10 ppm. The stated values do not reflect the necessary
analytical precision of determination. Precision should be
Residual solvents are typically determined using determined as part oí the validation of the method.
chromatographic techniques such as gas chromatography.
Any harmonised procedures for determining levels of residual Table 2. - Class 2 solvents in pharmaceutical products
solvents as described in the pharmacopoeias should be used, Solvent PDE Concentration lirnit
if feasible. Otherwise, manufacturers would be free to se!ect (rng/day) (ppm)
the most appropriate validated analytical procedure for a Acetonitrile 4.1 410
particular application. lf only Class 3 solvents are present, a
Chlorobenzene 3.6 360
non-specific method such as loss on drying may be used.
Validation of methods for residual solvents should conform to Chloroform 0.6 60
ICH guidelines "Text on Validation of Analytical Pro ce dures" Cyclohexane 38.8 3880
and "Extension of the ICH Text on Validation of Analytical
Procedures". 1,2-Dichloroethene 18.7 1870

3.5. REPORTING LEVELS OF RESIDUAL SOLVENTS Dichloromethane 6.0 600


Manufacturers of pharmaceutical products need certain 1,2-Dimethoxyethane 1.0 100
information about the content of residual solvents in excipients
or active substances in order to meet the criteria of this N.N- Dimethylacetamide 10.9 1090
guideline. The following statements are given as acceptable N, N-Dimethylformamide 8.8 880
examples of the information that could be provided from a
supplier of excipients or active substances to a pharmaceutical l,4-Dioxane 3.8 380
manufacturero The supplier might choose one of the following 2-EthoA7ethanol 1.6 160
as appropriate:
Ethyleneglycol 6.2 620
- Only Class 3 solvents are likely to be present. Loss on
drying is less than 0.5 per cent. Formamide 2.2 220

- Only Class 2 solvents X, Y, ". are like!y to be present. All Hexane 2.9 290
are below the Option 1 limit
Methanol 30.0 3000
(Here the supplier would name the Class 2 solvents
represented by X, Y, oo.) 2-Methoxyethanol 0.5 50

- Only Class 2 solvents X, Y, ." and Class 3 solvents are MethyJbutylketone 0.5 50
likely to be present. Residual Class 2 solvents are below
Methylcyclohexane 11.8 1180
the Option 1 limit and residual Class 3 solvents are below
0.5 per cent. N-Methylpyrrolidone 5.3 530
rf Class 1 solvents are likely to be present, they should be Nitromethane 0.5 50
identified and quantified. "Like!y to be present" refers to the
solvent used in the final manuÍacturing step and to solvents Pyridine 2.0 200
that are used in earlier manufacturing steps and not removed Sulfolane 1.6 160
consistently by a validated process.

General Notices (1) apply to all monographs and other texts 641
5.4. Residual solvents EUROPEAN PHARMACOPOEIA 8.0

Solvent PDE Concentration limit which to base a PDE was found. Manufacturers should
(mg/day) (ppm)
7.2 720
supply justification for residuallevels of these solvents in
Tetrahydrofuran
pharmaceutical products.
Tetralin 1.0 100
8.9 890
Table 4. - Solvents for which no adequate toxicological data
Toluene
wasfound
1,1,2-Trichloroethene 0.8 80
1,1-Diethoxypropane Methylisopropylketone
Xylene* 21.7 2170
1,1-Dimethoxymethane Methyltetrahydrofuran
*usually 60 per cent m-xylene, 14 per cent p-xylene, 9 per cent o-xylene
with 17 per cent ethyl benzene 2,2-Dimethoxypropane Petroleum ether
lsooctane Trichloroacetic acid
4.3. SOLVENTS WITH LOW TOXIC POTENTIAL
Solvents in Class 3 (shown in Table 3) may be regarded as les s lsopropyl ether Trifluoroacetic acid
toxic and of lower risk to human health. Class 3 includes no
solvent known as a human health hazard at levels normally
accepted in pharmaceuticals. However, there are no long-term
GLOSSARY
toxicity or carcinogenicity studies for many of the solvents in
Class 3. Available data indicate that they are less toxic in acute Genotoxic carcinogens: Carcinogens which produce cancer by
or short-term studies and negative in genotoxicity studies. It affecting genes or chromosomes.
is considered that amounts of these residual solvents of 50 mg
per day or less (corresponding to 5000 ppm or 0.5 per cent LOEL: Abbreviation for lowest-observed effect leve/o
under Option l) would be acceptable without justification. Lowest-observed effect level: The lowest dose of substance in a
Higher amounts may also be acceptable provided they are study or group of studies that produces biologically significant
realistic in relation to manufacturing capability and good increases in frequency or severity of any effects in the exposed
manufacturing practice. humans or animals.
Table 3. - Class 3 solvents which should be limited by GMP or
other quality-based requirements Modifying factor: A factor determined by professional
Acetic acid Heptane judgement of a toxicologist and applied to bioassay data to
relate that data safely to humans.
Acetone lsobutyl acetate
Neurotoxicity: The ability of a substance to cause adverse
Anisole Isopropyl acetate effects on the nervous system.
1-Butanol Methyl acetate NOEL: Abbreviation for no-observed-effect leve/o
2-Butanol 3-Methyl-l-butanol
No-observed-effect level: The highest dose of substance
Butyl acetate Methylethylketone at which there are no biologically significant increases in
frequency or severity of any effects in the exposed humans
tert- Butylmethyl ether Methylisobutylketone
or animals.
Cumene 2-Methyl-l-propanol
PDE: Abbreviation for permitted daily exposure.
Dimethyl sulfoxide Pentane
Permitted daily exposure: The maximum acceptable intake per
Ethanol 1-Pentanol day of residual solvent in pharmaceutical products.
Ethyl acetate ¡-Propanol
Reversible toxicity: The occurrence of harmful effects that are
Ethyl ether 2-Propanol caused by a substance and which disappear after exposure to
the substance ends.
Ethyl formate Propyl acetate
Strongly suspected human carcinogen: A substance for which
Formic acid
there is no epidemiological evidence of carcinogenesis but
there are positive genotoxicity data and clear evidence of
4.4. SOLVENTS POR WHICH NO ADEQUATE
carcinogenesis in rodents.
TOXICOLOGICAL DATA WAS POUND
The following solvents (Table 4) may also be of interest to Teratogenicity: The occurrence of structural malformations in
manufacturers of excipients, active substances, or medicinal a developing foetus when a substance is administered during
products. However, no adequate toxicological data on pregnancy.

642 See the information section on general monographs (caver pages)


EUROPEAN PHARMACOPOEIA 8.0 5.4. Residual solvents

APPENDIX 1. LIST OF SOLVENTS INCLUDED IN THE GUIDELINE


Solvent OtherNames Structure Class

Acetic acid Ethanoic acid CH,COOH Class 3

Acetone 2-Propanone CH 3 COCH, Class 3


Propan -2-one
Acetonitrile CH 3 CN Class 2

Anisole Methoxybenzene
O
1,#
OCH 3
Class 3

Benzene Benzol Class 1

1-Butanol n- Butyl alcohol


O
CH 3[CH 2 ],oH Class 3
Butan-1-o1
2-Butanol sec-Butyl alcohol CH 3 CH 2 CH(OH)CH 3 Class 3
Butan-2-o1
Butyl acetate Acetic acid butyl ester CH 3 COO[CH,],CH 3 Class 3

tert- Butylmethyl ether 2-Methoxy-2-methylpropane (CH3)3COCH3 Class 3

Carbon tetrachloride Tetrachloromethane CCI, Class 1

Chlorobenzene
OCI
1,#
Class 2

Chloroform Trichloromethane CHCl3 Class 2

Cumene Isopropylbenzene
CH Class 3

ifCH
3
(1-Methylethyl)benzene

Cyclohexane Hexamethylene Class 2

1,2-Dichloroethane sym- Dichloroethane


O
CH,CICH,CI Class 1
Ethylene dichloride
Ethylene chloride
1,1-Dichloroethene 1,1-Dichloroethylene H2C~CCI, Class 1
Vinylidene chloride
1,2-Dichloroethene 1,2-Dichloroethylene ClHC~CHCI Class 2
Acetylene dichloride
Dichloromethane Methylene chloride CH,Cl2 Class 2

1,2-Dimethoxyethane Ethyleneglycol dimethyl ether H,COCH,CH,OCH, Class 2


Monoglyme
Dimethyl cellosolve
N,N-Dimethylacetamide DMA CH 3CON(CH3), Class 2

N, N-Dimethylformamide DMF HCON(CH 3 ), Class 2

Dimethyl sulfoxide Methylsulfinylmethane (CH 3)2S0 Class 3


Methyl sulfoxide
DMSO
1,4-Dioxane p-Dioxane Class 2
[1,4] Dioxane (0)
Ethanol Ethyl alcohol °
CH 3CH 2 OH Class 3

2-Ethoxyethanol Cellosolve CH,CH 2 OCH 2 CH 2OH Class 2

Ethyl acetate Acetic acid ethyl ester CH 3COOCH,CH, Class 3

Ethyleneglycol 1,2-Dihydroxyethane HOCH,CH,OH Class 2


1,2-Ethanediol
Ethyl ether Diethyl ether CH 3 CH2 OCH 2 CH3 Class 3
Ethoxyethane
l,l'-Oxybisethane
Ethyl formate Formic acid ethyl ester HCOOCH 2 CH3 Class 3

Formamide Methanamide HCONH, Class 2

Formic acid HCOOH Class 3

General Natices (1) apply ta all managraphs and ather texts 643
5.4. Residual solvents EUROPEAN PHARMACOPOEIA 8.0

Solvent OtherNames Structure Class

Heptane n-Heptane CH 3[CH 2 l,CH 3 Class 3

Hexane n-Hexane CH 3[CH 2LCH 3 Class 2

Isobutyl acetate Acetic acid isobutyl ester CH 3COOCH 2CH(CH 3)2 Class 3

Isopropyl acetate Acetic acid isopropyl ester CH 3COOCH(CH 3), Class 3

Methanol Methyl alcohol CH 30H Class 2

2-Methoxyethanol Methyl cellosolve CH,oCH 2 CH2 OH Class 2

Methyl acetate Acetic acid methyl ester CH 3COOCH 3 Class 3

3-Methyl-l-butanol Isoamyl alcohol (CH,)2CHCH,CH,oH Class 3


Isopentyl alcohol
3-Methylbutan -1-01
Methylbutylketone 2-Hexanone CH3[CH,l3COCH3 Class 2
Hexan-2-one
Methylcyclohexane Cyclohexylmethane Class 2

Methylethylketone 2-Butanone
0
CH3CH2 COCH 3
CH3

Class 3
MEK
Butan -2-one
Methylisobutylketone 4-Methylpentan -2-one CH 3COCH2 CH(CH 3), Class 3
4-Methyl-2-pentanone
MIBK
2-Methyl-l-propanol Isobutyl alcohol (CH 3),CHCH,oH Class 3
2-Methylpropan-l-01
N-Methylpyrrolidone 1-Methylpyrrolidin -2-one Class 2
CH 3
1-Methyl-2-pyrrolidinone I

(Jo
Nitromethane CH 3NO, Class 2

Pentane n-Pentane CH 3[CH,l3CH 3 Class 3

l-Pentanol Amyl alcohol CH 3[CH,LCH,OH Class 3


Pentan -1-01
Pentyl alcohol
l-Propanol Propan-l-01 CH 3CH,CH,OH Class 3
Propyl alcohol
2-Propanol Propan-2-01 (CH3)2CHOH Class 3
Isopropyl alcohol
Propyl acetate Acetic acid propyl ester CH 3COOCH 2CH,CH 3 Class 3

Pyridine Class 2
N

Sulfonane Tetrahydrothiophene l,l-dioxide


O Class 2
°° \\ I¡

Ó
Tetrahydrofuran Tetramethylene oxide Class 2
Oxacyclopentane
O
ca
Tetralin 1,2,3,4-Tetrahydronaphthalene Class 2

a-
Toluene Methylbenzene Class 2

CH3
1#
1,1,1-Trichloroethane Methylchloroform CH3CCI3 Class 1

1,1,2-Trichloroethene Trichloroethene HCIC=CCI2 Class 2

a
Xylene* Dimethybenzene Class 2
Xylol
H3C - I CH3
~

*usually 60 per cent m-xylene, 14 per cent p-xylene, 9 per cent o-xylene with 17 per cent ethyl benzene.

644 See the information section on general monographs (cover pages)


EUROPEAN PHARMACOPOEIA 8.0 5.4. Residual solvents

APPENDIX 2. ADDITIONAL BACKGROUND The method is outlined here to give a better understanding of
A2.1. ENVIRONMENTAL REGULATION OF ORGANIC the origin of the PDE values. It is not necessary to perform
VOLATILE SOLVENTS these calculations in order to use the PDE values tabulated in
Section 4 of this document.
Several of the residual solvents frequently used in the
production of pharmaceuticals are listed as toxic chemicals PDE is derived from the no-observed-effect level (NOEL), or
in Environmental Health Criteria (EHC) monographs and the lowest-observed effect level (LOEL), in the most relevant
the Integrated Risk Information System (IRIS). The objectives animal study as follows:
of such groups as the International Programme on Chemical
PDE = NOEL x Weight Adjustment
Safety (IPCS), the United States Environmental Protection
Agency (USEPA) and the United States Food and Drug Fl x F2 x F3 x F4 x F5
Administration (USFDA) in dude the determination of The PDE is derived preferably from a NOEL. lf no NOEL
acceptable exposure levels. The goal is protection of human is obtained, the LOEL may be used. Modifying factors
health and maintenance of environmental integrity against proposed here, for relating the data to humans, are the same
the possible deleterious effects of chemicals resulting from kind of "uncertainty factors" used in Environmental Health
long-term environmental exposure. The methods involved in Criteria (Environmental Health Criteria 170, World Health
the estimation of maximum safe exposure limits are usuaHy Organization, Geneva, 1994), and "modifying factors" or
based on long-term studies. When long-term study data "safety factors" in Pharmacopoeial Forum. The assumption
are unavailable, shorter term study data can be used with of 100 per cent systemic exposure is used in aH calculations
modification of the approach such as use of larger safety regardless of route of administration.
factors. The approach described therein relates primarily to
long -term or life-time exposure of the general population in The modifying factors are as follows:
the ambient environment, i.e. ambient air, food, drinking F1 A factor to account for extrapolation between
water and other media. species
A2.2. RESIDUAL SOLVENTS IN PHARMACE UTICALS
Fl 2 for extrapolation from dogs to humans
Exposure limits in this guideline are established by referring
to methodologies and toxicity data described in EHC and FI 2.5 for extrapolation from rabbits to humans
IRIS monographs. However, sorne specific assumptions about
FI 3 for extrapolation from monkeys to human s
residual solvents to be used in the synthesis and formulation
of pharmaceutical products should be taken into account in Fl 5 for extrapolation from rats to humans
establishing exposure limits. They are:
FI 10 for extrapolation from other animals to
1) Patients (not the general population) use pharmaceuticals human s
to treat their diseases or for prophylaxis to prevent infection F1 12 for extrapolation from mice to humans
or disease.
2) The assumption of life-time patient exposure is not Fl takes into account the comparative surface area: body
necessary for most pharmaceutical products but may be weight ratios for the species concerned and Íor mano Surface
appropriate as a working hypothesis to reduce risk to human area (S) is calculated as:
health. s = kmO. 67
3) Residual solvents are unavoidable components in
pharmaceutical production and will often be a part of in which m = body mass, and the constant k has been taken to
medicinal products. be 10. The body weight used in the equation are those shown
below in Table A3.-1.
4) Residual solvents should not exceed recommended levels
except in exceptional circumstances. Table A3.-1. - Values used in the calculations in this document
5) Data from toxicological studies that are used to determine Rat body weight 425 g
acceptable levels for residual solvents should have been
generated using appropriate protocols such as those described Pregnant rat body weight 330 g
for example, by OECD and the FDA Red Book. Mouse body weight 28 g

Pregnant mouse body weight 30 g


APPENDIX 3. METHODS FOR ESTABLISHING EXPOSURE
LIMITS Guinea-pig body weight 500 g

The Gaylor-Kodell method of risk assessment (Gaylor, D. W Rhesus monkey body weight 2.5 kg
and Kodell, R. L. Linear Interpolation algorithm for low dose Rabbit body weight (pregnant or not) 4 kg
assessment of toxic substance. J. Environ. Pathology, 4, 305,
1980) is appropriate for Class 1 carcinogenic solvents. Only in Beagle dog body weight 11.5 kg
cases where reliable carcinogenicity data are available should Rat respiratory volume 290 L/day
extrapolation by the use of mathematical models be applied to
setting exposure limits. Exposure Iimits for Class 1 solvents Mouse respiratory volume 43 L/day
could be determined with the use of a large safety factor (i.e., Rabbit respiratory volume 1440 L/day
10000 to 100000) with respect to the no-observed-effect level
(NOEL). Detection and quantification of these solvents should Guinea-pig respiratory volume 430 L/day
be by state-of-the-art analytical techniques. Human respiratory volume 28800 L/day
Acceptable exposure levels in this guideline for Class 2 Dog respiratory volume 9000 L/day
solvents were established by calculation of PDE values
according to the procedures for setting exposure limits in Monkey respiratory volume 1150 L/day
pharmaceuticals (Pharmacopeial Forum, Nov-Dec 1989), Mouse water consumption 5 mL/day
and the method adopted by IPCS for Assessing Human
Health Risk of Chemicals (En viro n mental Health Criteria Rat water consumption 30 mL/day
170, WHO, 1994). These methods are similar to those used Rat food consumption 30 g/day
by the USEPA (IRIS) and the USFDA (Red Book) and others.

General Notices (1) apply to all monographs and other texts 645
5.4. Residual solvents EUROPEAN PHARMACOPOElA 8.0

F2 A factor of 10 to account for variability between weights of 60 kg or 70 kg that are often used in this type of
individuals. A factor of 10 is gene rally given for calculation. It is recognised that sorne adult patients weigh less
al! organic solvents, and 10 is used consistently in than 50 kg; these patients are considered to be accommodated
this guideline. by the built-in safety factors used to determine a PDE. lf the
solvent was present in a formulation speciflcally intended for
F3 A variable factor to account for toxicity studies of paediatric use, an adjustment for a lower body weight would
short -term exposure. be appropriate.
F3 1 for studies that last at least one half-lifetime (1 As an example of the application of this equation, consider the
year for rodents or rabbits; 7 years for cats, dogs toxicity study of acetonitrile in mice that is summarised in
and monkeys). Pharmeuropa, Vol. 9. No. 1, Supplement, April1997, page S24.
F3 1 for reproductive studies in which the whole The NOEL is calculated to be 50.7 mg kg-l day-l. The PDE for
period of organogenesis is covered. acetonitrile in this study is calculated as follows:
F3 2 for a 6 month study in rodents, or a 3.5 year
study in non-rodents.
PDE = 50.7mg kg- 1 day-1 x 50 kg = 4.22 m da -1
F3 5 for a 3 month study in rodents, or a 2 year 12 x 10 x 5 x 1 x 1 g Y
study in non-rodents.
F3 10 for studies of a shorter duration. In this example,
In al! cases, the higher factor has been used for study durations Fl 12 to account for the extrapolation from mice to
between the time points, e.g. a factor of 2 for a 9 month humans
rodent study.
F2 lOto account for differences between individual
F4 A factor that may be applied in cases of severe humans
toxicity, e.g. non-genotoxic carcinogenicity, F3 5 because the duration of the study was only
neurotoxicity or teratogenicity. In studies of 13 weeks
reproductive toxicity, the following factors are
used: F4 1 because no severe toxicity was encountered
F5 1 because the no-effect level was determined
F4 1 for foetal toxicity associated with maternal
toxicity
The equation for an ideal gas, PV = nRT, is used to convert
F4 5 for foetal toxicity without maternal toxicity concentrations of gases used in inhalation studies from units
F4 5 for a teratogenic effect with maternal toxicity of ppm to units of mg/L or mg/m 3 • Consider as an example
the rat reproductive toxicity study by inhalation of carbon
F4 10 for a teratogenic effect without maternal tetrachloride (molecular weight 153.84) summarised in
toxicity Pharmeuropa, Vol. 9, No. 1, Supplement, April1997, page S9.
F5 A variable factor that may be applied if the
n P 300 X 10- 6 atm x 153840 mg mol- 1
no-effect leve! was not established.
When only a LOEL is available, a factor of up to 10 can be
v RT 0.082 L atm K-1 mol- 1 x 298 K
used depending on the severity of the toxicity. = 46.15 mg = 1.89 m /L
The weight adjustment assumes an arbitrary adult human 24.45 L g
body weight for either sex of 50 kg. This relative!y low weight
provides an additional safety factor against the standard The relationship 1000 L = 1 m 3 is used to convert to mg/m 3•

646 See the informatíon sectíon on general monographs (caver pages)


EUROPEA N PHARMACOPOEIA 8.0

S.Se Alcoholimetric tables


5.5. Alcoholimetric tables ........................................................ 649

General Notices (1) apply to all monographs and other texts 647
EUROPEAN PHARMACOPOEIA 8.0

648 See the information section on general monographs (cover pages)


EUROPEAN PHARMACOPOEIA 8.0 5.5. Akoholimetric tables

0112008:50500 % v/v %m/m 1'20 (kg/m 3 )


3.9 3.10 992.55
5.5. ALCOHOLIMETRIC TABLES
The general formula agreed by the Council of the European
4.0 3.18 992.41
Communities in its Directive of 27 July 1976 on alcoholimetry
served as the basis for establishing the following tables. 4.1 3.26 992.28

% V/V %m/m 1'20 (kg/m 3 ) 4.2 3.34 992.14

0.0 0.0 998.20 4.3 3.42 992.00

0.1 0.08 998.05 4.4 3.50 99l.87

0.2 0.16 997.90 4.5 3.58 99l.73

0.3 0.24 997.75 4.6 3.66 991.59

0.4 0.32 997.59 4.7 3.74 99l.46

0.5 0.40 997.44 4.8 3.82 991.32

0.6 0.47 997.29 4.9 3.90 99l.l9

0.7 0.55 997.14

0.8 0.63 996.99 5.0 3.98 991.06

0.9 0.71 996.85 5.1 4.06 990.92

5.2 4.14 990.79

1.0 0.79 996.70 5.3 4.22 990.65

l.l 0.87 996.55 5.4 4.30 990.52

1.2 0.95 996.40 5.5 4.38 990.39

1.3 l.03 996.25 5.6 4.46 990.26

1.4 l.l1 996.11 5.7 4.54 990.12

l.5 l.l9 995.96 5.8 4.62 989.99

l.6 1.27 995.81 5.9 4.70 989.86

l.7 1.35 995.67

l.8 l.43 995.52 6.0 4.78 989.73

l.9 l.51 995.38 6.1 4.86 989.60

6.2 4.95 989.47

2.0 1.59 995.23 6.3 5.03 989.34

2.1 1.67 995.09 6.4 5.11 989.21

2.2 1.75 994.94 6.5 5.19 989.08

2.3 1.82 994.80 6.6 5.27 988.95

2.4 1.90 994.66 6.7 5.35 988.82

2.5 1.98 994.51 6.8 5.43 988.69

2.6 2.06 994.37 6.9 5.51 988.56

2.7 2.14 994.23

2.8 2.22 994.09 7.0 5.59 988.43

2.9 2.30 993.95 7.1 5.67 988.30

7.2 5.75 988.18

3.0 2.38 993.81 7.3 5.83 988.05

3.1 2.46 993.66 7.4 5.91 987.92

3.2 2.54 993.52 7.5 5.99 987.79

3.3 2.62 993.38 7.6 6.07 987.67

3.4 2.70 993.24 7.7 6.15 987.54

3.5 2.78 993.11 7.8 6.23 987.42

3.6 2.86 992.97 7.9 6.32 987.29

3.7 2.94 992.83

3.8 3.02 992.69 8.0 6.40 987.16

General Natices (1) apply ta all managraphs and ather texts 649
5.5. Akoholimetl'ic tables EUROPEAN PHARMACOPOEIA 8.0

% v/v %m/m 1"0 (kg/m3 ) % v/v %m/m 1'20 (kg/m 3 )


8.1 6.48 987.04 12.4 9.97 981.89

8.2 6.56 986.91 12.5 10.05 981.78

8.3 6.64 986.79 12.6 10.13 981.67

8.4 6.72 986.66 12.7 10.21 981.55

8.5 6.80 986.54 12.8 10.29 981.44

8.6 6.88 986.42 12.9 10.37 981.32

8.7 6.96 986.29

8.8 7.04 986.17 13.0 10.46 981.21

8.9 7.12 986.05 13.1 10.54 981.10

13.2 10.62 980.98

9.0 7.20 985.92 13.3 10.70 980.87

9.1 7.29 985.80 13.4 10.78 980.76

9.2 7.37 985.68 13.5 10.87 980.64

9.3 7.45 985.56 13.6 10.95 980.53

9.4 7.53 985.44 13.7 11.03 980.42

9.5 7.61 985.31 13.8 11.11 980.31

9.6 7.69 985.19 13.9 11.19 980.19

9.7 7.77 985.07

9.8 7.85 984.95 14.0 11.27 980.08

9.9 7.93 984.83 14.1 11.36 979.97

14.2 11.44 979.86

10.0 8.01 984.71 14.3 11.52 979.75

10.1 8.10 984.59 14.4 11.60 979.64

10.2 8.18 984.47 14.5 11.68 979.52

10.3 8.26 984.35 14.6 11.77 979.41

10.4 8.34 984.23 14.7 11.85 979.30

10.5 8.42 984.11 14.8 11.93 979.19

10.6 8.50 983.99 14.9 12.01 979.08

10.7 8.58 983.88

10.8 8.66 983.76 15.0 12.09 978.97

10.9 8.75 983.64 15.1 12.17 978.86

15.2 12.26 978.75

11.0 8.83 983.52 15.3 12.34 978.64

11.1 8.91 983.40 15.4 12.42 978.53

11.2 8.99 983.29 15.5 12.50 978.42

11.3 9.07 983.17 15.6 12.59 978.31

11.4 9.15 983.05 15.7 12.67 978.20

11.5 9.23 982.94 15.8 12.75 978.09

11.6 9.32 982.82 15.9 12.83 977.98

11.7 9.40 982.70

11.8 9.48 982.59 16.0 12.91 977.87

11.9 9.56 982.47 16.1 13.00 977.76

16.2 13.08 977.65

12.0 9.64 982.35 16.3 13.16 977.55

12.1 9.72 982.24 16.4 13.24 977.44

12.2 9.80 982.12 16.5 13.32 977.33

12.3 9.89 982.01 16.6 13.41 977.22

650 See the information section on general monographs (ca ver pages)
EUROPEAN PHARMACOPOEIA 8.0 5.5. Akohol:imetrk tables

% v/v %m/m P,o (kg/m3 ) % V/V %m/m 1'20 (kg/m 3 )


16.7 13.49 977.11

16.8 13.57 977.00 21.0 17.04 972.48

16.9 13.65 976.89 21.1 17.13 972.37

21.2 17.21 972.27

17.0 13.74 976.79 21.3 17.29 972.16

17.1 13.82 976.68 21.4 17.38 972.05

17.2 13.90 976.57 21.5 17.46 971.94

17.3 13.98 976.46 21.6 17.54 971.83

17.4 14.07 976.35 21.7 17.62 971.73

17.5 14.15 976.25 21.8 17.71 971.62

17.6 14.23 976.14 21.9 17.79 971.51

17.7 14.31 976.03

17.8 14.40 975.92 22.0 17.87 971.40

17.9 14.48 975.81 22.1 17.96 971.29

22.2 18.04 971.18

18.0 14.56 975.71 22.3 18.12 971.08

18.1 14.64 975.60 22.4 18.21 970.97

18.2 14.73 975.49 22.5 18.29 970.86

18.3 14.81 975.38 22.6 18.37 970.75

18.4 14.89 975.28 22.7 18.46 970.64

18.5 14.97 975.17 22.8 18.54 970.53

18.6 15.06 975.06 22.9 18.62 970.42

18.7 15.14 974.95

18.8 15.22 974.85 23.0 18.71 970.31

18.9 15.30 974.74 23.1 18.79 970.20

23.2 18.87 970.09

19.0 15.39 974.63 23.3 18.96 969.98

19.1 15.47 974.52 23.4 19.04 969.87

19.2 15.55 974.42 23.5 19.13 969.76

19.3 15.63 974.31 23.6 19.21 969.65

19.4 15.72 974.20 23.7 19.29 969.54

19.5 15.80 974.09 23.8 19.38 969.43

19.6 15.88 973.99 23.9 19.46 969.32

19.7 15.97 973.88

19.8 16.05 973.77 24.0 19.54 969.21

19.9 16.13 973.66 24.1 19.63 969.10

24.2 19.71 968.99

20.0 16.21 973.56 24.3 19.79 968.88

20.1 16.30 973.45 24.4 19.88 968.77

20.2 16.38 973.34 24.5 19.96 968.66

20.3 16.46 973.24 24.6 20.05 968.55

20.4 16.55 973.13 24.7 20.13 968.43

20.5 16.63 973.02 24.8 20.21 968.32

20.6 16.71 972.91 24.9 20.30 968.21

20.7 16.79 972.80

20.8 16.88 972.70 25.0 20.38 968.10

20.9 16.96 972.59 25.1 20.47 967.99

General Natices (1) apply ta all managraphs and ather texts 651
5.5. Akoholimetric tables EUROPEAN PHARMACOPOEIA 8.0

% v/v %m/m P20 (kg/m3 ) % v/v %m/m P20 (kg/m 3 )


25.2 20.55 967.87 29.5 24.18 962.83

25.3 20.63 967.76 29.6 24.27 962.7l

25.4 20.72 967.65 29.7 24.35 962.58

25.5 20.80 967.53 29.8 24.44 962.46

25.6 20.88 967.42 29.9 24.52 962.33

25.7 20.97 967.31

25.8 21.05 967.19 30.0 24.61 962.21

25.9 21.14 967.08 30.1 24.69 962.09

30.2 24.78 961.96

26.0 21.22 966.97 30.3 24.86 961.84

26.1 2l.31 966.85 30.4 24.95 961.7l

26.2 2l.39 966.74 30.5 25.03 961.59

26.3 21.47 966.62 30.6 25.12 961.46

26.4 21.56 966.51 30.7 25.20 96l.33

26.5 21.64 966.39 30.8 25.29 961.21

26.6 21.73 966.28 30.9 25.38 961.08

26.7 21.81 966.16

26.8 21.90 966.05 31.0 25.46 960.95

26.9 21.98 965.93 31.1 25.55 960.82

31.2 25.63 960.70

27.0 22.06 965.81 31.3 25.72 960.57

27.1 22.15 965.70 31.4 25.80 960.44

27.2 22.23 965.58 31.5 25.89 960.31

27.3 22.32 965.46 31.6 25.97 960.18

27.4 22.40 965.35 31.7 26.06 960.05

27.5 22.49 965.23 31.8 26.15 959.92

27.6 22.57 965.11 31.9 26.23 959.79

27.7 22.65 964.99

27.8 22.74 964.88 32.0 26.32 959.66

27.9 22.82 964.76 32.1 26.40 959.53

32.2 26.49 959.40

28.0 22.91 964.64 32.3 26.57 959.27

28.1 22.99 964.52 32.4 26.66 959.14

28.2 23.08 964.40 32.5 26.75 959.01

28.3 23.16 964.28 32.6 26.83 958.87

28.4 23.25 964.16 32.7 26.92 958.74

28.5 23.33 964.04 32.8 27.00 958.61

28.6 23.42 963.92 32.9 27.09 958.47

28.7 23.50 963.80

28.8 23.59 963.68 33.0 27.18 958.34

28.9 23.67 963.56 33.1 27.26 958.20

33.2 27.35 958.07

29.0 23.76 963.44 33.3 27.44 957.94

29.1 23.84 963.32 33.4 27.52 957.80

29.2 23.93 963.20 33.5 27.61 957.66

29.3 24.01 963.07 33.6 27.69 957.53

29.4 24.10 962.95 33.7 27.78 957.39

652 See the information section on general monographs (cover pages)


EUROPEAN PHARMACOPOEIA 8.0 5.5. Alcoholimetric tables

% v/v %m/m 1"0 (kg/m') % V/V %m/m 1"0 (kg/m 3 )

33.8 27.87 957.26 38.0 31.53 951.18

33.9 27.95 957.12 38.1 31.62 951.02

38.2 31.71 950.87

34.0 28.04 956.98 38.3 31.79 950.72

34.1 28.13 956.84 38.4 31.88 950.56

34.2 28.21 956.70 38.5 31.97 950.41

34.3 28.30 956.57 38.6 32.06 950.25

34.4 28.39 956.43 38.7 32.15 950.10

34.5 28.47 956.29 38.8 32.24 949.94

34.6 28.56 956.15 38.9 32.32 949.79

34.7 28.65 956.01

34.8 28.73 955.87 39.0 32.41 949.63

34.9 28.82 955.73 39.1 32.50 949.47

39.2 32.59 949.32

35.0 28.91 955.59 39.3 32.68 949.16

35.1 28.99 955.45 39.4 32.77 949.00

35.2 29.08 955.30 39.5 32.86 948.84

35.3 29.17 955.16 39.6 32.94 948.68

35.4 29.26 955.02 39.7 33.03 948.52

35.5 29.34 954.88 39.8 33.12 948.37

35.6 29.43 954.73 39.9 33.21 948.21

35.7 29.52 954.59

35.8 29.60 954.44 40.0 33.30 948.05

35.9 29.69 954.30 40.1 33.39 947.88

40.2 33.48 947.72

36.0 29.78 954.15 40.3 33.57 947.56

36.1 29.87 954.01 40.4 33.66 947.40

36.2 29.95 953.86 40.5 33.74 947.24

36.3 30.04 953.72 40.6 33.83 947.08

36.4 30.13 953.57 40.7 33.92 946.91

36.5 30.21 953.42 40.8 34.01 946.75

36.6 30.30 953.28 40.9 34.10 946.58

36.7 30.39 953.13

36.8 30.48 952.98 41.0 34.19 946.42

36.9 30.56 952.83 41.1 34.28 946.26

41.2 34.37 946.09

37.0 30.65 952.69 41.3 34.46 945.93

37.1 30.74 952.54 41.4 34.55 945.76

37.2 30.83 952.39 41.5 34.64 945.59

37.3 30.92 952.24 41.6 34.73 945.43

37.4 31.00 952.09 41.7 34.82 945.26

37.5 31.09 951.94 41.8 34.91 945.09

37.6 31.18 951.79 41.9 35.00 944.93

37.7 31.27 951.63

37.8 31.35 951.48 42.0 35.09 944.76

37.9 31.44 951.33 42.1 35.18 944.59

42.2 35.27 944.42

General Natices (1) apply ta all managraphs and ather texts 653
5.5. Alcoholimetric tables EUROPEAN PHARMACOPOEIA 8.0

% v/v %m/m P20 (kg/m') % v/v %m/m P20 (kg/m')


42.3 35.36 944.25 46.6 39.27 936.63

42.4 35.45 944.08 46.7 39.36 936.44

42.5 35.54 943.91 46.8 39.45 936.26

42.6 35.63 943.74 46.9 39.54 936.07

42.7 35.72 943.57

42.8 35.81 943.40 47.0 39.64 935.88

42.9 35.90 943.23 47.1 39.73 935.70

47.2 39.82 935.51

43.0 35.99 943.06 47.3 39.91 935.32

43.1 36.08 942.88 47.4 40.00 935.14

43.2 36.17 942.7l 47.5 40.10 934.95

43.3 36.26 942.54 47.6 40.19 934.76

43.4 36.35 942.37 47.7 40.28 934.57

43.5 36.44 942.19 47.8 40.37 934.38

43.6 36.53 942.02 47.9 40.47 934.19

43.7 36.62 941.84

43.8 36.71 941.67 48.0 40.56 934.00

43.9 36.80 941.49 48.1 40.65 933.81

48.2 40.75 933.62

44.0 36.89 941.32 48.3 40.84 933.43

44.1 36.98 941.14 48.4 40.93 933.24

44.2 37.07 940.97 48.5 41.02 933.05

44.3 37.16 940.79 48.6 41.12 932.86

44.4 37.25 940.61 48.7 41.21 932.67

44.5 37.35 940.43 48.8 41.30 932.47

44.6 37.44 940.26 48.9 41.40 932.28

44.7 37.53 940.08

44.8 37.62 939.90 49.0 41.49 932.09

44.9 37.7l 939.72 49.1 41.58 931.90

49.2 41.68 931.70

45.0 37.80 939.54 49.3 41.77 931.51

45.1 37.89 939.36 49.4 41.86 931.31

45.2 37.98 939.18 49.5 41.96 931.12

45.3 38.08 939.00 49.6 42.05 930.92

45.4 38.17 938.82 49.7 42.14 930.73

45.5 38.26 938.64 49.8 42.24 930.53

45.6 38.35 938.46 49.9 42.33 930.34

45.7 38.44 938.28

45.8 38.53 938.10 50.0 42.43 930.14

45.9 38.62 937.91 50.1 42.52 929.95

50.2 42.61 929.75

46.0 38.72 937.73 50.3 42.71 929.55

46.1 38.81 937.55 50.4 42.80 929.35

46.2 38.90 937.36 50.5 42.90 929.16

46.3 38.99 937.18 50.6 42.99 928.96

46.4 39.08 937.00 50.7 43.08 928.76

46.5 39.18 936.81 50.8 43.18 928.56

654 See the information section on general monographs (ca ver pages)
EUROPEAN PHARMACOPOEIA 8.0 5.5. Alcoholimetric taMes

% v/v %m/m 1'20 (kg/m') % V/V %m/m 1'20 (kg/m')


50.9 43.27 928.36 55.1 47.28 919.75

55.2 47.38 919.54

51.0 43.37 928.16 55.3 47.47 919.33

51.1 43.46 927.96 55.4 47.57 919.12

51.2 43.56 927.77 55.5 47.67 918.91

51.3 43.65 927.57 55.6 47.77 918.69

51.4 43.74 927.36 55.7 47.86 918.48

51.5 43.84 927.16 55.8 47.96 918.27

51.6 43.93 926.96 55.9 48.06 918.06

51.7 44.03 926.76

51.8 44.12 926.56 56.0 48.15 917.84

51.9 44.22 926.36 56.1 48.25 917.63

56.2 48.35 917.42

52.0 44.31 926.16 56.3 48.45 917.20

52.1 44.41 925.95 56.4 48.54 916.99

52.2 44.50 925.75 56.5 48.64 916.77

52.3 44.60 925.55 56.6 48.74 916.56

52.4 44.69 925.35 56.7 48.84 916.35

52.5 44.79 925.14 56.8 48.93 916.13

52.6 44.88 924.94 56.9 49.03 915.91

52.7 44.98 924.73

52.8 45.07 924.53 57.0 49.13 915.70

52.9 45.17 924.32 57.1 49.23 915.48

57.2 49.32 915.27

53.0 45.26 924.12 57.3 49.42 915.05

53.1 45.36 923.91 57.4 49.52 914.83

53.2 45.46 923.71 57.5 49.62 914.62

53.3 45.55 923.50 57.6 49.72 914.40

53.4 45.65 923.30 57.7 49.81 914.18

53.5 45.74 923.09 57.8 49.91 913.97

53.6 45.84 922.88 57.9 50.01 913.75

53.7 45.93 922.68

53.8 46.03 922.47 58.0 50.11 913.53

53.9 46.13 922.26 58.1 50.21 913.31

58.2 50.31 913.09

54.0 46.22 922.06 58.3 50.40 912.87

54.1 46.32 921.85 58.4 50.50 912.65

54.2 46.41 921.64 58.5 50.60 912.43

54.3 46.51 921.43 58.6 50.70 912.22

54.4 46.61 921.22 58.7 50.80 912.00

54.5 46.70 921.01 58.8 50.90 911.78

54.6 46.80 920.80 58.9 51.00 911.55

54.7 46.90 920.59

54.8 46.99 920.38 59.0 51.10 911.33

54.9 47.09 920.17 59.1 51.19 911.11

59.2 51.29 910.89

55.0 47.18 919.96 59.3 51.39 910.67

General Natices (1) apply ta all monographs and other texts 655
5.5. Akoholimetric tables EUROPEAN PHARMACOPOEIA 8.0

% v/v %m/m 1'20 (kg/m') % V/V %m/m 1'20 (kg/m')


59.4 5l.49 910.45 63.7 55.82 900.69

59.5 5l.59 910.23 63.8 55.92 900.46

59.6 5l.69 910.01 63.9 56.02 900.23

59.7 5l.79 909.78

59.8 5l.89 909.56 64.0 56.12 899.99

59.9 5l.99 909.34 64.1 56.23 899.76

64.2 56.33 899.53

60.0 52.09 909.11 64.3 56.43 899.29

60.1 52.19 908.89 64.4 56.53 899.06

60.2 52.29 908.67 64.5 56.64 898.83

60.3 52.39 908.44 64.6 56.74 898.59

60.4 52.49 908.22 64.7 56.84 898.36

60.5 52.59 908.00 64.8 56.94 898.12

60.6 52.69 907.77 64.9 57.05 897.89

60.7 52.79 907.55

60.8 52.89 907.32 65.0 57.15 897.65

60.9 52.99 907.10 65.1 57.25 897.42

65.2 57.36 897.18

6l.0 53.09 906.87 65.3 57.46 896.94

6l.l 53.19 906.64 65.4 57.56 896.71

61.2 53.29 906.42 65.5 57.67 896.47

61.3 53.39 906.19 65.6 57.77 896.23

6l.4 53.49 905.97 65.7 57.87 896.00

6l.5 53.59 905.74 65.8 57.98 895.76

6l.6 53.69 905.51 65.9 58.08 895.52

6l.7 53.79 905.29

6l.8 53.89 905.06 66.0 58.18 895.28

6l.9 53.99 904.83 66.1 58.29 895.05

66.2 58.39 894.81

62.0 54.09 904.60 66.3 58.49 894.57

62.1 54.19 904.37 66.4 58.60 894.33

62.2 54.30 904.15 66.5 58.70 894.09

62.3 54.40 903.92 66.6 58.81 893.85

62.4 54.50 903.69 66.7 58.91 893.61

62.5 54.60 903.46 66.8 59.01 893.37

62.6 54.70 903.23 66.9 59.12 893.13

62.7 54.80 903.00

62.8 54.90 902.77 67.0 59.22 892.89

62.9 55.00 902.54 67.1 59.33 892.65

67.2 59.43 892.41

63.0 55.11 902.31 67.3 59.54 892.17

63.1 55.21 902.08 67.4 59.64 89l.93

63.2 55.31 90l.85 67.5 59.74 89l.69

63.3 55.41 90l.62 67.6 59.85 89l.45

63.4 55.51 901.39 67.7 59.95 891.20

63.5 55.61 90 l.l 5 67.8 60.06 890.96

63.6 55.72 900.92 67.9 60.16 890.72

656 See the information section on general monographs (cover pages)


EUROPEAN PHARMACOPOEIA 8.0 5.5. Akoholimetrk tables

% v/v %m/m p,O (kg/m 3) % v/v %m/m P20(kg/m 3 )


72.2 64.75 880.03

68.0 60.27 890.48 72.3 64.86 879.78

68.1 60.37 890.23 72.4 64.97 879.52

68.2 60.48 889.99 72.5 65.08 879.27

68.3 60.58 889.75 72.6 65.19 879.01

68.4 60.69 889.50 72.7 65.29 878.75

68.5 60.80 889.26 72.8 65.40 878.50

68.6 60.90 889.01 72.9 65.51 878.24

68.7 6l.01 888.77

68.8 61.11 888.52 73.0 65.62 877.99

68.9 61.22 888.28 73.1 65.73 877.73

73.2 65.84 877.47

69.0 61.32 888.03 73.3 65.95 877.21

69.1 6l.43 887.79 73.4 66.06 876.96

69.2 61.54 887.54 73.5 66.17 876.70

69.3 61.64 887.29 73.6 66.28 876.44

69.4 61.75 887.05 73.7 66.39 876.18

69.5 61.85 886.80 73.8 66.50 875.92

69.6 61.96 886.55 73.9 66.61 875.66

69.7 62.07 886.31

69.8 62.17 886.06 74.0 66.72 875.40

69.9 62.28 885.81 74.1 66.83 875.14

74.2 66.94 874.88

70.0 62.39 885.56 74.3 67.05 874.62

70.1 62.49 885.31 74.4 67.16 874.36

70.2 62.60 885.06 74.5 67.27 874.10

70.3 62.71 884.82 74.6 67.38 873.84

70.4 62.81 884.57 74.7 67.49 873.58

70.5 62.92 884.32 74.8 67.60 873.32

70.6 63.03 884.07 74.9 67.71 873.06

70.7 63.13 883.82

70.8 63.24 883.57 75.0 67.82 872.79

70.9 63.35 883.32 75.1 67.93 872.53

75.2 68.04 872.27

71.0 63.46 883.06 75.3 68.15 872.00

71.1 63.56 882.81 75.4 68.26 871.74

71.2 63.67 882.56 75.5 68.38 871.48

71.3 63.78 882.31 75.6 68.49 871.21

71.4 63.89 882.06 75.7 68.60 870.95

71.5 63.99 881.81 75.8 68.71 870.68

71.6 64.10 881.55 75.9 68.82 870.42

71.7 64.21 88l.30

71.8 64.32 881.05 76.0 68.93 870.15

71.9 64.43 880.79 76.1 69.04 869.89

76.2 69.16 869.62

72.0 64.53 880.54 76.3 69.27 869.35

72.1 64.64 880.29 76.4 69.38 869.09

General Natices (1) apply ta all managraphs and ather texts 657
5.5. AlcohoHmetrk tables EUROPEAN PHARMACOPOEIA 8.0

% v/v %m/m 1"0 (kg/m3 ) % V/V %m/m 1'20 (kg/m3 )


76.5 69.49 868.82 80.8 74.41 857.03

76.6 69.61 868.55 80.9 74.53 856.75

76.7 69.72 868.28

76.8 69.83 868.02 81.0 74.64 856.46

76.9 69.94 867.75 8l.l 74.76 856.18

81.2 74.88 855.90

77.0 70.06 867.48 81.3 74.99 855.62

77.1 70.17 867.21 81.4 75.11 855.33

77.2 70.28 866.94 81.5 75.23 855.05

77.3 70.39 866.67 81.6 75.34 854.76

77.4 70.51 866.40 81.7 75.46 854.48

77.5 70.62 866.13 81.8 75.58 854.19

77.6 70.73 865.86 81.9 75.70 853.91

77.7 70.85 865.59

77.8 70.96 865.32 82.0 75.82 853.62

77.9 71.07 865.05 82.1 75.93 853.34

82.2 76.05 853.05

78.0 7l.l9 864.78 82.3 76.17 852.76

78.1 71.30 864.50 82.4 76.29 852.48

78.2 71.41 864.23 82.5 76.41 852.19

78.3 71.53 863.96 82.6 76.52 851.90

78.4 71.64 863.69 82.7 76.64 851.61

78.5 71.76 863.41 82.8 76.76 851.32

78.6 71.87 863.14 82.9 76.88 851.03

78.7 71.98 862.86

78.8 72.10 862.59 83.0 77.00 850.74

78.9 72.21 862.31 83.1 77.12 850.45

83.2 77.24 850.16

79.0 72.33 862.04 83.3 77.36 849.87

79.1 72.44 861.76 83.4 77.48 849.58

79.2 72.56 861.49 83.5 77.60 849.29

79.3 72.67 861.21 83.6 77.72 848.99

79.4 72.79 860.94 83.7 77.84 848.70

79.5 72.90 860.66 83.8 77.96 848.41

79.6 73.02 860.38 83.9 78.08 848.11

79.7 73.13 860.10

79.8 73.25 859.83 84.0 78.20 847.82

79.9 73.36 859.55 84.1 78.32 847.53

84.2 78.44 847.23

80.0 73.48 859.27 84.3 78.56 846.93

80.1 73.60 858.99 84.4 78.68 846.64

80.2 73.71 858.71 84.5 78.80 846.34

80.3 73.83 858.43 84.6 78.92 846.05

80.4 73.94 858.15 84.7 79.04 845.75

80.5 74.06 857.87 84.8 79.16 845.45

80.6 74.18 857.59 84.9 79.28 845.15

80.7 74.29 857.31

658 See the information section on general monographs (ca ver pages)
EUROPEAN PHARMACOPOEIA 8.0 5.5. Akoholimetrk tables

% v/v %m/m 1'20 (kg/m 3 ) % V/V %m/m 1"0 (kg/m 3 )

85.0 79.40 844.85 89.3 84.76 831.48

85.1 79.53 844.55 89.4 84.89 831.15

85.2 79.65 844.25 89.5 85.02 830.82

85.3 79.77 843.95 89.6 85.15 830.50

85.4 79.89 843.65 89.7 85.28 830.17

85.5 80.01 843.35 89.8 85.41 829.84

85.6 80.14 843.05 89.9 85.54 829.51

85.7 80.26 842.75

85.8 80.38 842.44 90.0 85.66 829.18

85.9 80.50 842.14 90.1 85.79 828.85

90.2 85.92 828.52

86.0 80.63 841.84 90.3 86.05 828.19

86.1 80.75 841.53 90.4 86.18 827.85

86.2 80.87 84l.23 90.5 86.31 827.52

86.3 81.00 840.92 90.6 86.44 827.18

86.4 81.12 840.62 90.7 86.57 826.85

86.5 8l.24 840.31 90.8 86.71 826.51

86.6 8l.37 840.00 90.9 86.84 826.17

86.7 81.49 839.70

86.8 81.61 839.39 91.0 86.97 825.83

86.9 81.74 839.08 91.1 87.10 825.49

91.2 87.23 825.15

87.0 81.86 838.77 9l.3 87.36 824.81

87.1 81.99 838.46 91.4 87.49 824.47

87.2 82.11 838.15 91.5 87.63 824.13

87.3 82.24 837.84 91.6 87.76 823.78

87.4 82.36 837.52 91.7 87.89 823.44

87.5 82.49 837.21 91.8 88.02 823.09

87.6 82.61 836.90 91.9 88.16 822.74

87.7 82.74 836.59

87.8 82.86 836.27 92.0 88.29 822.39

87.9 82.99 835.96 92.1 88.42 822.04

92.2 88.56 821.69

88.0 83.11 835.64 92.3 88.69 82l.34

88.1 83.24 835.32 92.4 88.83 820.99

88.2 83.37 835.01 92.5 88.96 820.63

88.3 83.49 834.69 92.6 89.10 820.28

88.4 83.62 834.37 92.7 89.23 819.92

88.5 83.74 834.05 92.8 89.37 819.57

88.6 83.87 833.73 92.9 89.50 819.21

88.7 84.00 833.41

88.8 84.13 833.09 93.0 89.64 818.85

88.9 84.25 832.77 93.1 89.77 818.49

93.2 89.91 818.12

89.0 84.38 832.45 93.3 90.05 817.76

89.1 84.51 832.12 93.4 90.18 817.40

89.2 84.64 831.80 93.5 90.32 817.03

General Natíces (1) apply ta all managraphs and ather texts 659
5.5. Alwholimetrk tables EUROPEAN PHARMACOPOEIA 800

% v/v %m/m 1'20 (kg/m3 ) % V/V %m/m 1'20 (kg/m 3 )

9306 90.46 816066 9609 95016 803.70

9307 90.59 816.30

9308 90.73 815093 9700 95.31 803.27

9309 90087 81555 9701 95.45 802085

97.2 95060 802.42

9400 91.01 815018 9703 95075 801.99

9401 91.15 814081 97.4 95090 801.55

9402 91.29 814.43 97.5 96.05 801.12

9403 91.43 814006 9706 96021 800068

94.4 91.56 813.68 9707 96.36 800024

9405 91.70 813.30 9708 96051 799080

9406 91.84 812092 9709 96066 799035

94.7 91.98 812.54

9408 92013 812.15 9800 96081 798090

9409 92.27 811.77 9801 96097 798.45

9802 97012 798000

9500 92.41 811.38 98.3 97028 797054

9501 92.55 810099 98.4 97.43 797008

95.2 92069 810060 98.5 97.59 796062

9503 92083 810.21 9806 97.74 796015

95.4 92098 809082 98.7 97090 795068

95.5 93012 809.42 9808 98006 795.21

9506 93026 809002 9809 98022 794073

95.7 93.41 808063

9508 9355 808023 9900 98.38 794.25

9509 93069 807082 9901 98.53 793.77

9902 98069 793028

9600 93084 807.42 99.3 98086 792.79

9601 93098 807.ül 99.4 99002 792.30

96.2 94013 806061 9905 99018 791.80

9603 94027 806.20 9906 99034 791.29

96.4 94.42 805078 9907 99.50 790079

96.5 94.57 805.37 9908 99067 790.28

9606 94071 804096 9909 99083 789.76

9607 94086 804.54

9608 95001 804012 10000 10000 789.24

660 See the infarmation sectian on general monagraphs (caver pages)


EUROPEAN PHARMACOPOEIA 8.0

5.6. Assay of interferons


5.6. Assay of interferons .......................................................... 663

General Natices (1) apply ta all managraphs and ather texts 661
EUROPEAN PHARMACOPOEIA 8.0

662 See the information section on general monographs (ca ver pages)
EUROPEAN PHARMACOPOEIA 8.0 5.6. Assay of interferons

01/2008:50600 - Incubate the flasks for 5 min to 10 min at 37 oc.


Microscopically or visually observe the cells for signs of
detachment. When viewed microscopically, the cells appear
5.6. ASSAY OF INTERFERONS rounded up or detached and free-floating. Shake the flask
vigorously to detach all the cells, add approximately 5 mL of
The following chapter is published for information. culture medium A. Shake vigorously to yield a suspension
of single cells.
l. INTRODUCTION - To prepare the cell suspensions for the assay procedure,
Monographs on human interferons gene rally contain a carefully disperse the ceUs by pipetting up and down to
bioassay based on the inhibitory activity of the interferon on disrupt cel! aggregates, count the cells and resuspend at a
the cytopathic action of a virus on a cellline in culture. In most concentration of 6 x 10 5 cells/mL.
cases, however, the virus, cellline and the assay details are not 3.2. PROPAGATION OF ENCEPHALOMYOCARDITIS
specified, in order to allow the appropriate flexibility, where VIRUS
the monograph covers more than one sub-class of interferon. Encephalomyocarditis virus is propagated in mouse L-929 cells
The present text is intended to provide outline information in order to produce a stock of progeny virus. L-929 cells are
for the analyst on how to design, optimise and validate such maintained by trypsin treatment and passage as described for
an assay once an appropriate combination of cellline and Hep2c cells (NOTE: it may be necessary ta substitute neanatal
cytopathic virus has been identified. A detailed procedure calf serum with foetal bovine serum if the cel/s show paar
for a particular cytopathic antiviral assay is described as an grawth).
example of a suitable method, together with information on Take several flasks containing confluent cultures ofL-929 cells.
other virus-cellline combinations and guidance on how to Pour off the medium from the flasks. Inoculate with 2 mL
adapt and validate the procedure for these other combinations. of the EMCV suspension appropriately diluted in culture
medium B so that it contains approximately 2.5 x 108 plaque
2. ANTIVIRAL (CYTOPATHIC EFFECT REDUCTION) forming units (PFU) per millilitre. Each flask will contain
ASSAYS 4-6 x 10 7 L-929 cells and therefore the multiplicity of infection
(m.oj.) will be approximately 10 PFU/cell. Carefully swirl the
The antiviral assay of human interferons is based on the
virus suspension over the entire cell monolayer and return
induction of a ceHular response in human cells, which
the flasks to the incubator for approximately 1 h. Maintain
prevents or reduces the cytopathic effect of an infectious
the medium at pH 7.4 to 7.8.
virus. The potency of interferon is estimated by comparing
its protective effect against a viral cytopathic effect with the After adsorption of the EMCV, add approximately 40 mL of
same effect of the appropriate reference preparation calibrated culture medium B to each flask and return the flasks to the
in International Units. incubator at 37 oC for about 30 h. Maintain the medium at
pH 7.4 to 7.8 to obtain a maximum virus yield. Remove the
3. INTERFERON ASSAY USING Hep2c CELLS AND culture fluid and sto re at approximately 4 oc.
INFECTIOUS ENCEPHALOMYOCARDITIS VIRUS Place the flasks at - 20 oC to freeze the cen monolayer. Then
thaw to room temperature. Add approximately 5 mL of culture
The antiviral assay of human interferons described is of
medium and shake the flask to disrupt the cel! walls. Transfer
the cytopathic effect reduction type. It uses human Hep2c
the contents of each flask to the container of culture fluid.
cells infected by encephalomyocarditis virus (EMCV) to
Transfer the culture fluid containing the EMCV to 50 mL
measure the potencies of different human interferon test
plastic centrifuge tubes and centrifuge at approximately 500 g
preparations. This assay has been used in three World Health
for about 10 min to remove ceH debris. Dispense the clarified
Organization (WHO) international collaborative studies of
culture fluid into glass screw-capped bottles, in quantities of
candidate International Standards for human interferon alpha,
20 mL, 10 mL, 5 mL, 1 mL, 0.5 mL or 0.2 mL, as appropriate.
human interferon beta and human interferon gamma and has
Store at - 70 oc. Larger volumes can be thawed, dispensed
repeatedly been demonstrated to be sensitive, reliable and
into smaller quantities and re-frozen when required. The
reproducible for potency estimations of the different types of
EMCV stock will retain its original titre if stored permanently
human interferon.
at approximately - 70 oC, but repeated freeze-thaw cycles or
For the culture of mammalian cells, all procedures are carried storage at higher temperatures, e.g. at approximately - 20 oC,
out using standard operating procedures for the maintenance results in progressive 10ss of titre.
of such celllines in culture. Volumes of reagents are indicated 3.3. ASSAY PROCEDURE
for cel! cultures carried out in 75 cm 2 flasks. Other types of
containers (flasks or plates) may be used but volumes must 3.3.1. Determination of the dose-response range
be adapted accordingly. Preparation oi the solutians
3.1. MAINTENANCE AND PREPARATION OF Hep2c CELLS Dilute the appropriate standard for interferon (for example
Hep2c cells are maintained and passaged in culture medium A. a specific WHO sub-type interferon standard) in culture
medium A, in 10-fold dose increments, to give doses covering
Cells are stored as frozen stocks using standard operating the range of 1000 - 0.001 IU/mL. Carry out the assay pro ce dure
procedures. Growing cells may be maintained in culture up to in 96-well microtitre plates. To each well add 100 flL of culture
a permitted passage number of 30, after which new cultures medium A. Add approximately 100 flL of each dilution of the
are established from frozen stocks. reference preparation to each well except for those intended
At the beginning of the assay procedure, harvest the cells from for virus controls. Using a multichannel pipette set at 100 flL,
the flasks showing 90 per cent confluent monolayers using the mix the contents of the wells.
trypsin-treatment pro ce dure described below. Dispensing of the ceU suspension
- Remove the culture medium from the flasks. Pour the ceH suspension of Hep2c cells, which has been
- To each flask, add 5 mL of trypsin solution heated at 37 oC adjusted to contain approximately 6 x 10 cells/mL of culture
5

(a trypsin stock solution contains 4 mg/mL of trypsin R medium A, into a plastic Petri-dish. Dispense the cel!
and 4 mg/mL of sodium edetate R; immediately before use, suspension from the Petri -dish into each well of the microtitre
dilute 50 times with phosphate buffered saline). Swirl the plates, using a multichannel pipette set at 100 flL.
capped flask to wash the cel! monolayer. Remove the excess Incubate the plates for about 24 h in an incubator set at 37 oC
of trypsin solution. and 5 per cent CO 2 •

General Natices (1) apply to all monographs and ather texts 663
5.6. Assay of interferons EUROPEAN PHARMACOPOEIA 8.0

Viral infection concentration (the log of the reciprocal of the interferon


At this stage, using an inverted microscope, check that the dilution) is plotted versus stain absorbance.
monolayers of Hep2c cells are confiuent, that they show a Plot the interferon concentration (log reciprocal of dilution)
relatively even distribution of cel!s, that they have correct versus the stain absorbance for the interferon reference
morphology and that they are healthy. preparation and for the interferon test solutions. Using the
linear portion of the curve, calculate the concentration of
Remove most of the culture medium from the wells by interferon in the sample by comparing the responses for test
inverting the plate and shaking it and blotting on a paper and reference solutions, using the usual statistical methods
towel (proceed in an identical way when discarding fiuids for a parallelline assay.
from micro-titre plates as described later). Dilute the EMCV
stock with fresh culture medium A to a titre of approximately 4. VALIDATION OF OTHER PROCEDURES
3 x 107 PFU/mL (NOTE: each plate requires approximately 4.1. CHOICE OF CELL LINE AND VIRUS
20 mL of diluted virus, plus 5 per cent to 10 per cent of extra
A number of other combinations of cellline and virus
volume). Dispense the diluted suspension from a 9 cm sterile
have been used in anti-viral assays for interferons. For
Petri -dish using a multichannel pipette set at 200 ¡.tL to al!
example, EMCV has been used in combination with the A549
wells including virus controls, but excluding cel! controls. Add
epitheliallung carcinoma cellline, Semliki Forest virus or
approximately 200 ¡.tL of culture medium A without virus to
Sindbis virus have been used with human fibroblasts, and
each of the cell control wells.
vesicular stomatitis virus has been used with either human
Return the plates to the incubator set at 37 oC and 5 per cent diploid fibroblasts, the human amnion WISH cellline or
CO 2 for approximately 24 h. the Madin-Darby bovine kidney cellline. In each case the
Staining choice of the cellline/virus combination is usually based on
that which gives the most sensitive response to the interferon
Examine the plates microscopically to check that the EMCV preparation to be assayed, and gives parallel responses when
has caused a cytopathic effect (c.p. e.) in the virus controls. comparing the test preparation and interferon standard.
The time interval for maximum c.p.e. mayvaryfrom one assay
to the next because of inherent variation of Hep2c cells to 4.2. CHOICE OF RESPONSE
virus challenge over a given period of continuous cultivation. The staining procedure described aboye measures remaining
viable cells. A number of other responses have been used,
Remove most of the culture medium from the wells by including methyl violet or crystal violet staining, or the
discarding into an appropriate decontaminating solution thiazolyl blue (MTT) conversion procedure. In each case, the
(sodium hypochlorite is suitable). Dispense phosphate method is selected on the basis of producing a suitably linear
buffered saline pH 7.4 R into each well. Discard the phosphate and sensitive relationship between response colour and viable
buffered saline pH 7.4 R into a decontaminating solution. cel! count.
Dispense into each well150 ¡.tL of staining solution. Stain
the cells for approximately 30 min at room temperature. 4.3. STATISTICAL VALIDATION
Discard the staining solution into a decontaminating solution. As with all parallelline bioassays, the assay must satisfy the
Dispense approximately 150 ¡.tL of fixing solution. Fix for usual statistical criteria of linearity of response, parallelism
10 min at room temperature. Discard the fixing solution into and variance.
a decontaminating solution and wash the cell monolayers by 4.4. VALIDATION OF ASSAY LAYOUT
immersing the assay plates in a plastic box containing running As with aH microtitre plate assay procedures, attention must
water. Discard the water and superficially dry the plates with be given to validating the assay layout. In particular, bias due
paper towels. Dry the assay pi ates at 20 oC to 37 oC until al! to non-random pipetting order or plate edge effects must be
moisture has evaporated. investigated and eliminated, by randomising the assay layout,
Add 150 ¡.tL of 0.1 M sodium hydroxide to each well. Elute or by avoiding the use of edge wells.
the stain by gentle agitation of the plates or by hitting them
REAGENTS AND CULTURE MEDIA
against the palm of the hand. Make sure that the stain is evenly
distributed in al! wells before making spectrophotometric Culture medium A (10 per cent neonatal calf serum)
readings. RPMI 1640 culture medium, supplemented with antibiotics if 450mL
necessary (penicillin 10 000 IU/mL; streptomycin 10 ng/mL)
Read the absorbance at 610 nm to 620 nm, using a microtitre L-Glutamine, 200 mM, sterile 5 mL
plate reader, taking as a blank a well or a column of wells
containing no cells and approximately 150 ¡.tL of 0.1 M sodium Neonatal calf serum 50 mL
hydroxide.
Culture medium B (2 per cent foetal bovine serum)
Estimate the concentrations of interferon standard that give 490 mL
RPMI 1640 culture medium, supplemented with antibiotics if
the maximum and minimum reduction of cytopathic effect. necessary (penicillin 10 000 IU/mL; streptomycin 10 ng/mL)
This is the dose response corresponding to the working range L-Glutamine, 200 mM, sterile 5 mL
of the assay.
Foetal bovine serum 10 mL
3.3.2. Assay procedure
Carry out the assay as described aboye, using: Staining solution
Naphthalene black 0.5 g
- as test solutions, the substance to be examined, diluted
in two-fold increments with culture medium A to give Acetic acid, glacial 90 mL
nominal concentrations covering the working range of the
Sodium aceta te, anhydrous 8.2 g
assay,
- as reference solutions, the appropriate standard for Water to 1000 mL
interferon (for example, a specific WHO sub-type
Fixing solution
interferon) in culture medium A, diluted in two-fold
increments to give nominal concentrations covering the Formaldehyde, 40 per cent 100mL
working range of the assay. Acetic acid, glacial 90 mL
3.3.3. Data analysis Sodium acetate, anhydrous 8.2 g
Results of the cytopathic effect reduction assay generally Water to 1000 mL
fit a sigmoidal dose-response curve, when the interferon

664 See the information section on general monographs (cover pages)


EUROPEAN PHARMACOPOEIA 8.0

5.7. Table of physical characteristics


of radionuclides mentioned in the
European Pharmacopoeia
5.7. Table of physical characteristics of radionuclides
mentioned in the European Pharmacopoeia ...................... 667

General Natices (1) apply ta all rnanagraphs and ather texts 665
EUROPEAN PHARMACOPOEIA 8.0

666 See the information section on general monographs (caver pages)


EUROPEAN PHARMACOPOEIA 8.0 5.7. Table of physkal chal'acteristks of radionudides

0112008:50700 ** PTB (Physikalisch-Technische Bundesanstalt,


Braunschweig, Germany),
*** NPL (National Physical Laboratory, Teddington,
5.7. TABLE OF PHYSICAL Middlesex, UK).
CHARACTERISTICS OF The uncertainty of the half-lives are given in parentheses.
RADIONUCLIDES MENTIONED IN In principIe the digits in parentheses are the standard
uncertainty of the corresponding last digits of the indicated
THE EUROPEAN PHARMACOPOEIA numerical value ('Guide to the Expression oí Uncertainty in
Measurement: International Organization for Standardization
The following table is given to complete the general (ISO), 1993, ISBN 92-67-10188-9).
monograph Radiopharmaceutical preparations (0125). The following abbreviations are used:
The values are obtained from the database of
eA Auger electrons,
the National Nuclear Data Center (NNDC) at
Brookhaven National Laboratory, Upton. N.Y., ce conversion electrons,
USA, directly accessible via Internet at the address:
'http://www.nndc.bnl.gov/nndc/nudat/radform.html'. ~- electrons,
In case another source of information is preferred (more
recent values), this source is explicitly mentioned. ~+ positrons,
Other data sources: y gamma rays,
* DAMRI (Département des Applications et de la Métrologie X X-rays.
des Rayonnements Ionisants, CEA Gif-sur-Yvette, France),
Electronic emission Photon emission

Emission Emission
probability probability
Radionllclide Half-life Type Energy (MeV) Type Energy (MeV)
(per 100 dis- (per 100 dis-
integrations) integrations)
Tritium eH) *12.33 (6) years '~- *0.006 (1) (max: 0.019) 'lOO
Carbon-ll (1lC) 20.385 (20) min ~' 0.386 (1) (max: 0.960) 99.8 Y 0.511 199.5 (11)

Nitrogen-13 C'N) 9.965 (4) min W 0.492 (1) (max: 1.198) 99.8 Y 0.511 199.6 (11)

Oxygen-15 ( 0)
15 122.24 (16) s W 0.735 (1) (max: 1.732) 99.9 Y 0.511 199.8 (11)

Pluorine-18 (IsP) 109.77 (5) min W 0.250 (1) (max: 0.633) 96.7 Y 0.511 193.5 (11)

Phosphorus-32 (32 P) 14.26 (4) days ~- 0.695 (1) (max: 1.71) 100

Phosphorus-33 (33P) 25.34 (12) days ~- 0.076 (I) (max: 0.249) 100

Sulfur-35 ("S) 87.51 (12) days V 0.049 (1) (max: 0.167) 100

27.7025 (24) days eA 0.004 67 X 0.005 22.3


Chromium-51 (SlCr)
y 0.320 9.9

77.27 (3) days e, 0.006 47 X 0.006-0.007 25

W 0.179 (1) 0.9 Y 0.511 38.0 (11)

0.631 (Il 18.1 0.847 100.0

1.038 14.1

1.175 2.2

1.238 66.1
Cobalt-56 (56 Co)
1.360 4.3

1.771 15.5

2.015 3.0

2.035 7.8

2.598 17.0

3.202 3.1

3.253 7.6

(1) Mean energy of the ~ spectrum.


(II) Maximum emission probability correspondi~ to a total annihilation in the source per 100 disintegrations.

General Notices (1) apply to all monographs and other texts 667
5.7. Table of physical characteristics of radionudides EUROPEAN PHARMACOPOEIA 8.0

Electronic emission Photon emission

Emission Emission
probability probability
Radionuclide Half-life Type Energy (MeV) Type Energy (MeV)
(per 100 dis- (per 100 dis-
integrations) integrations)
271.79 (9) days eA+ce 0.006-0.007 177.4 X 0.006-0.007 57

ce 0.014 7.4 Y 0.014 9.2


Cobalt-57 ("Co)
0.115 1.8 0.122 85.6

0.129 1.3 0.136 10.7

0.692 0.15

70.86 (7) days eA 0.006 49.4 X 0.006-0.007 26.3

~; 0.201 (1) 14.9 Y 0.511 29.9 (II)

Cobalt-58 (58 Co)


0.811 99.4

0.864 0.7

1.675 0.5

5.2714 (5) years ~- 0.096 (1) (max: 0.318) 99.9 Y 1.173 100.0
Cobalt-60 (60CO)
1.333 100.0

9.49 (7) hours eA 0.008 21 X 0.009-0.010 19.1

~+ 0.157 (1) 1 Y 0.511 112 IlI)

0.331 Ir) 0.7 0.834 5.9

0.397 Ir) 3.8 1.039 37

0.782 (1) 0.3 1.333 1.2

1.90 (1) 50 1.919 2.1

2.190 5.6
Gallium-66 (MiGa)
2.423 1.9

2.752 23.4

3.229 1.5

3.381 1.5

3.792 1.1

4.086 1.3

4.295 4.1

4.807 1.8

3.2612 (6) days eA 0.008 62 X 0.008-0.010 57

ce 0.082-0.084 30.4 Y 0.091-0.093 42.4

0.090-0.092 3.6 0.185 21.2


Gallium-67 (67 Ga)
0.175 0.3 0.209 2.4

0.300 16.8

0.394 4.7

0.888 0.15

270.82 (27) days eA 0.008 42.4 X 0.009-0.010 44.1

Germanium-68 (68 Ge)


in equilibrium with
Gallium-68 (68 Ga) W 0.353 Ir) 1.2 y 0.511 178.3
("Ga: 67.629 0.836 (1) 88.0 1.077 3.0
(24) min)
(I) Mean energy of the Pspectrurn.
(II) Maxirnurn ernission probability corresponding to a total annihilation in the source per 100 disintegrations.

668 See the information section on general monographs (caver pages)


EUROPEAN PHARMACOPOEIA 8.0 5.7. Table oí physical characteristics oí radionudides

Electronic emission Photon emission

Emission Emission
probability probability
Radionudide Half-life Type Energy (MeV)
(per 100 dis-
Type Energy (MeV)
(per 100 dis-
integrations) integrations)
67.629 (24) min eA 0.008 5.1 X 0.009-0.010 4.7

Gallium-68 (68 Ga)


W 0.353 (1) 1.2 Y 0.511 178.3
0.836 (1) 88.0 1.077 3.0

13.10(3)s ee 0.176 26.4 X 0.012-0.014 17.0

Krypton-81m (81m Kr) 0.189 4.6

Y 0.190 67.6

4.576 (5) hours eA 0.011 31.3 X 0.013-0.014 57.2

ee 0.176 25.0 Y 0.190 64

Rubidium -81 ('1 Rb) 0.188 4.3 0.446 23.2


in equilibrium with
Krypton-81m ('lmKr) 0.457 3.0

~+ 0.253 (l) 1.8 0.510 5.3


0.447 (1) 25.0 0.511 54.2

('lmKr: 13.10 (3) s) 0.538 2.2

50.53 (7) days ~- 0.583 (1) (max: 1.492) 99.99 Y 0.909 0.01
Strontium-89 (89 Sr)
in equilibrium with
Yttrium-89m (89my)
(89my: 16.06 (4) s)
28.74 (4) years ~- 0.196 (1) (max: 0.546) 100
Strontium-90 (9OSr)
in equilibrium with
Yttrium-90 ('"Y)
(9OY: 64.10 (8) hours)
64.10 (8) hours ~- 0.934 (1) (max: 2.280) 100
Yttrium-90 (90y)

65.94 (1) hours ~- 0.133 (1) 16.4 X 0.018-0.021 3.6

0.290 (l) 1.1

0.443 (I) 82.4 Y 0.041 1.1


Molybdene-99 (99Mo) 0.141 4.5
in equilibrium with
Technetium-99m 0.181 6
(99mTe)
0.366 1.2

('9mTe: 6.01 (1) hours) 0.740 12.1

0.778 4.3

6.01 (1) hours ce 0.002 74 X 0.018-0.021 7.3

eA 0.015 2.1 Y 0.141 89.1


Teehnetium-99m
(99mTe)

ee 0.120 9.4

0.137-0.140 1.3

2.11 x 10 5 years ~- 0.085 (1) (max: 0.294) 100


Technetium-99 (99Te)

(I) Mean energy of the ~ spectrum.


(n) Maximum emission probability corresponding to a total annihilation in the souree per 100 disintegrations.

General Notices (1) apply to all monographs and other texts 669
5.7. Table of physkal characteristic§ of radionudides EUROPEAN PHARMACOPOEIA 8.0

Electronic emissian PIlOtan emission

Emission Emission
probability probability
Radionudide Half-Iife Type Energy (MeV) Type Energy (MeV)
(per 100 dis- (per 100 dis-
integratians) integrations)
39.26 (2) days eA+ce 0.017 12 X 0.020-0.023 9.0

Ruthenium-103 C03 Ru) ce 0.030-0.039 88.3 Y 0.497 91


in eqnilibrium with
Rhodium -103m 0.610 5.8
C03 mRh)
~- 0.031 (l) 6.6
(I 03m Rh: 56.ll4 (20) 0.064 (1) 92.2
min)
4.9 (1) hours eA 0.019 13.4 X 0.023-0.026 70.5

Y 0.642 25.9

Indium-110 CIOIn) 0.658 98.3

0.885 92.9

0.938 68.4

0.997 10.5

69.1 (5) min eA. 0.019 5.3 X 0.023-0.026 27.8

l.015 (1) Y 123.4


Indium-llOm ('lOmln) W 61 0.5ll (11)

0.658 97.8

2.129 2.1

2.8047 (5) days eA 0.019 15.6 X 0.003 6.9

0.023-0.026 82.3
ce 0.145 7.8
Indium-l11 CllIn)
0.167-0.171 1.3 Y 0.171 90.2

0.219 4.9 0.245 94.0

0.241-0.245 l.0

49.51 (1) days ce 0.162 40 X 0.023-0.027 36.3

0.186-0.190 40
Indium-114m ('14mIn)
in equilibrium with y 0.190 15.6
Indium-114 (1l4In)
*~- 0.777 (1) (max: l.985) 95 0.558 3.2

(1l4In: 7l.9 (1) s) 0.725 3.2

154.0 (7) days eA 0.003 88.0 X 0.026-0.031 50.5

0.022-0.023 7.4

Tellurium-121m y 0.212 8l.4


(l2lmTe) in equilibrium
ce 0.050 33.2 1.102 2.5
with Tellure-l21 (,2lTe)
e Te: 19.16 (5) days)
2l 0.077 40.0

0.180 6.1

**19.16 (5) days eA 0.022 1l.6 X 0.026-0.030 75.6

Tellurium-121 e2l Te) y 0.470 1.4

0.508 17.7

0.573 80.3

(1) Mean energy of the ~ spectrum.


(11) Maximum ernission probability corresponding to a total annihilation in the source per 100 disintegrations.

670 See the information section on general monographs (caver pages)


EUROPEAN PHARMACOPOEIA 8.0 5.7. Table of physical charaderistics oí radionudides

Electronic emission Photon emission

Emission Emission
probability probability
Radionuclide Half-life Type Energy (MeV) Type Energy (MeV)
(per 100 dis- (per 100 dis-
integrations) integrations)
13.27 (8) hours eA 0.023 12.3 X 0.004 9.3

0.027-0.031 86.6
ce 0.127 13.6

0.154 1.8 Y 0.159 83.3

lodine-l23 (i23I) 0.158 0.4 0.346 0.1

0.440 0.4

0.505 0.3

0.529 1.4

0.538 0.4

59.402 (14) days e/\.+ce 0.004 80 X 0.004 15.5

0.023-0.035 33 0.027 114

lodine-125 [1 25 1) 0.031 26

Y 0.035 6.7

13.11 (5) days e,; 0.023 6 X 0.027-0.031 42.2

ce 0.354 0.5 Y 0.388 34

0.634 0.1 0.491 2.9

0.511 2.3 (JI)

lodine-126 (1 "'1)
~- 0.109 (1) 3.6 0.666 33

0.290 (1) 32.1 0.754 4.2

0.459 (J) 8.0 0.880 0.8

1.420 0.3

W 0.530 (l) 1

8.02070 (11) days ce 0.46 3.5 X 0.029-0.030 3.9

0.330 1.6

Y 0.080 2.6

lodine-131 [13(1) ~- 0.069 (J) 2.1 0.284 6.1

0.097 (1) 7.3 0.365 81.7

0.192 11) 89.9 0.637 7.2

0.723 1.8

11.84 (7) days eA 0.025 6.8 X 0.004 8.3

0.030 44.0
ce 0.129 61 0.034 10.2
Xenon-13lm (D1mXe)
0.159 28.5

0.163 8.3 Y 0.164 2.0

20.8 (1) hours ~- 0.140 II ) 3.8 Y 0.530 87

lodine-133 [1331) 0.162 (J) 3.2 0.875 4.5


(decays lo radio active
0.299 (1) 4.2 1.298 2.4
Xenon-133)
0.441 (1) 83

(I) Mean energy of the ~ spectrurn.


(I!) Maximurn emission probability corresponding to a total annihilation in the source per lOO disintegrations.

General Notices (1) apply to all monographs and other texts 671
5.7. Table of physical characíeristics of radionudides EUROPEAN PHARMACOPOEIA 8.0

Electronic emission Photon emission

Emission Emission
probability probability
Radionuclide Half-life Type Energy (MeV) Type Energy (MeV)
(per 100 dis- (per 100 dis-
integrations) integrations)
5.243 (1) days eA 0.026 5.8 X 0.004 6.3

0.031 40.3

ce 0.045 55.1 0.035 9.4


Xenon-133 ( 133Xe)
0.075-0.080 9.9

Y 0.080 38.3

~- 0.101 (1) 99.0

2.19 (1) days eA 0.025 7 X 0.004 7.8

0.030 45.9
Xenon-133m ('33mXe)
ce 0.199 64.0 0.034 10.6
(decays to radio active
Xenon-133)
0.228 20.7

0.232 4.6 Y 0.233 10.0

6.57 (2) honrs ~- 0.140 (J) 7.4 Y *0.527 13.8

0.237 (1) 8 0.547 7.2

0.307 (l) 8.8 0.837 6.7

0.352 (J) 21.9 1.039 8.0


lodine- 135 (' 35 1)
(decays to radio active 0.399 (1) 8 1.132 22.7
Xenon-135)
0.444 (1) 7.5 1.260 28.9

0.529 (J) 23.8 1.458 8.7

1.678 9.6

1.791 7.8

9.14 (2) honrs ce 0.214 5.5 X 0.031-0.035 5.0

Xenon-135 ('3'Xe)
~- 0.171 3.1 Y 0.250 90.2

0.308 96.0 0.608 2.9

30.04 (3) years eA 0.026 0.8 X 0.005 1

0.032-0.036 7

ce 0.624 8.0
Caesium-137 ( 137 Cs)
in equilibrium with 0.656 1.4 Y 0.662 85.1
Barium-137m ('37mBa)

~- 0.174 (1) 94.4

0.416 (J) 5.6


( 137m Ba: 2.552 (1) min)
26.1 (1) honrs ce 0.285 3.4 X 0.ül0 32.0

0.353 1.4 0.069-0.071 63.3

0.08 17.5

W 0.495 (1) 0.3

Y 0.368 87.2

0.579 13.8
Thallium-200 (200Tl)
0.828 10.8

1.206 29.9

1.226 3.4

1.274 3.3

1.363 3.4

1.515 4.0

(1) Mean energy of the ~ spectrum.


(11) Maximum emission probability corresponding to a total annihilation in the sonrce per 100 disintegrations.

672 See the information section on general monographs (ca ver pages)
EUROPEAN PHARMACOPOEIA 8.0 5.7. Table of physical characteristics off radionudides

Electronic emission Pnoton emission

Emission Emission
probability probability
Radionudide Half-Iife Type Energy (MeV) Type Energy (MeV)
(per 100 dis- (per 100 dis-
integrations) integrations)
9.33 (3) hours eA 0.055 3 X 0.070-0.073 69

0.083 19
ce 0.246 8.5

0.276 2 Y 0.331 79

0.316 2.3 0.361 9.9

0.406 2.0

Lead-201 (,oIPb) 0.585 3.6


(decays to radio active
Thallium-201) 0.692 4.3

0.767 3.2

0.826 2.4

0.908 5.7

0.946 7.9

1.099 1.8

1.277 1.6

72.912 (17) hours ce 0.016-0.0l7 17.7 X 0.010 46.0

0.027 -0.029 4.1 0.069-0.071 73.7

0.052 7.2 0.080 20.4


Thallium-201 (20I TI)
0.084 15.4

0.153 2.6 Y 0.135 2.6

0,167 10.0

12.23 (2) days e,l 0.054 2.8 X 0.010 31.0

0.069-0.071 61.6

Thallium-202 (20'TI) ce 0.357 2.4 0.080 17.1

Y 0.440 91.4

51.873 (9) hours eA 0.055 3.0 X 0.010 37.0

0.071-0.073 69.6
ce 0.194 13.3 0.083 19.4
Lead-Z03 ('03 Pb)

y 0.279 80.8

0.401 3.4

(l) Mean energy of the p spectrurn.


(Ir) Maxirnurn ernission probability corresponding to a total annihilation in the source per 100 disintegrations.

General Notices (1) apply to all monographs and other texts 673
EUROPEAN PHARMACOPOEIA 8.0

674 See the information section on general monographs (cover pages)


EUROPEAN PHARMACOPOEIA 8.0

5.8. Pharmacopoeial harmonisation


5.8. Pharmacopoeial harmonisation" """""."""""""""""." 677

General Notices (1) apply to all monographs and other texts 675
EUROPEAN PHARMACOPOEIA 8.0

676 See the information section on general monographs (caver pages)


EUROPEAN PHARMACOPOEIA 8.0 5.80 Pharmacopoeial harmonisation

0112014:50800 2.2.31. ELECTROPHORESIS


The following comparative commentary refers to the texts
5.8. PHARMACOPOEIAL 23. SDS-Polyacrylamide Gel Electrophoresis in the Japanese
Pharmacopoeia XV and <1056> Biotechnology-derived Articles
HARMONISATION - Polyacrylamide Gel Electrophoresis in the United States
This general chapter is included for guidance of users. Pharmacopeia USP31 NF26 2nd Supplement, and chapter
2.2.31. Electrophoresis in the European Pharmacopoeia.
It provides information on the degree of harmonisation of
In the Ph. Eur. the harmonised chapter has been included
various general chapters and monographs of the European
as a section entitled Sodium dodecyl sulfate polyacrylamide
Pharmacopoeia and those of the Japanese Pharmacopoeia and
gel electrophoresis (SDS-PAGE), within a more general
United States Pharmacopeia.
chapter entitled Electrophoresis. The general chapter includes
This information reflects: other parts: General principIe, Free or moving boundary
- for general methods, the status observed for a given edition electrophoresis, Zone electrophoresis using a supporting
of the corresponding regional texts and any notification of medium, and Polyacrylamide rod gel electrophoresis, which
regulatory acceptance by ICH (International Conference are not within the scope of pharmacopoeial harmonisation.
on Harmonisation); The corresponding parts have been placed between black
- for monographs on excipients, the decision taken by the diamonds ( •• ).
3 regional pharmacopoeias when the harmonised text was The aboye differences in the Ph. Eur. text do not affect
signed off. harmonisation as the general chapter provides additional
information.
However, it remains the ultimate responsibility of the user to
verify the current content of the texts in force in the respective The texts of the 3 pharmacopoeias are therefore considered
pharmacopoeias. harmonised.
The chapter does not affect in any way the status of the 2.2.47. CAPILLARY ELECTROPHORESIS
monographs and general chapters as the authoritative As a result of an evaluation of the texts 4. Capillary
reference in any case of doubt or dispute where compliance Electrophoresis in the Japanese Pharmacopoeia XV
with the European Pharmacopoeia is required. and <1053> Biotechnology-derived articles - Capillary
The European Pharmacopoeia Commission recognises the Electrophoresis in the Unites States Pharmacopoeia USP33
utility of working with other pharmacopoeial bodies to NF28, and chapter 2.2.47. Capillary electrophoresis in the
develop harmonised monographs and general chapters. Such European Pharmacopoeia, the texts of the 3 pharmacopoeias
harmonisation is fully compatible with the declared aims of are considered harmonised.
the Commission and has benefits of different kinds, notably
the simplification and rationalisation of quality control 2.2.54. ISOELECTRIC FOCUSING
methods and licensing procedures. Such harmonisation also As a result of an evaluation of the texts 9. Isoelectric
enhances the benefits of the work of ICH and the Veterinary Focusing in the Japanese Pharmacopoeia XV and <1054>
International Cooperation on Harmonisation (VICH) since Biotechnology-derived articles - Isoelectric Focusing in the
sorne of the guidelines developed depend on pharmacopoeial Unites States Pharmacopoeia USP33 NF28, and chapter 2.2.54.
general chapters for their application. Isoelectric focusing in the European Pharmacopoeia, the texts
Work on harmonisation is carried out by a well-defined but of the 3 pharmacopoeias are considered harmonisedo
informal process in the Pharmacopoeial Discussion Group
2.2.55. PEPTIDE MAPPING
(PDG), in which the European Pharmacopoeia, the Japanese
Pharmacopoeia and the United States Pharmacopeia are The following comparative commentary refers to the texts
associated. Information is given in this general chapter on 15. Peptide Mapping in the Japanese Pharmacopoeia XV and
items that have been dealt with by the PDG: <1055> Biotechnology-derived Articles - Peptide Mapping in
the United States Pharmacopeia USP31 NF26 2nd Supplement,
- where harmonisation of general chapters is carried out, the and chapter 2.2.55. Peptide mapping in the European
aim is to arrive at interchangeable methods or requirements Pharmacopoeia.
so that demonstration of compliance using a general
chapter from one of the 3 pharmacopoeias implies that the Validation (USP). The USP has entitled this part System
same result would be obtained using the general chapter Suitability. This terminology has been accepted by the
of either of the other pharmacopoeias; when a formal 3 pharmacopoeias.
declaration of interchangeability has been recommended The use of peptide mapping for genetic stability evaluation
by ICH, it will be indicated in this general chapter; (USP). This additional section does not impact harmonisation
- where harmonisation of monographs is carried out, the aim since it is used only in development.
is to arrive at identical requirements for aH attributes of a The aboye differences in the USP text do not affect
product; for sorne products it can be extremely difficult harmonisation.
to achieve complete harmonisation, for example because The texts of the 3 pharmacopoeias are therefore considered
of differences in legal status and interpretation; it has harmonised.
therefore appeared worthwhile to the PDG to approve
and publish monographs in which as many attributes as 2.2.56. AMINO ACID ANALYSIS
possible are harmonised. The following comparative commentary refers to the texts
Information on any non-harmonised attributes/provisions 1. Amino Acid Analysis in the Japanese Pharmacopoeia XV
and on any local requirements, Le. attributes/provisions that and <1052> Biotechnology-derived Artie/es - Amino Acid
are present only in the Ph. Eur. text, is included in this general Analysis in the United States Pharmacopeia USP31 NF26
chapter. The non-harmonised attributes/provisions are placed 1st Supplement, and chapter 2.2.56. Amino acid analysis in
between black diamonds (. +) in the corresponding Ph. Eur. the European Pharmacopoeia.
texts., while the local requirements are placed between white Methodologies of amina add analysis: general principIes
diamonds (<><». (USP). The USP has replaced '6-aminoquinolyl-N-
The 3 pharmacopoeias have undertaken not to make unilateral hydroxysuccinimidyl carbamate or o-phthalaldehyde' with
changes to harmonised monographs and general chapters '6-aminoquinolyl-N -hydroxysuccinimidyl carbonate'.
but rather to apply the agreed revision procedure whereby all These reagents are different but compatible and the use of one
partners adopt a revision simultaneously. or the other does not affect harmonisation.

General Notices (1) apply to all monographs and other texts 677
5.8. Pharmacopoeial harmonisation EUROPEAN PHARMACOPOEIA 8.0

The USP has added a detailed example to describe each 2.6.12. MICROBIOLOGICAL EXAMINATION OF
method listed below: NON-STERILE PRODUCTS: MICROBIAL ENUMERATION
- Method 1: post-column ninhydrin detection; TESTS
- Method 2: post-column OPA derivatisation; As a result of an eva!uation of the texts 4.05 Microbiological
Examination of Non-sterile Products: 1. Microbiological
- Method 3: pre-column PITC derivatisation; Examination of Non-sterile Products - Microbial Enumeration
- Method 4: pre-column AQC derivatisation; Tests in the Japanese Pharmacopoeia XV 1st Supplement
Method 5: pre-column OPA derivatisation; and <61> Microbiological Examination of Non-sterile
Products: Microbial Enumeration Tests in the United
- Method 6: pre-column DABS-Cl derivatisation;
States Pharmacopeia USP30 NF25, and chapter 2.6.12.
- Method 7: pre-column FMOC-Cl derivatisation; Microbiological examination of non-sterile products: microbial
- Method 8: pre-column NBD-F derivatisation. enumeration tests in the European Pharmacopoeia, the texts
of the 3 pharmacopoeias are considered harmonised.
The aboye examples are given for further information and
do not affect harmonisation. NOTE: ICH has declared this method interchangeable within
the ICH regions.
The texts of the 3 pharmacopoeias are therefore considered
harmonised. 2.6.13. MICROBIOLOGICAL EXAMINATION OF
NON-STERILE PRODUCTS: TEST FOR SPECIFIED
2.4.14. SULFATED ASH MICRO-ORGANISMS
The following comparative commentary refers to the texts 2.44 As a result of an evaluation of the texts 4.05 Microbiological
Residue on Ignition Test in the Japanese Pharmacopoeia XV Examination of Non-sterile Products: JI. Microbiological
and <281> Residue on Ignition in the United States Examination of Non-sterile Products Test for Specified
Pharmacopeia USP32 NF27 1't Supplement, and chapter Micro-organisms in the Japanese Pharmacopoeia XV 1't
2.4.14. Sulfated ash in the European Pharmacopoeia. Supplement and <62> Microbiological Examination of
The JP has added a non-harmonised introductory part, Non-sterile Products: Test for Specified Micro-organisms in the
included between black diamonds, at the beginning of this United States Pharmacopeia USP30 NF25, and chapter 2.6.13.
chapter. It is given for further information and therefore does Microbiological examination of non-sterile products: test for
not affect harmonisation. specified micro-organisms in the European Pharmacopoeia,
The USP text allows for the test to be performed at an the texts of the 3 pharmacopoeias are considered harmonised.
ignition temperature other than 600 ± 50 oC if prescribed in NOTE: ICH has declared this method interchangeable within
an individual monograph. In the same way, a sample mass the ICH regions.
different fram the usual quantity of 1-2 g can be used if
prescribed in an individual monograph. 2.9.1. DISINTEGRATION OF TABLETS AND CAPSULES
The USP has added a section, included between black The following comparative commentary refers to the texts
diamonds, on the use of a muffle furnace and its calibration. 6.09 Disintegration Test in the Japanese Pharmacopoeia
XV and <701> Disintegration in the United States
The aboye differences in the USP text do not affect Pharmacopeia USP32 NF27 pt Supplement, and chapter 2.9.1.
harmonisation.
Disintegration of tablets and capsules (Test A) in the European
The texts of the 3 pharmacopoeias are therefore considered Pharmacopoeia.
harmonised. In the Ph. Eur chapter, test A corresponds to the harmonised
NOTE: ICH has declared this method interchangeable within chapter while test B do es not and is intended for tablets and
the ICH regions. capsules that are greater than 18 mm long. Test Bis not within
the scope of pharmacopoeial harmonisation and has been
2.6.1. STERILITY placed between black diamonds (+ +).
The following comparative commentary refers to the texts The JP and USP specify procedures and acceptance crHeria for
4.06 Sterility Test in the partia! revision of the Japanese different types of dosage forms. The equivalent statements are
Pharmacopoeia XV made official March 31, 2009, by included in the Ph. Eur. general monographs on dosage forms.
the Ministry of Health, Labour and Welfare Ministerial These statements are not within the scope of pharmacopoeial
Notification No. 190 and <71> Sterility Tests in the United harmonisation.
States Pharmacopeia as presented in Pharmacapeial Forum,
In addition, the JP describes an auxiliar y tube, and a metal
Volume 34(6), Interim Revision Announcement No. 6,
plate to secure the glass tubes. This has been placed between
December 1, 2008, official on May 1, 2009, and chapter 2.6.1.
black diamonds ( •• ). The use of this tube and this plate
Sterility in the European Pharmacopoeia.
may have an impact on hydrodynamics and thus may affect
The USP has added requirements that cover either pharmacy harmonisation.
bulk packages of antibiotics (which are not of concern in The texts of the 3 pharmacopoeias are therefore considered
Europe and Japan) or medical devices (which are outside the harmonised.
scope of the Ph. Eur. and JP). The corresponding parts, which
are not within the scope of pharmacopoeial harmonisation, NOTE: ICH has declared this method interchangeable within
have been placed between black diamonds ( •• ). the ICH regions subject to the conditions detailed below.
The JP has deleted the requirements for 'Catgut and other For tablets and capsules larger than 18 mm long, for which
surgical sutures for veterinary use' in Table 2, in the section a different apparatus is used, the disintegration test is not
'Direct inoculation of the culture medium' and in Table 3. considered to be interchangeable in the 3 regions.
Catgut and other surgical sutures are outside the scope of The test for disintegration is not considered to be
the JP. interchangeable in the 3 regions for dosage forms referred to
The aboye differences in the JP and USP texts do not affect in the pharmacopoeias as delayed-release, gastro-resistant or
harmonisation. enteric-coated.
The texts of the 3 pharmacopoeias are therefore considered 2.9.7. FRIABILITY OF UNCOATED TABLETS
harmonised. As a result of an evaluation of the texts 26. Tablet Friability
NOTE: ICH has declared this method interchangeable within Test in the Japanese Pharmacopoeia XV and <1216> Tablet
the ICH regions. Friability in the United States Pharmacopeia USP31 NF26

678 See the information section on general monographs (caver pages)


EUROPEAN PHARMACOPOEIA 8.0 5.8. Pharmacopoeial harmonisation

1't Supplement, and chapter 2.9.7. Friability of uncoated Multi-point measurement UP). The JP does not state the
tablets in the European Pharmacopoeia, the texts of the meaning of the 22400 constant in the definition of the specific
3 pharmacopoeias are considered harmonised. surface are a S and does not require a test to determine the
linearity of the method.
2.9.17. TEST FOR EXTRACTABLE VOLUME OF
Single-point measurement UP). The JP does not state
PARENTERAL PREPARATIONS
the equivalent quantity of gas corresponding to the value
The following comparative commentary refers to the texts of PIPo> which is les s precise (0.30) than in the other
6.05 Test for Extractable Volume of Parenteral Preparations in pharmacopoeias (0.300).
the Japanese Pharmacopoeia XV and <1> Injections in the The JP does not as sume the material constant C to be invariant.
United States Pharmacopeia USP32 NF27 1't Supplement,
and chapter 2.9.17. Test for extractable volume of parenteral Measurements (IP). The JP does not specify the temperature
preparations in the European Pharmacopoeia. required to perform the test for either method.
The JP has added a non-harmonised introductory part, The JP limits its volumetric method to dassical instruments
included between black diamonds, at the beginning of this and does not take alternative instruments into account.
chapter. It is given for further information and does not affect The aboye differences in the JP text might affect harmonisation.
harmonisation. Therefore only the texts of the Ph. Eur. and the USP are
The USP has included this test in general chapter <1> considered harmonised.
Injections, under a specific part entitled Determination of
2.9.36. POWDER FLOW
Volume ofInjection in Containers. This does not affect
harmonisation. The following comparative commentary refers to the texts
18. Powder Flow in the Japanese Pharmacopoeia XV and
The texts of the 3 pharmacopoeias are therefore considered
<1174> Powder Flow in the United States Pharmacopeia
harmonised.
USP31 NF26 1't Supplement, and chapter 2.9.36. Powder flow
NOTE: ICH has declared this method interchangeable within in the European Pharmacopoeia.
the ICH regions.
Flow through an orifice UP). The JP limits the use of orifices
2.9.19. PARTICULATE CONTAMINATION: SUB-VISIBLE to dassical ones and does not allow vibrators or moving
PARTICLES orifices. A test result using the JP method will be compatible
with the Ph. Eur and the USP. A Ph. Eur. or USP test result
The following comparative commentary refers to the texts 6.07 will not comply with the JP when a vibrator or moving orifice
Insoluble Particulate Matter Test for Injections in the Japanese is used.
Pharmacopoeia XV (corrected version dated September 2007)
and <788> Particulate Matter in Injections in the United States 2.9.37. OPTICAL MICROSCOPY
Pharmacopeia USP32 NF27 2nd Supplement, and chapter As a result of an evaluation of the texts 3.04 Particle
2.9.19. Particulate contamination: sub-visible particles in the Size Determination in the Japanese Pharmacopoeia XV
European Pharmacopoeia. and <776> Optical Microscopy in the United States
The USP specifies that system suitability can be verified using Pharmacopeia USP31 NF26 2nd Supplement, and chapter
USP Particle Count RS. This statement is not within the scope 2.9.37. Optical microscopy in the European Pharmacopoeia,
of pharmacopoeial harmonisation. It has been placed between the texts of the 3 pharmacopoeias are considered harmonised.
black diamonds ( •• ) and it does not affect harmonisation.
2.9.38. PARTICLE-SIZE DISTRIBUTION ESTIMATION BY
The JP includes a detailed section on calibration of the ANALYTICAL SIEVING
apparatus. In particular, requirements for the quality of
The following comparative commentary refers to the
particle-free water are given, which differ from those stated in
texts 3.04 Particle Size Determination in the Japanese
the USP (see section Reagents, Indicators and Solutions) and
Pharmacopoeia XV and <786> Particle-size Distribution
in the Ph. Eur (see chapter 4.1.1). The section on calibration
Determination by Analytical Sieving in the United States
is not within the scope of pharmacopoeial harmonisation. It
Pharmacopeia USP31 NF26 1st Supplement, and chapter
has been placed between black diamonds ( •• ) and it does not
2.9.38. Particle-size distribution estimation by analytical
affect harmonisation.
sieving in the European Pharmacopoeia.
In addition, the JP describes more stringent acceptance
Sieving methods - Dry sieving method UP). The JP permits
criteria for parenteral preparations having a nominal
any powder on the down surface of the sieve to be brushed
volume of 100 mL. This was acknowledged by the PDG as
and combined with the fraction of the next sieve.
a non-harmonised item. It has been placed between black
diamonds (+.). The acceptance criteria for parenteral The aboye difference in the JP text might affect harmonisation.
preparations having a nominal volume of 100 mL are therefore Therefore only the texts of the Ph. Eur. and the USP are
considered non -harmonised. considered harmonised.
The texts of the 3 pharmacopoeias are therefore considered 5.1.4. MICROBIOLOGICAL QUALITY OF NON-STERILE
harmonised except for the acceptance criteria for parenteral PHARMACEUTICAL PREPARATIONS AND SUBSTANCES
preparations having a nominal volume of 100 mL. FOR PHARMACEUTICAL USE
NOTE: ICH has dedared this method interchangeable within The following comparative commentary refers to the texts 12.
the ICH regions except the acceptance criteria for parenteral Microbial Attributes of Non-sterile Pharmaceutical Products in
preparations having a nominal volume of 100 mL. the Japanese Pharmacopoeia XV 1't Supplement and <1111>
Microbiological Attributes of Non-sterile Pharmaceutical
2.9.26. SPECIFIC SURFACE AREA BY GAS ADSORPTION Products in the United States Pharmacopeia USP30 NF25,
The following comparative commentary refers to the texts and chapter 5.1.4. Microbiological qua lity of non-sterile
3.02 Specific Surface Area by Gas Adsorption in the Japanese pharmaceutical preparations and substances jor pharmaceutical
Pharmacopoeia XV and <846> Specific Surface Area in the use in the European Pharmacopoeia.
United States Pharmacopeia USP31 NF26 1't Supplement, and A special Ph. Eur. provision for oral dosage forms containing
chapter 2.9.26. Specific surface area by gas adsorption in the raw materials of natural origin is included within table 5.1.4.-1.
European Pharmacopoeia. Also, a reference to chapter 5.1.8 giving recommended
The JP has chosen to express all the temperatures of this acceptance criteria for the microbiological quality of herbal
chapter in degrees Celsius. medicinal products for oral use and extracts used in their

General Notices (1) apply to all monographs and other texts 679
5.8. Pharmacopoeial harmonisation EUROPEAN PHARMACOPOEIA 8.0

preparation is included in the texto The corresponding pH + + +

( ..
parts, which are not within the scope of pharmacopoeial
harmonisation, have been placed between black diamonds
).
The above differences in the Ph. Eur. text do not affect
Heavy metals

Loss on drying
+

+
+

+
+

harmonisation. Sulfated ash + + +

The texts of the 3 pharmacopoeias are therefore considered Assay + + +


harmonised.
Legend
NOTE: ICH has declared these texts interchangeable within
the lCH regions. + : will adopt and implement
: will not stipulate
ETHYLCELLULOSE (0822)
Non-harmonised attributes
As a result of the process of pharmacopoeial harmonisation,
the following reflects the agreement reached by the Ph. Eur., Characters, Packaging and storage
the JP and the USP. Local requirements
Harmonised attributes Appearance of solution (Ph. Eur.), Description OP)
Attribute Ph. Eur. JP USP Reagents and reference materials
Definition + + Each pharmacopoeia adapts the text to take account of local
reference materials and reagent specifications.
Identification by IR + +
(sign-off date: 6 ¡une 2012)
Acidity/alkalinity + +

Viscosity + + MAlZE STARCH (0344)


Acetaldehyde + + As a result of the process of pharmacopoeial harmonisation,
the following reflects the agreement settled by the Ph. Eur.,
Chlorides + +
the JP and the USP.
Loss on drying + + Harmonised attributes
Sulfated ash + +
Attribute Ph. Eur. JP usp
Assay + +
Definition + + +
Labelling + +
ldentification
- A + + +
Legend
- B + + +
+ : will adopt and implement - C + + +
- : will not stipulate pH + + +
Non-harmonised attributes + + +
lron
Characters, Heavy metals, Storage
Oxidising substances + + +
Local requirements
Sulfur dioxide + + +
Identification (compliance with limits of assay) (Ph. Eur.)
Loss on drying + + +
Reagents and reference materials
Sulfated ash + + +
Each pharmacopoeia adapts the text to take account of local
reference materials and reagent specifications. Microbial contamination + +
(sign-off date: 27 October 2009) Legend
HYPROMELLOSE (0348) + : will adopt and implement
As a result of the process of pharmacopoeial harmonisation, - : will not stipulate
the following reflects the agreement settled by the Ph. Eur., Non-harmonised attributes
the JP and the USP.
Characters, Storage
Harmonised attributes
Local requirements
Attribute Ph. Eur. JP usp Labelling (USP), Absence of Salmonella (Ph. Eur.), Absence of
Staphylococcus aureus and Pseudomonas aeruginosa when the
Definition + + +
product is intended for use in preparing Absorbable Dusting
Labelling + + + Powder (USP), Foreign MaUer (Ph. Eur., TP)
Reagents and reference materials
Identification
- A + + + Each pharmacopoeia adapts the text to take account of local
- B + + + reference materials and reagent specifications.
- C + + +
(sign-off date: 6 ¡une 2012)
- D + + +
- E + + +
MANNITOL (0559)
Viscosity As a result of the process of pharmacopoeial harmonisation,
- Method 1 + + + the following reflects the agreement settled by the Ph. Eur.,
- Method 2 + + + the JP and the USP.

680 See the information section on general monographs (cover pages)


EUROPEAN PHARMACOPOEIA 8.0 5.8. Pharmacopoeial harmonisation

Harmonised attributes Legend


Attribute Ph. Eur. IP USP
+ : will adopt and implement
Definition + + +

Identification by IR + + + -: will not stipulate

Appearance of solution + + + Non-harmonised attributes

Conductivity + + + Characters, Packaging and storage


Melting point + + + Local requirements
Reducing sugars + + +

You might also like